ONCH-1762; No. of Pages 10
ARTICLE IN PRESS
Critical Reviews in Oncology/Hematology xxx (2013) xxx–xxx
Lung cancer in never smokers: Disease characteristics and risk factors Athanasios G. Pallis ∗ , Konstantinos N. Syrigos Oncology Unit GPP, Athens School of Medicine, Athens, Greece Accepted 28 June 2013
Contents 1. 2. 3. 4. 5.
6.
7.
8. 9.
Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Search strategy and selection criteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Pathology and clinical characteristics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Risk factors for lung cancer development in never-smokers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.1. Second hand smoking . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.2. Hormonal factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.3. Genetic factors and lung cancer susceptibility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Inherited susceptibility to lung cancer in never smokers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.1. Carcinogen metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.2. DNA repair genes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.3. Inflammatory response genes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acquired somatic mutations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7.1. EGFR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7.2. HER-2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7.3. ALK rearrangements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7.4. ROS1 rearrangements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Prognostic and predictive role. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Reviewers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Biographies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00
Abstract It is estimated that approximately 25% of all lung cancer cases are observed in never-smokers and its incidence is expected to increase due to smoking prevention programs. Risk factors for the development of lung cancer described include second-hand smoking, radon exposure, occupational exposure to carcinogens and to cooking oil fumes and indoor coal burning. Other factors reported are infections (HPV and Mycobacterium tuberculosis), hormonal and diatery factors and diabetes mellitus. Having an affected relative also increases the risk for lung cancer while recent studies have identified several single nucleotide polymorphisms associated with increased risk for lung cancer development in never smokers. Distinct clinical, pathology and molecular characteristics are observed in lung cancer in never smokers; more frequently is observed in females and adenocarcinoma is the predominant histology while it has a different pattern of molecular alterations. The purpose of this review is to summarize our current knowledge of this disease. © 2013 Elsevier Ireland Ltd. All rights reserved. Keywords: Lung cancer; Smoking; Never smokers; NSCLC; EGFR ∗ Corresponding author at: Oncology Unit GPP, Athens School of Medicine, Sotiria General Hospital, Mesogion 152, 115 26 Athens, Greece. Tel.: +30 2107475034. E-mail address:
[email protected] (A.G. Pallis).
1040-8428/$ – see front matter © 2013 Elsevier Ireland Ltd. All rights reserved. http://dx.doi.org/10.1016/j.critrevonc.2013.06.011
Please cite this article in press as: Pallis AG, Syrigos KN. Lung cancer in never smokers: Disease characteristics and risk factors. Crit Rev Oncol/Hematol (2013), http://dx.doi.org/10.1016/j.critrevonc.2013.06.011
ONCH-1762; No. of Pages 10
2
ARTICLE IN PRESS A.G. Pallis, K.N. Syrigos / Critical Reviews in Oncology/Hematology xxx (2013) xxx–xxx
1. Introduction Lung cancer is the leading cause of cancer related death in Western world, with approximately 1.6 million new cases and 1.4 million deaths globally in 2008 [1]. Cigarette smoking is considered the major and most well-established risk factor for human lung cancer and there is a huge bank of both prospective and retrospective epidemiologic research data supporting this association [2–4]. The relative risk of lung cancer development in a long-term smoker compared with a never-smoker is estimated to be 10- to 30-fold [5]. However, lung cancer occurs also in individuals with no history of smoking. After the observation that gefitinib is very active in never-smokers non-small-cell lung cancer (NSCLC) patients [6] attention was turned toward this population. Lung cancer in never-smokers has been described recently and major clinic-pathological differences and differences on the molecular level have been observed, implying that this might be a different disease compared with lung cancer in smokers [7–9]. Patients who have smoked less than 100 cigarettes during their lifetime are considered as “never-smokers”, while people who have smoked more are considered as “ever-smokers” [7]. The purpose of this review is to summarize our current knowledge of the epidemiological, clinical and molecular characteristics of lung cancer in never-smokers.
2. Search strategy and selection criteria A bibliographic search of the Medline database was conducted for papers published from 2000 to 2012, with the keywords “lung cancer”, “lung neoplasm”, “chemotherapy”, “NSCLC”, “risk factors”, “smoking”, and “smoker”. The search was limited to articles written in English. The Medline search was supplemented by a manual search of meeting abstracts (World Conference on Lung Cancer, European Society of Medical Oncology Annual Congress, American Society of Clinical Oncology Annual Meeting, and European Lung Cancer Conference) as well as reference lists of original and review articles.
3. Epidemiology According to the World Health Organization the incidence of lung cancer in never-smokers is approximately 25% of all cases [10]. However, there is considerable variance in the proportions of lung cancer in never smokers (LCNS) ranging from approximately 10% in males in Western world [11], up to almost 40% in females in Asia [12]. If LCNS is considered as a separate entity then it is the seventh cause of cancer-related death in the world [7]. Wakelee et al. using data from six big epidemiological studies found that in these six cohorts age-adjusted incidence rates of lung cancer among never smokers age 40 to 79 years ranged from 14.4 to 20.8 per
100,000 person-years in women and 4.8 to 13.7 per 100,000 person-years in men [13]. A large epidemiological study from US with approximately 12,000 patients found that approximately 10% of them were never-smokers [14], while a large population-based study of 20,500 cases by Radzikowska et al. found that only 4.3% of all these cases were never-smokers [15]. There are conflicting data regarding changes in the incidence of LCNS. A report by Bofetta et al. observed a significant increase in LCNS incidence (from 1.5 to 5.4/100,000 of population) in a Swedish cohort study [16], while another large epidemiological study from US observed a non-significant increase in the prevalence of LCNS (bronchoalveolar subtype) from 1995–1999 to 1999–2003 [14]. Finally, two large case control studies from UK (hospital and community based) found no difference in the percentage of never-smokers among male lung cancer patients between 1950 and 1990 (0.5% in both cases), while for women the percentage of never-smokers among lung cancer patients decreased from 37% in 1950 to 7.6% in 1990 [17].
4. Pathology and clinical characteristics Small-cell lung cancer is rarely observed in never-smokers [18], while in NSCLC the most common histological type in never-smokers is adenocarcinoma [8,9,12,13,19–22]. A case–control study by Sobue et al. [23] demonstrated that the relative risk for incident cases associated with current smokers versus never-smokers according to histologic type was 12.7 (95% Confidence Interval [CI]: 4.7–34.7) and 17.5 (95% CI: 4.9–62.1) for squamous cell carcinoma and small cell carcinoma, while for adenocarcinoma it was 2.8 (95% CI: 1.6–4.9) and 2.0 (95% CI: 0.8–5.0) for men and women, respectively. Similarly, in a report by Toh et al., adenocarcinomas involved 69.9% of cases in never-smokers, versus 39.9% in current and 47.3% in former smokers (p-value < 0.001) [8]. It is also noteworthy that adenocarcinoma was also the most common histology in young patients population (<40 years of age), in which population the proportion of neversmokers was high [24]. An analysis of 17 trials by Sun et al. demonstrated that adenocarcinoma was 3.4 times more frequent than squamous carcinoma in never smokers [7], and this higher incidence of adenocarcinoma was also confirmed by Radzikowska et al. [15]. Significant gender variations are also observed in LCNS. Lung cancer in never smokers is more common in women and this is confirmed in numerous series [7–9,12,13,15,19–22,25]. However, it is not clear whether this higher incidence in females is due to inherent susceptibility or to a greater contribution of risk factors other than smoking, or this simply represents the fact that twice as many women as men are never smokers and this difference increases with age [26]. Furthermore, significant geographical variation is observed in the proportion of never-smokers among female lung cancer cases, with that proportion being
Please cite this article in press as: Pallis AG, Syrigos KN. Lung cancer in never smokers: Disease characteristics and risk factors. Crit Rev Oncol/Hematol (2013), http://dx.doi.org/10.1016/j.critrevonc.2013.06.011
ONCH-1762; No. of Pages 10
ARTICLE IN PRESS A.G. Pallis, K.N. Syrigos / Critical Reviews in Oncology/Hematology xxx (2013) xxx–xxx
approximately 10–15% in the Western world to 80% in south Asia [7]. On the contrary, an analysis of 13 cohorts by Thun et al., found no significant difference in the incidence rates between male (14.0 per 100,000) and female (13.8 per 100,000) never-smokers lung cancer patients of European descent, when the rates were standardized to all ages 40 years and above, although women had higher incidence rates in the age group of 40–59 years [27]. However, despite the higher incidence rates in females, prospective data have consistently reported higher death rates from lung cancer in male than female LCNS [26,27]. Thun et al. studied data from two large American Cancer Society Cancer Prevention Study cohorts (CPS I and II) and the age-standardized lung cancer death rates among never-smokers were 17.1 and 14.7 per 100,000, for men and women, respectively [26]. Several studies mostly from Asia support that LCNS is diagnosed in earlier age [8,12,21], although this was not confirmed in all cases [9,13,20]. An epidemiological study in a Caucasian population found a higher percentage of never smokers (23.9%) among a cohort of early onset (≤50 years) lung cancer patients than among patients diagnosed at ≥70 years of age (17.6%; p-value < 0.001) [28], although another study limited to patients with adenocarcinoma had the opposite result (median age of diagnosis 63.5 for LCNS patients versus 59.4 for smokers; p-value = 0.0005) [25]. Furthermore, retrospective reports found that LCNS is presented at a more advanced disease stage [8,21]. This is reasonable given that because lung cancer is a disease of smokers the clinical threshold for examining a symptomatic never-smoker potential lung cancer patient is higher, a fact that leads to delayed diagnosis and thus to a more advanced disease stage at presentation for never smokers. However, this observation has not been confirmed by other studies [12,19,20,22].
5. Risk factors for lung cancer development in never-smokers Although the most well established risk factor for lung cancer is smoking several other, unrelated to smoking, risk factors have been described however, the association is weak to moderate and the cause(s) of lung cancer in never-smokers remain to be found. 5.1. Second hand smoking Given the strong association between smoking and lung cancer the issue of second hand smoking has been widely studied. This assumption is supported also by a recent study supporting that tobacco metabolites can be found in 90% of urine samples of children whose parents smoke [29]. According to a retrospective analysis of data from 192 countries approximately 21,000 deaths of lung cancer could be attributed to second hand smoking [30]. Similarly a big European prospective study estimated the proportion of lung cancers in never- and ex-smokers attributable to
3
environmental tobacco smoke as between 16 and 24%, mainly due to the contribution of work-related exposure [31]. A meta-analysis published in 1997 demonstrated a 25% excess risk for lung cancer development in non-smokers who lived with a smoker [32]. The International Agency for Research on Cancer (IARC) analysis estimated the risk to develop lung cancer to be increased by 35% in men and 25% in women who were exposed to second hand smoking, compared to men and women who were not exposed [33]. An updated meta-analysis of the IARC analysis including data from 19 studies of women who did not smoke (n = 3588 lung cancer patients) demonstrated an increased risk of approximately 20% due to second hand smoking [34]. The current evidence supports that second-hand smoking plays a modest role to lung-cancer development and larger samples may be required to detect a strong relationship. 5.2. Hormonal factors The predominance of females among LCNS patients also implies a potential role for hormones. Estrogen receptors (ER ␣ and ) are expressed in both normal lung tissue and lung tumors [35]. Other studies also found that ER- is more frequently expressed in women and never smokers [36] and in certain types of adenocarcinomas [37]. Activation of ERs result in increased expression of genes involved in cellular proliferation and tumor growth [38,39]. Expression of estrogen receptors is also associated with prognosis. ER-␣ is a poor prognostic factor while the expression of ER- is associated with better prognosis [35]. The fact that ERs may be involved in carcinogenesis mechanisms and may have an impact on lung cancer prognosis has raised the question about the role of hormone replacement therapy (HRT). Current data do not support an association between HRT and increased risk of lung cancer, although it should be noted that available data is limited [40]. Several other risk factors are presented in Table 1. However, it should be emphasized that the association is weak to moderate highlighting the need for further research in this area. 5.3. Genetic factors and lung cancer susceptibility Although as stated before the major risk factor for lung cancer is smoking, there is a growing bank of data supporting that genetic factors may play a role in lung cancer susceptibility and development. Tokuhata and Lilienfeld [41] initially reported a 2.5-fold higher lung cancer risk in smoking first-degree relatives of lung cancer cases compared with smoking relatives of controls. They also demonstrated that the familial aggregation occurred irrespective of tobacco use history [41]. This observation was confirmed by several epidemiological studies that consistently demonstrated an approximately 2.5-fold higher risk for lung cancer attributable to a family history of lung cancer after controlling for tobacco smoke [42].
Please cite this article in press as: Pallis AG, Syrigos KN. Lung cancer in never smokers: Disease characteristics and risk factors. Crit Rev Oncol/Hematol (2013), http://dx.doi.org/10.1016/j.critrevonc.2013.06.011
ONCH-1762; No. of Pages 10
4
ARTICLE IN PRESS A.G. Pallis, K.N. Syrigos / Critical Reviews in Oncology/Hematology xxx (2013) xxx–xxx
Table 1 Smoking-unrelated risk factors for lung cancer development. Factor Second-hand smoking Radon exposure Occupational exposure to carcinogens Cooking oil vapors and indoor coal burning Hormonal factors Air pollution Infectious factors Diabetes mellitus Diatery factors
A meta-analysis on passive smoking found a higher cancer risk due to spousal exposure [32–34] Radon is an established risk factor for lung cancer [95] A list of potential lung carcinogens are recognized by the International Agency for Research on Cancer (asbestos, polycyclic aromatic hydrocarbons, silica, heavy metals) [96] Epidemiological studies in Asia have confirmed an association between exposure to cooking oil fumes and indoor coal burning and lung cancer development [97,98]. An IARC study in Caucasians found that solid fuel use for heating or cooking was associated with higher risk for lung cancer development (odds ratio 1.24 for heating and 1.37 for cooking) [99]. Epidemiological studies have suggested that exogenous and endogenous estrogen may be associated in lung carcinogenesis [40] An increase in lung cancer risk has been reported as a result of exposure to air pollution components [100] HPV infection might be associated with lung cancer development [101] M. tuberculosis is considered as a risk factor for lung cancer [102] It is considered as an independent risk factor for lung cancer [103] Healthy eating pattern (vegetables, fruits, and low-fat food items) is associated with significantly reduced risk of lung-cancer in never-smokers [104]
A recent review by Matakidou et al. using data from both case–control and cohort studies show a significantly increased lung cancer risk associated with having an affected relative [43]. Risk was greater in relatives of cases diagnosed at a young age and in those with multiple affected family members. A pooled analysis of all case–control studies (n = 28 with a total of 15,766 cases and 18,184 controls) demonstrated a significantly elevated relative risk (RR) of lung cancer associated with having an affected relative (RR: 1.82; 95% CI: 1.58–2.10). Pooled analysis of four cohort studies that have investigated the relationship between family history and lung cancer risk demonstrated a pooled RR of 2.01 (95% CI: 1.62–2.50).
6. Inherited susceptibility to lung cancer in never smokers Recent multistage genome-wide association (GWAS) studies identified a locus within chromosome region 15q24-15q25.1 that contributes to lung cancer risk [44,45]. This region contains several genes, including three that encode nicotinic acetylcholine receptor subunits (CHRNA5, CHRNA3 and CHRNB4). These subunits are expressed in alveolar epithelial cells, pulmonary neuroendocrine cells and lung cancer cell lines, and they bind to N -nitrosonornicotine and potential lung carcinogens. Two single-nucleotide polymorphisms (SNPs), (rs1051730 and rs8034191) were identified and associated with increased lung cancer risk [45]. For rs8034191 and rs1051730, the combined P values were 3.15 × 10−18 and 7.00 × 10−18 , respectively. Combined adjusted ORs for lung cancer associated with rs8034191 and rs1051730 were 1.32 (95% CI: 1.24–1.41) and 1.32 (95% CI: 1.23–1.39), respectively. Hung et al. [44] observed that these SNPs were associated with lung cancer risk and they were found to account for 14% (attributable risk) of lung cancer cases. Statistically similar risks were reported regardless of smoking status or propensity to smoke tobacco. A study by Amos et al. failed to confirm this
observation [45]. However, it should be noted that the latter study included only 125 never-smoking cases in their series. Genome-wide association studies have also identified chromosome 5p15.33 as one of the regions associated with lung cancer in non-smokers [46,47]. In these studies a highly significant association between the common SNP rs2736100 localized in the 5p15.33 chromosome and lung cancer was observed (p = 0.02 in the McKay study [46] and p = 2.6 × 10−20 in the Hsiung study [47]). The McKay study [46] included both smokers and never-smokers, while the Hsiung study [47] included only lung adenocarcinoma neversmoker females. This 5p15.33 region contains two candidate susceptibility genes, TERT and CLPTM1L. TERT encodes a catalytic subunit of telomerase that maintains telomere ends and its overexpression leads to prolongation of the life span of the cell. Although not detectable in most normal tissues, it is overexpressed in cancer cells. CLPTM1L is implicated in apoptosis and its upregulation results in cisplatin-resistant cell lines. The study by McKay et al. identified also a second SNP that was significantly associated with higher risk for lung cancer, the rs402710 (p = 4 × 10−6 ). Another region that has been implicated by GWAS in susceptibility to lung cancer is the HLA region at chromosome 6p21.33 [48]. The authors identified significant associations for two SNPs (rs3117582 and rs3131379) after adjustment for multiple testing. Two susceptibility genes have been identified; BAT3 and MSH5. BAT3 is implicated in apoptosis and the protein complexes with E1A-binding protein p300, required for acetylation of p53 in response to DNA damage [49]. MSH5 is involved in DNA mismatch repair (MMR) and meiotic recombination, and deficiency of MMR has been documented to have a role in lung cancer [50]. Li et al. also identified a SNP (rs2352028) within chromosome 13q31.3 that was significantly associated with risk for lung cancer in never smokers [51]. According to authors more than 10% of cases of lung cancer in never smokers could be attributed to genetic variation of the SNP rs2352028. This SNP was correlated with decreased GPC5 gene expression. GPC5 is a member of the glypican gene family, and glypicans (family of heparan sulphate proteoglycans (HSPGs) that
Please cite this article in press as: Pallis AG, Syrigos KN. Lung cancer in never smokers: Disease characteristics and risk factors. Crit Rev Oncol/Hematol (2013), http://dx.doi.org/10.1016/j.critrevonc.2013.06.011
ARTICLE IN PRESS
ONCH-1762; No. of Pages 10
A.G. Pallis, K.N. Syrigos / Critical Reviews in Oncology/Hematology xxx (2013) xxx–xxx
are linked to the exocytoplasmic surface of the plasma membrane) influence cell growth, differentiation, and the cellular response to the environment [52]. These studies imply that there may be some genetic predisposition for lung cancer development (even in the absence of smoking), however, the literature is still immature and further evidence is required. 6.1. Carcinogen metabolism Polymorphisms for genes encoding the metabolic enzymes that are implicated in carcinogen metabolism might contribute to the variability in individual susceptibility to cancer [53]. A pooled analysis of 14 case–control studies on lung cancer in Caucasian non-smokers (n = 302 cases and 1631 controls) was performed by Hung et al. to study CYP1A1 and GSTM1 genetic polymorphisms [54]. The odds ratio (OR) of lung cancer was 2.99 (95%CI: 1.51–5.91) for the variant CYP1A1 Ile(462)Val polymorphism and this effect was stronger on lung adenocarcinoma (OR 4.85: 95%CI 2.03–11.6). The authors also observed a combined effect of the CYP1A1 Ile(462)Val polymorphism and GSTM1 null genotype with an OR of 4.67 (95%CI: 2.00–10.9). Similarly, a meta-analysis by Chen et al. [55] studied the polymorphism of cytochrome P4501B1 (CYP1B1) codon 432 (rs1056836) (n = 6501 subjects) and found an association between the CYP1B1 codon 432 polymorphism and risk of lung cancer in Caucasians (Leu/Val variant vs. Leu/Leu variant, OR = 1.30; 95% CI = 1.03–1.64). This association was irrespective of smoking status. 6.2. DNA repair genes Polymorphisms of genes involved in DNA repair mechanisms have also been studied as risk factors for lung cancer in never-smokers. Zhou et al. [56] observed an increase in lung cancer risk in subjects with the Arg399Gln polymorphism of the XRCC1 gene (OR 2.4, 95% CI: 1.2–5.0). When this polymorphism was present in combination with two polymorphisms of the ERCC2 gene (Asp312Asn and Lys751Gln), the adjusted ORs versus wild genotype were 5.2 (95% CI, 1.7–16.6) for never-smokers. A higher lung cancer risk was also identified among non-smokers carrying the variant ERCC2 allele of Asp312Asn (OR = 2.10; 95% CI: 1.22–3.64) [57]. Finally, a study by Gorlova et al. demonstrated that suboptimal DNA repair capacity level (below the control median) was associated with a significantly increased lung cancer risk in never smokers (OR 1.92; 95% CI: 1.3–2.9) [58]. The risk was 3.38-fold higher for individuals with DNA repair capacity below the first quartile compared with individuals with DNA repair capacity above the third quartile. 6.3. Inflammatory response genes It is reported that chronic inflammation plays a role in tumor initiation and promotion most probably through
5
altered cytokine levels [59]. Oh et al. using data form three case-control studies demonstrated that the IL10 gene SNP rs1800871 is associated with lung cancer among never smokers (OR: 2.5, 95% CI: 1.3–5.1) while the TNF gene rs1799964 SNP is inversely associated with smoking-related cancer (CC vs. CT + TT OR: 0.36, 95% CI: 0.17–0.77) [59]. So far numerous studies have been reported studying the role of candidate susceptibility SNPs in LCNS, and those involved in carcinogen metabolism, DNA repair or in inflammatory response. However, their role remains still to be fully clarified and further studies are needed to better understand the role for genetic factors in LCNS development.
7. Acquired somatic mutations Several acquired molecular abnormalities have been observed in lung cancer that contribute to tumorigenesis and might be necessary for tumor maintenance. Some of these are majorly observed in patients with a never-smoking history. 7.1. EGFR EGFR pathway is one of the most well documented and investigated pathways in NSCLC. EGFR is over-expressed in the majority of NSCLC and plays a strong stimulatory effect on cell proliferation, survival, migration and angiogenesis [60]. The discovery of EGFR mutations in 2004 [61,62] and their direct association with response to gefitinib and erlotinib has highlighted the importance of this signaling pathway for many NSCLCs. Most common of these are inframe deletions in exon 19 (codons 746–750) and a missense mutation leading to a substitution of arginine for leucine at codon 858 (L858R) [63]. The incidence of these mutations is significantly higher in females compared with males, in Asians versus Caucasians, in adenocarcinoma histology versus other histological types and in non-smokers [64]. Mutations are inversely associated with smoking history with a higher prevalence in never-smokers [65]. The incidence of these mutations is approximately 17% in the Western population [66] while in the Asian population it is approximately 60% [67]. As pack-years smoked increases, the probability of detecting an activating EGFR mutation falls. One large nonconsecutive series of both East Asian and Western patients reported no mutations in patients with more than a 75 packyear exposure, with a prevalence of 51% in never smokers, 19% in former smokers, and 4% in current smokers [68]. 7.2. HER-2 The HER2 gene, encodes a transmembrane receptor that is involved in the regulation of cell proliferation. The HER2 receptor is a member of the Epidermal Growth Factor receptor (EGFR) family, does not have a known ligand, and is activated by homodimerization with other HER2 receptors or by heterodimerization, preferentially with either EGFR
Please cite this article in press as: Pallis AG, Syrigos KN. Lung cancer in never smokers: Disease characteristics and risk factors. Crit Rev Oncol/Hematol (2013), http://dx.doi.org/10.1016/j.critrevonc.2013.06.011
ONCH-1762; No. of Pages 10
6
ARTICLE IN PRESS A.G. Pallis, K.N. Syrigos / Critical Reviews in Oncology/Hematology xxx (2013) xxx–xxx
or HER3 [69]. The activation of HER2 triggers the initiation of the PI3K/AKT/mTOR and RAS/RAF/MEK cascades [69]. Heterodimers between EGFR and HER2 are more stable than complexes containing other Erb family members [70] and thus Her-2 plays a major role in the EGFR signaling network [71]. HER-2 is overexpressed in approximately 20% of non-small cell lung cancer (NSCLC) cases, while gene amplification and gene mutations are observed in approximately 2% of NSCLC patients [72]. These mutations usually involve in frame insertions in exon 20 and are more frequently observed in never-smokers, in women, and in Asian patients, and are more common in adenocarcinoma histology than in other histological types [73]. HER-2 mutations are mutually exclusive with EGFR or KRAS mutations and result in the constitutive activation of the receptor [74]. HER-2 mutations have been associated with response to treatment with tyrosine kinase inhibitors (TKIs) that target both EGFR and HER2 (e.g., lapatinib) but not to those that target EGFR alone [75]. 7.3. ALK rearrangements ALK gene encodes a transmembrane receptor tyrosine kinase that is part of the insulin receptor superfamily that is not expressed in normal lung tissue [76]. In lung cancer activating mutations of the ALK do not occur and the gene is activated by the formation of fusion genes [77]. Somatic rearrangement within the small arm of chromosome 2p leads to the formation of EML4-ALK fusion gene. This fusion gene results in constitutive activation of the kinase domain [77]. This rearrangement is relatively rare and observed in 3–7% of the original reporting series [77]. An increased incidence is observed in patients with adenocarcinoma histology, younger patients, and in people who are never-smokers or ex light-smokers (≤15 pack-years) [78]. EML4-ALK translocation is considered as mutually exclusive to other genotypes (EGFR mutation, and KRAS mutation) [78]. Presence of ALK rearrangement in lung tumors is related to significant response and clinical benefit with the use of ALK TKIs, such as crizotinib [79]. 7.4. ROS1 rearrangements ROS1 is a receptor tyrosine kinase of the insulin receptor family and several publications demonstrated that ROS1 gene is involved in chromosomal translocations in glioblastoma [80] and more recently lung cancer [81]. A study by Rikova et al. identified ROS1 fusions as potential driver mutations in NSCLC cell line and also in a NSCLC patient tumor sample (CD74-ROS1) [81]. These fusions lead to constitutive kinase activity and are associated with sensitivity in vitro to TKIs. A recent report by Bergethon et al. screened 1073 lung cancer patients and 18 (1.7%) were identified with ROS1 rearrangements by FISH, and 31 (2.9%) were ALK rearranged [82]. ROS1 rearrangements were more frequently observed in
younger and never-smokers patients (each p-value < 0.001). All of the ROS1-positive tumors were adenocarcinomas, with a tendency toward higher grade.
8. Prognostic and predictive role Several reports have demonstrated that LCNS patients have better survival compared with ever smoker patients [14,25,83]. Placebo controlled phase III trials of EGFR TKIs (erlotinib BR.21 study [84]; gefitinib ISEL study [85]) demonstrated that never-smoker patients in the placebo arm had a trend toward better outcome compared to ever smokers (median overall survival for never-smokers vs. ever smokers: BR.21 5.6 months vs. 4.6 months; ISEL 6.1 vs. 4.9), although this difference was not statistically significant. A retrospective analysis of 26,957 patients with NSCLC demonstrated a significant difference in median OS between never-smoker and ever smoker patients (median OS: never vs. ever-smokers: 30.0 months vs. 19.0 months, respectively; p-value < 0.0001). Multivariate analysis identified good PS, never smoker status, early stage, female gender, squamous cell carcinoma histology, and treatment as independent favorable prognostic factors [86]. Another retrospective review of 12,000 patients in USA demonstrated a worse prognosis for ever smokers vs. never smokers (HR: 1.09; p-value = 0.045) [14]. Other smaller series had similar results [9,20,21,87], although this observation was not confirmed in all studies [12,19,22]. However, it should be noted that although these differences in survival could represent differences in the biology of lung cancer between ever and never-smokers, they could also simply reflect the higher proportion of early stage disease in the never-smokers group which impacts prognosis [87]. On the other hand a prospective study in 2010 NSCLC patients with advanced disease (stage IIIB/IV) who completed a smoking questionnaire demonstrated that neversmokers had longer OS compared with ever-smokers (17.8 months vs. 11.3 months, respectively; p-value < 0.001) [88]. More cigarette smoking (as measured in pack-years) was associated with decreased survival. Similarly a retrospective analysis of a phase III trial by Scagliotti et al. observed that smoking was a negative prognostic factor for both OS (p-value < 0.05) and PFS (p-value < 0.05) [89]. The role of smoking as a predictive factor for treatment outcomes is not widely studied. Three chemotherapy trials in treatment naïve NSCLC patients reported subgroup results by smoking status [90–92]. All of these trials failed to demonstrate any difference in treatment effects between ever and never-smokers. A small retrospective report however, demonstrated a significantly higher response rate for neversmokers patients [93]. Response rates for never vs. former vs. current smokers were 19% vs. 8% vs. 12%, respectively (p-value = 0.004). Never-smoker patients had lower rates of progressive disease (p-value = 0.002) and higher OS rates (p-value < 0.0001).
Please cite this article in press as: Pallis AG, Syrigos KN. Lung cancer in never smokers: Disease characteristics and risk factors. Crit Rev Oncol/Hematol (2013), http://dx.doi.org/10.1016/j.critrevonc.2013.06.011
ONCH-1762; No. of Pages 10
ARTICLE IN PRESS A.G. Pallis, K.N. Syrigos / Critical Reviews in Oncology/Hematology xxx (2013) xxx–xxx
For targeted treatments there is a striking difference in treatment outcome of never-smokers vs. ever-smokers NSCLC patients when treated with EGFR TKIs, but this should be attributed to the higher incidence of EGFR activating mutations in the never-smokers population [6].
9. Conclusions The incidence of LCNS is expected to increase due to extensive and successful smoking prevention and cessation programs that are implemented, especially in the developed countries. Strong epidemiological data support a causal association between second-hand smoking and lung cancer development. Similarly, radon exposure, indoor coal burning and oil vapors, hormonal factors, air pollution, viral factors and dietary factors are reported as causes of lung caner in never smokers. Although there are a few published reports in the field familial aggregation and genetic susceptibility studies did not focus on LCNS as a separate entity. Similarly although GWAS studies demonstrated some very important observations, they primarily focused on genes associated with tobacco carcinogen metabolism which may be less important in LCNS. GWAS studies in never-smokers specifically could be very important and reveal genetic loci that predispose to LCNS. A very important finding however, is that a big percentage of these patients present with driver aberrations (EGFR and HER2 mutations, ALK and ROS1 rearrangements). These alterations could potentially be treated with targeted agents already in clinical use or under development. Impressive results have already been observed with erlotinib and gefitinib in EGFR mutated patients [94] and with crizotinib in patients with ALK and ROS1 rearrangements [79,82]. Therefore understanding the biology of LCNS and the distinct clinical and pathology characteristics of this disease is crucial.
Conflict of interest Both authors have no conflict of interest.
Reviewers Filippo de Marinis, MD, Director, San Camillo Hospital, 1st Puòmonary Oncological Division, Via Goffredo Mameli 3/1, IT-16122 Genova, Italy. Robert Pirker, MD, Medical University Vienna, Department of Medicine I, Währinger Gürtel 18, Vienna, Austria.
References [1] Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA: A Cancer Journal for Clinicians 2011;61:69–90.
7
[2] Doll R, Hill AB. Smoking and carcinoma of the lung; preliminary report. British Medical Journal 1950;2:739–48. [3] Doll R, Peto R, Boreham J, Sutherland I. Mortality in relation to smoking: 50 years’ observations on male British doctors. BMJ 2004;328:1519. [4] The Health Consequences of Smoking: A Report of the Surgeon General. Washington, DC: U.S. Department of Health and Human Services, Public Health Service, Centers for Disease Control and Prevention; 2004. Available at: wwwcdcgov/tobacco/sgr/sgr 2004/indexhtm [CDC Publication No. 7829]. [5] Samet JM. Health benefits of smoking cessation. Clinics in Chest Medicine 1991;12:669–79. [6] Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin–paclitaxel in pulmonary adenocarcinoma. New England Journal of Medicine 2009;361:947–57. [7] Sun S, Schiller JH, Gazdar AF. Lung cancer in never smokers—a different disease. Nature Reviews Cancer 2007;7:778–90. [8] Toh CK, Gao F, Lim WT, et al. Never-smokers with lung cancer: epidemiologic evidence of a distinct disease entity. Journal of Clinical Oncology 2006;24:2245–51. [9] Yano T, Miura N, Takenaka T, et al. Never-smoking nonsmall cell lung cancer as a separate entity: clinicopathologic features and survival. Cancer 2008;113:1012–8. [10] Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. International Journal of Cancer 2010;127:2893–917. [11] Scagliotti GV, Longo M, Novello S. Nonsmall cell lung cancer in never smokers. Current Opinion in Oncology 2009;21:99–104. [12] Toh CK, Wong EH, Lim WT, et al. The impact of smoking status on the behavior and survival outcome of patients with advanced non-small cell lung cancer: a retrospective analysis. Chest 2004;126:1750–6. [13] Wakelee HA, Chang ET, Gomez SL, et al. Lung cancer incidence in never smokers. Journal of Clinical Oncology 2007;25:472–8. [14] Zell JA, Ou SH, Ziogas A, Anton-Culver H. Epidemiology of bronchioloalveolar carcinoma: improvement in survival after release of the 1999 WHO classification of lung tumors. Journal of Clinical Oncology 2005;23:8396–405. [15] Radzikowska E, Glaz P, Roszkowski K. Lung cancer in women: age, smoking, histology, performance status, stage, initial treatment and survival. Population-based study of 20 561 cases. Annals of Oncology 2002;13:1087–93. [16] Boffetta P, Jarvholm B, Brennan P, Nyren O. Incidence of lung cancer in a large cohort of non-smoking men from Sweden. International Journal of Cancer 2001;94:591–3. [17] Peto R, Darby S, Deo H, Silcocks P, Whitley E, Doll R. Smoking, smoking cessation, and lung cancer in the UK since 1950: combination of national statistics with two case–control studies. BMJ 2000;321:323–9. [18] Antony GK, Bertino E, Franklin M, Otterson GA, Dudek AZ. Small cell lung cancer in never smokers: report of two cases. Journal of Thoracic Oncology 2010;5:747–8. [19] Subramanian J, Velcheti V, Gao F, Govindan R. Presentation and stage-specific outcomes of lifelong never-smokers with non-small cell lung cancer (NSCLC). Journal of Thoracic Oncology 2007;2: 827–30. [20] Santoro IL, Ramos RP, Franceschini J, Jamnik S, Fernandes AL. Non-small cell lung cancer in never smokers: a clinical entity to be identified. Clinics (Sao Paulo) 2011;66:1873–7. [21] Bryant A, Cerfolio RJ. Differences in epidemiology, histology, and survival between cigarette smokers and never-smokers who develop non-small cell lung cancer. Chest 2007;132:185–92. [22] Meguid RA, Hooker CM, Harris J, et al. Long-term survival outcomes by smoking status in surgical and nonsurgical patients with non-small cell lung cancer: comparing never smokers and current smokers. Chest 2010;138:500–9. [23] Sobue T, Yamamoto S, Hara M, Sasazuki S, Sasaki S, Tsugane S. Cigarette smoking and subsequent risk of lung cancer by histologic
Please cite this article in press as: Pallis AG, Syrigos KN. Lung cancer in never smokers: Disease characteristics and risk factors. Crit Rev Oncol/Hematol (2013), http://dx.doi.org/10.1016/j.critrevonc.2013.06.011
ONCH-1762; No. of Pages 10
8
ARTICLE IN PRESS A.G. Pallis, K.N. Syrigos / Critical Reviews in Oncology/Hematology xxx (2013) xxx–xxx
[24]
[25]
[26]
[27]
[28]
[29]
[30]
[31]
[32]
[33]
[34]
[35]
[36]
[37]
[38]
[39]
[40]
[41]
[42]
[43]
type in middle-aged Japanese men and women: the JPHC study. International Journal of Cancer 2002;99:245–51. Liu NS, Spitz MR, Kemp BL, et al. Adenocarcinoma of the lung in young patients: the M. D. Anderson experience. Cancer 2000;88:1837–41. Nordquist LT, Simon GR, Cantor A, Alberts WM, Bepler G. Improved survival in never-smokers vs current smokers with primary adenocarcinoma of the lung. Chest 2004;126:347–51. Thun MJ, Henley SJ, Burns D, Jemal A, Shanks TG, Calle EE. Lung cancer death rates in lifelong nonsmokers. Journal of the National Cancer Institute 2006;98:691–9. Thun MJ, Hannan LM, Adams-Campbell LL, et al. Lung cancer occurrence in never-smokers: an analysis of 13 cohorts and 22 cancer registry studies. PLOS Medicine 2008;5:e185. Etzel CJ, Lu M, Merriman K, Liu M, Vaporciyan A, Spitz MR. An epidemiologic study of early onset lung cancer. Lung Cancer 2006;52:129–34. Thomas JL, Guo H, Carmella SG, et al. Metabolites of a tobaccospecific lung carcinogen in children exposed to secondhand or thirdhand tobacco smoke in their homes. Cancer Epidemiology, Biomarkers and Prevention 2011;20:1213–21. Oberg M, Jaakkola MS, Woodward A, Peruga A, Pruss-Ustun A. Worldwide burden of disease from exposure to second-hand smoke: a retrospective analysis of data from 192 countries. Lancet 2011;377:139–46. Vineis P, Hoek G, Krzyzanowski M, et al. Lung cancers attributable to environmental tobacco smoke and air pollution in non-smokers in different European countries: a prospective study. Environmental Health 2007;6:7. Hackshaw AK, Law MR, Wald NJ. The accumulated evidence on lung cancer and environmental tobacco smoke. BMJ 1997;315: 980–8. International Agency for Research on Cancer. Tobacco smoke and involuntary smoking. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans 2004;83:1–1438. Vineis P, Alavanja M, Buffler P, et al. Tobacco and cancer: recent epidemiological evidence. Journal of the National Cancer Institute 2004;96:99–106. Kawai H, Ishii A, Washiya K, et al. Estrogen receptor alpha and beta are prognostic factors in non-small cell lung cancer. Clinical Cancer Research 2005;11:5084–9. Wu CT, Chang YL, Shih JY, Lee YC. The significance of estrogen receptor beta in 301 surgically treated non-small cell lung cancers. Journal of Thoracic and Cardiovascular Surgery 2005;130:979–86. Ali G, Donati V, Loggini B, et al. Different estrogen receptor beta expression in distinct histologic subtypes of lung adenocarcinoma. Human Pathology 2008;39:1465–73. Stabile LP, Davis AL, Gubish CT, et al. Human non-small cell lung tumors and cells derived from normal lung express both estrogen receptor alpha and beta and show biological responses to estrogen. Cancer Research 2002;62:2141–50. Omoto Y, Kobayashi Y, Nishida K, et al. Expression, function, and clinical implications of the estrogen receptor beta in human lung cancers. Biochemical and Biophysical Research Communications 2001;285:340–7. Samet JM, Avila-Tang E, Boffetta P, et al. Lung cancer in never smokers: clinical epidemiology and environmental risk factors. Clinical Cancer Research 2009;15:5626–45. Tokuhata GK, Lilienfeld AM. Familial aggregation of lung cancer in humans. Journal of the National Cancer Institute 1963;30: 289–312. Amos CI, Xu W, Spitz MR. Is there a genetic basis for lung cancer susceptibility. Recent Results in Cancer Research 1999;151: 3–12. Matakidou A, Eisen T, Houlston RS. Systematic review of the relationship between family history and lung cancer risk. British Journal of Cancer 2005;93:825–33.
[44] Hung RJ, McKay JD, Gaborieau V, et al. A susceptibility locus for lung cancer maps to nicotinic acetylcholine receptor subunit genes on 15q25. Nature 2008;452:633–7. [45] Amos CI, Wu X, Broderick P, et al. Genome-wide association scan of tag SNPs identifies a susceptibility locus for lung cancer at 15q251. Nature Genetics 2008;40:616–22. [46] McKay JD, Hung RJ, Gaborieau V, et al. Lung cancer susceptibility locus at 5p15.33. Nature Genetics 2008;40:1404–6. [47] Hsiung CA, Lan Q, Hong YC, et al. The 5p15.33 locus is associated with risk of lung adenocarcinoma in never-smoking females in Asia. PLOS Genetics 2010:6. [48] Wang Y, Broderick P, Webb E, et al. Common 5p15.33 and 6p21.33 variants influence lung cancer risk. Nature Genetics 2008;40:1407–9. [49] Sasaki T, Gan EC, Wakeham A, Kornbluth S, Mak TW, Okada H. HLA-B-associated transcript 3 (Bat3)/Scythe is essential for p300-mediated acetylation of p53. Genes and Development 2007;21:848–61. [50] Wang YC, Hsu HS, Chen TP, Chen JT. Molecular diagnostic markers for lung cancer in sputum and plasma. Annals of the New York Academy of Sciences 2006;1075:179–84. [51] Li Y, Sheu CC, Ye Y, et al. Genetic variants and risk of lung cancer in never smokers: a genome-wide association study. Lancet Oncology 2010;11:321–30. [52] Blackhall FH, Merry CL, Davies EJ, Jayson GC. Heparan sulfate proteoglycans and cancer. British Journal of Cancer 2001;85:1094–8. [53] Vineis P, Bartsch H, Caporaso N, et al. Genetically based N-acetyltransferase metabolic polymorphism and low-level environmental exposure to carcinogens. Nature 1994;369:154–6. [54] Hung RJ, Boffetta P, Brockmoller J, et al. CYP1A1 and GSTM1 genetic polymorphisms and lung cancer risk in Caucasian nonsmokers: a pooled analysis. Carcinogenesis 2003;24:875–82. [55] Chen B, Qiu LX, Li Y, et al. The CYP1B1 Leu432Val polymorphism contributes to lung cancer risk: evidence from 6501 subjects. Lung Cancer 2010;70:247–52. [56] Zhou W, Liu G, Miller DP, et al. Polymorphisms in the DNA repair genes XRCC1 and ERCC2, smoking, and lung cancer risk. Cancer Epidemiology, Biomarkers and Prevention 2003;12:359–65. [57] Qian B, Zhang H, Zhang L, Zhou X, Yu H, Chen K. Association of genetic polymorphisms in DNA repair pathway genes with non-small cell lung cancer risk. Lung Cancer 2011;73:138–46. [58] Gorlova OY, Weng SF, Zhang Y, Amos CI, Spitz MR, Wei Q. DNA repair capacity and lung cancer risk in never smokers. Cancer Epidemiology, Biomarkers and Prevention 2008;17:1322–8. [59] Oh SS, Chang SC, Cai L, et al. Single nucleotide polymorphisms of 8 inflammation-related genes and their associations with smoking-related cancers. International Journal of Cancer 2010;127: 2169–82. [60] Ciardiello F, Tortora G. EGFR antagonists in cancer treatment. New England Journal of Medicine 2008;358:1160–74. [61] Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of nonsmall-cell lung cancer to gefitinib. New England Journal of Medicine 2004;350:2129–39. [62] Paez JG, Janne PA, Lee JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 2004;304:1497–500. [63] Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nature Reviews Cancer 2007;7:169–81. [64] Pallis AG, Fennell DA, Szutowicz E, Leighl NB, Greillier L, Dziadziuszko R. Biomarkers of clinical benefit for anti-epidermal growth factor receptor agents in patients with non-small-cell lung cancer. British Journal of Cancer 2011;105:1–8. [65] 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 Research 2004;64:8919–23.
Please cite this article in press as: Pallis AG, Syrigos KN. Lung cancer in never smokers: Disease characteristics and risk factors. Crit Rev Oncol/Hematol (2013), http://dx.doi.org/10.1016/j.critrevonc.2013.06.011
ONCH-1762; No. of Pages 10
ARTICLE IN PRESS A.G. Pallis, K.N. Syrigos / Critical Reviews in Oncology/Hematology xxx (2013) xxx–xxx
[66] Rosell R, Moran T, Queralt C, et al. Screening for epidermal growth factor receptor mutations in lung cancer. New England Journal of Medicine 2009;361:958–67. [67] Fukuoka M, Wu YL, Thongprasert S, et al. Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia (IPASS). Journal of Clinical Oncology 2011;29:2866–74. [68] Pham D, Kris MG, Riely GJ, et al. Use of cigarette-smoking history to estimate the likelihood of mutations in epidermal growth factor receptor gene exons 19 and 21 in lung adenocarcinomas. Journal of Clinical Oncology 2006;24:1700–4. [69] Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nature Reviews Molecular Cell Biology 2001;2:127–37. [70] Lenferink AE, Pinkas-Kramarski R, van de Poll ML, et al. Differential endocytic routing of homo- and hetero-dimeric ErbB tyrosine kinases confers signaling superiority to receptor heterodimers. EMBO Journal 1998;17:3385–97. [71] Tzahar E, Waterman H, Chen X, et al. A hierarchical network of interreceptor interactions determines signal transduction by Neu differentiation factor/neuregulin and epidermal growth factor. Molecular and Cellular Biology 1996;16:5276–87. [72] Stephens P, Hunter C, Bignell G, et al. Lung cancer: intragenic ERBB2 kinase mutations in tumours. Nature 2004;431:525–6. [73] Pao W, Girard N. New driver mutations in non-small-cell lung cancer. Lancet Oncology 2011;12:175–80. [74] Shimamura T, Ji H, Minami Y, et al. Non-small-cell lung cancer and Ba/F3 transformed cells harboring the ERBB2 G776insV G/C mutation are sensitive to the dual-specific epidermal growth factor receptor and ERBB2 inhibitor HKI-272. Cancer Research 2006;66: 6487–91. [75] Wang SE, Narasanna A, Perez-Torres M, et al. HER2 kinase domain mutation results in constitutive phosphorylation and activation of HER2 and EGFR and resistance to EGFR tyrosine kinase inhibitors. Cancer Cell 2006;10:25–38. [76] Camidge DR, Doebele RC. Treating ALK-positive lung cancer—early successes and future challenges. Nature Reviews Clinical Oncology 2012;9:268–77. [77] Soda M, Choi YL, Enomoto M, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 2007;448:561–6. [78] Horn L, Pao W. EML4-ALK: honing in on a new target in non-small-cell lung cancer. Journal of Clinical Oncology 2009;27: 4232–5. [79] Kwak EL, Bang YJ, Camidge DR, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. New England Journal of Medicine 2010;363:1693–703. [80] Birchmeier C, O’Neill K, Riggs M, Wigler M. Characterization of ROS1 cDNA from a human glioblastoma cell line. Proceedings of the National Academy of Sciences of the United States of America 1990;87:4799–803. [81] Rikova K, Guo A, Zeng Q, et al. Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell 2007;131:1190–203. [82] Bergethon K, Shaw AT, Ignatius Ou SH, et al. ROS1 rearrangements define a unique molecular class of lung cancers. Journal of Clinical Oncology 2012;30:863–70. [83] Tammemagi CM, Neslund-Dudas C, Simoff M, Kvale P. Smoking and lung cancer survival: the role of comorbidity and treatment. Chest 2004;125:27–37. [84] Shepherd FA, Rodrigues PJ, Ciuleanu T, et al. Erlotinib in previously treated non-small-cell lung cancer. New England Journal of Medicine 2005;353:123–32. [85] Thatcher N, Chang A, Parikh P, et al. Gefitinib plus best supportive care in previously treated patients with refractory advanced nonsmall-cell lung cancer: results from a randomised, placebo-controlled,
[86]
[87]
[88]
[89]
[90]
[91]
[92]
[93]
[94]
[95]
[96]
[97]
[98]
[99]
[100]
[101]
[102]
[103]
9
multicentre study (Iressa Survival Evaluation in Lung Cancer). Lancet 2005;366:1527–37. Kawaguchi T, Takada M, Kubo A, et al. Performance status and smoking status are independent favorable prognostic factors for survival in non-small cell lung cancer: a comprehensive analysis of 26,957 patients with NSCLC. Journal of Thoracic Oncology 2010;5:620–30. Hanagiri T, Sugio K, Mizukami M, et al. Significance of smoking as a postoperative prognostic factor in patients with non-small cell lung cancer. Journal of Thoracic Oncology 2008;3:1127–32. Janjigian YY, McDonnell K, Kris MG, et al. Pack-years of cigarette smoking as a prognostic factor in patients with stage IIIB/IV nonsmall cell lung cancer. Cancer 2010;116:670–5. Scagliotti GV, De Marinis F, Rinaldi M, et al. The role of histology with common first-line regimens for advanced non-small cell lung cancer: a brief report of the retrospective analysis of a three-arm randomized trial. Journal of Thoracic Oncology 2009;4:1568–71. Syrigos KN, Vansteenkiste J, Parikh P, et al. Prognostic and predictive factors in a randomized phase III trial comparing cisplatin-pemetrexed versus cisplatin–gemcitabine in advanced non-small-cell lung cancer. Annals of Oncology 2010;21:556–61. Kubota K, Kawahara M, Ogawara M, et al. Vinorelbine plus gemcitabine followed by docetaxel versus carboplatin plus paclitaxel in patients with advanced non-small-cell lung cancer: a randomised, open-label, phase III study. Lancet Oncology 2008;9:1135–42. Yang CH, Simms L, Park K, Lee JS, Scagliotti G, Orlando M. Efficacy and safety of cisplatin/pemetrexed versus cisplatin/gemcitabine as first-line treatment in East Asian patients with advanced non-small cell lung cancer: results of an exploratory subgroup analysis of a phase III trial. Journal of Thoracic Oncology 2010;5:688–95. Tsao AS, Liu D, Lee JJ, Spitz M, Hong WK. Smoking affects treatment outcome in patients with advanced nonsmall cell lung cancer. Cancer 2006;106:2428–36. Pallis AG, Syrigos KN. Epidermal growth factor receptor tyrosine kinase inhibitors in the treatment of NSCLC. Lung Cancer 2013;80:120–30. Darby S, Hill D, Auvinen A, et al. Radon in homes and risk of lung cancer: collaborative analysis of individual data from 13 European case–control studies. BMJ 2005;330:223. Travis WD, Brambilla E, Muller-Hermelink HK, Harris CC. Pathology and genetics tumours of the lung, pleura, thymus and heart. In: Kleihues P, Sobin LH, editors. WHO Classification of Tumors. Lyon: IARC Press; 2004. Ko YC, Cheng LS, Lee CH, et al. Chinese food cooking and lung cancer in women nonsmokers. American Journal of Epidemiology 2000;151:140–7. Kleinerman RA, Wang Z, Wang L, et al. Lung cancer and indoor exposure to coal and biomass in rural China. Journal of Occupational and Environmental Medicine 2002;44:338–44. Lissowska J, Bardin-Mikolajczak A, Fletcher T, et al. Lung cancer and indoor pollution from heating and cooking with solid fuels: the IARC international multicentre case–control study in Eastern/Central Europe and the United Kingdom. American Journal of Epidemiology 2005;162:326–33. Vineis P, Forastiere F, Hoek G, Lipsett M. Outdoor air pollution and lung cancer: recent epidemiologic evidence. International Journal of Cancer 2004;111:647–52. Cheng YW, Chiou HL, Sheu GT, et al. The association of human papillomavirus 16/18 infection with lung cancer among nonsmoking Taiwanese women. Cancer Research 2001;61:2799–803. Liang HY, Li XL, Yu XS, et al. Facts and fiction of the relationship between preexisting tuberculosis and lung cancer risk: a systematic review. International Journal of Cancer 2009;125:2936–44. Lee MY, Lin KD, Hsiao PJ, Shin SJ. The association of diabetes mellitus with liver, colon, lung, and prostate cancer is independent of hypertension, hyperlipidemia, and gout in Taiwanese patients. Metabolism – Clinical and Experimental 2012;61:242–9.
Please cite this article in press as: Pallis AG, Syrigos KN. Lung cancer in never smokers: Disease characteristics and risk factors. Crit Rev Oncol/Hematol (2013), http://dx.doi.org/10.1016/j.critrevonc.2013.06.011
ONCH-1762; No. of Pages 10
10
ARTICLE IN PRESS A.G. Pallis, K.N. Syrigos / Critical Reviews in Oncology/Hematology xxx (2013) xxx–xxx
[104] Gorlova OY, Weng SF, Hernandez L, Spitz MR, Forman MR. Dietary patterns affect lung cancer risk in never smokers. Nutrition and Cancer 2011;63:842–9.
Biographies Athanasios Pallis, M.D., M.Sc., Ph.D. graduated from School of Medicine, University of Crete, in 1997. He was trained in Medical Oncology at the Medical Oncology Department of the University General Hospital of Heraklion, Crete (University of Crete). In 2003 he got his PhD degree from the School of Medicine, in University of Crete. He has served as an EORTC fellow in 2008–2009. Dr Pallis has served as a reviewer for several international journals. Ha has published more than 80 peer-reviewed, international articles with more than 1200 citations and contributed in 10 chapters in international books. Currently he serves on the editorial board of 3 scientific journals His main fields of interest are Lung Cancer, Targeted Therapies, and Geriatric Oncology. Konstantinos Syrigos, M.D., Ph.D. Assc. Professor and Head, Oncology Unit GPP, Athens School of Medicine. Visiting Professor of Thoracic Oncology, Yale School of Medicine, CT, USA. Assc. Professor Kostas Syrigos graduated from Athens School of Medicine in 1988. He was trained in Internal Medicine at the Laikon University
Hospital (Athens University) and in Medical Oncology at the Hammersmith Hospital (London University). He got his MD thesis with distinction from the Athens School of Medicine, in 1995 and his PhD thesis from the Imperial College of Science, Technology and Medicine, London University, in 2000. He worked as Medical Oncologist Senior Registrar at the Hammersmith and St Bartholomew’s Hospitals, in London and as consultant at the Sotiria General Hospital, in Athens. In 2002 he was appointed Assc. Professor of Oncology in Medicine and Head of the Sotiria Oncology Unit. From 2006 he is also Visiting Professor of Thoracic Oncology at Yale University, CT, USA. His main fields of interest are Targeted Therapies, drugs development as well as Thoracic and Head & Neck oncology. Dr Syrigos participated in several international clinical trials Phase I-IV in lung, colon, head & neck and pancreatic cancer. He is a member of numerous scientific societies, including the European Society of Medical Oncology (ESMO), the European Respiratory Society (ERS), the American Society of Clinical Oncology (ASCO), the American Association of Cancer Research (AACR) and the International Association for the Study of Lung Cancer (IASLC). He is a manuscript reviewer for 18 scientific journals and currently serves on the editorial board of 5 scientific journals. He is the editor of 8 International Scientific Volumes. He has contributed 80 chapters in international books and he is the author of 290 peer-reviewed, international articles, with more than 4900 citations. He is currently sitting as member of the ESMO Translational Research Group and of the MASCC Board of Directors.
Please cite this article in press as: Pallis AG, Syrigos KN. Lung cancer in never smokers: Disease characteristics and risk factors. Crit Rev Oncol/Hematol (2013), http://dx.doi.org/10.1016/j.critrevonc.2013.06.011