Abstract
Lung cancer is the leading cause of cancer-related mortality globally, accounting for 1.8 million deaths in 2020. While the vast majority are caused by tobacco smoking, 15%−25% of all lung cancer cases occur in lifelong never-smokers. The International Agency for Research on Cancer (IARC) has classified multiple agents with sufficient evidence for lung carcinogenesis in humans, which include tobacco smoking, as well as several environmental exposures such as radon, second-hand tobacco smoke, outdoor air pollution, household combustion of coal and several occupational hazards. However, the IARC evaluation had not been stratified based on smoking status, and notably lung cancer in never-smokers (LCINS) has different epidemiological, clinicopathologic and molecular characteristics from lung cancer in ever-smokers. Among several risk factors proposed for the development of LCINS, environmental factors have the most available evidence for their association with LCINS and their roles cannot be overemphasized. Additionally, while initial genetic studies largely focused on lung cancer as a whole, recent studies have also identified genetic risk factors for LCINS. This article presents an overview of several environmental factors associated with LCINS, and some of the emerging evidence for genetic factors associated with LCINS. An increased understanding of the risk factors associated with LCINS not only helps to evaluate a never-smoker’s personal risk for lung cancer, but also has important public health implications for the prevention and early detection of the disease. Conclusive evidence on causal associations could inform longer-term policy reform in a range of areas including occupational health and safety, urban design, energy use and particle emissions, and the importance of considering the impacts of second-hand smoke in tobacco control policy.
Keywords: Lung cancer, never-smoker, risk factor, environmental factor, genetic factor
Introduction
Lung cancer is the leading cause of cancer-related mortality in men and women in 93 and 25 individual countries respectively worldwide (1,2). It has been estimated that in 2020, lung cancer accounted for 1.8 million deaths worldwide, of which more than one-third occurred in China (1,3). Although smoking remains the leading risk factor for lung cancer, 15%−25% of all lung cancer cases globally occur in lifelong never-smokers (4-6), and it has been reported that this proportion is even higher in China, with 43.2% of lung cancer cases (483,040) occurring among never-smokers in 2005 (7). The International Agency for Research on Cancer (IARC) has classified multiple agents with sufficient evidence for lung carcinogenesis in humans as Group 1 carcinogens, including tobacco smoking, as well as several environmental exposures such as radon, second-hand tobacco smoke (SHS), outdoor air pollution, household combustion of coal and several occupational hazards (8-13). However, the IARC evaluation had not been stratified based on smoking status. Current evidence suggests that lung cancer in never-smokers (LCINS) has different epidemiological, clinicopathologic and molecular characteristics from lung cancer in ever-smokers (4,14). When compared with lung cancer in ever-smokers, LCINS occurs more frequently among women, individuals of East Asian descent, and younger age groups, and is predominantly adenocarcinoma in histology subtype (4,14,15). Therefore, a separate evaluation of environmental risk factors for LCINS is warranted. Similarly, while initial genetic association studies largely focused on risk factors for lung cancer as a whole, more recent studies have also identified genetic risk factors for LCINS.
This article reviews epidemiological and mechanistic evidence for several environmental and genetic factors which are found to be associated with LCINS.
Radon
Radon, as a naturally occurring radioactive noble gas, is present throughout the Earth’s crust with varying concentration in different parts of the world. It can readily diffuse through rocks and soil into any air space and accumulate in enclosed areas or unventilated environments. When radon is inhaled by humans, the decay products or radon progenies adhere to the airways of the lungs, where they are thought to continue to emit radioactive alpha particles which potentially induce lung carcinogenesis. Also, based on substantial epidemiological evidence showing a strong and consistent dose-response relationship between increased lung cancer risk and high-level occupational radon exposure amongst underground miners (16,17), radon has been identified as the second strongest risk factor for lung cancer after smoking and the leading risk factor for LCINS worldwide (18). It was classified as a Group 1 carcinogen by IARC in 1988 (8).
However, there are limitations to extrapolating the risk from occupational radon exposure among miners to residential radon exposure in the general population. To assess lung cancer risk attributable to residential radon exposure, large pooled collaborative studies (19-25) and several meta-analyses (26,27) have been conducted to compare the pooled risk estimates with extrapolations from the miner-based risk models (23). Notably, the residual confounding effect of smoking was a major limitation in these studies, as most assessed study populations of both ever-smokers and never-smokers. To establish a link between residential radon and lung cancer risk among never-smokers, we previously carried out a meta-analysis of 24 case-control studies which included 2,341 LCINS cases and 8,967 never-smoker controls, estimating an adjusted excess relative risk (RR) of 0.15 per 100 Bq/m3 [95% confidence interval (95% CI), 0.06−0.25] (28). Our meta-analysis also found that among never-smokers in radon-prone areas, men were at higher risk of lung cancer than women, although the potential mechanisms underlying this difference remain obscure.
There have been some research into the process by which exposure to radon and the subsequent radiation effect leads to lung carcinogenesis among never-smokers. Past research has suggested that radon exposure might induce fusions of the “rearranged during transfection” (RET) gene, caused by an inversion on chromosome 10 (observed in 2/37 lung cancer patients exposed to radiation, compared to 1/240 not exposed, P=0.044) (29). Other research also found that residential radon exposure increased the tumor mutation burden in never-smokers with lung adenocarcinoma (median 4.94 mutations per 1 million base pairs in radon-high group, n=24, vs. 2.62 in radon-low group, n=17, P=0.01) (30). An exploratory analysis of DNA methylation has also found that radon exposure may be associated with some epigenetic changes (31). Studies with larger sample size are warranted to analyze the carcinogenic mechanisms of radon exposure among never-smokers.
SHS
SHS has been one of the most widely studied risk factors for LCINS since tobacco smoke was found to be the major cause of lung cancer (14) and is increasingly factored into tobacco control policy (32). SHS is the combination of the exposure to mainstream smoke exhaled by smokers and side-stream smoke produced directly by tobacco containing products (cigarettes, cigars, and pipes) between the smoking puffs (9). Like tobacco smoke, SHS is a complex mixture of numerous compounds with concentrations varying with time and environment. Both mainstream smoke and side-stream smoke contain a similar range of chemicals, but they differ in the relative proportions and amounts. Many of these chemicals belong to the classes known to be genotoxic and carcinogenic, including the IARC group 1, 2A and 2B carcinogens (9); and among them, polycyclic aromatic hydrocarbon (PAH) and nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone are likely to play major roles (33).
Several meta-analyses have provided important epidemiological evidence of the relationship between SHS and LCINS. Taylor et al.’s meta-analysis of 55 studies evaluated lung cancer risk associated with SHS in never-smoking women exposed to smoking spouses in North America, Asia and Europe, and reported an overall pooled RR of 1.27 (95% CI, 1.17−1.37) (34). Another meta-analysis of 20 case-control studies among non-smoking adults in China found that the risk of lung cancer was significantly higher for those exposed to SHS: odds ratio (OR)=1.64 (95% CI, 1.34−2.01) (35). A recent meta-analysis of 28 case-control studies assessed the association between long-term exposures to SHS and lung cancer incidence in China, and reported that the pooled OR for exposure from parents (2.12; 95% CI, 1.63−2.76) was higher than that for exposure from a spouse (1.15; 95% CI 1.00−1.33) and from work (1.45; 95% CI, 1.31−1.62), suggesting higher lung cancer risk from childhood exposure than adulthood exposure (36). However, at least one meta-analysis reported that there was no evidence of an increased lung cancer risk for SHS exposure in childhood (summary RR=0.91; 95% CI, 0.80−1.05) (37).
In contrast to the evidence pointing to an association between SHS and lung cancer risk, several prospective cohort studies have found no association between SHS and LCINS (38-40). Also, in a recent meta-analysis of 41 studies of non-smoking women in Asia, Europe and North America, Ni et al. (2018) found a significant association between SHS and lung cancer in 34 case-control studies (pooled OR=1.35; 95% CI, 1.17−1.56), but the association did not reach statistical significance in the seven cohort studies (pooled RR=1.17; 95% CI, 0.94−1.44) (41). The discrepancy between the results of case-control and prospective cohort studies warrants reflection on the possibility of selection and recall biases in the previous case-control studies, and suggests that an update of the IARC evaluation may be informative. It is also important to note that all these studies are subject to exposure misclassification, with self-reported ascertainment of SHS exposure, under-reported exposure in the reference group, and potential confounding due to occupational or other environmental factors (9). Future studies using measurement of serum or urinary cotinine levels may help to identify SHS-attributable risk for lung cancer more accurately (42,43). Another emerging complementary approach is to identify exposure to SHS through the analysis of somatic “mutational signatures” observed within LCINS tumors (44,45). Nonetheless, before a conclusive role of SHS in lung cancer development is established, public health strategies should still highlight the importance of reducing SHS exposure in tobacco control policy and zero tolerance position of the World Health Organization (WHO).
As a higher incidence of epidermal growth factor receptor (EGFR) mutations has been observed in LCINS (up to 79%) compared with lung cancer in ever-smokers (10%−20%) (46,47), the relationship between SHS exposure and incidence of EGFR mutations in LCINS is another important area of interest for research. Lee et al. first reported that SHS exposure was inversely associated with the incidence of EGFR mutations in 179 incident cases of LCINS (48), and hypothesized that the mechanism of carcinogenesis in never-smokers exposed to SHS may be similar to that in active smokers, but different from the process involved in mutant-EGFR-dependent carcinogenesis (49). Therefore, SHS, like active smoking, could play a role as a negative predictive factor for EGFR mutations (50). However, there are also other conflicting findings. Liang et al. reported a direct association between SHS exposure duration and rate of EGFR mutation (51). Moreover, an extended multinational cohort including 498 LCINS cases found that increased SHS exposure was significantly associated with EGFR mutations in female never-smokers, but not in male subjects, suggesting a possible role for sex hormones in the development of lung cancer harbouring EGFR mutations (52). Both studies suggested that lung cancer harbouring EGFR mutations may be different between the SHS ever-exposed and never-exposed individuals, and it was hypothesized that among the SHS ever-exposed, a low but prolonged dose of tobacco carcinogens might be an inducing factor for EGFR mutations (53). In view of the relatively small sample sizes of these studies, caution should be taken when interpreting these results, and larger prospective studies are needed to verify the findings.
Air pollution
Air pollution is the contamination of air by harmful gases, dust and smoke, which are detrimental to human health and the planet as a whole. Air pollution represents a global environmental health problem, which is estimated to have contributed to 6.67 million deaths worldwide in 2019, with over 90% of the world’s population living in places where air pollution levels exceed the WHO limits (54). The specific composition of outdoor or ambient air pollution varies across the globe. It depends on the specific climatic conditions and various sources of air pollutants, which arise from many natural processes and anthropogenic activities, including household cooking and heating, biomass burning, transportation, power generation, and industrial processes (12). Among the most abundant and harmful pollutants are sulphur dioxide (SO2), nitrogen dioxide (NO2), carbon monoxide (CO), and ozone (O3), as well as certain microscopic solid or liquid particles suspended in the air, referred to as particulate matter (PM) (55). A table summarizing the most common outdoor and indoor air pollutants of health concern can be found in Turner et al.’s overview article (56). According to the IARC classifications, Group 1 carcinogens for lung cancer in humans include indoor emissions from household combustion of coal, PM from outdoor air pollution, and diesel in engine exhaust, while household biomass fuel (e.g. wood, charcoal) was classified as a Group 2A agent (10,12,57). PM represents a complex and heterogenous mixture of inorganic, organic and biological compounds of various size and composition, including dust, dirt, droplets, soot, PAHs, aromatic amines, bacterial products (endotoxins), and fungi (58). It is classified based on aerodynamic diameter into coarse (2.5−10 µm, PM 10), fine (0.1−2.5 µm, PM 2.5) and ultrafine (0.1 µm, PM 1), and generally arises from different sources and various human activities. Smaller PM is more hazardous to health and PM 2.5 has been widely used by monitoring networks as an indicator of the level of anthropogenic pollutants in ambient or outdoor air pollution (59).
There is substantial epidemiological evidence of the association between long-term exposure to ambient air pollution and lung cancer risk (56). In an analysis of 1,100 lung cancer deaths among 188,699 lifelong never-smokers in the large cohort Cancer Prevention Study-II of the American Cancer Society, each 10 µg/m3 increase in PM 2.5 concentration was associated with a 15%−27% increase in lung cancer mortality (60). A meta-analysis of 18 studies (17 cohorts and one case-control) in 2014 reported a meta-RR of 1.18 (95% CI, 1.00−1.39) for lung cancer risk among never-smokers associated with a 10 µg/m3 increase in exposure to PM 2.5 (61). A sub-group analysis in Huang et al.’s meta-analysis of 17 studies (16 cohorts and one case-control) found that the meta-estimates for lung cancer incidence and mortality associated with a 10 µg/m3 increase in exposure to PM 2.5 for never-smokers were 1.10 (95% CI, 0.76−1.59) and 1.16 (95% CI, 1.02−1.33), respectively (62). A recent study in China with 16,483 lung cancer cases also found an increased risk of lung cancer associated with a 10 µg/m3 increase in 3-year PM 2.5 exposure, reporting a RR of 1.12 (95% CI, 1.00−1.26) which adds to the currently limited evidence from studies conducted in low- and middle-income countries (63). Despite the assessment methods for PM exposure varying widely among these studies (61), they have reported consistent estimates for lung cancer risk or death associated with PM 2.5 exposure.
Measuring indoor or household air pollution (HAP) is complicated due to the interaction of multiple determinants related to the HAP source and the domestic environment (10). HAP is largely caused by incomplete combustion of household fuels for cooking and/or heating, with the most common fuels used in developing countries being biomass fuels (e.g. wood, charcoal) and coal, which are referred to as solid fuels (10). It was estimated that in 2019 about half of the world’s population (3.8 billion people) was still exposed to HAP from solid fuel use (54), and that 3.8 million deaths were attributable to HAP in 2016 (64). Epidemiological evidence of the association of solid fuel use with lung cancer risk has primarily come from case-control studies (65-68), summarized in two IARC monographs evaluating the effects of HAP (10,11). However, there has been substantial heterogeneity across studies (65-68), with pooled ORs of lung cancer risk associated with solid fuel use ranging from 1.17 (95% CI, 1.01−1.37) to 2.31 (95% CI, 1.94−2.76). These case-control studies examined different types of coal or biomass fuels used by both ever- and never-smokers to assess the association between HAP and lung cancer risk, and were limited by their retrospective nature, selection and recall biases, and a potential residual confounding from smoking, all of which may have led to overestimated risks. Even the two cohort studies (69,70) cited in the IARC monographs (10,11) were retrospective studies which only included residents in Xuanwei, Yunnan, where lung cancer mortality rates are among the highest in China (71), and no direct evidence of an association between solid fuel use and lung cancer risk was provided. Therefore, large-scale prospective cohort studies are needed to examine the association between HAP and lung cancer risk.
As PM 2.5 has generally been used to designate the level of anthropogenic pollutants in ambient air pollution (59), studies have explored the mechanism underlying the relationship between PM 2.5 exposure and lung cancer. PM 2.5 exposure has been found to be associated with increased levels of DNA adducts, suggesting that exposure to air pollution may induce a range of effects at the cellular level, including inflammation, DNA damage, and genomic instability, which could potentially drive the carcinogenic process (72,73). Also, PM 2.5 particles contain high concentrations of ubiquitous pollutants, PAHs, which are chemical compounds characterized by the presence of multiple aromatic rings containing only carbon and hydrogen. When activated, these compounds can bind covalently to DNA to form stable or depurinating adducts, and induce oxidative damage (74). Exposure to PAHs via various routes to the body has been shown to be associated with several cancers including lung cancer (75-77). The most studied PAH, benzo[a]pyrene (B[a]P), contributes more than 50% to the total carcinogenic potential, is often used as a marker of PAH exposure (78), and was classified as a Group 1 carcinogens by the IARC in 2010 (79).
Since PAHs are also emitted from cooking oils heated at high temperatures (80), there are growing concerns that exposure to cooking oil fumes could be a causative factor for LCINS, particularly among Asian women, who usually take on the task of domestic cooking (81). One of the major mutagenic compounds in cooking oil fumes, trans-trans-2,4-decadienal (t-t-2,4-DDE), has been shown to reduce the survival rate of human erythroleukemia cells and induce significant oxidative damage to DNA (82). Epidemiological studies conducted among Asian women in mainland China, Taiwan, China, and Singapore have reported that exposure to cooking oil fumes, especially in the absence of fume extractors, was significantly associated with an increased risk of LCINS (83-85). A recent meta-analysis of 23 studies (2 retrospective cohort studies and 21 case-control studies) (86) found that cooking oil fumes are associated with lung cancer risk among women regardless of smoking status, with a pooled OR of 1.98 (95% CI, 1.54−2.54) among non-smokers and 2.00 (95% CI, 1.46−2.74) among ever-smokers. It also suggested that stir frying was associated with an increased risk of lung cancer (pooled OR=1.89, 95% CI, 1.23−2.90). Additionally, different types of cooking oil have been studied, and an increased lung cancer risk has been reported with the use of rapeseed oil (compared with linseed oil) (84) and lard oil (compared with vegetable oil) (87). More research is warranted to investigate the carcinogenic effect of cooking oils.
Occupational hazards
Around 15% of lung cancer cases have been attributed to occupational exposures (88). It has been estimated that in 2000 eight occupational exposures (arsenic, asbestos, beryllium, cadmium, chromium, diesel fumes, nickel, and silica) were responsible for 10% (88,000) and 5% (14,300) of global lung cancer deaths in men and women, respectively (89,90). These agents were all classified as Group 1 carcinogens for lung cancer by the IARC (91). A table listing all carcinogenic agents related to occupational exposure with sufficient or limited evidence in humans is reproduced from Spyratos et al.’s article (92) (Table 1 ). Although there is considerable epidemiological evidence for the association between these occupational exposures and lung cancer, it could be difficult to identify their carcinogenic role due to the probable confounding effect of smoking among most of these occupational groups, and similar studies among never-smokers are very limited (93,94).
Table 1. Carcinogenic agents related to occupational exposure with sufficient (left column) or limited (right column) evidence in humans (92)* .
Sufficient evidence | Limited evidence |
*, Reproduced with permission from Ref. 92, Pioneer Bioscience Pub. Co. | |
1. Aluminum production | 1. Acid mists, strong inorganic |
2. Arsenic and inorganic arsenic compounds
3. Asbestos (all forms) 4. Beryllium and beryllium compounds |
2. Art glass, glass containers and pressed ware (manufacture of) |
5. Bis (chloromethyl) ether; chloromethyl methyl ether (technical grade)
6. Cadmium and cadmium compounds |
3. Biomass fuel (primarily wood), indoor emissions from household combustion of |
7. Chromium (VI) compounds
8. Coal, indoor emissions from household combustion 9. Coal gasification |
4. Bitumens, occupational exposure to oxidized bitumens and their emissions during roofing |
10. Coal-tar pitch
11. Coke production 12. Engine exhaust, diesel 13. Hematite mining (underground) |
5. Bitumens, occupational exposure to hard bitumens and their emissions during mastic asphalt work |
14. Iron and steel founding | 6. Carbon electrode manufacture |
15. MOPP (vincristine-prednisone-nitrogen mustard procarbazine mixture)
16. Nickel compounds |
7. alpha-Chlorinated toluenes and benzoyl chloride (combined exposures) |
17. Painting
18. Plutonium |
8. Cobalt metal with tungsten carbide |
19. Radon-222 and its decay products | 9. Creosotes |
20. Rubber production industry | 10. Frying, emissions from high temperature |
21. Silica dust, crystalline
22. Soot 23. Sulfur mustard |
11. Insecticides, non-arsenical (occupational exposures in spraying and application) |
24. Tobacco smoke, second-hand | 12. Printing processes |
25. Tobacco smoking
26. X-radiation, gamma-radiation |
13. 2,3,7,8-Tetrachlorodibenzopara-dioxin |
14. Welding fumes |
Occupational exposures to asbestos has been known to be of concern for lung carcinogenesis since the 1930s (95,96), and exposure is associated with five-fold higher risk of lung cancer (97). As a result, there have been extensive studies of the associated epidemiological, clinical, biological and medico-legal aspects. Asbestos is a generic term referring to six naturally occurring silicate minerals that can be generally grouped into the serpentine and amphibole class. All minerals are made up of long and thin fibrous crystals, and each fibre contains many microscopic “fibrils” that can be released into the atmosphere and easily inhaled by humans (98). Long-term inhalation of asbestos fibres can lead to various diseases including asbestosis. It also increases risk for asbestos-related lung cancer and mesothelioma, and the risk is dependent on both fibre type and level of exposure. Asbestos exposure is the only known cause of mesothelioma, but asbestos exposure causes six times more cases of lung cancer than mesothelioma (99,100).
Arsenic is a metallic element which is highly toxic in its inorganic form, and arsenic exposure affects more than 150 million people worldwide (98). It is found naturally throughout the environment in the air, water and soil, and long-term exposure to arsenic is known to cause cancer (101). Based on epidemiological evidence from ecological, case-control and cohort studies since the 1950s, lung cancer has been observed to be associated with arsenic intoxicated patients (102) via exposure to inhaled arsenic or ingestion of arsenic in drinking-water (98). According to a study in the United States (U.S.), arsenic exposure may have a significantly greater effect on lung cancer incidence than previously expected and may contribute to over 5,000 lung cancer cases (after adjusting for smoking and income) in the U.S. per year (103).
Diesel fumes from motor engine exhaust, another important source of PAHs (104), are also one of the most prevalent occupational lung carcinogens worldwide, especially in developed countries (105). Their ubiquitous nature also renders diesel fumes a public health risk for the general population. Diesel fumes were reclassified by the IARC from Group 2A (probable human carcinogen) to Group 1 (definite human carcinogen) in 2012 (106), based on re-analysis of data from the Diesel Exhaust in Miners study: a cohort mortality study and a nested case-control study (107,108). Although a critical review has pointed out several methodological flaws of the re-analysis and claimed that a causal link between diesel exhaust and lung cancer risk was not definite (109), aggregated evidence from experimental, epidemiologic, and mechanistic research provides strong evidence that diesel exhaust causes lung cancer in humans (106,110).
Other occupational exposures which are Group 1 carcinogens for lung cancer include exposure to PAHs as reported by millions of workers in industries like aluminium production, coal gasification and coke production (75,111), exposure to some environmental heavy metals such as arsenic, beryllium, chromium and nickel (91) in industries like wood preservation, ceramic production, stainless steel production and glass work, respectively (112), and exposure to crystalline silica, which can occur for millers, ceramic workers, glassmakers and granite workers (113). Although there is sufficient evidence for the carcinogenicity of these exposures in humans according to the IARC (13), the exact mechanism of pathogenesis leading to lung cancer has not been fully understood. Other occupations which have been associated with lung cancer include bartenders (likely due to SHS exposure), carpenters, bricklayers, painters, electricians, roofers (112) and hairdressers (114).
Familial aggregation
Since Tokuhata and Lilienfeld provided the first epidemiologic evidence for familial aggregation of lung cancer in 1963 (115), several subsequent studies have reported increased familial risk for lung cancer. Inherited susceptibility was found to contribute to increased familial risk (116-119). It is, however, extremely complex to disentangle the specific contribution of genetic predisposition from the shared environmental exposures within family members, with smoking being the most important shared risk factor.
In Matakidou et al.’s meta-analysis that included 11 case-control studies of never-smokers (116), a family history of lung cancer in one or more affected relatives was associated with an increased risk of LCINS (RR=1.51; 95% CI, 1.11−2.06). Lissowska et al.’s meta-analysis of 20 European studies (18 case-control and 2 cohort studies that provided estimates for a subgroup analysis of non-smokers) also showed that a family history of lung cancer was associated with increased lung cancer risk (OR=1.40; 95% CI, 1.17−1.68) (118). In a pooled analysis of 24 case-control studies from the International Lung Cancer Consortium with 3,301 LCINS cases, it was found that lung cancer risk among never-smokers was associated with a familial history of lung cancer in a sibling (OR=1.44; 95% CI, 1.07−1.93) but not with a history of lung cancer in a parent (117). In another recent meta-analysis that included 23 studies (including Asian/Western and cohort as well as case-control studies), the pooled summary estimate for familial risk of lung cancer in a subgroup analysis of never-smokers was 1.72 (95% CI, 1.39−2.14), and the estimate was greater in Asian studies (2.62; 95% CI, 2.25−3.06) as compared with Western studies (1.24; 95% CI, 1.05−1.47) (120). These large-scale analyses, which included some studies that overlapped, demonstrated a similar risk estimate (about 1.5-fold) of lung cancer among never-smokers with a family history of the disease, and the risk was generally higher for Asians and for those with an affected sibling than other first-degree relatives. While genomic research on lung cancer is developing and the role of genetic factors in lung carcinogenesis remains to be fully defined, family history assessment is still valuable and might additionally provide indirect information on shared environmental risk exposures.
Genetic factors
Genetic susceptibility plays an important role in lung cancer risk for both ever-smokers and never-smokers (121). With the advent of new genomics technologies, research on lung cancer has been expanding rapidly, with particular focus on two different areas: identifying inherited genetic variants that are associated with lung cancer risk, and investigating molecular tumour characteristics to elucidate carcinogenic mechanisms and/or predict response to treatment. Multiple studies have focused on never-smokers to gain insights into primary lung carcinogenesis rather than smoking-driven carcinogenesis (122), and potentially advance future development of personalised screening, diagnosis and treatment approaches for LCINS (123).
In the past decade, analyses based on large-scale multistage genome-wide association studies (GWAS) have used robust approaches to systematically test for association between lung cancer risk and hundreds of thousands of inherited genetic variants (often called “single-nucleotide polymorphisms” or SNPs). To date, these studies have identified 45 genetic regions (“susceptibility loci”) associated with lung cancer in different ethnic populations, and several of these loci were associated with LCINS (123). The first lung cancer GWAS were published in 2008, when three independent studies (124-126) consistently found strong associations of lung cancer with a genetic region (15q25) that contained three nicotinic acetylcholine receptor subunits (CHRNA5,CHRNA3, andCHRNB4), and was also found to be associated with tobacco consumption. A subsequent GWAS reported two other loci (5p15 and 6p21) associated with lung cancer risk, which were found to have no association with smoking behavior (127). In 2010, the first GWAS on LCINS found an association with the 13q31.3 region (128). Since then, several other loci associated with LCINS have been identified (with evidence for multiple independent risk variants in some regions) (123,129,130), including some loci that were also found to be associated in the original lung cancer GWAS (131-136). Several LCINS GWAS were carried out in Asian populations, so the underlying risk variants may also have different frequencies in populations with other ancestry (123,137).
Notably, the genetic associations detected to date only confer modest increases in LCINS risk (with per-allele OR of at most 1.30) (136). As for other cancers, so-called “polygenic risk scores” (PRS) can be constructed to combine risk information across many genetic regions to capture inherited lung cancer predisposition more fully. Dai et al. identified 19 susceptibility loci significantly associated with non-small cell lung cancer (NSCLC) risk (six of them were newly identified in the study) and used them as well as other known susceptibility loci to construct a PRS (138). Among 95,408 participants of the China Kadoorie Biobank cohort, the PRS was significantly associated with lung cancer incidence, with an adjusted hazard ratio of 1.96 (95% CI, 1.53−2.51) for individuals with the 10% highest compared to those with the 10% lowest PRS. Another study constructed a different PRS and evaluated it in the United Kingdom Biobank data (n=335,931), estimating an OR of 2.39 (95% CI, 1.92−3.00) for individuals with the 10% highest compared to those with the 10% lowest PRS (139). Both studies provide evidence that GWAS-derived PRS might support risk stratification and help identify groups at high risk of lung cancer. However, given the strong risk conferred by smoking, non-smokers with the 5% highest PRS still only had comparable risk to light smokers with the 5% lowest PRS, and much lower risk than heavy smokers (138).
Some studies have also investigated gene-environment interaction effects, which is challenging as it requires well-characterised environmental exposures and large sample sizes to ensure sufficient power (123). Studies have found suggestive evidence for interactions between multiple genetic regions and asbestos exposure (140), or with solid fuel burning for heating and cooking in never-smoking women from Asia (141). Exploratory analyses in other studies have also generated additional hypotheses, but have been limited by low statistical power and lack of independent validation. Consequently, very large cohort studies with accurately measured information for environmental exposures are needed to establish robust evidence in this area.
Genomic studies of lung cancer tumors to characterise their molecular features have found differences between LCINS tumors and lung cancer tumors from smokers, generally with a smaller number of mutations in LCINS and differences in the dominant mutation spectrum (predominantly C:G → T:A mutations for LCINS, but predominantly C:G → A:T mutations for lung cancer in smokers) (47,142). Lung carcinogenesis is a complex multistage process involving irreversible genetic changes that alter cellular processes such as proliferation and differentiation, and progressively leading to invasion and metastasis (143). Mutations in so-called “driver genes” can enable the unchecked proliferation, and some driver gene mutations are found more often in LCINS. In particular, mutations in EGFR and Kirsten rat sarcoma virus (KRAS), as well as anaplastic lymphoma kinase (ALK) rearrangements are the three major recurrent oncogenic alterations found in LCINS tumors, with EGFR mutations being the most frequently encountered in LCINS (144). EGFR gene mutations in NSCLC were first reported in 2004, when it was also found that tumors with EGFR mutations are highly sensitive to EGFR tyrosine kinase inhibitors (TKI) (145,146). Subsequently, clinical studies also confirmed that EGFR mutations are associated with better clinical response to targeted therapy with EGFR-TKI (146,147). EGFR mutations are overall more common in LCINS than lung cancer in smokers, in adenocarcinoma than other histology types, in women than men, and in people with East Asian ancestry (148). TP53 is another key driver gene with mutations present in 10%−48% of LCINS cases, although the risk of these mutations also increases with tobacco consumption (149). Moreover, molecular profiling of lung cancer tumors has also given rise to many clinical trials of other targeted treatments (150). Ongoing and future research for the identification of mutational signatures to predict clinical responses and/or more widespread molecular and genomic profiling of LCINS tumors is pivotal to the understanding of LCINS carcinogenesis and corresponding risk factors.
Conclusions
We have provided epidemiological and mechanistic evidence for some major environmental risk factors for LCINS. We have also discussed the new development in lung cancer genomics with a focus on never-smokers. This fast-expanding area aims to explore and comprehend the complex and sophisticated roles of environmental and genetic factors in lung cancer development among never-smokers. Future research with large sample sizes and refined exposure assessments is warranted to better understand the role of these factors. This understanding will not only help to evaluate a never-smoker’s personal risk for lung cancer, but also has important implications for public health, as it could inform longer-term policy reform in a range of areas including occupational health and safety, urban design, energy use and particle emissions. Meanwhile, it will provide a basis for well-informed recommendations for prevention, as well as potentially establishing practical and feasible criteria for lung cancer screening programmes.
Footnote
Conflicts of Interest: The authors have no conflicts of interest to declare.
Acknowledgements
Elvin Cheng thanks support from the 2019 Cancer Council NSW PhD Research Scholarship program. The funding agency played no role in the planning or writing of this review.
References
- 1.Sung H, Ferlay J, Siegel RL, et al Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49. doi: 10.3322/caac.21660. [DOI] [PubMed] [Google Scholar]
- 2.Siegel RL, Miller KD, Fuchs HE, et al Cancer Statistics, 2021. CA Cancer J Clin. 2021;71:7–33. doi: 10.3322/caac.21654. [DOI] [PubMed] [Google Scholar]
- 3.Li N, Wu P, Shen Y, et al Predictions of mortality related to four major cancers in China, 2020 to 2030. Cancer Commun (Lond) 2021;41:404–13. doi: 10.1002/cac2.12143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Sun S, Schiller JH, Gazdar AF Lung cancer in never smokers — A different disease. Nat Rev Cancer. 2007;7:778–90. doi: 10.1038/nrc2190. [DOI] [PubMed] [Google Scholar]
- 5.Torok S, Hegedus B, Laszlo V, et al Lung cancer in never smokers. Future Oncol. 2011;7:1195–211. doi: 10.2217/fon.11.100. [DOI] [PubMed] [Google Scholar]
- 6.Ferlay J, Colombet M, Soerjomataram I, et al Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 2019;144:1941–53. doi: 10.1002/ijc.31937. [DOI] [PubMed] [Google Scholar]
- 7.Wang JB, Jiang Y, Wei WQ, et al Estimation of cancer incidence and mortality attributable to smoking in China. Cancer Causes Control. 2010;21:959–65. doi: 10.1007/s10552-010-9523-8. [DOI] [PubMed] [Google Scholar]
- 8.International Agency for Research on Cancer. IARC monographs on the evaluation of carcinogenic risks to humans: Man-made mineral fibres and radon. Vol. 43. Lyon: IARC, 1988.
- 9.International Agency for Research on Cancer. IARC monographs on the evaluation of carcinogenic risks to humans: Tobacco smoke and involuntary smoking. Vol. 83. Lyon: IARC, 2004.
- 10.International Agency for Research on Cancer. IARC monographs on the evaluation of carcinogenic risks to humans: Household use of solid fuels and high-temperature frying. Vol. 95. Lyon: IARC, 2010.
- 11.International Agency for Research on Cancer. IARC monographs on the evaluation of carcinogenic risks to humans: A review of human carcinogens: Personal habits and indoor combustions. Vol. 100E. Lyon: IARC, 2012.
- 12.International Agency for Research on Cancer. IARC scientific publication No. 161: Air pollution and cancer. Lyon: IARC, 2013.
- 13.International Agency for Research on Cancer. List of classifications by cancer sites with sufficient or limited evidence in humans. IARC Monographs Vol. 1-128. Lyon: IARC, 2020.
- 14.Couraud S, Zalcman G, Milleron B, et al Lung cancer in never smokers — A review. Eur J Cancer. 2012;48:1299–311. doi: 10.1016/j.ejca.2012.03.007. [DOI] [PubMed] [Google Scholar]
- 15.Sun JM, Choi YL, Ji JH, et al Small-cell lung cancer detection in never-smokers: clinical characteristics and multigene mutation profiling using targeted next-generation sequencing. Ann Oncol. 2015;26:161–6. doi: 10.1093/annonc/mdu504. [DOI] [PubMed] [Google Scholar]
- 16.Lubin JH. Radon and lung cancer risk: a joint analysis of 11 underground miners studies. Washington: U. S. Department of Health and Human Services, Public Health Service, National Institutes of Health, 1994.
- 17.National Research Council, Committee on Health Risks of Exposure to Radon, Board on Radiation Effects Research and Commission on Life Sciences. Health effects of exposure to radon: Beir VI. Washington: National Academy Press, 1999.
- 18.World Health Organization. WHO handbook on indoor radon: a public health perspective. Geneva: WHO, 2009.
- 19.Lubin JH Studies of radon and lung cancer in North America and China. Radiat Prot Dosimetry. 2003;104:315–9. doi: 10.1093/oxfordjournals.rpd.a006194. [DOI] [PubMed] [Google Scholar]
- 20.Lubin JH, Wang ZY, Boice JD Jr, et al Risk of lung cancer and residential radon in China: pooled results of two studies. Int J Cancer. 2004;109:132–7. doi: 10.1002/ijc.11683. [DOI] [PubMed] [Google Scholar]
- 21.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. doi: 10.1136/bmj.38308.477650.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Darby S, Hill D, Deo H, et al Residential radon and lung cancer-detailed results of a collaborative analysis of individual data on 7148 persons with lung cancer and 14, 208 persons without lung cancer from 13 epidemiologic studies in Europe. Scand J Work Environ Health. 2006;32(suppl 1):1–83. [PubMed] [Google Scholar]
- 23.Krewski D, Lubin JH, Zielinski JM, et al Residential radon and risk of lung cancer: a combined analysis of 7 North American case-control studies. Epidemiology. 2005;16:137–45. doi: 10.1097/01.ede.0000152522.80261.e3. [DOI] [PubMed] [Google Scholar]
- 24.Krewski D, Lubin JH, Zielinski JM, et al A combined analysis of North American case-control studies of residential radon and lung cancer. J Toxicol Environ Health A. 2006;69:533–97. doi: 10.1080/15287390500260945. [DOI] [PubMed] [Google Scholar]
- 25.Lorenzo-González M, Ruano-Ravina A, Torres-Durán M, et al Lung cancer and residential radon in never-smokers: A pooling study in the Northwest of Spain. Environ Res. 2019;172:713–8. doi: 10.1016/j.envres.2019.03.011. [DOI] [PubMed] [Google Scholar]
- 26.Garzillo C, Pugliese M, Loffredo F, et al Indoor radon exposure and lung cancer risk: A meta-analysis of case-control studies. Transl Cancer Res. 2017;6:S934–43. doi: 10.21037/tcr.2017.05.42. [DOI] [Google Scholar]
- 27.Malinovsky G, Yarmoshenko I, Vasilyev A Meta-analysis of case-control studies on the relationship between lung cancer and indoor radon exposure. Radiat Environ Biophys. 2019;58:39–47. doi: 10.1007/s00411-018-0770-5. [DOI] [PubMed] [Google Scholar]
- 28.Cheng ES, Egger S, Hughes S, et al Systematic review and meta-analysis of residential radon and lung cancer in never-smokers. Eur Respir Rev. 2021;30:200230. doi: 10.1183/16000617.0230-2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Dacic S, Luvison A, Evdokimova V, et al RET rearrangements in lung adenocarcinoma and radiation. J Thorac Oncol. 2014;9:118–20. doi: 10.1097/jto.0000000000000015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Lim SM, Choi JW, Hong MH, et al Indoor radon exposure increases tumor mutation burden in never-smoker patients with lung adenocarcinoma. Lung Cancer. 2019;131:139–46. doi: 10.1016/j.lungcan.2019.04.002. [DOI] [PubMed] [Google Scholar]
- 31.de Vocht F, Suderman M, Ruano-Ravina A, et al Residential exposure to radon and DNA methylation across the lifecourse: an exploratory study in the ALSPAC birth cohort. Wellcome Open Res. 2019;4:3. doi: 10.12688/wellcomeopenres.14991.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.World Health Organization. WHO Framework Convention on Tobacco Control WHO. Available online: http://apps.who.int/iris/bitstream/handle/10665/42811/9241591013.pdf?sequence=1
- 33.Wogan GN, Hecht SS, Felton JS, et al Environmental and chemical carcinogenesis. Semin Cancer Biol. 2004;14:473–86. doi: 10.1016/j.semcancer.2004.06.010. [DOI] [PubMed] [Google Scholar]
- 34.Taylor R, Najafi F, Dobson A Meta-analysis of studies of passive smoking and lung cancer: effects of study type and continent. Int J Epidemiol. 2007;36:1048–59. doi: 10.1093/ije/dym158. [DOI] [PubMed] [Google Scholar]
- 35.Sheng L, Tu JW, Tian JH, et al A meta-analysis of the relationship between environmental tobacco smoke and lung cancer risk of nonsmoker in China. Medicine (Baltimore) 2018;97:e11389. doi: 10.1097/MD.0000000000011389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Li M, Liu X, Zhang L The relationship of indoor coal use and environmental tobacco smoke exposure with lung cancer in China: A meta-analysis. J Cancer Res Ther. 2018;14(Supplement):S7–S13. doi: 10.4103/0973-1482.168965. [DOI] [PubMed] [Google Scholar]
- 37.Boffetta P, Trédaniel J, Greco A Risk of childhood cancer and adult lung cancer after childhood exposure to passive smoke: A meta-analysis. Environ Health Perspect. 2000;108:73–82. doi: 10.1289/ehp.0010873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Enstrom JE, Kabat GC Environmental tobacco smoke and tobacco related mortality in a prospective study of Californians, 1960-98. BMJ. 2003;326:1057. doi: 10.1136/bmj.326.7398.1057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hill SE, Blakely T, Kawachi I, et al Mortality among lifelong nonsmokers exposed to secondhand smoke at home: cohort data and sensitivity analyses. Am J Epidemiol. 2007;165:530–40. doi: 10.1093/aje/kwk043. [DOI] [PubMed] [Google Scholar]
- 40.Wang A, Kubo J, Luo J, et al Active and passive smoking in relation to lung cancer incidence in the Women’s Health Initiative Observational Study prospective cohort. Ann Oncol. 2015;26:221–30. doi: 10.1093/annonc/mdu470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Ni X, Xu N, Wang Q Meta-analysis and systematic review in environmental tobacco smoke risk of female lung cancer by research type. Int J Environ Res Public Health. 2018;15:1348. doi: 10.3390/ijerph15071348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Flores RM, Liu B, Taioli E Association of serum cotinine levels and lung cancer mortality in non-smokers. Carcinogenesis. 2016;37:1062–9. doi: 10.1093/carcin/bgw094. [DOI] [PubMed] [Google Scholar]
- 43.Thomas CE, Wang R, Adams-Haduch J, et al Urinary cotinine is as good a biomarker as serum cotinine for cigarette smoking exposure and lung cancer risk prediction. Cancer Epidemiol Biomarkers Prev. 2020;29:127–32. doi: 10.1158/1055-9965.Epi-19-0653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Alexandrov LB, Ju YS, Haase K, et al Mutational signatures associated with tobacco smoking in human cancer. Science. 2016;354:618–22. doi: 10.1126/science.aag0299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Landi MT, Synnott NC, Rosenbaum J, et al Tracing lung cancer risk factors through mutational signatures in never-smokers. Am J Epidemiol. 2021;190:962–76. doi: 10.1093/aje/kwaa234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Sun Y, Ren Y, Fang Z, et al Lung adenocarcinoma from East Asian never-smokers is a disease largely defined by targetable oncogenic mutant kinases. J Clin Oncol. 2010;28:4616–20. doi: 10.1200/jco.2010.29.6038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Cancer Genome Atlas Research Network Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511:543–50. doi: 10.1038/nature13385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lee YJ, Cho BC, Jee SH, et al Impact of environmental tobacco smoke on the incidence of mutations in epidermal growth factor receptor gene in never-smoker patients with non-small-cell lung cancer. J Clin Oncol. 2010;28:487–92. doi: 10.1200/JCO.2009.24.5480. [DOI] [PubMed] [Google Scholar]
- 49.Gazdar AF, Shigematsu H, Herz J, et al. Mutations and addiction to EGFR: the Achilles “heal” of lung cancers? Trends Mol Med 2004;10:481-6.
- 50.Zhang Y, Kang S, Fang W, et al Impact of smoking status on EGFR-TKI efficacy for advanced non-small-cell lung cancer in EGFR mutants: a meta-analysis. Clin Lung Cancer. 2015;16:144–51.e1. doi: 10.1016/j.cllc.2014.09.008. [DOI] [PubMed] [Google Scholar]
- 51.Liang W, Liang H, Liu Z, et al Environmental tobacco smoke exposure and EGFR mutations in non-smokers with lung cancer: A dose-response analysis of published data. J Thorac Oncol. 2018;13(suppl 10):S779. doi: 10.1016/j.jtho.2018.08.1346. [DOI] [Google Scholar]
- 52.Soo RA, Kubo A, Ando M, et al Association between environmental tobacco smoke exposure and the occurrence of EGFR mutations and ALK rearrangements in never-smokers with non-small-cell lung cancer: Analyses from a prospective multinational ETS registry. Clin Lung Cancer. 2017;18:535–42. doi: 10.1016/j.cllc.2017.01.005. [DOI] [PubMed] [Google Scholar]
- 53.Kawaguchi T, Ando M, Kubo A, et al Long exposure of environmental tobacco smoke associated with activating EGFR mutations in never-smokers with non-small cell lung cancer. Clin Cancer Res. 2011;17:39–45. doi: 10.1158/1078-0432.CCR-10-1773. [DOI] [PubMed] [Google Scholar]
- 54.Health Effects Institute. State of Global Air 2020. Special Report. Boston: Health Effects Institute, 2020.
- 55.McGranahan G, Murray F, editors. Air pollution & health in rapidly developing countries — Introduction chapter p. 1-10. London: Earthscan, 2003.
- 56.Turner MC, Andersen ZJ, Baccarelli A, et al Outdoor air pollution and cancer: An overview of the current evidence and public health recommendations. CA Cancer J Clin. 2020;70:460–79. doi: 10.3322/caac.21632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.International Agency for Research on Cancer. IARC monographs on the evaluation of carcinogenic risks to humans: Outdoor air pollution. Vol. 109. Lyon: IARC, 2016.
- 58.Harrison RM, Yin J. Particulate matter in the atmosphere: which particle properties are important for its effects on health? Sci Total Environ 2000;249:85-101.
- 59.World Health Organization. Health aspects of air pollution. Chapter 5. Particulate matter (PM), Section 5.1 Introduction. Geneva: WHO, 2003.
- 60.Turner MC, Krewski D, Pope CA 3rd, et al Long-term ambient fine particulate matter air pollution and lung cancer in a large cohort of never-smokers. Am J Respir Crit Care Med. 2011;184:1374–81. doi: 10.1164/rccm.201106-1011OC. [DOI] [PubMed] [Google Scholar]
- 61.Hamra GB, Guha N, Cohen A, et al Outdoor particulate matter exposure and lung cancer: a systematic review and meta-analysis. Environ Health Perspect. 2014;122:906–11. doi: 10.1289/ehp.140809210.1289/ehp/1408092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Huang F, Pan B, Wu J, et al Relationship between exposure to PM2. 5 and lung cancer incidence and mortality:A meta-analysis. Oncotarget. 2017;8:43322–31. doi: 10.18632/oncotarget.17313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zhang Z, Zhu D, Cui B, et al Association between particulate matter air pollution and lung cancer. Thorax. 2020;75:85–7. doi: 10.1136/thoraxjnl-2019-213722. [DOI] [PubMed] [Google Scholar]
- 64.World Health Organization. Burden of disease from household air pollution for 2016. Geneva: WHO, 2018.
- 65.Hosgood HD 3rd, Wei H, Sapkota A, et al Household coal use and lung cancer: systematic review and meta-analysis of case-control studies, with an emphasis on geographic variation. Int J Epidemiol. 2011;40:719–28. doi: 10.1093/ije/dyq259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Kurmi OP, Arya PH, Lam KB, et al Lung cancer risk and solid fuel smoke exposure: a systematic review and meta-analysis. Eur Respir J. 2012;40:1228–37. doi: 10.1183/09031936.00099511. [DOI] [PubMed] [Google Scholar]
- 67.Bruce N, Dherani M, Liu R, et al Does household use of biomass fuel cause lung cancer? A systematic review and evaluation of the evidence for the GBD 2010 study. Thorax. 2015;70:433–41. doi: 10.1136/thoraxjnl-2014-206625. [DOI] [PubMed] [Google Scholar]
- 68.Gordon SB, Bruce NG, Grigg J, et al Respiratory risks from household air pollution in low and middle income countries. Lancet Respir Med. 2014;2:823–60. doi: 10.1016/S2213-2600(14)70168-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lan Q, Chapman RS, Schreinemachers DM, et al Household stove improvement and risk of lung cancer in Xuanwei, China. J Natl Cancer Inst. 2002;94:826–35. doi: 10.1093/jnci/94.11.826. [DOI] [PubMed] [Google Scholar]
- 70.Hosgood HD 3rd, Chapman R, Shen M, et al Portable stove use is associated with lower lung cancer mortality risk in lifetime smoky coal users. Br J Cancer. 2008;99:1934–9. doi: 10.1038/sj.bjc.6604744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Mumford JL, He XZ, Chapman RS, et al Lung cancer and indoor air pollution in Xuan Wei, China. Science. 1987;235:217–20. doi: 10.1126/science.3798109. [DOI] [PubMed] [Google Scholar]
- 72.Shen M, Bin P, Li H, et al Increased levels of etheno-DNA adducts and genotoxicity biomarkers of long-term exposure to pure diesel engine exhaust. Sci Total Environ. 2016;543(PtA):267–73. doi: 10.1016/j.scitotenv.2015.10.165. [DOI] [PubMed] [Google Scholar]
- 73.de Oliveira Alves N, Martins Pereira G, Di Domenico M, et al Inflammation response, oxidative stress and DNA damage caused by urban air pollution exposure increase in the lack of DNA repair XPC protein. Environ Int. 2020;145:106150. doi: 10.1016/j.envint.2020.106150. [DOI] [PubMed] [Google Scholar]
- 74.Xue W, Warshawsky D Metabolic activation of polycyclic and heterocyclic aromatic hydrocarbons and DNA damage: a review. Toxicol Appl Pharmacol. 2005;206:73–93. doi: 10.1016/j.taap.2004.11.006. [DOI] [PubMed] [Google Scholar]
- 75.Boffetta P, Jourenkova N, Gustavsson P Cancer risk from occupational and environmental exposure to polycyclic aromatic hydrocarbons. Cancer Causes Control. 1997;8:444–72. doi: 10.1023/a:1018465507029. [DOI] [PubMed] [Google Scholar]
- 76.Brüske-Hohlfeld I, Möhner M, Pohlabeln H, et al Occupational lung cancer risk for men in Germany: results from a pooled case-control study. Am J Epidemiol. 2000;151:384–95. doi: 10.1093/oxfordjournals.aje.a010218. [DOI] [PubMed] [Google Scholar]
- 77.Armstrong B, Hutchinson E, Unwin J, et al Lung cancer risk after exposure to polycyclic aromatic hydrocarbons: a review and meta-analysis. Environ Health Perspect. 2004;112:970–8. doi: 10.1289/ehp.6895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.World Health Organization. WHO guidelines for indoor air quality: Selected pollutants. Geneva: WHO, 2010.
- 79.IARC Working Group on the Evaluation of Carcinogenic Risks to Humans Some non-heterocyclic polycyclic aromatic hydrocarbons and some related exposures. IARC Monogr Eval Carcinog Risks Hum. 2010;92:1–853. [PMC free article] [PubMed] [Google Scholar]
- 80.Chiang TA, Wu PF, Wang LF, et al Mutagenicity and polycyclic aromatic hydrocarbon content of fumes from heated cooking oils produced in Taiwan. Mutat Res. 1997;381:157–61. doi: 10.1016/s0027-5107(97)00163-2. [DOI] [PubMed] [Google Scholar]
- 81.Zhou F, Zhou C Lung cancer in never smokers-the East Asian experience. Transl Lung Cancer Res. 2018;7:450–63. doi: 10.21037/tlcr.2018.05.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Wu SC, Yen GC Effects of cooking oil fumes on the genotoxicity and oxidative stress in human lung carcinoma (A-549) cells. Toxicol In Vitro. 2004;18:571–80. doi: 10.1016/j.tiv.2004.01.004. [DOI] [PubMed] [Google Scholar]
- 83.Ko YC, Cheng LS, Lee CH, et al Chinese food cooking and lung cancer in women nonsmokers. Am J Epidemiol. 2000;151:140–7. doi: 10.1093/oxfordjournals.aje.a010181. [DOI] [PubMed] [Google Scholar]
- 84.Metayer C, Wang Z, Kleinerman RA, et al Cooking oil fumes and risk of lung cancer in women in rural Gansu, China. Lung Cancer. 2002;35:111–7. doi: 10.1016/s0169-5002(01)00412-3. [DOI] [PubMed] [Google Scholar]
- 85.Wang G, Bai Y, Fu W, et al Daily cooking duration and its joint effects with genetic polymorphisms on lung cancer incidence: Results from a Chinese prospective cohort study. Environ Res. 2019;179(PtA):108747. doi: 10.1016/j.envres.2019.108747. [DOI] [PubMed] [Google Scholar]
- 86.Jia PL, Zhang C, Yu JJ, et al The risk of lung cancer among cooking adults: a meta-analysis of 23 observational studies. J Cancer Res Clin Oncol. 2018;144:229–40. doi: 10.1007/s00432-017-2547-7. [DOI] [PubMed] [Google Scholar]
- 87.Chen TY, Fang YH, Chen HL, et al Impact of cooking oil fume exposure and fume extractor use on lung cancer risk in non-smoking Han Chinese women. Sci Rep. 2020;10:6774. doi: 10.1038/s41598-020-63656-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Rushton L, Hutchings SJ, Fortunato L, et al Occupational cancer burden in Great Britain. Br J Cancer. 2012;107 Suppl 1(Suppl 1):S3–7. doi: 10.1038/bjc.2012.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Driscoll T, Nelson DI, Steenland K, et al The global burden of disease due to occupational carcinogens. Am J Ind Med. 2005;48:419–31. doi: 10.1002/ajim.20209. [DOI] [PubMed] [Google Scholar]
- 90.Fingerhut M, Nelson DI, Driscoll T, et al The contribution of occupational risks to the global burden of disease: summary and next steps. Med Lav. 2006;97:313–21. [PubMed] [Google Scholar]
- 91.International Agency for Research on Cancer. IARC monographs on the evaluation of carcinogenic risks to humans: Some metals and metallic cornpounds. Vol. 23. Lyon: IARC, 1980.
- 92.Spyratos D, Zarogoulidis P, Porpodis K, et al Occupational exposure and lung cancer. J Thorac Dis. 2013;5 Suppl 4(Suppl 4):S440–5. doi: 10.3978/j.issn.2072-1439.2013.07.09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Brownson RC, Alavanja MC, Chang JC Occupational risk factors for lung cancer among nonsmoking women: a case-control study in Missouri (United States) Cancer Causes Control. 1993;4:449–54. doi: 10.1007/bf00050864. [DOI] [PubMed] [Google Scholar]
- 94.Zeka A, Mannetje A, Zaridze D, et al Lung cancer and occupation in nonsmokers: a multicenter case-control study in Europe. Epidemiology. 2006;17:615–23. doi: 10.1097/01.ede.0000239582.92495.b5. [DOI] [PubMed] [Google Scholar]
- 95.Lynch KM, Smith WA Pulmonary asbestosis III: Carcinoma of lung in asbesto-silicosis. Am J Cancer. 1935;24:56–64. doi: 10.1158/ajc.1935.56. [DOI] [Google Scholar]
- 96.International Agency for Research on Cancer. IARC monographs on the evaluation of carcinogenic risks of chemicals to man: Asbestos. Vol 14. Lyon: IARC, 1977.
- 97.Alberg AJ, Brock MV, Ford JG, et al Epidemiology of lung cancer: Diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2013;143(suppl 5):e1S–e29S. doi: 10.1378/chest.12-2345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.IARC Working Group on the Evaluation of Carcinogenic Risks to Humans Arsenic, metals, fibres, and dusts. IARC Monogr Eval Carcinog Risks Hum. 2012;100(PtC):11–465. [PMC free article] [PubMed] [Google Scholar]
- 99.Kamp DW Asbestos-induced lung diseases: an update. Transl Res. 2009;153:143–52. doi: 10.1016/j.trsl.2009.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Carbone M, Kratzke RA, Testa JR The pathogenesis of mesothelioma. Semin Oncol. 2002;29:2–17. doi: 10.1053/sonc.2002.30227. [DOI] [PubMed] [Google Scholar]
- 101.Hughes MF, Beck BD, Chen Y, et al Arsenic exposure and toxicology: a historical perspective. Toxicol Sci. 2011;123:305–32. doi: 10.1093/toxsci/kfr184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Sommers SC, McManus RG Multiple arsenical cancers of skin and internal organs. Cancer. 1953;6:347–59. doi: 10.1002/1097-0142(195303)6:2<347::aid-cncr2820060219>3.0.co;2-l. [DOI] [PubMed] [Google Scholar]
- 103.Putila JJ, Guo NL Association of arsenic exposure with lung cancer incidence rates in the United States. PLoS One. 2011;6:e25886. doi: 10.1371/journal.pone.0025886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Boström CE, Gerde P, Hanberg A, et al Cancer risk assessment, indicators, and guidelines for polycyclic aromatic hydrocarbons in the ambient air. Environ Health Perspect. 2002;110 Suppl 3(Suppl 3):451–88. doi: 10.1289/ehp.110-1241197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Morfeld P, Spallek M Diesel engine exhaust and lung cancer risks — evaluation of the meta-analysis by Vermeulen et al. 2014. J Occup Med Toxicol. 2015;10:31. doi: 10.1186/s12995-015-0073-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.International Agency for Research on Cancer. IARC monographs on the evaluation of carcinogenic risks to humans: Diesel and gasoline engine exhausts and some nitroarenes. Vol. 105. Lyon: IARC, 2014.
- 107.Silverman DT, Samanic CM, Lubin JH, et al The Diesel Exhaust in Miners study: a nested case-control study of lung cancer and diesel exhaust. J Natl Cancer Inst. 2012;104:855–68. doi: 10.1093/jnci/djs034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Attfield MD, Schleiff PL, Lubin JH, et al The Diesel Exhaust in Miners study: a cohort mortality study with emphasis on lung cancer. J Natl Cancer Inst. 2012;104:869–83. doi: 10.1093/jnci/djs035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Möhner M, Wendt A A critical review of the relationship between occupational exposure to diesel emissions and lung cancer risk. Crit Rev Toxicol. 2017;47:185–224. doi: 10.1080/10408444.2016.1266598. [DOI] [PubMed] [Google Scholar]
- 110.Silverman DT Diesel exhaust and lung cancer-aftermath of becoming an IARC group 1 carcinogen. Am J Epidemiol. 2018;187:1149–52. doi: 10.1093/aje/kwy036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.International Agency for Research on Cancer. IARC monographs on the evaluation of carcinogenic risks to humans: Polynuclear aromatic compounds, Part 3, Industrial exposures in aluminium production, coal gasification, coke production, and iron and steel founding. Vol. 32. Lyon: IARC, 1984.
- 112.Shankar A, Dubey A, Saini D, et al Environmental and occupational determinants of lung cancer. Transl Lung Cancer Res. 2019;8(Suppl 1):S31–S49. doi: 10.21037/tlcr.2019.03.05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Steenland K, Ward E Silica: a lung carcinogen. CA Cancer J Clin. 2014;64:63–9. doi: 10.3322/caac.21214. [DOI] [PubMed] [Google Scholar]
- 114.Takkouche B, Regueira-Méndez C, Montes-Martínez A Risk of cancer among hairdressers and related workers: a meta-analysis. Int J Epidemiol. 2009;38:1512–31. doi: 10.1093/ije/dyp283. [DOI] [PubMed] [Google Scholar]
- 115.Tokuhata GK, Lilienfeld AM Familial aggregation of lung cancer in humans. J Natl Cancer Inst. 1963;30:289–312. [PubMed] [Google Scholar]
- 116.Matakidou A, Eisen T, Houlston RS Systematic review of the relationship between family history and lung cancer risk. Br J Cancer. 2005;93:825–33. doi: 10.1038/sj.bjc.6602769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Coté ML, Liu M, Bonassi S, et al Increased risk of lung cancer in individuals with a family history of the disease: a pooled analysis from the International Lung Cancer Consortium. Eur J Cancer. 2012;48:1957–68. doi: 10.1016/j.ejca.2012.01.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Lissowska J, Foretova L, Dabek J, et al Family history and lung cancer risk: international multicentre case-control study in Eastern and Central Europe and meta-analyses. Cancer Causes Control. 2010;21:1091–104. doi: 10.1007/s10552-010-9537-2. [DOI] [PubMed] [Google Scholar]
- 119.Lin H, Huang YS, Yan HH, et al A family history of cancer and lung cancer risk in never-smokers: A clinic-based case-control study. Lung Cancer. 2015;89:94–8. doi: 10.1016/j.lungcan.2015.05.017. [DOI] [PubMed] [Google Scholar]
- 120.Ang L, Chan CPY, Yau WP, et al Association between family history of lung cancer and lung cancer risk: a systematic review and meta-analysis. Lung Cancer. 2020;148:129–37. doi: 10.1016/j.lungcan.2020.08.012. [DOI] [PubMed] [Google Scholar]
- 121.Wang J, Liu Q, Yuan S, et al Genetic predisposition to lung cancer: comprehensive literature integration, meta-analysis, and multiple evidence assessment of candidate-gene association studies. Sci Rep. 2017;7:8371. doi: 10.1038/s41598-017-07737-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Subramanian J, Govindan R Molecular profile of lung cancer in never smokers. EJC Suppl. 2013;11:248–53. doi: 10.1016/j.ejcsup.2013.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Bossé Y, Amos CI A decade of GWAS results in lung cancer. Cancer Epidemiol Biomarkers Prev. 2018;27:363–79. doi: 10.1158/1055-9965.Epi-16-0794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.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. doi: 10.1038/nature06885. [DOI] [PubMed] [Google Scholar]
- 125.Thorgeirsson TE, Geller F, Sulem P, et al A variant associated with nicotine dependence, lung cancer and peripheral arterial disease. Nature. 2008;452:638–42. doi: 10.1038/nature06846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Amos CI, Wu X, Broderick P, et al Genome-wide association scan of tag SNPs identifies a susceptibility locus for lung cancer at 15q25. 1. Nat Genet. 2008;40:616–22. doi: 10.1038/ng.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Wang Y, Broderick P, Webb E, et al Common 5p15. 33 and 6p21. 33 variants influence lung cancer risk. Nat Genet. 2008;40:1407–9. doi: 10.1038/ng.273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Li Y, Sheu CC, Ye Y, et al Genetic variants and risk of lung cancer in never smokers: a genome-wide association study. Lancet Oncol. 2010;11:321–30. doi: 10.1016/s1470-2045(10)70042-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Seow WJ, Matsuo K, Hsiung CA, et al Association between GWAS-identified lung adenocarcinoma susceptibility loci and EGFR mutations in never-smoking Asian women, and comparison with findings from Western populations. Hum Mol Genet. 2017;26:454–65. doi: 10.1093/hmg/ddw414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Hung RJ, Spitz MR, Houlston RS, et al Lung cancer risk in never-smokers of european descent is associated with genetic variation in the 5(p)15. 33 TERT-CLPTM1Ll region. J Thorac Oncol. 2019;14:1360–9. doi: 10.1016/j.jtho.2019.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.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 Genet. 2010;6:e1001051. doi: 10.1371/journal.pgen.1001051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Spitz MR, Gorlov IP, Amos CI, et al Variants in inflammation genes are implicated in risk of lung cancer in never smokers exposed to second-hand smoke. Cancer Discov. 2011;1:420–9. doi: 10.1158/2159-8290.Cd-11-0080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Ahn MJ, Won HH, Lee J, et al The 18p11. 22 locus is associated with never smoker non-small cell lung cancer susceptibility in Korean populations. Hum Genet. 2012;131:365–72. doi: 10.1007/s00439-011-1080-z. [DOI] [PubMed] [Google Scholar]
- 134.Lan Q, Hsiung CA, Matsuo K, et al Genome-wide association analysis identifies new lung cancer susceptibility loci in never-smoking women in Asia. Nat Genet. 2012;44:1330–5. doi: 10.1038/ng.2456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Kim SC, Jung Y, Park J, et al A high-dimensional, deep-sequencing study of lung adenocarcinoma in female never-smokers. PLoS One. 2013;8:e55596. doi: 10.1371/journal.pone.0055596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Wang Z, Seow WJ, Shiraishi K, et al Meta-analysis of genome-wide association studies identifies multiple lung cancer susceptibility loci in never-smoking Asian women. Hum Mol Genet. 2016;25:620–9. doi: 10.1093/hmg/ddv494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Park SL, Cheng I, Haiman CA Genome-wide association studies of cancer in diverse populations. Cancer Epidemiol Biomarkers Prev. 2018;27:405–17. doi: 10.1158/1055-9965.Epi-17-0169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Dai J, Lv J, Zhu M, et al Identification of risk loci and a polygenic risk score for lung cancer: a large-scale prospective cohort study in Chinese populations. Lancet Respir Med. 2019;7:881–91. doi: 10.1016/s2213-2600(19)30144-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Hung RJ, Warkentin MT, Brhane Y, et al Assessing lung cancer absolute risk trajectory based on a polygenic risk model. Cancer Res. 2021;81:1607–15. doi: 10.1158/0008-5472.Can-20-1237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Liu CY, Stücker I, Chen C, et al Genome-wide gene-asbestos exposure interaction association study identifies a common susceptibility variant on 22q13. 31 associated with lung cancer risk. Cancer Epidemiol Biomarkers Prev. 2015;24:1564–73. doi: 10.1158/1055-9965.Epi-15-0021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Hosgood HD, Song M, Hsiung CA, et al Interactions between household air pollution and GWAS-identified lung cancer susceptibility markers in the Female Lung Cancer Consortium in Asia (FLCCA) Hum Genet. 2015;134:333–41. doi: 10.1007/s00439-014-1528-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Govindan R, Ding L, Griffith M, et al Genomic landscape of non-small cell lung cancer in smokers and never-smokers. Cell. 2012;150:1121–34. doi: 10.1016/j.cell.2012.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Pitot HC The molecular biology of carcinogenesis. Cancer. 1993;72(3 Suppl):962–70. doi: 10.1002/1097-0142(19930801)72:3+<962::aid-cncr2820721303>3.0.co;2-h. [DOI] [PubMed] [Google Scholar]
- 144.Riely GJ Lung cancer in “Never-smokers”: molecular factors trump risk factors. Oncology (Williston Park) 2010;24:38,40. [PubMed] [Google Scholar]
- 145.Lynch TJ, Bell DW, Sordella R, et al Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129–39. doi: 10.1056/NEJMoa040938. [DOI] [PubMed] [Google Scholar]
- 146.Paez JG, Jänne PA, Lee JC, et al EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304:1497–500. doi: 10.1126/science.1099314. [DOI] [PubMed] [Google Scholar]
- 147.Miller VA, Kris MG, Shah N, et al Bronchioloalveolar pathologic subtype and smoking history predict sensitivity to gefitinib in advanced non-small-cell lung cancer. J Clin Oncol. 2004;22:1103–9. doi: 10.1200/jco.2004.08.158. [DOI] [PubMed] [Google Scholar]
- 148.Ha SY, Choi SJ, Cho JH, et al Lung cancer in never-smoker Asian females is driven by oncogenic mutations, most often involving EGFR. Oncotarget. 2015;6:5465–74. doi: 10.18632/oncotarget.2925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Le Calvez F, Mukeria A, Hunt JD, et al TP53 and KRAS mutation load and types in lung cancers in relation to tobacco smoke: distinct patterns in never, former, and current smokers. Cancer Res. 2005;65:5076–83. doi: 10.1158/0008-5472.Can-05-0551. [DOI] [PubMed] [Google Scholar]
- 150.Politi K, Herbst RS Lung cancer in the era of precision medicine. Clin Cancer Res. 2015;21:2213–20. doi: 10.1158/1078-0432.Ccr-14-2748. [DOI] [PMC free article] [PubMed] [Google Scholar]