Purpose: To evaluate the efficacy and safety of gefitinib plus simvastatin (GS) versus gefitinib alone (G) in previously treated patients with advanced non–small cell lung cancer (NSCLC).

Experimental Design: Between May 2006 and September 2008, 106 patients (51% men, 75% adenocarcinoma, 50% never smoker) were randomly assigned to G alone (250 mg/d, n = 54) or GS (250 and 40 mg/d, respectively, n = 52). One cycle was 4 weeks of treatment. Therapy was continued until disease progression or intolerable toxicity was observed. The primary endpoint was response rate (RR). Secondary endpoints included toxicity, progression-free survival (PFS), and overall survival (OS).

Results: The RR was 38.5% (95% CI, 25.3–51.7) for GS and 31.5% (95% CI, 19.1–43.9) for G. The median PFS was 3.3 months [M] (95% CI, 1.4–5.2M) for GS and 1.9M (95% CI, 1.0–2.8M) for G. The median OS was 13.6M (95% CI, 7.1–20.1M) for GS and 12.0M (95% CI, 7.8–16.2M) for G. In exploratory subgroup analysis, GS showed higher RR (40% vs. 0%, P = 0.043) and longer PFS (3.6M vs. 1.7M, P = 0.027) compared with G alone in patients with wild-type epidermal growth factor receptor (EGFR) nonadenocarcinomas. Adverse events in both arms were generally mild and mainly consisted of skin rashes.

Conclusions: Although no superiority of GS to G was demonstrated in this unselected NSCLC population, GS showed higher RR and longer PFS compared with G alone in patients with wild-type EGFR nonadenocarcinomas. Simvastatin may improve the efficacy of gefitinib in that subgroup of gefitinib-resistant NSCLC patients. Clin Cancer Res; 17(6); 1553–60. ©2011 AACR.

Translational Relevance

The impact of statins on epidermal growth factor receptor (EGFR) function and signaling and in vivo activity against tumor cells has generated interest in studying statins as a potential EGFR-targeted therapeutic intervention. This is the first randomized phase II study comparing the efficacy of gefitinib plus simvastatin versus gefitinib alone in previously treated patients with advanced non–small cell lung cancer. The results show that in unselected patients, no significant difference in response rate and survival was observed between the 2 arms. However, gefitinib plus simvastatin improved the response rate and PFS compared with gefitinib alone in the exploratory subgroup analysis of the patients with wild-type EGFR nonadenocarcinomas. This finding suggests that inhibiting the mevalonate pathway using simvastatin may enhance the efficacy of gefitinib in this relatively gefitinib-resistant subpopulation.

The epidermal growth factor receptor (EGFR) is a key regulator of proliferation, differentiation, and survival of epithelial cells and has been implicated in the oncogenesis of epithelial cancers, including lung cancer (1). Because 50% and 80% of non–small cell lung cancer (NSCLC) cases reportedly express the EGFR protein, it is expected that an efficient EGFR inhibitor would be effective for the majority of patients with NSCLC (2). However, both gefitinib and erlotinib, orally active EGFR tyrosine kinase inhibitors (TKIs), produce durable treatment responses in a surprisingly small fraction of patients with NSCLC (3–6). DNA sequencing studies in primary lung tumor samples have shown a convincing association between responses to EGFR–TKIs and the presence of somatic mutations in the EGFR gene (7–9). Recently, several randomized phase III studies have demonstrated that gefitinib is superior to conventional chemotherapy in patients with EGFR-mutant tumors (10–12). Patients treated with gefitinib have displayed significantly longer progression-free survival (PFS) and higher response rates compared with chemotherapy among those genetically selected patients. Thus, activating somatic mutations in the EGFR gene are regarded as the strongest predictors of better response to gefitinib. However, the overall frequency of EGFR mutations is 5% to 20%, depending on geographic location (13). The remaining majority of NSCLC patients have wild-type EGFR and hardly benefit from gefitinib. To date, several studies have reported efficacy data for relapsed patients with wild-type EGFR tumors in the subgroup analysis of phase III trials. In those patients treated with gefitinib, the response rate, median progression-free and overall survival (OS) times were less than 10%, around 2 and 6 months, respectively (14–16). Although these results are similar to those observed with docetaxel monotherapy (14), several questions remain to be answered to improve the efficacy of gefitinib in those genetically unselected patients.

Statins inhibit 3-hydroxy-3-methylglutaryl-coenzyme A reductase, which is the rate-limiting enzyme of the mevalonate pathway and is required for the synthesis of cholesterol and isoprenoids such as farnesylpyrophosphate and geranylgeranylpyrophosphate (17). The impact of statins on cholesterol and isoprenoid synthesis may have anticancer effects through at least 2 primary mechanisms, which include impairment of protein prenylation and interference with the formation of cholesterol-rich lipid microdomains called lipid rafts within the cell membrane (18). Both of these processes are critical for the function of EGFR and the activity of numerous proteins important for EGFR signaling, such as Ras (18, 19). The impact of statins on EGFR function and signaling and in vivo activity against tumor cells has generated interest in studying statins as a potential EGFR-targeted therapeutic intervention. Some in vitro studies have reported that the combination of gefitinib and lovastatin has synergistic cytotoxicity and enhances EGFR inhibition in squamous cell head and neck carcinoma, NSCLC, and colon carcinoma cell lines (20–22). Interestingly, not all of the studied cell lines possess EGFR mutations, which confer increased sensitivity to gefitinib. We also showed in a previous study that the combination of gefitinib and lovastatin induces synergistic cytotoxicity in gefitinib-resistant NSCLC cells (23). Lovastatin effectively downregulates Ras protein and suppresses the phosphorylation of RAF, ERK1/2, AKT, and EGFR in NSCLC cells, which enhances gefitinib-induced apoptosis. These promising preclinical data led to the present randomized phase II study.

Patients

The main eligibility criteria were histologic or cytologic confirmation of locally advanced or metastatic (stage IIIB/IV) NSCLC after failure of at least 1 platinum-based chemotherapy, age ≥18 years, an Eastern Cooperative Oncology Group (ECOG) performance status (PS) of less than 3, and a life expectancy ≥12 weeks. Patients were required to have measurable disease by Response Evaluation Criteria in Solid Tumors (RECIST, ref. 24). Patients with brain metastases were permitted if clinically stable without steroid treatment. Exclusion criteria included significant hematologic, hepatic, renal, or cardiac dysfunction and any previous treatment with EGFR signaling inhibitors and statins.

Study design and treatment

This randomized, open-label, phase II study of gefitinib and simvastatin (CJ Pharmaceutical Co.) versus gefitinib alone was conducted at a single institution (National Cancer Center) from May 2006 to September 2008. In both study arms, patients received once-daily oral doses of 250 mg gefitinib alone (G) or in combination with 40 mg/d simvastatin (GS) in 28-day cycles. Study treatment continued until disease progression (PD) or until another termination criterion was met: unacceptable toxicity, consent withdrawal, loss to follow-up, death, major protocol violation, or noncompliance. The protocol was approved by an independent ethics committee/institutional review board and was conducted in accordance with the Declaration of Helsinki, Good Clinical Practice. Each patient provided written informed consent.

Assessment

The primary endpoint was tumor response rate (complete response [CR] + partial response [PR] using RECIST). Disease assessment was performed by investigators every 8 weeks until PD. Secondary endpoints included PFS, OS, and safety. Adverse events (AEs) were assessed throughout treatment and for 30 days after the last treatment dose and were graded using NCI CTCAE 3.0.

Detection of EGFR mutations

Whole-blood and tissue samples were collected immediately before treatment. Plasma was separated within 2 hours after the sample collection and stored at −80°C until used. Genomic DNA was extracted from plasma and paraffin-embedded tissues by using the QIAamp DNA mini kit (Qiagen). We used peptide nucleic acid (PNA) clamping-based asymmetric PCR with melting curve analysis using unlabeled probes (25). A capillary PCR machine (Light Cycler; Roche) was used instead of plate PCR, and the melting carve analysis for the probe peak was performed in the same machine. Forward and reverse primers were designed to amplify the commonly mutated portions of exon fragments 19 and 21, and the amplicon sizes were 91 and 89 base pairs, respectively. Locked nucleic acids were incorporated into the forward primer of exon 19 to increase the annealing temperature. The forward primer for exon 19 antisense PNAs and sense mutation probes were designed to span the mutation sites of exons 19 and 21 of the EGFR gene. The antisense PNA complementary to the wild-type sequence was used to clamp PCR for wild type but not mutant alleles. The sense mutation probes that were complementary to mutant alleles were used to detect both wild-type and mutant alleles. The mutation probe for exon 19 was complementary to E746-A750del type 1 (2235-2249del) and was used to detect wild-type and E746-A750del type 2 (2236-2250del) mutant as well as E746-A750del type 1. The mutation probe for exon 21 is complementary to L858R (T2573G) and was used to detect both wild-type and L858R mutant alleles.

Statistical analysis

The primary endpoint was objective tumor response rate. This study employed a “pick up the winner” design based on the randomized phase II clinical trial approach proposed by Simon and colleagues, which gives a 90% chance of selecting the better treatment if the difference is at least 15% and the smaller response rate is assumed to be 10% (26). To ensure that at least 84 patients (42 per arm) enrolled, approximately 106 patients overall were planned for. Patients were stratified based on sex (female vs. male), performance status (PS) by ECOG (0/1 vs. 2/3), and the number of prior regimens (1 vs. 2 or more). Secondary endpoints included toxicity profile, PFS, and OS. PFS was defined as the interval between the start date of treatment and the date of occurrence of progressive disease or death. OS was measured from the date of study entry until the date of death. If a patient was lost to follow-up, the patient was censored on the date of last contact. PFS and OS were evaluated by using the Kaplan-Meier method. All treated patients were included in the analysis of efficacy and safety. Although no formal statistical comparison between the 2 arms was planned, the χ2 or Fisher's exact test was performed on the response rate, and the log-rank test was applied to survival curves for exploratory purpose. Cox proportional hazards regression models were used to estimate hazard ratios (HRs). All expressed P values are 2-sided.

Patient characteristics

Between May 2006 and September 2008, 106 patients were randomly assigned and received treatment with G alone (n = 54) or GS (n = 52). All patients were assessable for efficacy and safety (Fig. 1). There were 82 deaths (77%) at the time of data cutoff (July 5, 2010): 43 patients randomly assigned to G and 39 randomly assigned to GS.

Figure 1.

CONSORT diagram.

Figure 1.

CONSORT diagram.

Close modal

Patient characteristics and baseline demographics were generally similar in both arms (Table 1). The median age was 60 years (range, 20–84), and 51% of the patients were men. Forty-nine percent of patients were never smokers, 31% were former smokers, and 20% were current smokers. Among ever (current or former) smokers, the median pack-year was 32 (range, 0.5–127.5). The majority of patients (75%) had adenocarcinoma.

Table 1.

Demographic characteristics of patients

CharacteristicsGefitinib alone (n = 54)Gefitinib and simvastatin (n = 52)
N(%)N(%)
Median age, y 60  58  
Range (32–84)  (20–76)  
Sex     
 Male 29 (54) 25 (48) 
 Female 25 (46) 27 (52) 
ECOG performance status     
 0 16 (30) 19 (36) 
 1 32 (59) 30 (58) 
 2 (11) (4) 
 3 (0) (2) 
Stage     
 IIIB (11) (6) 
 IV 48 (89) 49 (94) 
Prior chemotherapy     
 1 24 (44) 25 (48) 
 2 30 (56) 25 (48) 
 3 (0) (4) 
Histology     
 Adenocarcinoma 41 (76) 39 (75) 
 Squamous (15) (17) 
 NOSa (9) (8) 
Smoking status     
 Never smoker 27 (50) 26 (50) 
 Former smoker 16 (30) 16 (31) 
 Current smoker 11 (20) 10 (19) 
 Pack-year, median (range) 33 (2–60) 32.5 (0.5–127.5) 
EGFR mutations (n = 94)     
 Negative 31 (69) 37 (76) 
 Positive 14 (31) 12 (25) 
CharacteristicsGefitinib alone (n = 54)Gefitinib and simvastatin (n = 52)
N(%)N(%)
Median age, y 60  58  
Range (32–84)  (20–76)  
Sex     
 Male 29 (54) 25 (48) 
 Female 25 (46) 27 (52) 
ECOG performance status     
 0 16 (30) 19 (36) 
 1 32 (59) 30 (58) 
 2 (11) (4) 
 3 (0) (2) 
Stage     
 IIIB (11) (6) 
 IV 48 (89) 49 (94) 
Prior chemotherapy     
 1 24 (44) 25 (48) 
 2 30 (56) 25 (48) 
 3 (0) (4) 
Histology     
 Adenocarcinoma 41 (76) 39 (75) 
 Squamous (15) (17) 
 NOSa (9) (8) 
Smoking status     
 Never smoker 27 (50) 26 (50) 
 Former smoker 16 (30) 16 (31) 
 Current smoker 11 (20) 10 (19) 
 Pack-year, median (range) 33 (2–60) 32.5 (0.5–127.5) 
EGFR mutations (n = 94)     
 Negative 31 (69) 37 (76) 
 Positive 14 (31) 12 (25) 

aNon–small cell lung cancer, not otherwise specified.

Safety

The safety profiles of both arms were similar (Table 2). The most commonly observed AE was rash. Overall, the grade 3 or 4 toxicity rates were low in both arms. There were no treatment-related deaths.

Table 2.

Common adverse events

Gefitinib alone (n = 54)Gefitinib+Simvastatin (n = 52)
Grade 1Grade 2Grade 3Grade 4Grade 1Grade 2Grade 3Grade 4
N(%)N(%)N(%)N(%)N(%)N(%)N(%)N(%)
Rash 21 (39) 17 (32) (2) (0) 14 (27) 21 (40) (4) (0) 
Dry skin 30 (56) (4) (0) (0) 26 (50) (12) (0) (0) 
Stomatitis 11 (20) (2) (0) (0) 21 (40) (2) (0) (0) 
Anorexia 20 (37) (9) (0) (0) 16 (31) (7) (0) (0) 
Diarrhea 18 (33) (6) (0) (0) 17 (33) (8) (0) (0) 
Asthenia (13) (4) (0) (0) (13) (4) (0) (0) 
Nausea (9) (0) (0) (0) (12) (0) (0) (0) 
Gefitinib alone (n = 54)Gefitinib+Simvastatin (n = 52)
Grade 1Grade 2Grade 3Grade 4Grade 1Grade 2Grade 3Grade 4
N(%)N(%)N(%)N(%)N(%)N(%)N(%)N(%)
Rash 21 (39) 17 (32) (2) (0) 14 (27) 21 (40) (4) (0) 
Dry skin 30 (56) (4) (0) (0) 26 (50) (12) (0) (0) 
Stomatitis 11 (20) (2) (0) (0) 21 (40) (2) (0) (0) 
Anorexia 20 (37) (9) (0) (0) 16 (31) (7) (0) (0) 
Diarrhea 18 (33) (6) (0) (0) 17 (33) (8) (0) (0) 
Asthenia (13) (4) (0) (0) (13) (4) (0) (0) 
Nausea (9) (0) (0) (0) (12) (0) (0) (0) 

Response and survival

The response rate was 31.5% (95% CI, 19.1–43.9%) for G and 38.5% (95% CI, 25.3–51.7%) for GS (Table 3). Median PFS was 1.9 months (95% CI, 1.0–2.8 months) for G and 3.3 months (95% CI, 1.4–5.2 months) for GS (Fig. 2A). The estimated HR was 0.891 (95% CI, 0.604–1.315, P = 0.549). After a median follow-up of 30 months, the median OS was 12 months (95% CI, 7.8–16.2 months) for G and 13.6 months (95% CI, 7.1–20.1 months) for GS (Fig. 2B). The 1-year survival rate was 50% for G and 56% for GS; the 2-year survival rates were 30% and 28%, respectively. The estimated HR was 0.876 (95% CI, 0.567–1.354, P = 0.491).

Figure 2.

Progression-free survival (A) and overall survival (B).

Figure 2.

Progression-free survival (A) and overall survival (B).

Close modal
Table 3.

Overall and subset analysis of response according to treatment

ResponseGefitinib alone (n = 54)Gefitinib and simvastatin (n = 52)P
N(%)N(%)
Overall PR 17 (31.5) 20 (38.5) .666 
 SD 10 (18.5) (13.5)  
 PD 27 (50.0) 25 (48)  
       
Subset analysis  RR (NRR (NP 
Histology Adenocarcinoma 39% (16/41) 39% (15/39) .959 
 Nonadenocarcinoma 8% (1/13) 39% (5/13) .063 
Smoking status Never-smoker 44% (12/27) 62% (16/26) .213 
 Ever-smoker 19% (5/27) 15% (4/26) .761 
Sex Male 21% (6/29) 16% (4/25) .658 
 Female 44% (11/25) 59% (16/27) .271 
EGFR mutations Positive 64% (9/14) 75% (9/12) .555 
 Negative 13% (4/31) 27% (10/37) .151 
Adenocarcinoma EGFR mutation-positive 62% (8/13) 80% (8/10) .340 
 EGFR mutation-negative 19% (4/21) 22% (6/27) .788 
Nonadenocarcinoma EGFR mutation-positive 100% (1/1) 50% (1/2) .667 
 EGFR mutation-negative 0% (0/10) 40% (4/10) .043 
ResponseGefitinib alone (n = 54)Gefitinib and simvastatin (n = 52)P
N(%)N(%)
Overall PR 17 (31.5) 20 (38.5) .666 
 SD 10 (18.5) (13.5)  
 PD 27 (50.0) 25 (48)  
       
Subset analysis  RR (NRR (NP 
Histology Adenocarcinoma 39% (16/41) 39% (15/39) .959 
 Nonadenocarcinoma 8% (1/13) 39% (5/13) .063 
Smoking status Never-smoker 44% (12/27) 62% (16/26) .213 
 Ever-smoker 19% (5/27) 15% (4/26) .761 
Sex Male 21% (6/29) 16% (4/25) .658 
 Female 44% (11/25) 59% (16/27) .271 
EGFR mutations Positive 64% (9/14) 75% (9/12) .555 
 Negative 13% (4/31) 27% (10/37) .151 
Adenocarcinoma EGFR mutation-positive 62% (8/13) 80% (8/10) .340 
 EGFR mutation-negative 19% (4/21) 22% (6/27) .788 
Nonadenocarcinoma EGFR mutation-positive 100% (1/1) 50% (1/2) .667 
 EGFR mutation-negative 0% (0/10) 40% (4/10) .043 

Abbreviations: PR, partial response; PD, progressive disease by RECIST1.0; RR, response rate; SD, stable disease.

EGFR mutation analysis and exploratory subgroup analyses

Among a total of 106 patients, 94 plasma DNA samples were adequate for EGFR mutation analysis. Activating mutations were detected in 26 of 94 (28%) cases (23 exon 19 deletions and 3 exon 21 L858R mutations, Table 1). To validate the plasma EGFR mutation results, we tested the 2 major EGFR mutations, the exon 19 deletion and exon 21 (L858R), in the paired tumor tissues. In the 15 paired specimens of plasma and tumor tissues, 11 (73%) revealed concordant results. The comparison between EGFR mutation status in plasma and tumor samples is summarized in Table 4. Although they were not statistically significant, trends toward higher frequency of EGFR mutations were observed in adenocarcinoma (32% [23/71] vs. 13% [3/23] for nonadenocarcinoma, P = 0.107), women (36% [16/45] vs. 20% [10/49] for men, P = 0.101), and never smokers (33% [15/46] vs. 23% [11/48] for ever smokers, P = 0.294). Among the entire population, patients with EGFR-mutant tumors showed significantly higher response rates (69% [18/26] vs. 21% [14/68] for wild-type EGFR, P < 0.0001), and longer PFS (HR = 0.386 [95% CI, 0.238–0.627], P < 0.0001) and OS (HR = 0.540 [95% CI, 0.316–922], P = 0.024) compared with those with wild-type EGFR tumors.

Table 4.

Comparison of EGFR mutation status in the paired specimens of plasma and tumor samples (n = 15)

Tumor tissuePlasma sample
PositiveNegative
Positive 
Negative 
Tumor tissuePlasma sample
PositiveNegative
Positive 
Negative 

Exploratory subgroup analyses (Table 3) suggested a trend toward an improved response rate in the GS group compared with the G group among patients with nonadenocarcinoma histology (39% vs. 8%, respectively, P = 0.063) and wild-type EGFR (27% vs. 13%, respectively, P = 0.151). We therefore subdivided tumor histology according to EGFR mutation status and compared the efficacy of G versus GS. Among patients with nonadenocarcinomas, those with wild-type EGFR showed a higher response rate (40% vs. 0%, respectively, P = 0.043) and longer median PFS (3.7 months vs. 1.7 months, respectively, P = 0.027; Fig. 3) when treated with GS compared with G alone.

Figure 3.

Progression-free survival in nonadenocarcinoma with EGFR wild type.

Figure 3.

Progression-free survival in nonadenocarcinoma with EGFR wild type.

Close modal

This is the first randomized phase II study comparing the efficacy of GS versus G in previously treated patients with advanced NSCLC. The results show that in unselected patients, no significant difference in response rate and survival was observed between the 2 arms. However, GS improved the response rate and PFS compared with G in the exploratory subgroup analysis of the patients with wild-type EGFR nonadenocarcinomas. This finding suggests that inhibiting the mevalonate pathway using simvastatin may enhance the efficacy of gefitinib in this relatively gefitinib-resistant subpopulation.

To date, several preclinical studies have shown that the combination of statins with gefitinib induces a potent synergistic cytotoxicity in a variety of tumors without EGFR mutations (20, 21). Recently, Zhao and colleagues reported that lovastatin inhibits ligand-induced EGFR dimerization in squamous cell head and neck carcinoma cells, which results in inhibition of AKT activation along with its downstream targets that regulate protein translation initiation. Lovastatin induces actin cytoskeletal disorganization and increases the expression of inactive rhoA, which also inhibits EGFR dimerization and activation. Furthermore, they retrospectively evaluated the effect of statin on the OS and disease-specific survival (DSS) of NSCLC patients enrolled in the NCIC Clinical Trials Group phase III clinical trial BR21 (erlotinib vs. placebo). Although it was not statistically significant, erlotinib-treated patients with statin use showed a trend toward improved OS and DSS compared with patients without statin use (22). In our study, patients with wild-type EGFR nonadenocarcinomas treated with GS showed higher response rates and longer PFS compared with G. Although the number of these patients was small, these clinical observations may support the preclinical evidence of the effective EGFR-inhibitory activity of statins in wild-type EGFR non-adenocarcinoma tumors. Moreover, the combination treatment was generally well tolerated, and the AEs observed were similar to those reported with gefitinib alone.

In our study, some inconsistency of EGFR mutation results was found between tissue and plasma DNA. The significance of a negative result for an EGFR mutation is highly dependent upon samples tested as well as methods performed. Direct DNA sequencing is a common detection method but has well-known sensitivity limitations depending on the proportion of tumor cells present in the material available for DNA extraction (27). The peptide nucleic acid-locked nucleic acid (PNA–LNA) PCR clamp is capable of detecting EGFR mutations in the presence of 100-fold background levels of wild-type EGFR from normal cells. Because of its high sensitivity and specificity, PNA–LNA PCR clamp is considered suitable to detect EGFR mutations in cytology samples (28). However, this method uses mutation-specific primers and therefore can miss rare mutations (e.g., L861Q or exon 18 mutations). In addition, the rate of detection of L858R in our study was very low compared with the rate of E746_A750del. Similar results were also reported previously using Scorpion Amplified Refractory Mutation System technology detect EGFR mutations in serum DNA (29). Further analyses in much larger groups of patients will be necessary to clarify the low-frequency L858R mutation could be due to assay-related false-negative findings.

Although this study did not demonstrate the superiority of gefitinib plus simvastatin as a salvage treatment in unselected NSCLC patients, the improved efficacy of this combination in the patients with wild-type EGFR nonadenocarcinomas may provide a novel, promising therapeutic approach using gefitinib as a salvage treatment in those gefitinib-resistant patients. Further studies of this combination as a salvage treatment for patients with wild-type EGFR nonadenocarcinomas are warranted.

No potential conflicts of interest were disclosed.

This study was supported by a grant from the National Cancer Center 0810130.

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.

1.
Mendelsohn
J
,
Baselga
J
. 
The EGF receptor family as targets for cancer therapy
.
Oncogene
2000
;
19
:
6550
65
.
2.
Gazdar
AF
. 
Epidermal growth factor receptor inhibition in lung cancer: the evolving role of individualized therapy
.
Cancer Metastasis Rev
2010
;
29
:
37
48
.
3.
Fukuoka
M
,
Yano
S
,
Giaccone
G
,
Tamura
T
,
Nakagawa
K
,
Douillard
JY
, et al
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
.
4.
Kris
MG
,
Natale
RB
,
Herbst
RS
,
Lynch
TJ
 Jr
,
Prager
D
,
Belani
CP
, 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
.
5.
Thatcher
N
,
Chang
A
,
Parikh
P
,
Rodrigues
Pereira J
,
Ciuleanu
T
,
von Pawel
J
, et al
Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: results from a randomised, placebo-controlled, multicentre study (Iressa Survival Evaluation in Lung Cancer)
.
Lancet
2005
;
366
:
1527
37
.
6.
Shepherd
FA
,
Rodrigues
Pereira J
,
Ciuleanu
T
,
Tan
EH
,
Hirsh
V
,
Thongprasert
S
, et al
National Cancer Institute of Canada Clinical Trials Group. Erlotinib in previously treated non-small-cell lung cancer
.
N Engl J Med
2005
;
353
:
123
32
.
7.
Lynch
TJ
,
Bell
DW
,
Sordella
R
,
Gurubhagavatula
S
,
Okimoto
RA
,
Brannigan
BW
, 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
.
8.
Paez
JG
,
Jänne
PA
,
Lee
JC
,
Tracy
S
,
Greulich
H
,
Gabriel
S
, et al
EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy
.
Science
2004
;
304
:
1497
500
.
9.
Pao
W
,
Miller
V
,
Zakowski
M
,
Doherty
J
,
Politi
K
,
Sarkaria
I
, 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
.
10.
Mok
TS
,
Wu
YL
,
Thongprasert
S
,
Yang
CH
,
Chu
DT
,
Saijo
N
, et al
Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma
.
N Engl J Med
2009
;
361
:
947
57
.
11.
Maemondo
M
,
Inoue
A
,
Kobayashi
K
,
Sugawara
S
,
Oizumi
S
,
Isobe
H
, et al
Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR
.
N Engl J Med
2010
;
362
:
2380
8
.
12.
Mitsudomi
T
,
Morita
S
,
Yatabe
Y
,
Negoro
S
,
Okamoto
I
,
Tsurutani
J
, 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
2010
;
11
:
121
8
.
13.
Gazdar
AF
. 
Activating and resistance mutations of EGFR in non-small-cell lung cancer: role in clinical response to EGFR tyrosine kinase inhibitors
.
Oncogene
2009
;
28
(
Suppl 1
):
S24
31
.
14.
Kim
ES
,
Hirsh
V
,
Mok
T
,
Socinski
MA
,
Gervais
R
,
Wu
YL
, et al
Gefitinib versus docetaxel in previously treated non-small-cell lung cancer (INTEREST): a randomised phase III trial
.
Lancet
2008
;
372
:
1809
18
.
15.
Zhu
CQ
,
da Cunha Santos
G
,
Ding
K
,
Sakurada
A
,
Cutz
JC
,
Liu
N
, et al
Role of KRAS and EGFR as biomarkers of response to erlotinib in National Cancer Institute of Canada Clinical Trials Group Study BR.21
.
J Clin Oncol
2008
;
26
:
4268
75
.
16.
Hirsch
FR
,
Varella-Garcia
M
,
Bunn
PA
 Jr
,
Franklin
WA
,
Dziadziuszko
R
,
Thatcher
N
, et al
Molecular predictors of outcome with gefitinib in a phase III placebo-controlled study in advanced non-small-cell lung cancer
.
J Clin Oncol
2006
;
24
:
5034
42
.
17.
Endo
A
. 
The discovery and development of HMG-CoA reductase inhibitors
.
J Lipid Res
1992
;
33
:
1562
82
.
18.
Grzegorz
S.
,
Nowakowski
GS
,
Maurer
MJ
, et al
Statin use and prognosis in patients with diffuse large b-cell lymphoma and follicular lymphoma in the rituximab era
.
J Clin Oncol
2010
;
28
:
412
7
.
19.
Jakobisiak
M
,
Golab
J
. 
Potential antitumor effects of statins
.
Int J Oncol
2003
;
23
:
1055
69
.
20.
Mantha
AJ
,
Hanson
JE
,
Goss
G
,
Lagarde
AE
,
Lorimer
IA
,
Dimitroulakos
J
. 
Targeting the mevalonate pathway inhibits the function of the epidermal growth factor receptor
.
Clin Cancer Res
2005
;
11
:
2398
407
.
21.
Dimitroulakos
J
,
Lorimer
IA
,
Goss
G
. 
Strategies to enhance epidermal growth factor inhibition: targeting the mevalonate pathway
.
Clin Cancer Res
2006
;
12
:
4426s
31s
.
22.
Zhao
TT
,
Le Francois
BG
,
Goss
G
,
Ding
K
,
Bradbury
PA
,
Dimitroulakos
J
. 
Lovastatin inhibits EGFR dimerization and AKT activation in squamous cell carcinoma cells: potential regulation by targeting rho proteins
.
Oncogene
2010
;
29
:
4682
92
.
23.
Park
IH
,
Kim
JY
,
Jung
JI
,
Han
JY
. 
Lovastatin overcomes gefitinib resistance in human non-small cell lung cancer cells with K-Ras mutations
.
Invest New Drugs
2009 Sep 17
.
[Epub ahead of print]
.
24.
Therasse
P
,
Arbuck
SG
,
Eisenhauer
EA
,
Wanders
J
,
Kaplan
RS
,
Rubinstein
L
, 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
.
25.
Oh
JE
,
Lim
HS
,
An
CH
,
Jeong
EG
,
Han
JY
,
Lee
SH
, et al
Detection of low-level KRAS mutations using PNA-mediated asymmetric PCR clamping and melting curve analysis with unlabeled probes
.
J Mol Diagn
2010
;
12
:
418
24
.
26.
Simon
R
,
Wittes
RE
,
Ellenberg
SS
. 
Randomized phase II clinical trials
.
Cancer Treat Rep
1985
;
69
:
1375
81
.
27.
Pao
W
,
Ladanyi
M
. 
Epidermal growth factor receptor mutation testing in lung cancer: searching for the ideal method
.
Clin Cancer Res
2007
;
13
:
4954
5
.
28.
Sutani
A
,
Nagai
Y
,
Udagawa
K
,
Uchida
Y
,
Koyama
N
,
Murayama
Y
, et al
Gefitinib for non-small-cell lung cancer patients with epidermal growth factor receptor gene mutations screened by peptide nucleic acid-locked nucleic acid PCR clamp
.
Br J Cancer
2006
;
95
:
1483
9
.
29.
Kimura
H
,
Kasahara
K
,
Kawaishi
M
,
Kunitoh
H
,
Tamura
T
,
Holloway
B
, 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
.