Purpose: Erlotinib has proven activity in pretreated patients with advanced non–small cell lung cancer (NSCLC). We evaluated erlotinib in the frontline treatment of advanced NSCLC and assessed biological predictors of outcome.

Experimental Design: In this phase II study, chemotherapy-naive patients with stage IIIB/IV NSCLC received oral erlotinib (150 mg/d) until disease progression or unacceptable toxicity occurred. Tumor response was assessed every 6 weeks, and samples were analyzed for potential molecular markers of treatment response and survival. The primary end point was the proportion of patients without disease progression after 6 weeks of treatment.

Results: Fifty-three patients were eligible. The overall rate of nonprogression at 6 weeks was 52.8% (28 of 53 patients). Tumor response rate was 22.7%, with 1 complete response, 11 partial responses, and 16 cases of stable disease. Responses were seen across most patient clinical characteristics. The median duration of tumor response was 333 days; median overall survival was 391 days; and median time to disease progression was 84 days. Erlotinib was well tolerated, the main treatment-related adverse events being mild-to-moderate rash and diarrhea. Histologic material for biological studies was available in 29 cases. Four of five responders and one patient with stable disease had a classic epidermal growth factor receptor tyrosine kinase mutation. Two progressing patients exhibited epidermal growth factor receptor point mutations (one with T790M mutation), and K-ras mutations were detected in 10 nonresponders.

Conclusions: Erlotinib shows significant antitumor activity in the first-line treatment of advanced NSCLC and may be a viable alternative to chemotherapy. Patient selection cannot easily be based on clinical or biological variables.

There are ∼1.2 million new cases of lung cancer diagnosed every year worldwide, and 1.1 million patients die of the disease (1). Non–small cell lung cancer (NSCLC) represents 80% of lung cancers. Surgery is the most important curative modality in the treatment of early-stage NSCLC; however, only ∼20% to 30% of patients are diagnosed at an operable stage.

Platinum-based doublet chemotherapy, the standard treatment for advanced NSCLC, achieves mostly partial responses in only 30% to 40% of patients and a modest survival increase (2, 3); in general, single agents are inferior to combinations. Median survival and 1-year survival after first-line chemotherapy are 8 to 10 months and 30% to 40%, respectively (4). These regimens are associated with considerable toxicity, particularly myelosuppression. More effective and less toxic treatments are urgently needed for advanced NSCLC.

Among the new drugs that have been introduced in the treatment of advanced NSCLC after failure of first-line chemotherapy are docetaxel, pemetrexed, and, recently, epidermal growth factor receptor (EGFR) tyrosine kinase (TK) inhibitors. All of these agents produce a low response rate (below 10%) and have different toxicity profiles. In a large randomized study, erlotinib improved survival versus best supportive care in second-line and third-line treatment (5). Erlotinib is a targeted agent that inhibits EGFR by competing with ATP at the intracellular TK domain of the receptor. Erlotinib is given orally continuously and is devoid of bone marrow and major organ toxicities (6). Common side effects are diarrhea and skin toxicity.

Despite promising preclinical data, the combination of small-molecule inhibitors of EGFR with first-line chemotherapy failed to improve survival, compared with chemotherapy alone in four large randomized studies that investigated erlotinib or gefitinib in combination with either carboplatin-paclitaxel or cisplatin-gemcitabine (6).

We did a phase II study of single-agent erlotinib as first-line therapy in patients with advanced NSCLC. We also did biological studies on tumor samples of patients entered in the study to identify patients who benefited most from treatment.

Patients

Patients with a histologic or cytologic diagnosis of advanced NSCLC not amenable to radical surgery or radiotherapy were eligible for the study. No prior chemotherapy or other systemic treatment was allowed. Other eligibility criteria were measurable disease according to the Response Evaluation Criteria in Solid Tumors, ages ≥ 18 years, performance status of 0 to 2, life expectancy ≥ 12 weeks, at least 4 weeks since any prior surgery or radiotherapy, granulocyte count ≥1.5 × 109/L, platelet count ≥100 × 109/L, bilirubin and transaminases ≤1.5 × upper limit of normal, and serum creatinine ≤1.5 × upper limit of normal, or creatinine clearance ≥60 mL/min. Females of childbearing potential had to have a negative pregnancy test. Patients were excluded if they had unstable systemic disease (active infection, uncontrolled hypertension, unstable angina, congestive heart failure, myocardial infarction within the previous year, and serious cardiac arrhythmia requiring medication), any other malignancies within 5 years except for adequately treated carcinoma in situ of the cervix, or basal or squamous cell skin cancer. Patients with brain metastases were only allowed in the study if there was no evidence of progression in the brain and in the absence of corticosteroid treatment. Patients with significant eye disorders were excluded (severe dry eye syndrome, Sjogren syndrome, severe exposure keratinitis, and any other eye disorder likely to increase the risk of corneal epithelial lesions). Signed informed consent was required for all patients. The study was conducted according to the latest version of the Declaration of Helsinki, and the protocol was approved by the independent Ethics Committee and the Review Board from each participating institution.

Treatment

Erlotinib 150-mg tablets were given orally daily, in the morning with 200 mL of water, and at least 1 hour before or 2 hours after ingestion of food or medication. Caution was taken to avoid medications that interact with CYP 450-3A4 enzyme, and concomitant warfarin was not permitted. Assessment of toxicity was according to National Cancer Institute Common Toxicity Criteria version 2.0. Dose adjustments, in 50-mg decrements, were done if grade 3 or 4 toxicity was encountered.

Before starting treatment, patients were assessed by physical examination, electrocardiogram, disease measurement by appropriate radiological techniques, complete blood cell counts, serum chemistries, and urine pregnancy test in females of childbearing potential. Patients were evaluated every 3 weeks, and hematology and blood chemistry were done; tumor measurements were assessed every 6 weeks. An electrocardiogram was repeated after 18 weeks of treatment and as clinically indicated afterwards. Response assessment was according to the Response Evaluation Criteria in Solid Tumors (7). All patients received the Lung Cancer Symptoms Score patient scale questionnaire at baseline and again at 3 and 6 weeks and every 6 weeks thereafter.

Mutation analysis and chromogenic in situ hybridization

Mutation analysis of EGFR, PIK3CA, and K-ras. Paraffin-embedded tumor material was cut into 4-μm-thick sections and placed onto glass slides, stained with H&E, and the presence of tumor cells was verified by a pathologist. Tumor cells were microdissected, and genomic DNA was isolated using the QIAamp DNA Micro kit (Qiagen, Venlo, The Netherlands). Nested PCRs were carried out using primers (Table 1) to amplify exons 18 to 21 of EGFR, exons 9 and 20 of PIK3CA, and exons 1 to 2 of K-ras. To facilitate sequencing, internal primers incorporated an M13Tag. Sequencing of PCR products was done with the ABI PRISM 310 Genetic analyzer (Applied Biosystems, Foster City, CA). Mutations were confirmed by sequencing independent PCR products.

Table 1.

Primers used in EGFR, PIK3CA, and K-ras mutation analysis

GeneSense (5′-3′)Antisense (5′-3′)
EGFR exon 18   
    Internal CTGAGGTGACCCTTGTCTCT CCATGAGAGGCCCTA 
    External ATGGTGAGGGCTGAGGTGAC TCCCCACCAGACCATGAGAG 
EGFR exon 19   
    Internal TCTCACAATTGCCAGTTAAC TGAGGTTCAGAGCCATGGAC 
    External GTGGCACCATCTCACAATTG AAGGTGGGCCTGAGGTTCAG 
EGFR exon 20   
    Internal CCCCTCCTTCTGGCCACCAT ATCCTGGCTCCTTATCTCCC 
    External GTCCATGTGCCCCTCCTTC GCATGTGAGGATCCTGGCTC 
EGFR exon 21   
    Internal AATTCGGATGCAGAGCTTCT CTGGTGTCAGGAAAATGCTG 
    External ATGACCCTGAATTCGGATG AGCCTGGTCCCTGGTGTCAG 
PIK3CA exon 9   
    Internal ATCATCTGTGAATCCAGAGG TTTTAGCACTTACCTGTGAC 
    External TCTGTAAATCATCTGTGAAT AATTCTGCTTTATTTATTCC 
PIK3CA exon 20   
    Internal GCTCCAAACTGACCAAACTG ATCCAGAGTGAGCTTTCATT 
    External AACATCATTTGCTCCAAACT TGTGGAATCCAGAGTGAGCT 
K-ras exon 1   
    Internal AGTCACATTTTCATTATTTT AGAAACCTTTATCTGTATCAAAGAATG 
    External GTTCTAATATAGTCACATTT ACTCATGAAAATGGTCAGAGAAACCTTTAT 
K-ras exon 2   
    Internal GTGCACTGTAATAATCCAGA ACTCCTTAATGTCAGCTTAT 
    External GAAGTAAAAGGTGCACTGTA AACTATAATTACTCCTTAAT 
M1 3   
 TGTAAAACGACGGCCAGT CAGGAAACAGCTATGACC 
GeneSense (5′-3′)Antisense (5′-3′)
EGFR exon 18   
    Internal CTGAGGTGACCCTTGTCTCT CCATGAGAGGCCCTA 
    External ATGGTGAGGGCTGAGGTGAC TCCCCACCAGACCATGAGAG 
EGFR exon 19   
    Internal TCTCACAATTGCCAGTTAAC TGAGGTTCAGAGCCATGGAC 
    External GTGGCACCATCTCACAATTG AAGGTGGGCCTGAGGTTCAG 
EGFR exon 20   
    Internal CCCCTCCTTCTGGCCACCAT ATCCTGGCTCCTTATCTCCC 
    External GTCCATGTGCCCCTCCTTC GCATGTGAGGATCCTGGCTC 
EGFR exon 21   
    Internal AATTCGGATGCAGAGCTTCT CTGGTGTCAGGAAAATGCTG 
    External ATGACCCTGAATTCGGATG AGCCTGGTCCCTGGTGTCAG 
PIK3CA exon 9   
    Internal ATCATCTGTGAATCCAGAGG TTTTAGCACTTACCTGTGAC 
    External TCTGTAAATCATCTGTGAAT AATTCTGCTTTATTTATTCC 
PIK3CA exon 20   
    Internal GCTCCAAACTGACCAAACTG ATCCAGAGTGAGCTTTCATT 
    External AACATCATTTGCTCCAAACT TGTGGAATCCAGAGTGAGCT 
K-ras exon 1   
    Internal AGTCACATTTTCATTATTTT AGAAACCTTTATCTGTATCAAAGAATG 
    External GTTCTAATATAGTCACATTT ACTCATGAAAATGGTCAGAGAAACCTTTAT 
K-ras exon 2   
    Internal GTGCACTGTAATAATCCAGA ACTCCTTAATGTCAGCTTAT 
    External GAAGTAAAAGGTGCACTGTA AACTATAATTACTCCTTAAT 
M1 3   
 TGTAAAACGACGGCCAGT CAGGAAACAGCTATGACC 

Because of concerns about the sensitivity of direct sequencing, DNA from nine independent samples from Dana-Farber Cancer Institute and 13 from the Vrije Universiteit Medical Center were analyzed by the other institution, in a blinded fashion, for EGFR mutations in exons 18 to 21. At the Dana-Farber Cancer Institute, specimens were analyzed using Surveyor DNA endonuclease, combined with the WAVE HS DHPLC system (Transgenomic, Inc., Omaha, NE) as described (8). DNA variants detected using Surveyor were subjected to fractionation and sequencing.

EGFR gene copy number. Chromogenic in situ hybridization (CISH) was used to study EGFR gene copy number. Tissue sections were incubated at 64°C for 2 to 4 hours, deparaffinized, and rehydrated. Samples were boiled in 1 mmol/L EDTA/Tris (pH 9) and digested with 0.01% pepsin/0.2 N HCl at 37°C. After dehydration in alcohol, the EGFR probe was added (Zymed Spotlight EGFR amplification probe 84-1300; Zymed, San Francisco, CA). Samples were denatured at 80°C for 10 minutes and incubated overnight in a humidifying chamber at 37°C. The next day, samples were washed for 5 minutes in 0.5 × SSC at 75°C to 80°C and incubated in 3% H2O2/PBS for 10 minutes to block endogenous peroxidase activity. After blocking the slides in pre-immune normal goat serum for 10 minutes, slides were incubated with mouse-α-digoxigenin antibody (1:100 dilution) for 60 minutes. CISH signals were visualized using the DAKO Envision system (DakoCytomation, Heverlee, Belgium). CISH signals were counted in 200 cells by two observers who were blinded to the patient's clinical characteristics. The scoring system was as described by Cappuzzo et al. for fluorescence in situ hybridization (9).

Statistical considerations

The primary end point of the first part of the study was the rate of nonprogression at 6 weeks, defined as the number of patients whose tumor did not progress (<19% tumor increase to total disappearance of tumor, according to the Response Evaluation Criteria in Solid Tumors). Deterioration of performance status was considered progression. This end point was considered an early conservative estimate of biological activity of the compound and was based on results obtained with single-agent first-line chemotherapy (10, 11).

Sixteen patients were to be entered, and of these, at least eight were to be nonprogressive in order for the study to continue accrual to the second part. If nine or more patients progressed during the first 6 weeks, the study was to be stopped (Simon's optimal design: p0 = 40%, p1 = 60%, α = 0.05, β = 0.20). The second part of the study had a targeted total accrual of an additional 30 patients, with a total accrual to the study of 46 patients. If at least 24 patients did not progress, the study would be considered of interest and promising.

Secondary objectives of the study were objective response rate, disease control rate, duration of response, time to disease progression or death, survival, and safety. Patients who did not receive at least 6 weeks of treatment were considered as progressors. Exploratory studies included quality-of-life assessment and the correlation of biological markers with treatment outcome. Survival, time to progression, and duration of response were calculated from day 1 of treatment. Survival curves were constructed using the Kaplan-Meier method, and survival was compared using the log-rank test. The SPSS statistical package for Windows version 9.01 was used.

From January to July 2004, 54 patients were enrolled in the study from three institutions in Europe. One patient was not eligible because of unstable brain metastases and did not start treatment. Table 2 summarizes the main characteristics of the eligible patients.

Table 2.

Patient characteristics (N = 53)

Variablen (%)
Median age (range), y 60 (30-80) 
Gender  
    Male 22 (41.5) 
    Female 31 (58.5) 
Performance status  
    0 13 (24.5) 
    1 32 (60.4) 
    2 8 (15.1) 
Histology  
    Adenocarcinoma 24 (45.3) 
    Bronchioloalveolar carcinoma 6 (11.3) 
    Squamous cell carcinoma 8 (15.1) 
    Large cell carcinoma 9 (17) 
    Others 6 (11.3) 
Smoking history  
    Never smoker 16 (30.2) 
    Former or current smoker 37 (69.8) 
Stage  
    IIIB 11 (21) 
    IV 42 (79) 
Prior therapy  
    Surgery 8 (15.1) 
    Radiotherapy 5 (9.4) 
    Surgery + radiotherapy 3 (5.7) 
    None 37 (69.8) 
Variablen (%)
Median age (range), y 60 (30-80) 
Gender  
    Male 22 (41.5) 
    Female 31 (58.5) 
Performance status  
    0 13 (24.5) 
    1 32 (60.4) 
    2 8 (15.1) 
Histology  
    Adenocarcinoma 24 (45.3) 
    Bronchioloalveolar carcinoma 6 (11.3) 
    Squamous cell carcinoma 8 (15.1) 
    Large cell carcinoma 9 (17) 
    Others 6 (11.3) 
Smoking history  
    Never smoker 16 (30.2) 
    Former or current smoker 37 (69.8) 
Stage  
    IIIB 11 (21) 
    IV 42 (79) 
Prior therapy  
    Surgery 8 (15.1) 
    Radiotherapy 5 (9.4) 
    Surgery + radiotherapy 3 (5.7) 
    None 37 (69.8) 

Response. Of 16 patients entered into the first part of the study, 10 had not progressed after 6 weeks of treatment; therefore, the study was opened to further accrual. Thirteen patients had <6 weeks of treatment; 12 were classified as early progressions, and one refused further treatment. The response rate based on intent to treat was 22.7% (95% confidence interval, 12.3-36.2%; Table 3). The percentage of nonprogressors at 6 weeks was 52.8% (28 of 53; 95% confidence interval, 38.6-66.7%). The median duration of response was 333 days.

Table 3.

Response to erlotinib

ResponsePatients (%)
Complete 1 (2) 
Partial 11 (21) 
Stable disease 16 (30) 
Progression 12 (23) 
Not evaluable* 13 (25) 
    Progression 12 (23) 
    Refusal 1 (2) 
Total 53 (100) 
ResponsePatients (%)
Complete 1 (2) 
Partial 11 (21) 
Stable disease 16 (30) 
Progression 12 (23) 
Not evaluable* 13 (25) 
    Progression 12 (23) 
    Refusal 1 (2) 
Total 53 (100) 
*

Less than 6 weeks of treatment.

Responses were significantly correlated with histology (six responses in adenocarcinomas, four in bronchioloalveolar carcinomas, two in squamous carcinomas, and none in large cell and other histologies; Pearson χ2, P = 0.025), smoking history (seven responses in never smokers and five in former/current smokers; Pearson χ2, P = 0.016), but not gender, performance status, stage of disease, or age.

When patients progressed, they were offered chemotherapy. Chemotherapy was given to 31 patients, 28 of whom received carboplatin or cisplatin in combination with gemcitabine; the response rate was 28.6% in these patients. Three patients received single-agent chemotherapy, and one patient was rechallenged with erlotinib after a partial response and interruption of treatment for 8 months at the patient's request; this patient again experienced a partial response. No further therapy was given to 13 patients who progressed on first-line erlotinib (24.5%), and four had only palliative radiotherapy. The median survival of progressive disease patients was 234 days versus 592 days for those with disease control (P = 0.004). The 13 patients who did not receive any form of treatment at progression had a significantly worse performance status at start (likelihood ratio χ2, P = 0.020). The median survival of the 28 patients who received platinum-based chemotherapy was 350 days, calculated from the date of progression on erlotinib.

Toxicity. The most frequent drug-related side effects were mild to moderate skin toxicity and diarrhea (Table 4). All responders had some degree of rash, whereas only seven nonresponders developed rash (likelihood ratio χ2, P = 0.048). Emesis was usually mild. Liver toxicity (transaminase and bilirubin increase) was observed in just >10% of patients and was of mild or moderate severity and reversible. Dose reduction to 100 mg/d was necessary in five cases due to increased bilirubin and in one due to skin toxicity. Dose interruptions for >1 day were necessary in 13 patients; in five patients, this was due to hyperbilirubinemia.

Table 4.

Treatment-related side effects (N = 53)

ToxicityGrade
1234
Skin 17 (32%) 28 (53%) 2 (4%) 
Diarrhea 30 (57%) 5 (9%) 2 (4%) 1(2%) 
Hair loss 11 (21%) 5 (9%) — — 
Nails 3 (6%) 7 (13%) — — 
Mucositis 7 (13%) 5 (9%) — — 
Ocular 10 (19%) 1 (2%) 1 (2%) — 
Nausea 4 (8%) 8 (15%) 2 (4%) — 
Vomiting 4 (8%) 2 (4%) 1 (2%) — 
Transaminases 6 (11%) 3 (6%) — — 
Bilirubin 4 (8%) 2 (4%) — — 
ToxicityGrade
1234
Skin 17 (32%) 28 (53%) 2 (4%) 
Diarrhea 30 (57%) 5 (9%) 2 (4%) 1(2%) 
Hair loss 11 (21%) 5 (9%) — — 
Nails 3 (6%) 7 (13%) — — 
Mucositis 7 (13%) 5 (9%) — — 
Ocular 10 (19%) 1 (2%) 1 (2%) — 
Nausea 4 (8%) 8 (15%) 2 (4%) — 
Vomiting 4 (8%) 2 (4%) 1 (2%) — 
Transaminases 6 (11%) 3 (6%) — — 
Bilirubin 4 (8%) 2 (4%) — — 

Survival. Median follow-up was 518 days. At the time of this analysis, 28 patients were dead; two were lost to follow-up; and 23 were alive. Median survival time for the 53 eligible patients was 391 days. The 1-year survival rate was 54%.

By univariate analysis, the following variables were significantly correlated with survival: smoking history, age, histology, and response to erlotinib (Table 5). The severity of skin toxicity was also significantly correlated with survival.

Table 5.

Univariate analysis of survival

VariableMedian survival (d)P
Gender   
    Male 592 NS 
    Female 391  
Stage   
    IIIB 592 NS 
    IV 347  
Performance status   
    0 >585 0.077 
    1 389  
    2 82  
Smoking status   
    Never >644 0.014 
    Former/current 347  
Age   
    <70 592 0.027 
    >70 234  
Histology   
    Bronchioloalveolar carcinoma >607 0.006 
    Adenocarcinoma 478  
    Squamous 391  
    Others 180  
Response   
    CR + PR >627 0.0035 
    SD 389  
    PD 234  
Rash   
    0 82 0.0018 
    1 270  
    2-3 592  
EGFR   
    Wild type 377 0.15 
    Mutant classic >627  
K-ras   
    Wild type >627 0.0059 
    Mutant 170  
EGFR-CISH   
    Amplification + high polysomy 188 0.5 
    Other 180  
VariableMedian survival (d)P
Gender   
    Male 592 NS 
    Female 391  
Stage   
    IIIB 592 NS 
    IV 347  
Performance status   
    0 >585 0.077 
    1 389  
    2 82  
Smoking status   
    Never >644 0.014 
    Former/current 347  
Age   
    <70 592 0.027 
    >70 234  
Histology   
    Bronchioloalveolar carcinoma >607 0.006 
    Adenocarcinoma 478  
    Squamous 391  
    Others 180  
Response   
    CR + PR >627 0.0035 
    SD 389  
    PD 234  
Rash   
    0 82 0.0018 
    1 270  
    2-3 592  
EGFR   
    Wild type 377 0.15 
    Mutant classic >627  
K-ras   
    Wild type >627 0.0059 
    Mutant 170  
EGFR-CISH   
    Amplification + high polysomy 188 0.5 
    Other 180  

Abbreviations: CR, complete response; SD, stable disease; NS, not significant; PD, progressive disease; PR, partial response.

Median time to progression was 84 days, with three patients who had not yet progressed. Two patients remain on treatment (after 22 and 20 months, respectively), and one patient is being rechallenged with erlotinib. One patient underwent an R0 resection after a response on erlotinib. The percentage of patients not progressing at 1 year was 16%.

Quality of life. The Lung Cancer Symptoms Score was analyzed by assessing the visual analogue averages to the first six questions, concerning disease-related symptoms (appetite, fatigue, coughing, shortness of breath, hemoptysis, and pain). A decrease of 25% of the score compared with basal level was considered symptomatic progression. Although there was a difference in time to symptomatic progression between responding patients and stable disease and progressing patients, this was not significant (not shown).

EGFR, K-ras, and PIK3CA mutations and EGFR copy number. Histologic material from 29 patients was available to perform mutation analyses. EGFR mutations were detected in seven cases, five of which were deletions in exon 19; four of these cases responded to therapy, and one had long-lasting stable disease. One major response was detected in a patient with wild-type EGFR. Furthermore, two point mutations were detected in exon 20: one not previously described (V802I) and one that has been described in resistant disease (T790M; refs. 12, 13). In the latter sample, another point mutation in exon 21 was also recognized (R832H). These point mutations, which were only detectable using the WAVE system, but not direct sequencing, were observed in progressing patients and in tumors also bearing a K-ras mutation. The exon 19 deletions were only observed in females (Fisher's exact test, P = 0.047), mainly in never smokers (Fisher's exact test, P = 0.030). No patients with K-ras mutations had an exon 19 deletion (likelihood ratio, P = 0.026). There was no association between classic EGFR mutations and histology, performance status, stage, age, or appearance of rash upon treatment.

K-ras mutations were detected in 10 patients, all smokers (Fisher's exact test, P = 0.061), and nonresponders. There was a trend for a correlation between K-ras mutations and lack of development of rash (Fisher's exact test, P = 0.052) and no association with gender, histology (although more frequent in adenocarcinomas), performance status, stage, or age.

The presence of classic EGFR mutations was significantly correlated with response to treatment (Fisher's exact test, P = 0.001), and K-ras mutations were more commonly associated with lack of response (Fisher's exact test, P = 0.125). One PIK3CA mutation was detected in a patient who had stable disease on treatment. A summary of the mutation analyses is reported in Table 6. Survival was longer in patients with classic EGFR mutations than in the other patients, although the difference did not reach statistical significance (Table 5; Fig. 1A). Conversely, survival of patients with K-ras mutations was significantly shorter than that of patients with wild-type K-ras (Table 5; Fig. 1B). Of the two patients still on treatment, one did not have tumor available for mutation analysis; the other patient's tumor contained an exon 19 deletion of EGFR.

Table 6.

Mutation analysis and patient characteristics

CharacteristicEGFR
PIK3CA
K-ras
WTDel MUT (all Del 746-750)Point MUTWTMUTWTMUT
Gender        
    Female 10 1(T790M*, R832H*) 14 5 (G13V, Q61L, G12C, 2xG12V) 
    Male 12 1 (V802I*) 10 1 (E545Q) 5 (G12A, G12V, 2xG12C, G12D) 
Histology        
    Adenocarcinoma 10 1(T790M*, R832H*) 12 1 (E545Q) 5 (G12A, Q61L, G12D, 2xG12V) 
    Bronchioloalveolar carcinoma 1 (G12C) 
    Squamous cell carcinoma 1 (V802I*) 1 (G13V) 
    Large cell carcinoma 1 (G12C) 
    Others 2 (G12V, G12C) 
Smoking status        
    Never-smoker 
    Current/former smoker 20 2 (T790M*, R832H*,V802I*) 20 1 (E545Q) 10 10 (G12A, 3xG12C, G12D, G13V, Q61L, 3xG12V) 
Response        
    Partial response 
    Stable disease 11 10 1 (E545Q) 3 (G12V, G12C, Q61L) 
    Progressive disease 10 2 (T790M*, R832H*,V802I*) 11 7 (G12A, 2xG12C, G12D, G13V, 2xG12V) 
CharacteristicEGFR
PIK3CA
K-ras
WTDel MUT (all Del 746-750)Point MUTWTMUTWTMUT
Gender        
    Female 10 1(T790M*, R832H*) 14 5 (G13V, Q61L, G12C, 2xG12V) 
    Male 12 1 (V802I*) 10 1 (E545Q) 5 (G12A, G12V, 2xG12C, G12D) 
Histology        
    Adenocarcinoma 10 1(T790M*, R832H*) 12 1 (E545Q) 5 (G12A, Q61L, G12D, 2xG12V) 
    Bronchioloalveolar carcinoma 1 (G12C) 
    Squamous cell carcinoma 1 (V802I*) 1 (G13V) 
    Large cell carcinoma 1 (G12C) 
    Others 2 (G12V, G12C) 
Smoking status        
    Never-smoker 
    Current/former smoker 20 2 (T790M*, R832H*,V802I*) 20 1 (E545Q) 10 10 (G12A, 3xG12C, G12D, G13V, Q61L, 3xG12V) 
Response        
    Partial response 
    Stable disease 11 10 1 (E545Q) 3 (G12V, G12C, Q61L) 
    Progressive disease 10 2 (T790M*, R832H*,V802I*) 11 7 (G12A, 2xG12C, G12D, G13V, 2xG12V) 
*

Only detectable by WAVE.

Fig. 1.

A, survival curves of EGFR classic mutations (exon 19 deletions) versus wild-type EGFR. B, survival curves of K-ras mutations versus wild-type K-ras.

Fig. 1.

A, survival curves of EGFR classic mutations (exon 19 deletions) versus wild-type EGFR. B, survival curves of K-ras mutations versus wild-type K-ras.

Close modal

EGFR CISH analysis could only be done in 13 tumors due to insufficient tumor tissue remaining after mutation analyses. Amplification was detected in two cases and high polysomy in four cases. One of the two samples with amplified EGFR also contained an exon 19 deletion, whereas the other sample contained a K-ras mutation. No clear correlation was observed between EGFR copy number and response to treatment (Fisher's exact test, P = 0.192) or survival.

EGFR TK inhibitors are active drugs in advanced NSCLC, but in unselected patients, the response rate in second-line and third-line for both erlotinib and gefitinib is in the range of ≤10% (6). However, response to EGFR TK inhibitors is higher in women, never smokers, East Asians, adenocarcinoma and bronchioloalveolar carcinomas, and those who develop rash (14). Furthermore, activating mutations in the EGFR TK domain and EGFR amplification have been shown to be predictors of response and survival in several reports (9, 1517).

In the present study, the primary end point was met, with >50% of patients showing no progression after 6 weeks of treatment. The response rate of 22.7% is similar to that obtained with most single chemotherapy agents. Although the protocol did not prescribe any type of patient selection, a higher percentage of women, never smokers, and patients with bronchioloalveolar carcinoma histology were included in the study than one would expect in a typical sample of patients with advanced NSCLC. This was a result of the growing evidence, accumulating during the conduct of the study, that these characteristics are more often associated with benefit from EGFR TK inhibitors.

Although response to erlotinib was more common in patients with adenocarcinoma (including bronchioloalveolar carcinoma histology) and in never smokers, like shown in other studies (5, 18), responses were also observed in patients with tumors of other histology and in former/current smokers. For example, a complete response was reported in a male patient who was a smoker. These results are consistent with those observed in the BR.21 study, in which erlotinib produced a significant survival benefit compared with best supportive care in most of the patient subgroups analyzed (5).

In the present study, the median overall survival (∼13 months) and the 1-year survival rate of 54% were better than those reported for several platinum-based doublet chemotherapy regimens. The use of erlotinib in first-line did not seem to have a detrimental effect on the response to subsequent chemotherapy. Our results are similar to those obtained with first-line gefitinib in Japanese patients (19). Promising results with erlotinib 150 mg/d monotherapy have also been achieved in an ongoing phase II study of patients ages >70 years with previously untreated advanced NSCLC (20).

Erlotinib was well tolerated in this study with the main toxicities being rash and diarrhea. Both response and survival were correlated with the grade of rash, in agreement with several other trials (18).

Several studies have assessed integration of EGFR inhibitors into first-line treatment of advanced NSCLC. Although the concomitant administration of erlotinib (21, 22) and gefitinib (23, 24) with chemotherapy failed to improve survival in the overall population, a survival benefit was observed in never smokers who received erlotinib and chemotherapy (25). This suggests that patient selection may identify groups of patients who may derive a large benefit from erlotinib treatment in combination with chemotherapy.

Certain mutations in the TK domain of EGFR are associated with increased sensitivity to EGFR TK inhibitors (16, 17, 26). In our study, four of five responding patients had EGFR mutations represented by well-documented deletions in exon 19. However, the tumor of one responding patient did not possess an EGFR mutation, and a mutation was found in a patient with long-lasting stable disease. In addition to exon 19 deletions, two point mutations were identified in patients who progressed, including the known resistant mutant T790M and a novel point mutation. Interestingly, in these samples, K-ras mutations were also found, suggesting that some EGFR point mutations can be associated with K-ras mutations and are related to resistance. The T790M EGFR mutation has been described in patients who were initially sensitive to EGFR TK inhibitors as a result of an exon 19 deletion and who later became resistant (12, 13). However, the T790M mutation has also been described in a patient with primary resistance (27), and germ line inheritance of this variant has been proposed in one family (28). The use of irreversible EGFR TK inhibitors or monoclonal antibodies directed to the extracellular domain of EGFR might circumvent resistance in these cases (12, 2932). In our study, the presence of classic EGFR mutations was correlated with longer survival, although, possibly due to the small number of samples analyzed, this did not reach statistical significance. We also analyzed the EGFR copy number using CISH but were unable to confirm the predictive value of this variable in our series, although the number of samples analyzed was too small to draw any firm conclusion.

In the BR.21 study, several techniques were used to assess EGFR status: mutation analysis, fluorescence in situ hybridization, and immunohistochemistry (33). In addition, fluorescence in situ hybridization seemed to be the strongest predictor of response and survival, although by multivariate analysis, none of the techniques were able to significantly predict outcome. In a retrospective analysis of patients treated with gefitinib, alone or in combination with chemotherapy, EGFR amplification (detected by a PCR method) and EGFR mutations identified two distinct groups of patients, and mutations were better predictors of outcome than amplification (34).

Technological issues are still very important for both the detection of mutations and the assessment of EGFR copy number (14). In a cross-validation analysis (data not shown), it was apparent that simple direct sequencing does not provide sufficient sensitivity for detection of all mutations; however, the use of more sensitive methods may identify mutations of uncertain significance (8, 14). Tumor heterogeneity and possibly the presence of mutations in a proportion of tumor cells may in fact potentially play a role in the level of detectability and biological relevance.

K-ras mutations seem to be associated with primary resistance to EGFR TK inhibitors (35). In our study, K-ras mutations were noted only in patients who did not respond to erlotinib, and survival was also significantly shorter. Poorer clinical outcomes were shown also for K-ras mutant patients treated with erlotinib plus chemotherapy (36), suggesting that K-ras mutations not only confer a poor prognosis but may also constitute a negative factor in patients treated with EGFR TK inhibitors. Interestingly, the addition of erlotinib to gemcitabine chemotherapy has been shown to significantly increase survival in patients with advanced pancreatic cancer (37), a tumor type in which K-ras mutations are very common, raising the question of whether the negative findings in NSCLC are limited to this tumor type. Potentially, the simultaneous inhibition of EGFR and ras signaling may be of interest in NSCLC (38).

The results of several studies show that EGFR mutations and K-ras mutations are mutually exclusive (39, 40). Interestingly, in our study, classic EGFR mutations and K-ras mutations were mutually exclusive; however, tumors from two refractory patients possessed both EGFR point mutations and K-ras mutations, indicating that the mutual exclusivity may only apply to activating mutations of EGFR. In the attempt to select patients for EGFR TK inhibitor, the exclusion of patients with K-ras mutations should be considered.

Given the difficulty of combining erlotinib and gefitinib with concomitant chemotherapy in first-line, other strategies need to be developed. The promising efficacy of erlotinib monotherapy in this study suggests that such therapy is a potential option for the first-line treatment of advanced NSCLC. Studies investigating the optimal sequence of erlotinib and chemotherapy are warranted. Selection of patients based on clinical and biological characteristics will allow a significant improvement of overall results. Combinations of targeted therapies also warrant investigation in this setting: erlotinib plus bevacizumab produced promising antitumor activity in a phase I/II trial involving patients with previously treated advanced NSCLC (41), and this combination is now also being investigated in first line.

In summary, the results of this study show that erlotinib has significant antitumor activity in the first-line treatment of advanced NSCLC, which may be comparable with single-agent chemotherapy, and is devoid of significant side effects. However, these results have to be seen in the context of the patient population entered, which is weighed toward individuals with higher chances of having EGFR mutations. Although responses to erlotinib were more common in some subgroups of patients, clinical benefit was not restricted to these subgroups. Further investigation of erlotinib monotherapy in this setting is warranted and the identification of patients who most benefit from the treatment is of paramount importance.

Grant support: Hofmann-La Roche Ltd., Basel, Switzerland (G. Giaccone); AstraZeneca (G. Giaccone); and Genentech (P. Janne).

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: N. Thatcher did an expert testimony for Roche.

We thank Dr. Gerrit Meijer and Karijn Floor for the CISH analysis, Alison J. Holmes for the cross-validation of EGFR mutations, and Dr. M. Smith (Thomson Gardiner-Caldwell Communications) for his assistance in drafting the article.

1
Jemal A, Murray T, Ward E, et al. Cancer statistics, 2005.
CA Cancer J Clin
2005
;
55
:
10
–30.
2
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
2004
;
22
:
330
–53.
3
D'Addario G, Pintilie M, Leighl NB, Feld R, Cerny T, Shepherd FA. Platinum-based versus non-platinum-based chemotherapy in advanced non-small-cell lung cancer: a meta-analysis of the published literature.
J Clin Oncol
2005
;
23
:
2926
–36.
4
Giaccone G, Smit E. Lung cancer.
Cancer Chemother Biol Response Modif
2003
;
21
:
445
–83.
5
Shepherd FA, Rodrigues PJ, Ciuleanu T, et al. Erlotinib in previously treated non-small-cell lung cancer.
N Engl J Med
2005
;
353
:
123
–32.
6
Giaccone G. Epidermal growth factor receptor inhibitors in the treatment of non-small-cell lung cancer.
J Clin Oncol
2005
;
23
:
3235
–42.
7
Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada.
J Natl Cancer Inst
2000
;
92
:
205
–16.
8
Janne 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.
9
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.
10
Lilenbaum RC, Herndon JE, List MA, et al. Single-agent versus combination chemotherapy in advanced non-small-cell lung cancer: the cancer and leukemia group B (study 9730).
J Clin Oncol
2005
;
23
:
190
–6.
11
Georgoulias V, Ardavanis A, Agelidou A, et al. Docetaxel versus docetaxel plus cisplatin as front-line treatment of patients with advanced non-small-cell lung cancer: a randomized, multicenter phase III trial.
J Clin Oncol
2004
;
22
:
2602
–9.
12
Kobayashi S, Boggon TJ, Dayaram T, et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib.
N Engl J Med
2005
;
352
:
786
–92.
13
Pao W, Miller VA, Politi KA, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain.
PLoS Med
2005
;
2
:
e73
.
14
Giaccone G, Rodriguez JA. EGFR inhibitors: what have we learned from the treatment of lung cancer?
Nat Clin Pract Oncol
2005
;
2
:
554
–61.
15
Janne PA, Engelman JA, Johnson BE. Epidermal growth factor receptor mutations in non-small-cell lung cancer: implications for treatment and tumor biology.
J Clin Oncol
2005
;
23
:
3227
–34.
16
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.
17
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.
18
Perez-Soler R, Chachoua A, Hammond LA, et al. Determinants of tumor response and survival with erlotinib in patients with non-small-cell lung cancer.
J Clin Oncol
2004
;
22
:
3238
–47.
19
Niho S, Kubota K, Goto K, et al. First-line single agent treatment with gefitinib in patients with advanced non-small-cell lung cancer: a phase II study.
J Clin Oncol
2006
;
24
:
64
–9.
20
Jackman D, Lucca J, Fidias P, et al. Phase II study of the EGFR tyrosine kinase inhibitor erlotinib in patients >70 years of age with previously untreated advanced non-small cell lung carcinoma [abstract].
J Clin Oncol
2006
;
23
:
657
.
21
Herbst RS, Prager D, Hermann R, et al. TRIBUTE: a phase III trial of erlotinib hydrochloride (OSI-774) combined with carboplatin and paclitaxel chemotherapy in advanced non-small-cell lung cancer.
J Clin Oncol
2005
;
23
:
5892
–9.
22
Gatzemeier U, Pluzanska A, Szczesna A, et al. Results of a phase III trial of erlotinib (OSI-774) combined with cisplatin and gemcitabine (GC) chemotherapy in advanced non-small cell lung cancer (NSCLC) [abstract].
J Clin Oncol
2004
;
22
:
14
–619s.
23
Giaccone G, Herbst RS, Manegold C, et al. Gefitinib in combination with gemcitabine and cisplatin in advanced non-small-cell lung cancer: a phase III trial-INTACT 1.
J Clin Oncol
2004
;
22
:
777
–84.
24
Herbst RS, Giaccone G, Schiller JH, et al. Gefitinib in combination with paclitaxel and carboplatin in advanced non-small-cell lung cancer: a phase III trial - INTACT 2.
J Clin Oncol
2004
;
22
:
785
–94.
25
Miller V, Herbst RS, Prager D, et al. Long survival of never smoking non-small cell lung cancer (NSCLC) patients (pts) treated with erlotinib HCl (OSI-774) and chemotherapy: sub-group analysis of TRIBUTE [abstract].
J Clin Oncol
2004
;
22
:
14
–631s.
26
Pao W, Miller V, Zakowski M, et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib.
Proc Natl Acad Sci U S A
2004
;
101
:
13306
–11.
27
Shih JY, Gow CH, Yang PC. EGFR mutation conferring primary resistance to gefitinib in non-small-cell lung cancer.
N Engl J Med
2005
;
353
:
207
–8.
28
Bell DW, Gore I, Okimoto RA, et al. Inherited susceptibility to lung cancer may be associated with the T790M drug resistance mutation in EGFR.
Nat Genet
2005
;
37
:
1315
–6.
29
Kobayashi S, Ji H, Yuza Y, et al. An alternative inhibitor overcomes resistance caused by a mutation of the epidermal growth factor receptor.
Cancer Res
2005
;
65
:
7096
–101.
30
Kwak EL, Sordella R, Bell DW, et al. Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib.
Proc Natl Acad Sci U S A
2005
;
102
:
7665
–70.
31
Tsuchihashi Z, Khambata-Ford S, Hanna N, Janne PA. Responsiveness to cetuximab without mutations in EGFR.
N Engl J Med
2005
;
353
:
208
–29.
32
Mukohara T, Engelman JA, Hanna NH, et al. Differential effects of gefitinib and cetuximab on non-small-cell lung cancers bearing epidermal growth factor receptor mutations.
J Natl Cancer Inst
2005
;
97
:
1185
–94.
33
Tsao MS, Sakurada A, Cutz JC, et al. Erlotinib in lung cancer: molecular and clinical predictors of outcome.
N Engl J Med
2005
;
353
:
133
–44.
34
Bell DW, Lynch TJ, Haserlat SM, et al. Epidermal growth factor receptor mutations and gene amplification in non-small-cell lung cancer: molecular analysis of the IDEAL/INTACT gefitinib trials.
J Clin Oncol
2005
;
23
:
8081
–92.
35
Pao W, Wang TY, Riely GJ, et al. KRAS mutations and primary resistance of lung adenocarcinomas to gefitinib or erlotinib.
PLoS Med
2005
;
2
:
e17
.
36
Eberhard DA, Johnson BE, Amler LC, et al. Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib.
J Clin Oncol
2005
;
23
:
5900
–9.
37
Moore MJ, Goldstein D, Hamm J, et al. Erlotinib plus gemcitabine compared to gemcitabine alone in patients with advanced pancreatic cancer. A phase III trial of the National Cancer Institute of Canada Clinical Trials Group [NCIC-CTG] [abstract].
J Clin Oncol
2006
;
23
:
1s
.
38
Janmaat ML, Rodriguez JA, Gallegos-Ruiz M, Kruyt FA, Giaccone G. Enhanced cytotoxicity induced by gefitinib and specific inhibitors of the Ras or phosphatidyl inositol-3 kinase pathways in non-small cell lung cancer cells.
Int J Cancer
2006
;
118
:
209
–14.
39
Shigematsu H, Lin L, Takahashi T, et al. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers.
J Natl Cancer Inst
2005
;
97
:
339
–46.
40
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.
41
Herbst RS, Johnson DH, Mininberg E, et al. Phase I/II trial evaluating the anti-vascular endothelial growth factor monoclonal antibody bevacizumab in combination with the HER-1/epidermal growth factor receptor tyrosine kinase inhibitor erlotinib for patients with recurrent non-small-cell lung cancer.
J Clin Oncol
2005
;
23
:
2544
–55.