Next Article in Journal
Recent Advance in Biosensors for microRNAs Detection in Cancer
Next Article in Special Issue
Tomato Lycopene and Lung Cancer Prevention: From Experimental to Human Studies
Previous Article in Journal
Biomarkers in Advanced Colorectal Cancer: Challenges in Translating Clinical Research into Practice
Previous Article in Special Issue
The Role of Epigenetics in Resistance to Cisplatin Chemotherapy in Lung Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Epidemiology of Female Lung Cancer

1
Integrated Research Center for Genome Polymorphism, The Catholic University of Korea School of Medicine, 505 Banpo-dong, Seocho-gu, Seoul 137-701, Korea
2
Department of Medical Humanities and Social Sciences, The Catholic University of Korea School of Medicine, 505 Banpo-dong, Seocho-gu, Seoul 137-701, Korea
3
Department of Microbiology, The Catholic University of Korea School of Medicine, 505 Banpo-dong, Seocho-gu, Seoul 137-701, Korea
*
Author to whom correspondence should be addressed.
Cancers 2011, 3(2), 1861-1876; https://doi.org/10.3390/cancers3021861
Submission received: 4 January 2011 / Revised: 18 March 2011 / Accepted: 31 March 2011 / Published: 1 April 2011
(This article belongs to the Special Issue Lung Cancer)

Abstract

: Lung cancer is still a leading cause of cancer mortality in the world. The incidence of lung cancer in developed countries started to decrease mainly due to global anti-smoking campaigns. However, the incidence of lung cancer in women has been increasing in recent decades for various reasons. Furthermore, since the screening of lung cancer is not as yet very effective, clinically applicable molecular markers for early diagnosis are much required. Lung cancer in women appears to have differences compared with that in men, in terms of histologic types and susceptibility to environmental risk factors. This suggests that female lung cancer can be derived by carcinogenic mechanisms different from those involved in male lung cancer. Among female lung cancer patients, many are non-smokers, which could be studied to identify alternative carcinogenic mechanisms independent from smoking-related ones. In this paper, we reviewed molecular susceptibility markers and genetic changes in lung cancer tissues observed in female lung cancer patients, which have been validated by various studies and will be helpful to understand the tumorigenesis of lung cancer.

1. Introduction

Lung cancer has been the most common cancer in the world for several decades. In 2008, there were an estimated 1.61 million new cases, representing 12.7% of all new cancers and lung cancer was the most common cause of death from cancer with 1.38 million deaths (18.2% of the total) [1]. Lung cancer was still more common in men worldwide (1.1 million cases, 16.5% of the total). However, it became the fourth most frequent cancer of women (513,000 cases, 8.5% of all cancers) and the second most common cause of death from cancer (427,000 deaths, 12.8% of the total) [1]. In Western countries, the incidence and mortality for lung cancer has reached the peak in men, and seems to now be declining. In women, however, incidence and mortality are still approaching the plateau.

Clinically, lung cancer is classified as small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). NSCLC accounts for approximately 80% of all cases of lung cancer and is histopathologically subclassified as squamous cell carcinoma (SCC), adenocarcinoma (AC), and large cell carcinoma, for which prognosis and management are similar [2]. In 1984, based on a study involving interviews with 7,804 cases and 15,207 hospital-based controls, Lubin and Blot reported that among those who never smoked there were remarkable cell type differences by sex, with a greater proportion of AC compared to SCC in females (45% vs. 25%) than in males (35% vs. 33%) [3]. Since then, the incidence of AC has been reported to be increasing in both genders, which has been attributed to changes in the composition of cigarettes and the implementation of filters [4-6]. In 2009, Egleston et al. reported the epidemiology of lung cancer using SEER 9-registry data over the past 35 years [7]. AC has remained the most prevalent tumor type in women and its incidence rates have been increasing over time. In contrast, SCC has been predominant in men and its incidence has declined and converged with the rates in women. The rate of large cell carcinoma in women has been similar to that of men, with rates decreasing slightly over time, but SCLC has remained relatively constant in both genders [7]. Due to the confounding effect of smoking and related factors, it is difficult to see whether there is a genuine difference between sexes in histologic types of lung cancer.

There have been many studies that reported the possible higher susceptibility of women to lung cancer compared with men regardless of smoking status. Brownson et al. reported higher odds ratios (OR) of ever-smoking and level of smoking in women for all histologic types of lung cancer except AC compared with men based on 14,596 cases and 36,438 age-matched controls [8]. Risch et al. reported that a risk of lung cancer increased with smoking in both sexes, but the association was significantly stronger for females than for males in each of the major histologic types [9]. The OR for women with a history of 40 pack-years was 27.9 (95% confidence interval [CI] 14.9–52.0) and that for men was 9.6 (95% CI 5.6–16.3) compared with lifelong nonsmokers [9]. Harris et al. also reported that women are at higher risk than men for a given level of smoking (OR 1.7, 95% CI 1.2–2.2) [10]. Zang and Wydner reported that dose-dependent ORs over cumulative exposure to cigarette smoking were 1.2-fold to 1.7-fold higher in women than in men for the three major histologic types (squamous/epidermoid, large-cell, and AC type) and concluded that this gender difference cannot be explained by differences in baseline exposure, smoking history, or body size, but it is more likely due to the higher susceptibility to tobacco carcinogens in women [11].

However, there are studies that have reported conflicting results. In a large prospective population based study (30,874 subjects included, 422,606 person-years of observation, 867 new lung cancer cases), Prescott et al. found that incidence rates of lung cancer among female and male never-smokers were similar and, after being adjusted for pack-years, age, and study population, the rate ratio between female and male smokers was 0.8 (95% CI 0.3–2.1), which means the incidence rates were also similar in smokers regardless of sex [12]. Bain et al. analyzed prospective data from former and current smokers in two large cohorts—the Nurses' Health Study and the Health Professionals Follow-up Study—and calculated incidence rates and hazard ratios of lung cancer in women compared with men [13]. After adjusting for age, number of cigarettes smoked per day, age at start of smoking, and time since quitting, the hazard ratio in women ever smokers compared with men was 1.11 (95% CI 0.95–1.31), which suggests that women do not seem to have a greater susceptibility to lung cancer than men, given equal smoking exposure [13]. Studies reporting higher susceptibility of women to lung cancer are mostly case-control studies, which are well known to be vulnerable to various biases. In contrast, two studies reporting null results are based on large prospective cohorts. Furthermore, exposure measurement cannot be said to be objective and accurate, because most studies used questionnaires for exposure measurement. It is necessary to apply biomarkers for accurate exposure measurement to determine whether the susceptibility to lung cancer is really different between sexes.

Apart from incidence, gender disparities have been observed in cancer progression, survival, and therapeutic response [14]. A variety of mechanisms, biological, behavioral and environmental, should be behind these differences. Oncogenesis is a complex, multifactorial process which involves various factors; host factors such as xenobiotic metabolism, sex, age, and hormones; and environmental factors such as tobacco smoke, asbestos, diet, and air pollution, etc. In this review, we examined germ-line genetic polymorphisms and somatic alterations in cancer tissues as host factors, especially ones distributed differently between sexes, which may explain clinical differences in female and male lung cancer.

2. Sex-specific Characteristics

2.1. Metabolism-Related Genetic Polymorphisms

Cigarettes contain a mixture of carcinogens, including a small dose of polycyclic aromatic hydrocarbons (PAHs) and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). Carcinogens such as NNK and PAHs require metabolic activation to exert their carcinogenic effects; there are competing detoxification pathways, and the balance between metabolic activation and detoxification differs among individuals and will affect cancer risk. Many (pro)carcinogens are converted to biologically reactive metabolites by Phase I enzymes such as those encoded by the cytochrome P450 (CYP) super family (CYP1, CYP2, CYP3, and CYP4 isoforms included) [15]. The metabolic activation process leads to the formation of DNA adducts, which are carcinogen metabolites bound covalently to DNA [2,16]. If DNA adducts escape cellular repair mechanisms and persist, they may result in permanent mutations and initiate carcinogenesis. If a permanent mutation occurs in a critical region of an oncogene or tumor suppressor gene, it can lead to activation of the oncogene or deactivation of the tumor suppressor gene. Multiple events of this type lead to aberrant cells with loss of normal growth control and, ultimately, to lung cancer [17]. Following the Phase I reaction, Phase II enzymes such as glutathione S-transferases (GSTs) are responsible for detoxifying the activated forms of PAH epoxides. The GSTs also form a supergene family and the major isoforms are GSTM1, GSTM3, GSTT1, and GSTP1 [18]. Genetic polymorphisms have been found in both Phase I and Phase II enzymes and some of them show sex-specific differences in the effect. Many papers have been reporting the association between polymorphisms and lung cancer, but a few of them analyzed differences in the effects of polymorphisms on lung cancer risk between sexes.

Tang et al. reported based on a case-control study that GSTM1 null genotype (inherited deletion) was significantly associated with NSCLC (OR 2.04, 95% CI 1.13-3.68) [19]. The ORs for GSTM1 null genotype were significant in female smokers (OR 3.03, 95% CI 1.09–8.40) and not in males (OR 1.42, 95% CI 0.53–4.06), which suggests that the effect of the GSTM1 null genotype on NSCLC is greater in female smokers than in male smokers with an equal smoking history [19]. Dresler et al. genotyped GSTM1 and CYP1A1 (Ile462Val, rs1048943) in a case-control study and found that although the OR for the CYP1A1 polymorphism in females and males were not significantly different, the effect of the CYP1A1 polymorphism may be stronger in female than in male smokers (OR 4.98 in women, 95% CI 1.50-16.39 vs. 1.37 in men, 95% CI 0.44–4.31). Although lung cancer risk did not increase significantly for GSTM1 null for either sex in the multivariate analysis, the effect of the CYP1A1 polymorphism may be stronger in GSTM1 null females (OR 6.54 in women, 95% CI 1.1–40.0 vs. 2.36 in men, 95% CI 0.49–11.49), independent of age or smoking history. These data suggest that polymorphisms in CYP1A1 and GSTM1 contribute to the increased risk of females for lung cancer [20]. In 2006, one meta-analysis of 130 studies (over 23,000 cases and 30,000 controls) reported the weakly positive, but not significant association between the GSTM1 null genotype and the risk of lung cancer [21]. Assuming that this conclusion is true, it will also be far more difficult to identify the sex-specific effect of GSTM1 polymorphism.

Hung et al. performed a pooled analysis of 14 case-control studies on lung cancer in Caucasian non-smokers and reported the OR of lung cancer for the CYP1A1 Ile462Val polymorphism was 2.99 (95% CI 1.51–5.91) [22]. The gender-specific ORs were 3.52 (95%CI 1.35–9.17) for men and 2.57 (95% CI 0.58–11.5) for women, which is different from the ORs from Dresler et al. Furthermore, the OR for women did not achieve statistical significance. Le Marchand et al. carried out a pooled analysis of the data submitted to the Genetic Susceptibility to Environmental Carcinogens (GSEC) database [23]. They reported that the pooled ORs for CYP1A1 Ile462Val heterozygote and homozygote polymorphisms were 1.15 (95% CI 0.95–1.39) and 1.54 (95% CI 0.97–1.46), respectively, and that the CYP1A1 Ile462Val polymorphism may confer an increased risk of lung cancer in women (OR 2.70, 95% CI 1.79–4.08 vs. OR 1.08, 95% CI 0.88–1.32 in men) [23]. Timofeeva et al. conducted a case-control study (638 Caucasian patients under the age of 51 with primary lung cancer and 1,300 cancer-free control individuals) and examined the effect of 13 polymorphisms in the CYP1A1, CYP1B1, CYP2A13, CYP3A4 and CYP3A5 genes [24]. No significant association was found between any of the analyzed polymorphisms and lung cancer risk overall. However, among women, a significantly increased risk was observed for carriers of the minor allele of CYP1B1 SNP rs1056836 (OR 1.97, 95% CI 1.32–2.94) and the effect was shown to be modified by smoking. The results suggest that the CYP1B1 polymorphism may contribute to increased susceptibility to early-onset lung cancer in women [24]. Recently, one meta-analysis suggested the protective effect of CYP2E1 RsaI/PstI and DraI polymorphism for lung cancer, but there is no information on frequency of the polymorphism by sex [25].

Regarding the GSTP1 polymorphisms, a lot of studies have been reporting inconsistent results. Among the studies, four meta- or pooled analysis studies have reported no significant association between the GSTP1 polymorphisms GSTP1 exon 5 (Ile105Val) and the risk of lung cancer and have not provided the gender-specific information [21,26-28]. Some studies have suggested the existence of differences by ethnicity, age of diagnosis, smoking status, and gender in the effect of the GSTP1 polymorphisms on the risk of lung cancer, but evidence is far from enough to draw any conclusions [29-34].

Myeloperoxidase (MPO) is a phase I metabolic enzyme that converts the metabolites of benzo[a]pyrene from tobacco smoke into highly reactive epoxides. A polymorphism in the promoter region of MPO (463G>A) has been found to be inversely associated with lung cancer and differences in the association with age and sex have been suggested. Taioli et al. conducted a pooled analysis (3,688 cases and 3,874 controls) and found the OR for lung cancer was 0.88 (95% CI 0.80–0.97) for the A/G variant of MPO 463G>A polymorphism, and 0.71 (95% CI 0.57–0.88) for the A/A variant after adjusting for smoking, age, gender, and ethnicity [35]. The inverse association between lung cancer and MPO 463G>A polymorphism was observed equally in males and females in a different study. However, more recent studies have been reporting no association between polymorphism and lung cancer [36,37].

Wang et al. screened common variants in 5′ flanking and 3′ untranslated regions of the adenosine triphosphate-binding cassette B1 (ABCB1) and ABCC1 candidate transporter genes which are the genetic components in the metabolism and disposition of NNK in Chinese population, and examined the association with lung cancer [38]. Compared with the wild A/A genotype, ABCB1 rs3842 A/G or G/G (OR 1.36, 95% CI 1.06–1.76) and ABCC1 rs212090 A/T or T/T (OR 1.37, 95% CI 1.03–1.83) genotypes were associated with an increased risk of lung cancer. The association of ABCB1 rs3842 with the risk of lung cancer was stronger in women (OR 2.57, 95% CI, 1.36–4.85) than in men (OR 1.19, 95% CI 0.89–1.58), but ABCC1 did not show sex difference [38].

Reported polymorphisms associated with female lung cancer risk are listed in the Table 1.

2.2. DNA Repair-Related Genetic Polymorphisms

DNA repair capacity (DRC) is an important host factor that may influence lung carcinogenesis. Tobacco smoke contains many carcinogens and reactive oxygen species that produce DNA adducts, cross-links, DNA damage and DNA strand breaks requiring repair through multiple pathways, including the following: base excision repair, nucleotide excision repair, mismatch repair, single-strand break and double-strand break mechanisms. A positive and consistent association was observed between the reduced DRC and cancer occurrence (ORs in the range of 1.4–75.3) [39]. It has been reported that DRC is relatively lower in female lung cancer patients than in male patients [40] and a significant sex difference in antibodies to oxidative DNA damage from cigarette smoking has also been observed [32]. Given the same duration of exposure, women had a higher level of antibodies to oxidative DNA damage and also reached peak levels at a lower cumulative smoking exposure (30 years) compared with male smokers (40 years) [41].

O-alkylated bases, such as O6-methyguanine, are the major carcinogenic lesions in DNA induced by alkylating mutagens. O6-methylguanine, a methylated damage lesion in DNA, correlates with spontaneous G:C>A:T transition mutations and leads to activation of oncogene K-ras or dysfunction of the tumor suppressor gene TP53. This DNA adduct is removed by the repair protein, O(6)-methylguanine-DNA methyltransferase (MGMT) [42]. MGMT promoter methylation is a common event in primary human neoplasms including lung cancer [43-46]. The highest prevalence of MGMT promoter methylation was found in male nonsmokers followed by male smokers and female nonsmokers, which does not support the hypothesis that the lung cancer risk is higher in women [47]. Although the possible association between MGMT polymorphisms and lung cancer risk has been examined in several studies, most results have been inconclusive and not included information on sex difference [48-53]. Only one study reported the sex difference in the effect of MGMT polymorphisms. Wang et al. examined whether genetic variants of MGMT are associated with increased lung cancer risk in a case-control study consisting of 1,121 Caucasian lung cancer patients and 1,163 matched cancer-free controls [54]. They genotyped four potentially functional SNPs of MGMT: exon 3 codon 84C>T (L84F), exon 5 codon 143A>G (I143V), and two promoter SNPs 135G> T and 485C>A. They categorized the MGMT genotypes as either 0 variants (84CC-143AA) or 1–4 variants. Compared with 0 variants, those with 1–4 variants showed a significantly increased risk of lung cancer (OR 1.19, 95% CI 1.01–1.41) and this increased risk was more prominent in women (OR 1.31, 95% 1.03–1.66) than in men (OR 1.09, 95% CI 0.86–1.83) [54].

Hung et al. examined the associations between lung cancer risk and 18 polymorphisms in 12 DNA repair genes, including APEX1, OGG1, XRCC1, XRCC2, XRCC3, ERCC1, XPD, XPF, XPG, XPA, MGMT, and TP53 [55]. Two of the variants were found to be significantly associated with lung cancer risk; XRCC3 T241M (heterozygote OR, 0.89, 95% CI 0.79–0.99 and homozygote OR 0.84, 95% CI 0.71–1.00), and XPD K751Q (heterozygote OR 0.99, 95% CI 0.89–1.10 and homozygote OR 1.19, 95% CI 1.02–1.39), but there was no sex differences in the stratified estimates of XRCC3 T241M [55].

2.3. Cell Proliferation-Related Genetic Changes

There are many signaling pathways known to be involved with lung carcinogenesis. Multistep accumulation of genetic alterations occurs at the genomic level, leading to initiation, development and maintenance of lung cancer. We focused on genetic polymorphisms and somatic DNA alterations in the genes which have been frequently suggested to be associated with lung cancer.

2.3.1. KRAS

KRAS gene encodes an oncogenic protein when mutated or overexpressed, which is known to act as signaling switches that relay growth signals from the cell surface to the mitogen-activated protein (MAP) kinase cascade. Mutations in KRAS are specific to AC, and occur rarely in SCC or SCLC. Most KRAS mutations in lung AC are smoking-related G to T transversions and affect exon 12 or exon 13, suggesting that KRAS mutations are induced primarily by tobacco smoke carcinogenesis [56,57].

There have been studies reporting no association between the KRAS point mutation in lung cancer tissue and sex [58-60]. However, Nelson et al. reported a significant association between female sex and KRAS mutation in lung AC tissue after adjustment for carcinogen exposures (OR 3.3, 95% CI 1.3–7.9) and mutations were found only in smokers [61]. They suggested a possible role of estrogen exposure in either the initiation or the selection of KRAS mutant clones in AC [61]. Boldrini et al. also reported that among 411 lung AC patients KRAS mutations were more frequent in males (23.7% vs. 10.1%, P = 0.0007) [62]. More studies will be required to verify the association between KRAS mutations and sex.

2.3.2. TP53

Malfunction of the p53 pathway is a phenomenon observed in most human tumors. Somatic mutation of TP53 is one of the most common mechanisms by which the p53 pathway is damaged during tumorigenesis. Although several studies have reported on sex difference in TP53 mutations, the results are inconsistent in terms of frequency and mutation types.

Chiba et al. examined mutations changing the p53 coding sequence from 51 NSCLC. Mutations were found in 45% of tumor specimens [63]. In multivariate analysis the presence of TP53 mutations was associated with younger age and squamous histology, but not with tumor stage, nodal status or sex [54]. Kure et al. also investigated sex differences in TP53 mutations from the tumor tissue of NSCLC patients and levels of DNA adducts in non-tumor lung tissue from the patients [64]. Although TP53 mutations seemed to be more frequent in male patients (53% vs. 36%), G:C>T:A mutations were more frequent in females (40%) than in male patients (28%). However, these differences were not statistically significant [64].

One meta-analysis reported that somatic TP53 gene mutations were observed more often in males than in females (OR 1.59, 95% CI 1.16–2.19) [65]. However, this result could be confounded by the fact that males have relatively more SCCs, which have a higher proportion of TP53 mutations than do ACs, which occur more frequently in women. They could not determine whether TP53 gene mutations are associated with sex independently of histologic types. There was no information on the spectrum of TP53 mutation types, either [65]. Using the International Agency for Research on Cancer TP53 database, Toyooka et al. reported the significant differences in TP53 mutational spectra in lung cancer tissue between male and female never-smokers [66]. The G:C>T:A transversions were significantly frequent in female smokers (36%) than in female never-smokers (11%), but there was no such difference in the mutational spectra of male never-smokers (31%) and smokers (27%). A very similar pattern was found in the analysis limited to AC. In other words, cancers arising in women smokers seemed to obtain significantly more tobacco-related mutations, which may contribute to the higher susceptibility of women to tobacco carcinogens [66].

Marrogi et al. reported a similar frequency and type of somatic TP53 mutations between men and women based on a case series (102 women and 201 men). The percentage of TP53 G:C>T:A mutations in women (41%) was slightly higher than in men (38%), but this was not statistically significant [67]. A sex difference in TP53 somatic mutations seems to exist, but more studies will be required to confirm the association between TP53 mutations and sexes.

Besides the somatic mutations, numerous SNPs and other sequence variations have been reported at the TP53 locus. However, there is no report on sex difference in TP53 polymorphisms. Fang et al. reported a 58% higher lung cancer risk in TP53 germ-line mutation carriers with the MDM2 SNP309 GG+GT alleles compared with TT homozygotes [68]. MDM2 SNP309 was located in the promoter of the MDM2 gene and result in higher levels of MDM2 RNA and protein, which consequently induce the attenuation of the p53 pathway [69]. A significant effect of SNP309 G allele was observed in women (OR 1.60, 95% CI 1.08–2.36) but not in men (OR 1.47, 95% CI 0.82–2.60), which may be related to biological regulation of MDM2 by estrogen [68]. In germ-line TP53 mutation carriers, SNP309 was reported to accelerate tumor onset and to be associated with the development of multiple primary tumors [70,71]. Bond et al. also showed that the same polymorphism accelerated tumor formation in women, but not in men and depended upon estrogen signaling [72].

2.3.3. EGFR

Epidermal growth factor receptor (EGFR) plays an important role in the development and progression of a variety of malignant tumors and mutated EGFR status is a known predictor of response to tyrosine kinase inhibitors (TKIs). The EGFR mutations are distributed throughout the kinase domain, but a deletion in exon 19 and the point mutation L858R in exon 21 account for approximately 90%, which confer a greater response to gefitinib treatment, compared with other types of EGFR mutations. There have been many reports on the sex-specific incidence or prevalence of EGFR mutations and the results are inconsistent.

Marchetti et al. examined somatic EGFR mutations in 860 NSCLC patients and found EGFR mutations to be independently associated with female sex (OR 2.9, 95% CI 1.3-6.7) [73]. Another study on 35 Taiwanese AC patients also reported a very strong association between somatic EGFR mutations and female sex (OR 10.9, 95% CI 1.8-67.0) [74]. Shigematsu et al. reported that the frequency of somatic EGFR mutations was greater for females versus males (42% vs. 14%, P < 0.001), never smokers versus ever smokers (51% vs. 10%, P < 0.001), and for AC versus other histologic types (40% vs. 3%, P < 0.001) and that results of multivariate analyses confirmed these findings [75]. There are more studies which reported more frequent somatic EGFR mutations in women, but most results were based on simple comparison between sexes, which makes it hard to draw meaningful conclusions [62,76-81].

There have been studies which reported no sex difference in EGFR mutations. Toyooka et al. examined the EGFR mutational spectrum in exons 18 to 21 in tumor tissue from 1,467 NSCLC patients [82]. In never smokers, there was no difference between female and male cases. In ever smokers, exon 19 mutations were significantly less frequent in male compared with female cases (OR 0.34, 95% CI 0.16–0.70). In the analysis restricted to ACs, similar results were obtained; in ever smokers, exon 19 mutations were significantly less frequent in males (OR 0.32, 95% CI 0.15–0.67). This finding suggests both sex and smoking status could influence the somatic EGFR mutational spectrum [82]. Tanaka et al. analyzed the EGFR mutation analysis from 1,176 Japanese NSCLC patients and reported that the frequency of EGFR mutation was significantly higher in AC (OR 3.18, 95% CI 1.39–7.23) and in light-smokers (OR 3.84, 95% CI 1.92–7.65), but not associated with sex in multivariate analysis [83]. Although the frequency of EGFR mutations seemed to be higher in females (P < 0.0001 for a chi-square test), after adjusting effects of age, sex, histology, staging and smoking history through logistic regression, only the deletions in exon 19 were found to be more frequent in males (P = 0.0011 from logistic regression). It suggested that reportedly more frequent EGFR mutations in females may be a reflection of a higher frequency of AC in females [83]. To confirm the existence of the association between sex and the frequency of somatic EGFR mutations, it is necessary to perform more detailed studies which can consider potential confounding factors such as tumor differentiation. Together with somatic mutations, increased EGFR gene copy number in tumor emerged as another important predictor for TKI sensitivity [84]. However, in various clinical trials, the frequency of EGFR gene amplification was not significantly different between sexes [76,85-88].

There are reports on germ-line polymorphisms and other variants in EGRF. To explore the association between germ-line polymorphisms of the EGFR and the lung cancer susceptibility, Jou et al. genotyped 14 SNPs in EGFR and found that a SNP 8227G>A (rs763317) showed statistically significant difference between lung cancer patients and control subjects, and the AA genotype of 8227G>A polymorphism had a significantly increased risk of developing lung cancer compared with the G/G genotype (OR 2.40, 95% CI 1.34–4.33) [89]. Although there was no statistically significant differences between the genotype distribution of EGFR 8227G>A polymorphism and cancer histology within the male population, in female population, ORs for 8227G>A were significantly increased in AC subtype (OR for G/A genotype 1.23, 95% CI 0.87–1.75; OR for A/A genotype 3.52, 95% CI 1.32–9.37), but not that in SCC. Haplotype analyses revealed that haplotype comprising the rare allele of 8227G>A, and the common allele of the other 13 SNPs, was associated with a significantly increased risk of female AC (OR 2.81, 95% CI 1.02–7.77) [89]. This result suggests that polymorphisms and haplotypes of the EGFR affect the development of lung cancer differently according to sex or histologic types.

3. Conclusions

There are many studies which reported sex-specific genomic characteristics, both constitutional and somatic, in the risk of lung cancer, but some of the studies seem to be vulnerable to various biases and confounding factors, which makes the whole picture somewhat confusing. Recent meta-analysis results have reflected this point; quite a few of the previously reported associations between polymorphisms and the risk of lung cancer have been negated. Many studies used information on histologic types and smoking status to adjust their confounding effects, but it is far from complete. Especially smoking status was determined by various methods based on different criteria. To confirm the existence of sex difference in lung cancer, it would be necessary to perform more detailed studies which consider potential confounding factors such as tumor differentiation and stage. It is far from complete to only include histologic types and basic smoking status.

Table 1. Reported polymorphisms associated with female lung cancer risk.
Table 1. Reported polymorphisms associated with female lung cancer risk.
FunctionsGenesPolymorphismsRemarksRef.
MetabolismCYP1A1rs1048943,Higher risk in female non-smoker[20,22]

CYP1B1rs1056836Higher risk in female smoker[24]
rs1056827Higher risk in female smoker
rs2567206Higher risk in female smoker

CYP2A13rs8192784Higher risk in male non-smoker[24]
rs1709084Higher risk in female non-smoker

GSTM1Null typeHigher risk in female and smoker[19]

ABCB1rs3842Higher risk in female[38]

DNA repairMGMTrs1711646,
rs1625649,
rs12917,
rs2308321
Higher risk in female[54]

Cell proliferationEGFRrs763317Significantly associated with AC in female but not in male[89]

Acknowledgments

This work was supported by a grant from the Korea Health 21 R&D Project, Ministry of Health and Welfare, Republic of Korea (A010250).

References

  1. IARC GLOBOCAN 2008. http://globocan.iarc.fr/ (accessed on 1 December 2010).
  2. Kiyohara, C.; Ohno, Y. Sex differences in lung cancer susceptibility: A review. Gend Med. 2010, 7, 381–401. [Google Scholar]
  3. Lubin, J.H.; Blot, W.J. Assessment of lung cancer risk factors by histologic category. J. Natl. Cancer Inst. 1984, 73, 383–389. [Google Scholar]
  4. Toh, C.K. The changing epidemiology of lung cancer. Methods Mol. Biol. 2009, 472, 397–411. [Google Scholar]
  5. Stellman, S.D.; Muscat, J.E.; Thompson, S.; Hoffmann, D.; Wynder, E.L. Risk of squamous cell carcinoma and adenocarcinoma of the lung in relation to lifetime filter cigarette smoking. Cancer 1997, 80, 382–388. [Google Scholar]
  6. Thun, M.J.; Lally, C.A.; Flannery, J.T.; Calle, E.E.; Flanders, W.D.; Heath, C.W., Jr. Cigarette smoking and changes in the histopathology of lung cancer. J. Natl. Cancer Inst. 1997, 89, 1580–1586. [Google Scholar]
  7. Egleston, B.L.; Meireles, S.I.; Flieder, D.B.; Clapper, M.L. Population-based trends in lung cancer incidence in women. Semin. Oncol. 2009, 36, 506–515. [Google Scholar]
  8. Brownson, R.; Chang, J.; Davis, J. Gender and histologic type cariations in smoking-related risk of lung cancer. Epidemiology 1992, 3, 61–64. [Google Scholar]
  9. Risch, H.A.; Howe, G.R.; Jain, M.; Burch, J.D.; Holowaty, E.J.; Miller, A.B. Are female smokers at higher risk for lung cancer than male smokers? A case-control analysis by histologic type. Am. J. Epidemiol. 1993, 138, 281–293. [Google Scholar]
  10. Harris, R.E.; Zang, E.A.; Anderson, J.I.; Wynder, E.L. Race and sex differences in lung cancer risk associated with cigarette smoking. Int. J. Epidemiol 1993, 22, 592–599. [Google Scholar]
  11. Zang, E.A.; Wynder, E.L. Differences in lung cancer risk between men and women: Examination of the evidence. J. Natl. Cancer Inst. 1996, 88, 183–192. [Google Scholar]
  12. Prescott, E.; Osier, M.; Rein, H.O.; Borch-Johnsen, K.; Lange, P.; Schnohr, P.; Vestbo, J. Gender and smoking-related risk of lung cancer. The Copenhagen Center for Prospective Population Studies. Epidemiology 1998, 9, 79–83. [Google Scholar]
  13. Bain, C.; Feskanich, D.; Speizer, F.E.; Thun, M.; Hertzmark, E.; Rosner, B.A.; Colditz, G.A. Lung cancer rates in men and women with comparable histories of smoking. J. Natl. Cancer Inst. 2004, 96, 826–834. [Google Scholar]
  14. Paggi, M.G.; Vona, R.; Abbruzzese, C.; Malorni, W. Gender-related disparities in non-small cell lung cancer. Cancer Lett. 2010, 298, 1–8. [Google Scholar]
  15. Roos, P.H.; Bolt, H.M. Cytochrome P450 interactions in human cancers: new aspects considering CYP1B1. Expert Opin. Drug Metab. Toxicol. 2005, 1, 187–202. [Google Scholar]
  16. Gasperino, J. Gender is a risk factor for lung cancer. Med. Hypotheses 2011, 76, 328–331. [Google Scholar]
  17. Hecht, S.S. Tobacco smoke carcinogens and lung cancer. J. Natl. Cancer Inst. 1999, 91, 1194–1210. [Google Scholar]
  18. Meyer, U.A. Overview of enzymes of drug metabolism. J. Pharmacokinet. Biopharm. 1996, 24, 449–459. [Google Scholar]
  19. Tang, D.L.; Rundle, A.; Warburton, D.; Santella, R.M.; Tsai, W.Y.; Chiamprasert, S.; Hsu, Y.Z.; Perera, F.P. Associations between both genetic and environmental biomarkers and lung cancer: evidence of a greater risk of lung cancer in women smokers. Carcinogenesis 1998, 19, 1949–1953. [Google Scholar]
  20. Dresler, C.M.; Fratelli, C.; Babb, J.; Everley, L.; Evans, A.A.; Clapper, M.L. Gender differences in genetic susceptibility for lung cancer. Lung Cancer 2000, 30, 153–160. [Google Scholar]
  21. Ye, Z.; Song, H.; Higgins, J.P.; Pharoah, P.; Danesh, J. Five glutathione s-transferase gene variants in 23,452 cases of lung cancer and 30,397 controls: Meta-analysis of 130 studies. PLoS Med. 2006, 3, e91. [Google Scholar]
  22. Hung, R.J.; Boffetta, P.; Brockmoller, J.; Butkiewicz, D.; Cascorbi, I.; Clapper, M.L.; Garte, S.; Haugen, A.; Hirvonen, A.; Anttila, S.; Kalina, I.; Le Marchand, L.; London, S.J.; Rannug, A.; Romkes, M.; Salagovic, J.; Schoket, B.; Gaspari, L.; Taioli, E. CYP1A1 and GSTM1 genetic polymorphisms and lung cancer risk in Caucasian non-smokers: A pooled analysis. Carcinogenesis 2003, 24, 875–882. [Google Scholar]
  23. Le Marchand, L.; Guo, C.; Benhamou, S.; Bouchardy, C.; Cascorbi, I.; Clapper, M.L.; Garte, S.; Haugen, A.; Ingelman-Sundberg, M.; Kihara, M.; Rannug, A.; Ryberg, D.; Stucker, I.; Sugimura, H.; Taioli, E. Pooled analysis of the CYP1A1 exon 7 polymorphism and lung cancer (United States). Cancer Causes Control 2003, 14, 339–346. [Google Scholar]
  24. Timofeeva, M.N.; Kropp, S.; Sauter, W.; Beckmann, L.; Rosenberger, A.; Illig, T.; Jager, B.; Mittelstrass, K.; Dienemann, H.; Bartsch, H.; Bickeboller, H.; Chang-Claude, J.C.; Risch, A.; Wichmann, H.E. CYP450 polymorphisms as risk factors for early-onset lung cancer: gender-specific differences. Carcinogenesis 2009, 30, 1161–1169. [Google Scholar]
  25. Wang, Y.; Yang, H.; Li, L.; Wang, H.; Zhang, C.; Yin, G.; Zhu, B. Association between CYP2E1 genetic polymorphisms and lung cancer risk: a meta-analysis. Eur. J. Cancer 2010, 46, 758–764. [Google Scholar]
  26. Langevin, S.M.; Ioannidis, J.P.; Vineis, P.; Taioli, E. Assessment of cumulative evidence for the association between glutathione S-transferase polymorphisms and lung cancer: application of the Venice interim guidelines. Pharmacogenet Genomics 2010, 20, 586–597. [Google Scholar]
  27. Cote, M.L.; Chen, W.; Smith, D.W.; Benhamou, S.; Bouchardy, C.; Butkiewicz, D.; Fong, K.M.; Gene, M.; Hirvonen, A.; Kiyohara, C.; Larsen, J.E.; Lin, P.; Raaschou-Nielsen, O.; Povey, A.C.; Reszka, E.; Risch, A.; Schneider, J.; Schwartz, A.G.; Sorensen, M.; To-Figueras, J.; Tokudome, S.; Pu, Y.; Yang, P.; Wenzlaff, A.S.; Wikman, H.; Taioli, E. Meta- and pooled analysis of GSTP1 polymorphism and lung cancer: a HuGE-GSEC review. Am. J. Epidemiol. 2009, 169, 802–814. [Google Scholar]
  28. Hosgood, H.D., 3rd; Berndt, S.I.; Lan, Q. GST genotypes and lung cancer susceptibility in Asian populations with indoor air pollution exposures: a meta-analysis. Mutat. Res. 2007, 636, 134–143. [Google Scholar]
  29. Timofeeva, M.; Kropp, S.; Sauter, W.; Beckmann, L.; Rosenberger, A.; Illig, T.; Jager, B.; Mittelstrass, K.; Dienemann, H.; Bartsch, H.; Bickeboller, H.; Chang-Claude, J.; Risch, A.; Wichmann, H.E. Genetic polymorphisms of MPO, GSTT1, GSTM1, GSTP1, EPHX1 and NQO1 as risk factors of early-onset lung cancer. Int. J. Cancer 2010, 127, 1547–1561. [Google Scholar]
  30. Cote, M.L.; Yoo, W.; Wenzlaff, A.S.; Prysak, G.M.; Santer, S.K.; Claeys, G.B.; Van Dyke, A.L.; Land, S.J.; Schwartz, A.G. Tobacco and estrogen metabolic polymorphisms and risk of non-small cell lung cancer in women. Carcinogenesis 2009, 30, 626–635. [Google Scholar]
  31. Yang, M.; Choi, Y.; Hwangbo, B.; Lee, J.S. Combined effects of genetic polymorphisms in six selected genes on lung cancer susceptibility. Lung Cancer 2007, 57, 135–142. [Google Scholar]
  32. Cote, M.L.; Kardia, S.L.; Wenzlaff, A.S.; Land, S.J.; Schwartz, A.G. Combinations of glutathione S-transferase genotypes and risk of early-onset lung cancer in Caucasians and African Americans: a population-based study. Carcinogenesis 2005, 26, 811–819. [Google Scholar]
  33. Miller, D.P.; Neuberg, D.; de Vivo, I.; Wain, J.C.; Lynch, T.J.; Su, L.; Christiani, D.C. Smoking and the risk of lung cancer: susceptibility with GSTP1 polymorphisms. Epidemiology 2003, 14, 545–551. [Google Scholar]
  34. Kiyohara, C.; Yamamura, K.I.; Nakanishi, Y.; Takayama, K.; Hara, N. Polymorphism in GSTM1, GSTT1, and GSTP1 and Susceptibility to Lung Cancer in a Japanese Population. Asian Pac. J. Cancer Prev. 2000, 1, 293–298. [Google Scholar]
  35. Taioli, E.; Benhamou, S.; Bouchardy, C.; Cascorbi, I.; Cajas-Salazar, N.; Dally, H.; Fong, K.M.; Larsen, J.E.; Le Marchand, L.; London, S.J.; Risch, A.; Spitz, M.R.; Stucker, I.; Weinshenker, B.; Wu, X.; Yang, P. Myeloperoxidase G-463A polymorphism and lung cancer: a HuGE genetic susceptibility to environmental carcinogens pooled analysis. Genet. Med. 2007, 9, 67–73. [Google Scholar]
  36. Yoon, K.A.; Kim, J.H.; Gil, H.J.; Hwang, H.; Hwangbo, B.; Lee, J.S. CYP1B1, CYP1A1, MPO, and GSTP1 polymorphisms and lung cancer risk in never-smoking Korean women. Lung Cancer 2008, 60, 40–46. [Google Scholar]
  37. Wang, H.; Jin, G.; Liu, G.; Qian, J.; Jin, L.; Wei, Q.; Shen, H.; Huang, W.; Lu, D. Genetic susceptibility of lung cancer associated with common variants in the 3′ untranslated regions of the adenosine triphosphate-binding cassette B1 (ABCB1) and ABCC1 candidate transporter genes for carcinogen export. Cancer 2009, 115, 595–607. [Google Scholar]
  38. Berwick, M.; Vineis, P. Markers of DNA repair and susceptibility to cancer in humans: an epidemiologic review. J. Natl. Cancer. Inst. 2000, 92, 874–897. [Google Scholar]
  39. Wei, Q.; Cheng, L.; Amos, C.I.; Wang, L.E.; Guo, Z.; Hong, W.K.; Spitz, M.R. Repair of tobacco carcinogen-induced DNA adducts and lung cancer risk: a molecular epidemiologic study. J. Natl. Cancer Inst. 2000, 92, 1764–1772. [Google Scholar]
  40. Mooney, L.A.; Perera, F.P.; Van Bennekum, A.M.; Blaner, W.S.; Karkoszka, J.; Covey, L.; Hsu, Y.; Cooper, T.B.; Frenkel, K. Gender differences in autoantibodies to oxidative DNA base damage in cigarette smokers. Cancer Epidemiol. Biomarkers Prev. 2001, 10, 641–648. [Google Scholar]
  41. Kaina, B.; Christmann, M.; Naumann, S.; Roos, W.P. MGMT: key node in the battle against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents. DNA Repair (Amst.) 2007, 6, 1079–1099. [Google Scholar]
  42. Licchesi, J.D.; Westra, W.H.; Hooker, C.M.; Herman, J.G. Promoter hypermethylation of hallmark cancer genes in atypical adenomatous hyperplasia of the lung. Clin. Cancer Res. 2008, 14, 2570–2578. [Google Scholar]
  43. Esteller, M.; Hamilton, S.R.; Burger, P.C.; Baylin, S.B.; Herman, J.G. Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia. Cancer Res. 1999, 59, 793–797. [Google Scholar]
  44. Zochbauer-Muller, S.; Fong, K.M.; Virmani, A.K.; Geradts, J.; Gazdar, A.F.; Minna, J.D. Aberrant promoter methylation of multiple genes in non-small cell lung cancers. Cancer Res. 2001, 61, 249–255. [Google Scholar]
  45. Uccella, S.; Cerutti, R.; Placidi, C.; Marchet, S.; Carnevali, I.; Bernasconi, B.; Proserpio, I.; Pinotti, G.; Tibiletti, M.G.; Furlan, D.; Capella, C. MGMT methylation in diffuse large B-cell lymphoma: validation of quantitative methylation-specific PCR and comparison with MGMT protein expression. J. Clin. Pathol. 2009, 62, 715–723. [Google Scholar]
  46. Wu, J.Y.; Wang, J.; Lai, J.C.; Cheng, Y.W.; Yeh, K.T.; Wu, T.C.; Chen, C.Y.; Lee, H. Association of O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation with p53 mutation occurrence in non-small cell lung cancer with different histology, gender, and smoking status. Ann. Surg. Oncol. 2008, 15, 3272–3277. [Google Scholar]
  47. Kaur, T.B.; Travaline, J.M.; Gaughan, J.P.; Richie, J.P., Jr.; Stellman, S.D.; Lazarus, P. Role of polymorphisms in codons 143 and 160 of the O6-alkylguanine DNA alkyltransferase gene in lung cancer risk. Cancer Epidemiol. Biomarkers Prev. 2000, 9, 339–342. [Google Scholar]
  48. Krzesniak, M.; Butkiewicz, D.; Samojedny, A.; Chorazy, M.; Rusin, M. Polymorphisms in TDG and MGMT genes - epidemiological and functional study in lung cancer patients from Poland. Ann. Hum. Genet. 2004, 68, 300–312. [Google Scholar]
  49. Cohet, C.; Borel, S.; Nyberg, F.; Mukeria, A.; Bruske-Hohlfeld, I.; Constantinescu, V.; Benhamou, S.; Brennan, P.; Hall, J.; Boffetta, P. Exon 5 polymorphisms in the O6-alkylguanine DNA alkyltransferase gene and lung cancer risk in non-smokers exposed to second-hand smoke. Cancer Epidemiol. Biomarkers Prev. 2004, 13, 320–323. [Google Scholar]
  50. Yang, M.; Coles, B.F.; Caporaso, N.E.; Choi, Y.; Lang, N.P.; Kadlubar, F.F. Lack of association between Caucasian lung cancer risk and O6-methylguanine-DNA methyltransferase-codon 178 genetic polymorphism. Lung Cancer 2004, 44, 281–286. [Google Scholar]
  51. Chae, M.H.; Jang, J.S.; Kang, H.G.; Park, J.H.; Park, J.M.; Lee, W.K.; Kam, S.; Lee, E.B.; Son, J.W.; Park, J.Y. O6-alkylguanine-DNA alkyltransferase gene polymorphisms and the risk of primary lung cancer. Mol. Carcinog. 2006, 45, 239–249. [Google Scholar]
  52. Zhong, Y.; Huang, Y.; Zhang, T.; Ma, C.; Zhang, S.; Fan, W.; Chen, H.; Qian, J.; Lu, D. Effects of O6-methylguanine-DNA methyltransferase (MGMT) polymorphisms on cancer: a meta-analysis. Mutagenesis 2010, 25, 83–95. [Google Scholar]
  53. Wang, L.; Liu, H.; Zhang, Z.; Spitz, M.R.; Wei, Q. Association of genetic variants of O6-methylguanine-DNA methyltransferase with risk of lung cancer in non-Hispanic Whites. Cancer Epidemiol. Biomarkers Prev. 2006, 15, 2364–2369. [Google Scholar]
  54. Hung, R.J.; Christiani, D.C.; Risch, A.; Popanda, O.; Haugen, A.; Zienolddiny, S.; Benhamou, S.; Bouchardy, C.; Lan, Q.; Spitz, M.R.; et al. International Lung Cancer Consortium: pooled analysis of sequence variants in DNA repair and cell cycle pathways. Cancer Epidemiol. Biomarkers Prev. 2008, 17, 3081–3089. [Google Scholar]
  55. Riely, G.J.; Kris, M.G.; Rosenbaum, D.; Marks, J.; Li, A.; Chitale, D.A.; Nafa, K.; Riedel, E.R.; Hsu, M.; Pao, W.; Miller, V.A.; Ladanyi, M. Frequency and distinctive spectrum of KRAS mutations in never smokers with lung adenocarcinoma. Clin. Cancer Res. 2008, 14, 5731–5734. [Google Scholar]
  56. Sun, S.; Schiller, J.H.; Gazdar, A.F. Lung cancer in never smokers--a different disease. Nat. Rev. Cancer 2007, 7, 778–790. [Google Scholar]
  57. Slebos, R.J.; Kibbelaar, R.E.; Dalesio, O.; Kooistra, A.; Stam, J.; Meijer, C.J.; Wagenaar, S.S.; Vanderschueren, R.G.; van Zandwijk, N.; Mooi, W.J.; et al. K-ras oncogene activation as a prognostic marker in adenocarcinoma of the lung. N. Engl. J. Med. 1990, 323, 561–565. [Google Scholar]
  58. Keohavong, P.; DeMichele, M.A.; Melacrinos, A.C.; Landreneau, R.J.; Weyant, R.J.; Siegfried, J.M. Detection of K-ras mutations in lung carcinomas: relationship to prognosis. Clin. Cancer Res. 1996, 2, 411–418. [Google Scholar]
  59. Ahrendt, S.A.; Decker, P.A.; Alawi, E.A.; Zhu Yr, Y.R.; Sanchez-Cespedes, M.; Yang, S.C.; Haasler, G.B.; Kajdacsy-Balla, A.; Demeure, M.J.; Sidransky, D. Cigarette smoking is strongly associated with mutation of the K-ras gene in patients with primary adenocarcinoma of the lung. Cancer 2001, 92, 1525–1530. [Google Scholar]
  60. Nelson, H.H.; Christiani, D.C.; Mark, E.J.; Wiencke, J.K.; Wain, J.C.; Kelsey, K.T. Implications and prognostic value of K-ras mutation for early-stage lung cancer in women. J. Natl. Cancer Inst. 1999, 91, 2032–2038. [Google Scholar]
  61. Boldrini, L.; Ali, G.; Gisfredi, S.; Ursino, S.; Baldini, E.; Melfi, F.; Lucchi, M.; Comin, C.E.; Maddau, C.; Tibaldi, C.; Camacci, T.; Servadio, A.; Mussi, A.; Fontanini, G. Epidermal growth factor receptor and K-RAS mutations in 411 lung adenocarcinoma: a population-based prospective study. Oncol. Rep. 2009, 22, 683–691. [Google Scholar]
  62. Chiba, I.; Takahashi, T.; Nau, M.M.; D'Amico, D.; Curiel, D.T.; Mitsudomi, T.; Buchhagen, D.L.; Carbone, D.; Piantadosi, S.; Koga, H.; et al. Mutations in the p53 gene are frequent in primary, resected non-small cell lung cancer. Lung Cancer Study Group. Oncogene 1990, 5, 1603–1610. [Google Scholar]
  63. Kure, E.H.; Ryberg, D.; Hewer, A.; Phillips, D.H.; Skaug, V.; Baera, R.; Haugen, A. p53 mutations in lung tumours: relationship to gender and lung DNA adduct levels. Carcinogenesis 1996, 17, 2201–2205. [Google Scholar]
  64. Tammemagi, M.C.; McLaughlin, J.R.; Bull, S.B. Meta-analyses of p53 tumor suppressor gene alterations and clinicopathological features in resected lung cancers. Cancer Epidemiol. Biomarkers Prev. 1999, 8, 625–634. [Google Scholar]
  65. Toyooka, S.; Tsuda, T.; Gazdar, A.F. The TP53 gene, tobacco exposure, and lung cancer. Hum. Mutat. 2003, 21, 229–239. [Google Scholar]
  66. Marrogi, A.J.; Mechanic, L.E.; Welsh, J.A.; Bowman, E.D.; Khan, M.A.; Enewold, L.; Shields, P.G.; Harris, C.C. TP53 mutation spectrum in lung cancer is not different in women and men. Cancer Epidemiol. Biomarkers Prev. 2005, 14, 1031–1033. [Google Scholar]
  67. Fang, S.; Krahe, R.; Lozano, G.; Han, Y.; Chen, W.; Post, S.M.; Zhang, B.; Wilson, C.D.; Bachinski, L.L.; Strong, L.C.; Amos, C.I. Effects of MDM2, MDM4 and TP53 codon 72 polymorphisms on cancer risk in a cohort study of carriers of TP53 germline mutations. PLoS One 2010, 5, e10813. [Google Scholar]
  68. Bond, G.L.; Hu, W.; Levine, A.J. MDM2 is a central node in the p53 pathway: 12 years and counting. Curr. Cancer Drug Targets 2005, 5, 3–8. [Google Scholar]
  69. Bond, G.L.; Hu, W.; Bond, E.E.; Robins, H.; Lutzker, S.G.; Arva, N.C.; Bargonetti, J.; Bartel, F.; Taubert, H.; Wuerl, P.; Onel, K.; Yip, L.; Hwang, S.J.; Strong, L.C.; Lozano, G.; Levine, A.J. A single nucleotide polymorphism in the MDM2 promoter attenuates the p53 tumor suppressor pathway and accelerates tumor formation in humans. Cell 2004, 119, 591–602. [Google Scholar]
  70. Bougeard, G.; Baert-Desurmont, S.; Tournier, I.; Vasseur, S.; Martin, C.; Brugieres, L.; Chompret, A.; Bressac-de Paillerets, B.; Stoppa-Lyonnet, D.; Bonaiti-Pellie, C.; Frebourg, T. Impact of the MDM2 SNP309 and p53 Arg72Pro polymorphism on age of tumour onset in Li- Fraumeni syndrome. J. Med. Genet. 2006, 43, 531–533. [Google Scholar]
  71. Bond, G.L.; Hirshfield, K.M.; Kirchhoff, T.; Alexe, G.; Bond, E.E.; Robins, H.; Bartel, F.; Taubert, H.; Wuerl, P.; Hait, W.; Toppmeyer, D.; Offit, K.; Levine, A.J. MDM2 SNP309 accelerates tumor formation in a gender-specific and hormone-dependent manner. Cancer Res. 2006, 66, 5104–5110. [Google Scholar]
  72. Marchetti, A.; Martella, C.; Felicioni, L.; Barassi, F.; Salvatore, S.; Chella, A.; Camplese, P.P.; Iarussi, T.; Mucilli, F.; Mezzetti, A.; Cuccurullo, F.; Sacco, R.; Buttitta, F. 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, 857–865. [Google Scholar]
  73. Hsieh, R.K.; Lim, K.H.; Kuo, H.T.; Tzen, C.Y.; Huang, M.J. Female sex and bronchioloalveolar pathologic subtype predict EGFR mutations in non-small cell lung cancer. Chest 2005, 128, 317–321. [Google Scholar]
  74. Shigematsu, H.; Lin, L.; Takahashi, T.; Nomura, M.; Suzuki, M.; Wistuba, II; Fong, K.M.; Lee, H.; Toyooka, S.; Shimizu, N.; Fujisawa, T.; Feng, Z.; Roth, J.A.; Herz, J.; Minna, J.D.; Gazdar, A.F. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. J. Natl. Cancer Inst. 2005, 97, 339–346. [Google Scholar]
  75. Bell, D.W.; Lynch, T.J.; Haserlat, S.M.; Harris, P.L.; Okimoto, R.A.; Brannigan, B.W.; Sgroi, D.C.; Muir, B.; Riemenschneider, M.J.; Iacona, R.B.; Krebs, A.D.; Johnson, D.H.; Giaccone, G.; Herbst, R.S.; Manegold, C.; Fukuoka, M.; Kris, M.G.; Baselga, J.; Ochs, J.S.; Haber, D.A. 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, 8081–8092. [Google Scholar]
  76. Sasaki, H.; Shimizu, S.; Endo, K.; Takada, M.; Kawahara, M.; Tanaka, H.; Matsumura, A.; Iuchi, K.; Haneda, H.; Suzuki, E.; Kobayashi, Y.; Yano, M.; Fujii, Y. EGFR and erbB2 mutation status in Japanese lung cancer patients. Int. J. Cancer 2006, 118, 180–184. [Google Scholar]
  77. Sasaki, H.; Okuda, K.; Kawano, O.; Endo, K.; Yukiue, H.; Yokoyama, T.; Yano, M.; Fujii, Y. ErbB4 expression and mutation in Japanese patients with lung cancer. Clin. Lung Cancer 2007, 8, 429–433. [Google Scholar]
  78. Matsuo, K.; Ito, H.; Yatabe, Y.; Hiraki, A.; Hirose, K.; Wakai, K.; Kosaka, T.; Suzuki, T.; Tajima, K.; Mitsudomi, T. Risk factors differ for non-small-cell lung cancers with and without EGFR mutation: assessment of smoking and sex by a case-control study in Japanese. Cancer Sci. 2007, 98, 96–101. [Google Scholar]
  79. Rosell, R.; Moran, T.; Queralt, C.; Porta, R.; Cardenal, F.; Camps, C.; Majem, M.; Lopez-Vivanco, G.; Isla, D.; Provencio, M.; Insa, A.; Massuti, B.; Gonzalez-Larriba, J.L.; Paz-Ares, L.; Bover, I.; Garcia-Campelo, R.; Moreno, M.A.; Catot, S.; Rolfo, C.; Reguart, N.; Palmero, R.; Sanchez, J.M.; Bastus, R.; Mayo, C.; Bertran-Alamillo, J.; Molina, M.A.; Sanchez, J.J.; Taron, M. Screening for epidermal growth factor receptor mutations in lung cancer. N. Engl. J. Med. 2009, 361, 958–967. [Google Scholar]
  80. McMillen, E.; Ye, F.; Li, G.; Wu, Y.; Yin, G.; Liu, W. Epidermal growth factor receptor (EGFR) mutation and p-EGFR expression in resected non-small cell lung cancer. Exp. Lung Res. 2010, 36, 531–537. [Google Scholar]
  81. Toyooka, S.; Matsuo, K.; Shigematsu, H.; Kosaka, T.; Tokumo, M.; Yatabe, Y.; Ichihara, S.; Inukai, M.; Suehisa, H.; Soh, J.; Kiura, K.; Fong, K.M.; Lee, H.; Wistuba, II; Gazdar, A.F.; Mitsudomi, T.; Date, H. The impact of sex and smoking status on the mutational spectrum of epidermal growth factor receptor gene in non small cell lung cancer. Clin. Cancer Res. 2007, 13, 5763–5768. [Google Scholar]
  82. Tanaka, T.; Matsuoka, M.; Sutani, A.; Gemma, A.; Maemondo, M.; Inoue, A.; Okinaga, S.; Nagashima, M.; Oizumi, S.; Uematsu, K.; Nagai, Y.; Moriyama, G.; Miyazawa, H.; Ikebuchi, K.; Morita, S.; Kobayashi, K.; Hagiwara, K. Frequency of and variables associated with the EGFR mutation and its subtypes. Int. J. Cancer 2010, 126, 651–655. [Google Scholar]
  83. Zhu, C.Q.; da Cunha Santos, G.; Ding, K.; Sakurada, A.; Cutz, J.C.; Liu, N.; Zhang, T.; Marrano, P.; Whitehead, M.; Squire, J.A.; Kamel-Reid, S.; Seymour, L.; Shepherd, F.A.; Tsao, M.S. 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, 4268–4275. [Google Scholar]
  84. Hirsch, F.R.; Varella-Garcia, M.; Bunn, P.A., Jr.; Di Maria, M.V.; Veve, R.; Bremmes, R.M.; Baron, A.E.; Zeng, C.; Franklin, W.A. Epidermal growth factor receptor in non-small-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis. J. Clin. Oncol. 2003, 21, 3798–3807. [Google Scholar]
  85. Hirsch, F.R.; Varella-Garcia, M.; Bunn, P.A., Jr.; Franklin, W.A.; Dziadziuszko, R.; Thatcher, N.; Chang, A.; Parikh, P.; Pereira, J.R.; Ciuleanu, T.; von Pawel, J.; Watkins, C.; Flannery, A.; Ellison, G.; Donald, E.; Knight, L.; Parums, D.; Botwood, N.; Holloway, B. 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, 5034–5042. [Google Scholar]
  86. Jeon, Y.K.; Sung, S.W.; Chung, J.H.; Park, W.S.; Seo, J.W.; Kim, C.W.; Chung, D.H. Clinicopathologic features and prognostic implications of epidermal growth factor receptor (EGFR) gene copy number and protein expression in non-small cell lung cancer. Lung Cancer 2006, 54, 387–398. [Google Scholar]
  87. Sasaki, H.; Shimizu, S.; Okuda, K.; Kawano, O.; Yukiue, H.; Yano, M.; Fujii, Y. Epidermal growth factor receptor gene amplification in surgical resected Japanese lung cancer. Lung Cancer 2009, 64, 295–300. [Google Scholar]
  88. Jou, Y.S.; Lo, Y.L.; Hsiao, C.F.; Chang, G.C.; Tsai, Y.H.; Su, W.C.; Chen, Y.M.; Huang, M.S.; Chen, H.L.; Chen, C.J.; Yang, P.C.; Hsiung, C.A. Association of an EGFR intron 1 SNP with never-smoking female lung adenocarcinoma patients. Lung Cancer 2009, 64, 251–256. [Google Scholar]

Share and Cite

MDPI and ACS Style

Yim, S.-H.; Chung, Y.-J. Molecular Epidemiology of Female Lung Cancer. Cancers 2011, 3, 1861-1876. https://doi.org/10.3390/cancers3021861

AMA Style

Yim S-H, Chung Y-J. Molecular Epidemiology of Female Lung Cancer. Cancers. 2011; 3(2):1861-1876. https://doi.org/10.3390/cancers3021861

Chicago/Turabian Style

Yim, Seon-Hee, and Yeun-Jun Chung. 2011. "Molecular Epidemiology of Female Lung Cancer" Cancers 3, no. 2: 1861-1876. https://doi.org/10.3390/cancers3021861

Article Metrics

Back to TopTop