Purpose: Lung adenocarcinomas among never-smokers are more common in females than in males. This implies that gender-dependent hormones promote smoking unrelated lung adenocarcinoma. We therefore investigated mRNA expression of aromatase, an intrinsic estrogen synthetase, in lung adenocarcinoma and assessed its correlation to clinicopathologic factors, including EGFR mutations and postsurgical prognosis.

Experimental Design: Aromatase mRNA expression in primary tumor samples from 110 patients with lung adenocarcinoma was evaluated with qRT-PCR. Inhibitory effects of the aromatase inhibitor exemestane were assessed in lung adenocarcinoma cell lines (11-18 and HCC4006), which have EGFR mutations, separately and combined with EGFR tyrosine kinase inhibitor erlotinib.

Results: Aromatase gene expression was not correlated with patients' clinicopathologic factors, including EGFR mutation status. High aromatase expression was associated with poor prognosis for both recurrence-free survival (P = 0.004) and overall survival (P = 0.003). In addition, the prognostic significance of aromatase expression was limited to females, never-smokers, and patients with EGFR mutations, but not in their counterparts. HCC4006, which has a low aromatase mRNA expression level, was not sensitive to exemestane, either alone or combined with erlotinib. In contrast, growth of 11-18 cells, which have high aromatase expression, was significantly inhibited by exemestane, both alone and combined with erlotinib.

Conclusions: Aromatase is a candidate prognostic factor in patients with lung adenocarcinoma, especially in those with EGFR mutations, and may also be a beneficial therapeutic target in those patients. Clin Cancer Res; 20(13); 3613–22. ©2014 AACR.

Translational Relevance

The proportion of females among patients with lung cancer with no smoking history is reportedly increasing, which implies that female hormones may affect the development of lung cancer. We examined mRNA expression of aromatase (CYP19A1), a possible intrinsic estrogen-synthetase, in patients with primary lung adenocarcinoma, and assessed its correlation with clinicopathologic factors including EGFR mutation status and prognosis. High aromatase gene expression was associated with poor outcomes. The prognostic significance of aromatase expression was also demonstrated in females, never-smokers, and patients with EGFR mutations, whereas such significance was not observed in their counterparts. In vitro analysis showed an antitumor effect of aromatase inhibitor in a lung adenocarcinoma cell line with an EGFR mutation and high aromatase expression. Our findings suggest aromatase is a possible therapeutic target in lung adenocarcinomas with EGFR mutations.

Worldwide, lung cancer is the leading cause of cancer death in males, and the second leading cause of cancer death in females (1). Although tobacco smoking is the predominant risk factor for lung cancer, approximately 25% of lung cancer cases are not attributable to tobacco use (2). The proportion of never-smokers among patients with non–small cell lung cancer (NSCLC) has significantly increased for decades. NSCLC in never-smokers is more frequent in females and the adenocarcinoma cell type, and has a better prognosis compared with NSCLC in ever smokers (3, 4). Furthermore, frequencies of oncogenic drivers, such as mutations in KRAS or epidermal growth factor receptor (EGFR), or echinoderm microtubule–associated protein-like 4 (EML4)–anaplastic lymphoma kinase (ALK) fusion are different between lung cancers in never-smokers and those in smokers (5–7). These striking differences in epidemiologic, clinical, and molecular characteristics suggest that lung cancers associated with smoking and those unassociated with smoking are separate entities (2, 3).

The higher proportion of females among patients with lung cancer who have never smoked suggests a possible role for gender-dependent hormones in lung cancer development. Estrogen reportedly affects differentiation and maturation of the normal lungs (8) and stimulates lung tumor growth in both laboratory-based (9–12) and clinical studies (13–16). Epidemiologic studies also have suggested that endogenous and exogenous estrogen affect development of lung cancer (17). A post hoc analysis of a randomized controlled trial in postmenopausal women showed that hormone replacement therapy (HRT) may increase the risk of death from lung cancer (18). A prospective cohort study confirmed dose-dependent increase in lung cancer risk among women who received HRT (19). A decreased incidence of lung cancer was observed in patients with breast cancer treated with an aromatase inhibitor, exemestane, after tamoxifen therapy compared with patients who continued tamoxifen therapy (20). These data strongly support an important effect of female hormones in lung cancer development.

Aromatase (CYP19A1) is a cytochrome P-450 enzyme that converts androstenedione and testosterone to estrone and estradiol, respectively, and supports breast and endometrial cancer growth via autocrine and paracrine stimulation (21, 22). In not only gonadal tissue, but also in lung tissue, estrogen is synthesized mainly by aromatase (12, 23). Treatment with aromatase inhibitor has been found to suppress growth in lung cancer cell lines and mice tumor xenografts (12). In early-stage lung cancer, high aromatase expression reportedly correlates with poor prognosis in women ages ≥65 years (13). These studies suggest that aromatase affects lung cancer development, although the precise pathway is unclear.

After somatic EGFR mutations were discovered in NSCLC, many studies found higher mutation frequencies in East Asians, women, never-smokers, and adenocarcinomas (5, 6). The prevalence of EGFR mutation among female patients implies interactions between female hormones and EGFR mutations. Interactions between estrogen receptor (ER) and EGFR pathways have been extensively investigated in vitro (10, 11, 24, 25) and in tumor specimens (14, 15, 26, 27), but the influence of aromatase on EGFR-dependent growth is not clear.

We studied expression and prognostic significance of aromatase, with regard to EGFR mutation status, in patients with primary lung adenocarcinoma. We also examined growth inhibition by aromatase inhibitor combined with EGFR tyrosine kinase inhibitor (TKI) on lung cancer cell lines with EGFR mutations.

Human tissue samples

Primary tumor and corresponding nonneoplastic lung specimens were collected from 110 consecutive patients who underwent complete resections (R0) for primary lung adenocarcinoma from April 2007 to March 2011 at the Department of Surgery and Science, Kyushu University Hospital (Fukuoka, Japan), for whom surgical specimens were available and EGFR mutation status were determined (Table 1). This study included 44 men and 66 women, with a mean age of 67.7 years (range: 37–85 years) at surgical resection. Almost all of the women were postmenopausal. Histologic tumor diagnoses were based on hematoxylin and eosin-stained preparations, using the WHO 2004 classification (28). Pathologic staging was performed according to the 7th edition of the TNM Classification of Malignant Tumors (29). EGFR mutation tests used the peptide nucleic acid–locked nucleic acid (PNA-LNA; Mitsubishi Chemical Medience, Tokyo, Japan) polymerase chain reaction (PCR) clamp method (30) with formalin-fixed paraffin-embedded sections of surgical specimens. No patient was treated with chemotherapy or radiotherapy before surgery. Thirty-nine (35.5%) patients received postoperative chemotherapy: 21 received oral tegafur and uracil, 17 were enrolled into a clinical trial for the postoperative adjuvant chemotherapy (S-1 or cisplatin-S-1), and 1 received paclitaxel. A routine check-up with a physical examination, blood cell counts, serum chemistry, serum tumor markers including carcinoembryonic antigen and cytokeratin fragment 19, and chest X-rays were performed on an outpatient basis 4 times a year for the first 3 years, and thereafter twice annually. Computed tomography was performed twice a year for the first 3 years, and thereafter at least annually. Brain magnetic resonance imaging, and bone scintigram or fluorodeoxyglucose positron-emission tomography were performed annually. This study was approved by the Kyushu University Institutional Review Board for Clinical Research (no. 24–173).

Table 1.

Clinicopathologic characteristics by aromatase expression (n = 110)

Aromatase expression
CharacteristicNumber (n = 110)Low (n = 83)High (n = 27)P
Age (y) < 70 58 44 14 0.92 
 ≥ 70 52 39 13  
Sex Male 44 33 11 0.93 
 Female 66 50 16  
Smoking history Never 60 46 14 0.75 
 Current or former 50 37 13  
EGFR mutation Negative 56 44 12 0.44 
 Positive 54 39 15  
SUVmaxa   6.1 ± 4.7 6.4 ± 4.5 0.76 
Tumor size (cm)   2.9 ± 1.7 3.0 ± 1.5 0.47 
Histologic grade G1 53 44 0.21 
 G2 41 28 13  
 G3 16 11  
 G4  
Pleural invasionb Negative 89 68 21 0.55 
 Positive 20 14  
Lymphatic invasion Negative 97 73 24 0.90 
 Positive 13 10  
Vascular invasion Negative 78 61 17 0.30 
 Positive 32 22 10  
Pathologic stage 81 64 17 0.15 
 II, IIIA 29 19 10  
Aromatase expression
CharacteristicNumber (n = 110)Low (n = 83)High (n = 27)P
Age (y) < 70 58 44 14 0.92 
 ≥ 70 52 39 13  
Sex Male 44 33 11 0.93 
 Female 66 50 16  
Smoking history Never 60 46 14 0.75 
 Current or former 50 37 13  
EGFR mutation Negative 56 44 12 0.44 
 Positive 54 39 15  
SUVmaxa   6.1 ± 4.7 6.4 ± 4.5 0.76 
Tumor size (cm)   2.9 ± 1.7 3.0 ± 1.5 0.47 
Histologic grade G1 53 44 0.21 
 G2 41 28 13  
 G3 16 11  
 G4  
Pleural invasionb Negative 89 68 21 0.55 
 Positive 20 14  
Lymphatic invasion Negative 97 73 24 0.90 
 Positive 13 10  
Vascular invasion Negative 78 61 17 0.30 
 Positive 32 22 10  
Pathologic stage 81 64 17 0.15 
 II, IIIA 29 19 10  

aData not available for 19 of the aromatase-low patients and 5 of the aromatase-high patients.

bData not available for one of the aromatase-low patients.

Tumor samples and corresponding nonneoplastic lung tissues (most distant from tumor) were obtained immediately after resection, frozen in liquid nitrogen, and stored at −80°C.

Cells and reagents

We obtained 21 lung adenocarcinoma cell lines and the breast cancer line MCF-7. A549, LK87, PC-9, and 11-18 cell lines were the kind gift of Dr. M. Takeshita. HCC4006 cell line was the kind gift of Dr. A.F. Gazdar, and was confirmed by identification of the rare EGFR deletion mutation (del L747_E749, A750P) in this cell line (31). The ACC-LC-319 cell line was a kind gift from Dr. T. Hida. Total RNAs from other cell lines were extracted in previous analyses (32, 33) or were the kind gifts of Dr. K. Tomizawa and Dr. T. Mitsudomi.

Driver mutations of the cell lines were KRAS mutations: A549, ACC-LC-94, H23, H358, H2009, LK87, and SK-LU1; EGFR mutations: H3255, HCC827, HCC4006, PC-9, and 11–18; MET mutation: H596; MET amplifications: ACC-LC-319 and H1993; Ros fusion: HCC78; HER2 mutation: H1781; EML4/ALK fusion, H2228; unknown: HCC193, SK-LC-3, and VMRC-LCD.

Cells were maintained in RPMI 1640 medium (Life Technologies) containing 10% fetal bovine serum (Life Technologies), 100 IU/mL penicillin, and 100 μg/mL streptomycin. The cells were maintained in a humidified atmosphere of 5% CO2 in air at 37 °C.

EGFR-TKI erlotinib and aromatase inhibitor exemestane were purchased from Selleck Chemicals and LKT Laboratories, respectively.

RNA extraction and quantitative RT-PCR

The aromatase mRNA expression levels were evaluated by quantitative RT-PCR. Total RNA was extracted from resected lung tissues and cell lines using ISOGEN (Nippon Gene) according to the manufacturer's protocol. cDNA was synthesized using a SuperScript III First-Strand Synthesis SuperMix (Invitrogen) according to the manufacturer's protocol. Quantitative PCR amplification was performed using Applied Biosystems StepOnePlus real-time PCR system (Life Technologies). TaqMan gene expression assay (Applied Biosystems) for CYP19A1 (Hs00903413_m1) was used and β-actin (Hs99999903_m1) was used as an internal control. The BD qPCR total RNA human reference (Clontech Laboratories, Inc.), corresponding to a standardized mixture of total RNAs from a collection of adult human tissues, was used as a standard for quantitation. Relative aromatase mRNA expression levels of each sample (tissue and cell line) were standardized to those of β-actin and calculated relative to that of the total RNA human reference. Each sample was tested with triplicate measurements, and the mean value of the triplicate measurements was defined as a final value. We divided patients with adenocarcinoma into 2 groups based on the expression level of aromatase compared with human reference; high aromatase expression was defined as being above the human reference expression, whereas low expression was defined as being below it.

Cell-proliferation assay

HCC4006, 11-18, H358, H2228, and ACC-LC-319 cells (5 × 103) were plated into each well of 96-well flat-bottomed plates and grown in phenol red-free RPMI 1640 (Life Technologies) containing 10% dextran-coated charcoal-stripped fetal bovine serum (Biological Industries). Twenty-four hours later, dimethyl sulfoxide (DMSO), erlotinib, exemestane, or a combination of these drugs was added to achieve the indicated drug concentration, and cells were incubated for an additional 72 hours. The viability of drug-treated cells was determined by a WST-8 method using Cell Count Reagent SF (Nacalai Tesque) according to the manufacturer's instructions. Percent growth was determined relative to DMSO-treated controls.

Statistical analysis

Statistical analysis was performed using JMP statistical software version 9.0.2 (SAS Institute Inc.). All variables are expressed as the mean ± standard deviation (SD). Qualitative variables were compared using χ2 tests, and quantitative variables were compared using Wilcoxon tests. Multivariate models were constructed using logistic regression, including sex, smoking history, and EGFR mutation status, with aromatase expression (high/low) as the outcome of interest. Survival curves were drawn using the Kaplan–Meier method. Significant differences among subgroups were compared using the log-rank test. The Cox proportional hazard regression model was used to explore the effects of the clinicopathologic variables and aromatase expression on survival. Factors showing prognostic significance in the univariate analyses were adopted as variables in multivariate analysis. P < 0.05 was considered statistically significant.

Expression of aromatase mRNA in lung adenocarcinoma tissues and corresponding nonneoplastic lung tissues

We first examined the mRNA expression level of aromatase in lung adenocarcinoma and corresponding nonneoplastic lung tissues, using quantitative RT-PCR. Relative aromatase mRNA expression in carcinoma tissues (0.83 ± 1.06) was significantly higher than in corresponding nonneoplastic lung tissues (0.55 ± 0.46; P = 0.025; Fig. 1). Aromatase mRNA expression in nonneoplastic lung tissues did not significantly differ among subgroups divided by age (<70 vs. ≥70), sex, smoking history, or EGFR mutation status (P = 0.07, 0.58, 0.46, and 0.61, respectively).

Figure 1.

Differences in aromatase mRNA expression levels between carcinoma tissues and corresponding nonneoplastic lung tissues (n = 94). Each value is shown in an open circle; paired values of the same patient are connected by a line. Two data groups are shown as box-and-whisker plots, with the bottom and top of the box at the first and third quartiles, and the band inside the box at the median. Upper and lower whiskers indicate 90th and 10th percentiles, respectively. Aromatase mRNA level for the human reference RNA is set as 1. Statistical difference was determined by a Wilcoxon matched-pair signed-rank test.

Figure 1.

Differences in aromatase mRNA expression levels between carcinoma tissues and corresponding nonneoplastic lung tissues (n = 94). Each value is shown in an open circle; paired values of the same patient are connected by a line. Two data groups are shown as box-and-whisker plots, with the bottom and top of the box at the first and third quartiles, and the band inside the box at the median. Upper and lower whiskers indicate 90th and 10th percentiles, respectively. Aromatase mRNA level for the human reference RNA is set as 1. Statistical difference was determined by a Wilcoxon matched-pair signed-rank test.

Close modal

Relationship between aromatase expression and clinicopathologic factors

In univariate analysis, no significant correlation was identified between tumor aromatase expression level and clinicopathologic factors that are associated with smoking-unrelated lung cancer, such as sex, smoking history, and EGFR mutation status (Table 1). The same results were observed for other clinicopathologic characteristics; age, maximum standardized uptake value (SUVmax), tumor size, histologic grade, pleural invasion, lymphatic invasion, vascular invasion, and pathologic stage. In addition, in multivariate analysis, no significant association between high aromatase expression and sex, smoking history, or EGFR mutation status was identified (Supplementary Table S1). We also examined tumor aromatase expression as a continuous variable. However, any statistically significant correlation was still not found between aromatase expression level and clinicopathologic factors, although tendencies were seen in vascular invasion (P = 0.06) and pathologic stage (I vs. ≥II, P = 0.051).

Influence of aromatase gene expression level on survival

A survival analysis was performed in 110 patients who underwent curative resections. Median follow-up time was 35 months (range: 9–66 months). High expression of aromatase was associated with poor prognosis in terms of both recurrence-free survival (RFS; P = 0.004; Fig. 2A) and overall survival (OS; P = 0.003; Fig. 2B).

Figure 2.

Kaplan–Meier postoperative RFS and OS curves according to aromatase expression level. N, number of patients in each category. RFS (A) and OS (B) curves for all cohort patients. RFS (C) and OS (D) curves for females. RFS (E) and OS (F) curves for never-smokers. RFS (G) and OS (H) curves for patients with EGFR mutations.

Figure 2.

Kaplan–Meier postoperative RFS and OS curves according to aromatase expression level. N, number of patients in each category. RFS (A) and OS (B) curves for all cohort patients. RFS (C) and OS (D) curves for females. RFS (E) and OS (F) curves for never-smokers. RFS (G) and OS (H) curves for patients with EGFR mutations.

Close modal

Cox regression analyses of clinical variables for RFS and OS are shown in Table 2. Among the variables, sex, aromatase expression, and pathologic stage were identified as potential predictors of RFS. A multivariate analysis that included the above variables also showed aromatase expression to be a significant prognostic factor, with a relative risk of 2.37 [95% confidence interval (CI), 1.05–5.31; P = 0.039] for RFS. Multivariate analysis for OS was not performed because of the small number of events (deaths).

Table 2.

Cox proportion hazards model for RFS and OS (n = 110)

UnivariateMultivariate
VariableHR (95% CI)PHR (95% CI)P
RFS 
 Age (≥70 vs. <70) 1.49 (0.71–3.17) 0.296   
 Sex (male vs. female) 2.30 (1.09–5.06) 0.029 2.49 (1.13–5.74) 0.024 
 Smoking (current or former vs. never) 1.05 (0.49–2.21) 0.907   
EGFR mutation (positive vs. negative) 0.72 (0.32–1.51) 0.382   
 Aromatase (high vs. low) 3.01 (1.35–6.64) 0.008 2.37 (1.05–5.31) 0.039 
 Pathologic stage (≥ II vs. I) 5.33 (2.53–11.5) <0.001 3.36 (1.50–7.70) 0.003 
OS 
 Age (≥70 vs. <70) 2.85 (1.06–8.97) 0.041   
 Sex (male vs. female) 2.74 (1.04–7.97) 0.038   
 Smoking (current or former vs. never) 0.76 (0.28–1.98) 0.573   
EGFR mutation (positive vs. negative) 0.25 (0.06–0.78) 0.015   
 Aromatase (high vs. low) 4.20 (1.49–12.1) 0.007   
 Pathologic stage (≥II vs. I) 3.54 (1.35–9.46) 0.011   
UnivariateMultivariate
VariableHR (95% CI)PHR (95% CI)P
RFS 
 Age (≥70 vs. <70) 1.49 (0.71–3.17) 0.296   
 Sex (male vs. female) 2.30 (1.09–5.06) 0.029 2.49 (1.13–5.74) 0.024 
 Smoking (current or former vs. never) 1.05 (0.49–2.21) 0.907   
EGFR mutation (positive vs. negative) 0.72 (0.32–1.51) 0.382   
 Aromatase (high vs. low) 3.01 (1.35–6.64) 0.008 2.37 (1.05–5.31) 0.039 
 Pathologic stage (≥ II vs. I) 5.33 (2.53–11.5) <0.001 3.36 (1.50–7.70) 0.003 
OS 
 Age (≥70 vs. <70) 2.85 (1.06–8.97) 0.041   
 Sex (male vs. female) 2.74 (1.04–7.97) 0.038   
 Smoking (current or former vs. never) 0.76 (0.28–1.98) 0.573   
EGFR mutation (positive vs. negative) 0.25 (0.06–0.78) 0.015   
 Aromatase (high vs. low) 4.20 (1.49–12.1) 0.007   
 Pathologic stage (≥II vs. I) 3.54 (1.35–9.46) 0.011   

NOTE: Multivariate analysis for OS was not performed because of the small number of events (deaths).

Prognostic significance of aromatase expression in lung adenocarcinomas with EGFR mutations

Next, we compared survival between subgroups divided by clinicopathologic factors that are related to smoking-unrelated lung cancer such as sex, smoking history, and EGFR mutation status. High aromatase expression was associated with a poor prognosis in females (P = 0.008 for RFS and P < 0.001 for OS; Fig. 2C and D), in never-smokers (P = 0.009 for RFS and P < 0.001 for OS; Fig. 2E and F), and in patients with EGFR mutations (P = 0.005 for RFS and P = 0.003 for OS; Fig. 2G and H), but not in males (P = 0.14 for RFS and P = 0.65 for OS; Supplementary Fig. S1A and S1B), not in current or former smokers (P = 0.16 for RFS and P = 0.58 for OS; Supplementary Fig. S1C and S1D) and not in patients without EGFR mutations (P = 0.19 for RFS and P = 0.07 for OS; Supplementary Fig. S1E and S1F). Eleven patients with EGFR mutations had recurrent disease, and among them 8 patients received EGFR-TKI gefitinib after recurrence. There was no patient who received EGFR-TKI before recurrence.

Cox regression analyses for potential predictors of survival in patients with EGFR mutations are shown in Table 3. Among the parameters, aromatase expression and pathologic stage were identified as potential predictors of RFS. Multivariate analysis was not performed because of the small number of recurrences.

Table 3.

Cox proportion hazards model for RFS in patients with EGFR mutations (n = 54)

Univariate
VariableHR (95% CI)P
Age: ≥70 (n = 21) vs. <70 (n = 33) 0.85 (0.22–2.82) 0.795 
Sex: male (n = 12) vs. female (n = 42) 3.48 (0.99–11.6) 0.050 
Smoking: current/former (n = 15) vs. never (n = 39) 1.60 (0.42–5.30) 0.466 
Aromatase: high (n = 15) vs. low (n = 39) 4.97 (1.50–19.0) 0.009 
Pathologic stage: ≥ II (n = 13) vs. I (n = 41) 6.55 (1.98–25.0) 0.002 
Univariate
VariableHR (95% CI)P
Age: ≥70 (n = 21) vs. <70 (n = 33) 0.85 (0.22–2.82) 0.795 
Sex: male (n = 12) vs. female (n = 42) 3.48 (0.99–11.6) 0.050 
Smoking: current/former (n = 15) vs. never (n = 39) 1.60 (0.42–5.30) 0.466 
Aromatase: high (n = 15) vs. low (n = 39) 4.97 (1.50–19.0) 0.009 
Pathologic stage: ≥ II (n = 13) vs. I (n = 41) 6.55 (1.98–25.0) 0.002 

NOTE: Multivariate analysis was not performed because of the small number of recurrences.

Growth inhibition of lung adenocarcinoma cell line by aromatase inhibitor

We examined aromatase mRNA expression in 21 human lung adenocarcinoma cell lines (Fig. 3A). No correlation was demonstrated between aromatase expression level and driver mutation type. Because patients with high aromatase expression had worse prognoses than those with low aromatase expression among those with EGFR mutations (Fig. 2G and H), we next investigated whether aromatase had therapeutic potential in lung adenocarcinomas with EGFR mutations. We chose 11-18 as high-aromatase mRNA-expressing cell line and HCC4006 as low-aromatase mRNA-expressing cell line, both of which have EGFR mutations. To test the growth inhibitory effects of the aromatase inhibitor exemestane, we conducted an MTT assay. HCC4006 was not sensitive to exemestane, either alone or combined with erlotinib (Fig. 3B). By contrast, 11-18 was sensitive to exemestane alone (Fig. 3C), and its cell growth was significantly inhibited by the combination of exemestane with erlotinib. We further tested the growth inhibitory effects of exemestane in high-aromatase mRNA-expressing cell lines without EGFR mutations: H358 (Supplementary Fig. S2A), H2228 (Supplementary Fig. S2B), and ACC-LC-319 (Supplementary Fig. S2C). The antitumor effect of exemestane in H358, H2228, and ACC-LC-319 was much weaker than in 11-18.

Figure 3.

Effect of exemestane alone and in combination with erlotinib on EGFR mutant lung adenocarcinoma cell line proliferation. A, mRNA expression levels of aromatase in 21 human lung adenocarcinoma cell lines. Quantitative real-time RT-PCR was performed with validated TaqMan probes; assays were done in triplicate. The expression value for each cell line was calculated relative to that of human reference. MCF-7: breast carcinoma cell line. B, HCC4006 cells were not sensitive to exemestane alone or in combination with erlotinib. C, 11-18 cells were sensitive to exemestane alone and in combination with erlotinib. HCC4006 and 11-18 cells were incubated for 24 hours and for an additional 72 hours with the indicated concentrations of exemestane or erlotinib; cell growth was then measured. Independent experiments were repeated 3 times. *, P < 0.05 vs. erlotinib alone; †, P < 0.05 vs. control (both erlotinib and exemestane were free) by Dunnett test. All data represent the mean ± SD from 3 independent experiments.

Figure 3.

Effect of exemestane alone and in combination with erlotinib on EGFR mutant lung adenocarcinoma cell line proliferation. A, mRNA expression levels of aromatase in 21 human lung adenocarcinoma cell lines. Quantitative real-time RT-PCR was performed with validated TaqMan probes; assays were done in triplicate. The expression value for each cell line was calculated relative to that of human reference. MCF-7: breast carcinoma cell line. B, HCC4006 cells were not sensitive to exemestane alone or in combination with erlotinib. C, 11-18 cells were sensitive to exemestane alone and in combination with erlotinib. HCC4006 and 11-18 cells were incubated for 24 hours and for an additional 72 hours with the indicated concentrations of exemestane or erlotinib; cell growth was then measured. Independent experiments were repeated 3 times. *, P < 0.05 vs. erlotinib alone; †, P < 0.05 vs. control (both erlotinib and exemestane were free) by Dunnett test. All data represent the mean ± SD from 3 independent experiments.

Close modal

Although increasing evidence indicates that female hormones affect development of lung cancer (34–36), to our knowledge, this study is the first report to elucidate the prognostic significance of aromatase expression in patients with lung adenocarcinomas with EGFR mutations. We found that aromatase mRNA expression level was not correlated with clinicopathologic factors, including EGFR mutation status. However, high aromatase expression was associated with poor prognosis in terms of both RFS and OS. Moreover, the prognostic significance of aromatase expression was limited to females, never-smokers, and patients with EGFR mutations, whereas such significance was not observed in their counterparts.

Aromatase is an enzyme that catalyzes the conversion from androgens to estrogens. In NSCLC cells, estrogen is reported to be mainly produced by intrinsic aromatase (23), and stimulates the ER signaling pathway, resulting in tumor development and progression (9–11, 13, 24). Here, we found that aromatase mRNA expression levels in carcinoma tissues were significantly higher than in corresponding nonneoplastic lung tissues. Niikawa and colleagues reported that the estradiol concentration in NSCLC was significantly higher than that in the nonneoplastic lung tissues, and intratumoral estradiol concentration in NSCLC was positively associated with aromatase mRNA expression (23). In another study, levels of aromatase activity tested by radioassay were significantly greater in tumors compared with those in nearby normal tissue (12). These studies indicate that intrinsic aromatase expression levels are closely associated with the estrogen levels in the lung cancer cells. Thus, increased aromatase level may have profound influence in carcinoma tissues through estrogen function.

Most estrogenic actions are mediated by ER, which exists in 2 forms, ERα and ERβ (37). Although immunohistochemical expressions of ERα and/or ERβ has been associated with clinical outcome in some studies (14–16, 26, 38), the findings for expression frequency and subcellular localization (nuclear or cytoplasm) of ERs are inconsistent (14–16, 27, 36, 38, 39). These differences could be because of variation in such factors as (a) antibodies and dilutions, (b) scoring systems for staining, and (c) patient cohort characteristics (16). This discrepancy may obscure the significance of hormone receptor expression in patients' clinicopathologic characteristics or prognoses. For this reason, we found it difficult to clarify the effect of estrogen on the development of lung cancer using immunohistochemical analysis.

In this study, no significant correlation was identified between tumor aromatase expression and clinicopathologic factors, including EGFR mutations, when analyzed as either dichotomized or continuous variables. These results are consistent with previously reports (27, 39, 40). However, a correlation between EGFR mutation and ER expression, both ERα (14) and nuclear ERβ (15), in lung adenocarcinoma, was reported in previous studies, suggesting that some interaction between ER and the EGFR signaling pathway may exist.

High aromatase expression was associated with a poor prognosis in patients who underwent curative resections for lung adenocarcinoma. We also demonstrated that the prognostic significance of aromatase expression was limited to females, never-smokers, and patients with EGFR mutations, whereas such significance was not observed in their counterparts. We conducted Cox proportional hazards analysis in patients with EGFR mutations by RFS, not by OS, because 8 of 11 patients (73%) with EGFR mutations had received EGFR-TKI treatment for recurrence. EGFR-TKI prolonged the survival of such patients; therefore, the follow-up period may not have been enough to evaluate OS in this study. Aromatase expression level and EGFR mutation status did not directly correlate, but aromatase expression only held prognostic significance for lung adenocarcinomas with EGFR mutations, which suggests that estrogenic signaling augments growth that depends on the EGFR pathway. Mah and colleagues reported that lower aromatase levels predicted greater chances of survival in women 65 years and older with NSCLC, particularly among women who had no smoking history (13). Although they did not investigate EGFR mutation status, their results are consistent with ours. Nose and colleagues reported that strong nuclear expression of ERβ correlated with better disease-free survival in patients with EGFR mutations, but found no such prognostic significance in patients without EGFR mutations (15). They also suggested that strong ERβ expression was a surrogate marker for good response to EGFR-TKI (26). These results, along with our own, indicate that female hormone-related factors, such as aromatase and ERβ, affect outcomes only in lung adenocarcinomas with EGFR mutation, which suggests that hormonal and EGFR pathways may contribute in concert to progression of lung adenocarcinoma.

To investigate the influence of differences in aromatase expression between the tumor and normal tissues on patient survival, we classified patients into the following 2 groups: T > N, in whom aromatase expression in adenocarcinoma tissue was higher than in nonneoplastic lung tissue (n = 51); and T < N, in whom it was lower (n = 43). We performed survival analyses between the 2 groups, but saw no significant survival difference in RFS (P = 0.22) or OS (P = 0.27). We further compared RFS and OS between the 2 groups in subsets divided by sex, smoking history, and EGFR mutation status, but saw no significant difference in these analyses (data not shown).

Recently, interactions between the ER and EGFR pathways have been investigated in vitro. A nonnuclear ER pool has been proposed that works via rapid signaling through various kinase cascades, including EGFR pathway and its downstream effectors in the lungs, such as MAPK (10, 11). However, the ER and EGFR pathways seem to act as alternate signaling pathways, with one upregulating when the other is inactivated (10, 41). This bi-directional crosstalk between ER and EGFR signaling suggests that simultaneous or combined therapy that targets both pathways could exert higher antitumor effect in patients with NSCLC.

Both in vitro and in vivo reports have demonstrated that estrogen downmodulator, alone or combined with EGFR-TKI, resulted in enhanced antitumor activity in NSCLCs (9–12, 23, 25, 41–44). Exemestane, an irreversible steroidal inactivator, either alone (41) or in combination with cisplatin (43) showed significant antitumor effects in 2 separate studies. Both letrozole and anastrozole, reversible steroidal inactivators, demonstrated similar antitumor activity in NSCLCs (12, 23, 44). However, lung cancer cell lines used in almost all of these studies were without EGFR mutations. We showed that the 11-18 cell line, which has an EGFR mutation accompanied with a high aromatase mRNA expression, was sensitive to exemestane alone and cell growth was significantly inhibited by the combination of exemestane and erlotinib. We also demonstrated that exemestane's antitumor effects in H358, H2228, and ACC-LC-319, which have high aromatase expressions without EGFR mutations, were much weaker than in 11-18. These results suggest that sensitivity to the aromatase inhibitor may depend on crosstalk between ER and the EGFR pathway; reducing estrogenic signaling by inhibiting aromatase might inhibit cell growth that depends on the EGFR pathway. Therefore, our result suggests that selecting patients with high aromatase expression accompanied by EGFR mutation might improve clinical responses to the combination of EGFR-TKI and aromatase inhibitor. However, we tested the growth inhibitory effects of only one cell line with high aromatase expression and EGFR mutation, thus this study remains limited. Further studies using in vivo and clinical models are needed to elucidate the therapeutic potential of aromatase inhibitor in lung adenocarcinomas with EGFR mutations. Traynor and colleagues reported a pilot study of gefitinib and fulvestrant in the treatment of 22 postmenopausal women diagnosed as NSCLC (45). No significant results were obtained in this small study, but combination therapy was well tolerated. Three of 12 patients tested for EGFR mutation status detected EGFR mutation. A trial of EGFR-TKI and estrogen downmodulator in patients with NSCLC with EGFR mutations may be therefore needed in the future.

In conclusion, high aromatase expression is correlated with poor outcome in patients with lung adenocarcinoma, including those with EGFR mutations. Aromatase may be a useful therapeutic target in lung adenocarcinomas with high aromatase expression and EGFR mutations. Although our results provide potential insights into the influence of aromatase expression in lung cancer, further studies are required to better understand the mechanisms of aromatase expression and interaction with EGFR signaling, and to determine the clinical applicability of aromatase inhibitors.

Y. Maehara reports receiving other research grants from Chugai and Pfizer. No potential conflicts of interest were disclosed by the other authors.

Conception and design: M. Kohno, T. Okamoto, K. Suda

Development of methodology: M. Kohno, K. Suda, T. Yano

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): M. Kohno, T. Yano

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): M. Kohno, K. Suda, M. Shimokawa

Writing, review, and/or revision of the manuscript: M. Kohno, T. Okamoto, M. Shimokawa, M. Takenoyama, T. Yano

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): K. Suda, H. Kitahara, S. Shimamatsu, H. Konishi, T. Yoshida

Study supervision: M. Takenoyama, T. Yano, Y. Maehara

The authors thank Dr. A.F. Gazdar, Dr. T. Hida, Dr. K. Tomizawa, Dr. T. Mitsudomi, and Dr. M. Takeshita for kindly providing cell lines.

This study was supported in part by a Grant-in-Aid for Scientific Research (C) from the Japan Society for the Promotion of Science (21591816).

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.
Jemal
A
,
Bray
F
,
Center
MM
,
Ferlay
J
,
Ward
E
,
Forman
D
. 
Global cancer statistics.
CA Cancer J Clin
2011
;
61
:
69
90
.
2.
Sun
S
,
Schiller
JH
,
Gazdar
AF
. 
Lung cancer in never smokers—a different disease.
Nat Rev Cancer
2007
;
7
:
778
90
.
3.
Yano
T
,
Miura
N
,
Takenaka
T
,
Haro
A
,
Okazaki
H
,
Ohba
T
, et al
Never-smoking nonsmall cell lung cancer as a separate entity: clinicopathologic features and survival.
Cancer
2008
;
113
:
1012
8
.
4.
Yano
T
,
Haro
A
,
Shikada
Y
,
Maruyama
R
,
Maehara
Y
. 
Non-small cell lung cancer in never smokers as a representative ‘non-smoking-associated lung cancer': epidemiology and clinical features.
Int J Clin Oncol
2011
;
16
:
287
93
.
5.
Shigematsu
H
,
Lin
L
,
Takahashi
T
,
Nomura
M
,
Suzuki
M
,
Wistuba
II
, 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
.
6.
Kosaka
T
,
Yatabe
Y
,
Endoh
H
,
Kuwano
H
,
Takahashi
T
,
Mitsudomi
T
. 
Mutations of the epidermal growth factor receptor gene in lung cancer: biological and clinical implications.
Cancer Res
2004
;
64
:
8919
23
.
7.
Shaw
AT
,
Yeap
BY
,
Mino-Kenudson
M
,
Digumarthy
SR
,
Costa
DB
,
Heist
RS
. 
Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4-ALK.
J Clin Oncol
2009
;
27
:
4247
53
.
8.
Patrone
C
,
Cassel
TN
,
Pettersson
K
,
Piao
YS
,
Cheng
G
,
Ciana
P
, et al
Regulation of postnatal lung development and homeostasis by estrogen receptor β.
Mol Cell Biol
2003
;
23
:
8542
52
.
9.
Stabile
LP
,
Davis
AL
,
Gubish
CT
,
Hopkins
TM
,
Luketich
JD
,
Christie
N
, et al
Human non-small cell lung tumors and cells derived from normal lung express both estrogen receptor α and β and show biological responses to estrogen.
Cancer Res
2002
;
62
:
2141
50
.
10.
Stabile
LP
,
Lyker
JS
,
Gubish
CT
,
Zhang
W
,
Grandis
JR
,
Siegfried
JM
. 
Combined targeting of the estrogen receptor and the epidermal growth factor receptor in non-small cell lung cancer shows enhanced antiproliferative effects.
Cancer Res
2005
;
65
:
1459
70
.
11.
Pietras
RJ
,
Marquez
DC
,
Chen
HW
,
Tsai
E
,
Weinberg
O
,
Fishbein
M
. 
Estrogen and growth factor receptor interactions in human breast and non-small cell lung cancer cells.
Steroids
2005
;
70
:
372
81
.
12.
Weinberg
OK
,
Marquez-Garban
DC
,
Fishbein
MC
,
Goodglick
L
,
Garban
HJ
,
Dubinett
SM
, et al
Aromatase inhibitors in human lung cancer therapy.
Cancer Res
2005
;
65
:
11287
91
.
13.
Mah
V
,
Seligson
DB
,
Li
A
,
Márquez
DC
,
Wistuba
II
,
Elshimali
Y
, et al
Aromatase expression predicts survival in women with early-stage non-small cell lung cancer.
Cancer Res
2007
;
67
:
10484
90
.
14.
Raso
MG
,
Behrens
C
,
Herynk
MH
,
Liu
S
,
Prudkin
L
,
Ozburn
NC
, et al
Immunohistochemical expression of estrogen and progesterone receptors identifies a subset of NSCLCs and correlates with EGFR mutation.
Clin Cancer Res
2009
;
15
:
5359
68
.
15.
Nose
N
,
Sugio
K
,
Oyama
T
,
Nozoe
T
,
Uramoto
H
,
Iwata
T
, et al
Association between estrogen receptor-β expression and epidermal growth factor receptor mutation in the postoperative prognosis of adenocarcinoma of the lung.
J Clin Oncol
2009
;
27
:
411
7
.
16.
Stabile
LP
,
Dacic
S
,
Land
SR
,
Lenzner
DE
,
Dhir
R
,
Acquafondata
M
, et al
Combined analysis of estrogen receptor β-1 and progesterone receptor expression identifies lung cancer patients with poor outcome.
Clin Cancer Res
2011
;
17
:
154
64
.
17.
Liu
Y
,
Inoue
M
,
Sobue
T
,
Tsugane
S
. 
Reproductive factors, hormone use and the risk of lung cancer among middle-aged never-smoking Japanese women: a large-scale population-based cohort study.
Int J Cancer
2005
;
117
:
662
6
.
18.
Chlebowski
RT
,
Schwartz
AG
,
Wakelee
H
,
Anderson
GL
,
Stefanick
ML
,
Manson
JE
, et al
Oestrogen plus progestin and lung cancer in postmenopausal women (Women's Health Initiative trial): a post-hoc analysis of a randomised controlled trial.
Lancet
2009
;
374
:
1243
51
.
19.
Slatore
CG
,
Chien
JW
,
Au
DH
,
Satia
JA
,
White
E
. 
Lung cancer and hormone replacement therapy: association in the vitamins and lifestyle study.
J Clin Oncol
2010
;
28
:
1540
6
.
20.
Coombes
RC
,
Hall
E
,
Gibson
LJ
,
Paridaens
R
,
Jassem
J
,
Delozier
T
, et al
A randomized trial of exemestane after two to three years of tamoxifen therapy in postmenopausal women with primary breast cancer.
N Engl J Med
2004
;
350
:
1081
92
.
21.
Simpson
ER
,
Mahendroo
MS
,
Means
GD
,
Kilgore
MW
,
Hinshelwood
MM
,
Graham-Lorence
S
, et al
Aromatase cytochrome P450, the enzyme responsible for estrogen biosynthesis.
Endocr Rev
1994
;
15
:
342
55
.
22.
Smith
IE
,
Dowsett
M
. 
Aromatase inhibitors in breast cancer.
N Engl J Med
2003
;
348
:
2431
42
.
23.
Niikawa
H
,
Suzuki
T
,
Miki
Y
,
Suzuki
S
,
Nagasaki
S
,
Akahira
J
, et al
Intratumoral estrogens and estrogen receptors in human non-small cell lung carcinoma.
Clin Cancer Res
2008
;
14
:
4417
26
.
24.
Hershberger
PA
,
Stabile
LP
,
Kanterewicz
B
,
Rothstein
ME
,
Gubish
CT
,
Land
S
, et al
Estrogen receptor β (ERβ) subtype-specific ligands increase transcription, p44/p42 mitogen activated protein kinase (MAPK) activation and growth in human non-small cell lung cancer cells.
J Steroid Biochem Mol Biol
2009
;
116
:
102
9
.
25.
Garon
EB
,
Pietras
RJ
,
Finn
RS
,
Kamranpour
N
,
Pitts
S
,
Márquez-Garbán
DC
, et al
Antiestrogen fulvestrant enhances the antiproliferative effects of epidermal growth factor receptor inhibitors in human non-small-cell lung cancer.
J Thorac Oncol
2013
;
8
:
270
8
.
26.
Nose
N
,
Uramoto
H
,
Iwata
T
,
Hanagiri
T
,
Yasumoto
K
. 
Expression of estrogen receptor β predicts a clinical response and longer progression-free survival after treatment with EGFR-TKI for adenocarcinoma of the lung.
Lung Cancer
2011
;
71
:
350
5
.
27.
Sun
HB
,
Zheng
Y
,
Ou
W
,
Fang
Q
,
Li
P
,
Ye
X
, et al
Association between hormone receptor expression and epidermal growth factor receptor mutation in patients operated on for non-small cell lung cancer.
Ann Thorac Surg
2011
;
91
:
1562
7
.
28.
Travis
WD
,
Brambilla
E
,
Muller-Hermelink
HK
,
Harris
CC
. 
Tumours of the lung, pleura, thymus and heart. Pathology and genetics.
Lyon
:
IARC Press
; 
2004
.
29.
Goldstraw
P
,
Crowley
J
,
Chansky
K
,
Giroux
DJ
,
Groome
PA
,
Rami-Porta
R
, et al
International Association for the Study of Lung Cancer International Staging Committee; Participating Institutions. The IASLC Lung Cancer Staging Project: proposals for the revision of the TNM stage groupings in the forthcoming (seventh) edition of the TNM Classification of Malignant Tumours.
J Thorac Oncol
2007
;
2
:
706
14
.
30.
Nagai
Y
,
Miyazawa
H
,
Huqun
,
Tanaka
T
,
Udagawa
K
,
Kato
M
, 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
.
31.
Suda
K
,
Tomizawa
K
,
Fujii
M
,
Murakami
H
,
Osada
H
,
Maehara
Y
, et al
Epithelial to mesenchymal transition in an epidermal growth factor receptor-mutant lung cancer cell line with acquired resistance to erlotinib.
J Thorac Oncol
2011
;
6
:
1152
61
.
32.
Tomizawa
K
,
Suda
K
,
Onozato
R
,
Kuwano
H
,
Yatabe
Y
,
Mitsudomi
T
. 
Analysis of ERBB4 mutations and expression in Japanese patients with lung cancer.
J Thorac Oncol
2010
;
5
:
1859
61
.
33.
Suda
K
,
Murakami
I
,
Katayama
T
,
Tomizawa
K
,
Osada
H
,
Sekido
Y
, et al
Reciprocal and complementary role of MET amplification and EGFR T790M mutation in acquired resistance to kinase inhibitors in lung cancer.
Clin Cancer Res
2010
;
16
:
5489
98
.
34.
Siegfried
JM
,
Hershberger
PA
,
Stabile
LP
. 
Estrogen receptor signaling in lung cancer.
Semin Oncol
2009
;
36
:
524
31
.
35.
Verma
MK
,
Miki
Y
,
Sasano
H
. 
Aromatase in human lung carcinoma.
Steroids
2011
;
76
:
759
64
.
36.
Miki
Y
,
Abe
K
,
Suzuki
S
,
Suzuki
T
,
Sasano
H
. 
Suppression of estrogen actions in human lung cancer.
Mol Cell Endocrinol
2011
;
340
:
168
74
.
37.
Cheng
G
,
Weihua
Z
,
Warner
M
,
Gustafsson
JA
. 
Estrogen receptors ER α and ER β in proliferation in the rodent mammary gland.
Proc Natl Acad Sci U S A
2004
;
101
:
3739
46
.
38.
Ishibashi
H
,
Suzuki
T
,
Suzuki
S
,
Niikawa
H
,
Lu
L
,
Miki
Y
, et al
Progesterone receptor in non-small cell lung cancer—a potent prognostic factor and possible target for endocrine therapy.
Cancer Res
2005
;
65
:
6450
8
.
39.
Abe
K
,
Miki
Y
,
Ono
K
,
Mori
M
,
Kakinuma
H
,
Kou
Y
, et al
Highly concordant coexpression of aromatase and estrogen receptor β in non-small cell lung cancer.
Hum Pathol
2010
;
41
:
190
8
.
40.
Oyama
T
,
Kagawa
N
,
Sugio
K
,
Uramoto
H
,
Hatano
O
,
Harada
N
, et al
Expression of aromatase CYP19 and its relationship with parameters in NSCLC.
Front Biosci
2009
;
14
:
2285
92
.
41.
Koutras
A
,
Giannopoulou
E
,
Kritikou
I
,
Antonacopoulou
A
,
Evans
TR
,
Papavas-siliou
AG
, et al
Antiproliferative effect of exemestane in lung cancer cells.
Mol Cancer
2009
;
24
:
109
.
42.
Marquez-Garban
DC
,
Chen
HW
,
Fishbein
MC
,
Goodglick
L
,
Pietras
RJ
. 
Estrogen receptor signaling pathways in human non-small cell lung cancer.
Steroids
2007
;
72
:
135
43
.
43.
Márquez-Garbán
DC
,
Chen
HW
,
Goodglick
L
,
Fishbein
MC
,
Pietras
RJ
. 
Targeting aromatase and estrogen signaling in human non-small cell lung cancer.
Ann NY Acad Sci
2009
;
1155
:
194
205
.
44.
Miki
Y
,
Suzuki
T
,
Abe
K
,
Suzuki
S
,
Niikawa
H
,
Iida
S
, et al
Intratumoral localization of aromatase and interaction between stromal and parenchymal cells in the non-small cell lung carcinoma microenvironment.
Cancer Res
2010
;
70
:
6659
69
.
45.
Traynor
AM
,
Schiller
JH
,
Stabile
LP
,
Kolesar
JM
,
Eickhoff
JC
,
Dacic
S
, et al
Pilot study of gefitinib and fulvestrant in the treatment of post-menopausal women with advanced non-small cell lung cancer.
Lung Cancer
2009
;
64
:
51
9
.