Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Sep 1.
Published in final edited form as: Eur J Cancer. 2012 Mar 19;48(13):1957–1968. doi: 10.1016/j.ejca.2012.01.038

Increased risk of lung cancer in individuals with a family history of the disease: A pooled analysis from the International Lung Cancer Consortium

Michele L Coté 1,*, Mei Liu 2,*, Stefano Bonassi 3, Monica Neri 3, Ann G Schwartz 1, David C Christiani 4, Margaret R Spitz 2, Joshua E Muscat 5, Gad Rennert 6, Katja K Aben 7, Angeline S Andrew 8, Vladimir Bencko 9, Heike Bickeböller 10, Paolo Boffetta 11, Paul Brennan 12, Hermann Brenner 13, Eric J Duell 14, Eleonora Fabianova 15, John K Field 16, Lenka Foretova 17, Søren Friis 18, Curtis C Harris 19, Ivana Holcatova 9, Yun-Chul Hong 20, Dolores Isla 21, Vladimir Janout 22, Lambertus A Kiemeney 23, Chikako Kiyohara 24, Qing Lan 25, Philip Lazarus 26, Jolanta Lissowska 27, Loic Le Marchand 28, Dana Mates 29, Keitaro Matsuo 30, Jose I Mayordomo 21, John R McLaughlin 31, Hal Morgenstern 32, Heiko Müeller 13, Irene Orlow 33, Bernard J Park 34, Mila Pinchev 6, Olaide Y Raji 16, Hedy S Rennert 6, Peter Rudnai 35, Adeline Seow 36, Isabelle Stucker 37, Neonila Szeszenia-Dabrowska 38, M Dawn Teare 39, Anne Tjønnelan 18, Donatella Ugolini 40, Henricus FM van der Heijden 23, Erich Wichmann 41,46, John K Wiencke 42, Penella J Woll 37, Ping Yang 43, David Zaridze 44, Zuo-Feng Zhang 45, Carol J Etzel 2,**, Rayjean J Hung 31,**
PMCID: PMC3445438  NIHMSID: NIHMS365912  PMID: 22436981

Abstract

Background and Methods

Familial aggregation of lung cancer exists after accounting for cigarette smoking. However, the extent to which family history affects risk by smoking status, histology, relative type and ethnicity is not well described. This pooled analysis included 24 case-control studies in the International Lung Cancer Consortium. Each study collected age of onset/interview, gender, race/ethnicity, cigarette smoking, histology and first-degree family history of lung cancer. Data from 24,380 lung cancer cases and 23,305 healthy controls were analyzed. Unconditional logistic regression models and generalized estimating equations were used to estimate odds ratios and 95% confidence intervals.

Results

Individuals with a first-degree relative with lung cancer had a 1.51-fold increase in risk of lung cancer, after adjustment for smoking and other potential confounders(95% CI: 1.39, 1.63). The association was strongest for those with a family history in a sibling, after adjustment (OR=1.82, 95% CI: 1.62, 2.05). No modifying effect by histologic type was found. Never smokers showed a lower association with positive familial history of lung cancer (OR=1.25, 95% CI: 1.03, 1.52), slightly stronger for those with an affected sibling (OR=1.44, 95% CI: 1.07, 1.93), after adjustment.

Conclusions

The increased risk among never smokers and similar magnitudes of the effect of family history on lung cancer risk across histological types suggests familial aggregation of lung cancer is independent of those associated with cigarette smoking. While the role of genetic variation in the etiology of lung cancer remains to be fully characterized, family history assessment is immediately available and those with a positive history represent a higher risk group.

Introduction

Lung cancer is the leading cause of cancer-related deaths worldwide (1). Current and former smokers are at greatest risk, but lung cancer does occur among nonsmokers, with varying rates across countries (2). The association between cigarette smoking and increased risk of lung cancer is now undisputed. Despite this, less than 20% of smokers develop lung cancer, suggesting that the effect of tobacco smoke exposure is modified by other variables, including individual susceptibility (3). The search for a gene or genes associated with susceptibility is still nascent. Genome wide association studies have independently reported chromosomal region 15q24-25.1, which contains nicotinic acetylcholine receptor sub-unit genes, to be associated with increased risk of lung cancer in ever smokers (4-6). These findings have been replicated among individuals with a family history of lung cancer, and the relative risk of lung cancer associated with markers in this region are much higher for familial cases compared to the relative risk observed among sporadic cases (7). Linkage analysis in families with aggregation of lung cancer also described a region on chromosome 6q23-25 associated with risk of lung cancer (8). The clinical significance of these findings is still unclear. In the meantime, lung cancer risk models using epidemiologic data have been developed, and the most parsimonious models for both ever and never smokers include a family history of cancer variable (9).

Developing risk models for lung cancer are vital, given the report from the National Lung Screening Trial (NLST), which suggests that low-dose helical computed tomography (CT) scans may cut deaths from lung cancer by 20% (10). It should be noted these findings were among individuals 55-74 years of age, with a smoking history of 30+ pack years, and should not be extended to other populations (11). A crucial component to a successful screening program is defining a population at high risk. Until screening protocols have been developed, the main recommendations to reduce risk of lung cancer remain smoking avoidance, cessation, and limiting exposure to known carcinogens (e.g. radon, environmental tobacco smoke)(12).

Relatives of individuals with lung cancer may be at higher risk of lung cancer than the general population. Various smaller studies have provided evidence that familial aggregation of lung cancer exists after adjusting for the aggregation of cigarette smoking and type of family relatedness (13, 14). In this study, we performed a pooled analysis of data contributed to the International Lung Cancer Consortium, in order to describe in greater detail familial aggregation of lung cancer. Given the large sample size of this analysis, we were able to perform subgroup analyses examining risk by gender, race/ethnicity, histologic type, age at diagnosis, and smoking status.

Materials and Methods

Study Population

The International Lung Cancer Consortium (ILCCO) was established in 2004, with the goal of sharing compatible data from lung cancer epidemiology studies to achieve greater power than from single studies alone. To date, 57 lung cancer studies are included in ILCCO. Further details regarding the aims, guidelines and policies of ILCCO are described in Hung et al.(15).

To be included in the main analysis of familial aggregation of lung cancer, the following minimum criteria were set. Each study must have collected data regarding the lung cancer status, age at diagnosis, smoking status of the proband, and vital status (living/deceased) for the mother, father and siblings of every case and control proband. Additional analyses were performed in 5 studies that also collected smoking status on each relative in addition to the minimum variables. Institutional approval and written informed consent from all subjects were obtained by the investigator at each study site for each original study and their de-identified data were submitted to ILCCO for pooling.

Statistical Methods

The individual-level data from each study included the following demographic variables for the case and control subjects (referred to as probands): gender, ethnicity, age at diagnosis (cases) or interview (controls), education, smoking status, and pack years smoked, and histological classification (cases only). Never smokers were individuals who reported smoking less than 100 cigarettes in their lifetime. Former smokers were individuals who reported smoking cessation at least 2 years prior to interview. Questions regarding these data were resolved by the original study investigator.

Relative risks of lung cancer associated with having a first-degree relative with lung cancer were assessed using unconditional logistic regression models to calculate odds ratios (ORs) and 95% confidence intervals. Estimates were adjusted for variables that differed significantly between probands or have been previously shown to be associated with lung cancer (gender, education, ethnicity, smoking status, pack years) and for study site. Logistic regression models were created for having any first degree relative with lung cancer, and then stratified by relative type (father, mother, sibling). To examine interactions between family history and race, a multiplicative variable of these two factors was added to the regression model. For all subgroup analyses except for histology, the controls were restricted to those from the same population (e.g., in the Asian subgroup analysis, only Asian controls were used). For histology subgroups, all controls (n=23305) were used. For the sub-set of studies where individual-level information was collected on both affected and unaffected fathers, mothers, and siblings, in addition to the probands, generalized estimating equation (GEE) regression models were created to determine whether familial risk of lung cancer was present after adjustment for risk factors among the relatives of the probands. These models correct for the correlation between family members (16), and adjusted for gender and smoking status of each first-degree relative. First, the dataset was restricted to first-degree relatives, and a variable to indicate whether each individual was related to a case or control proband was assigned to each subject. GEE models were then constructed for estimating relative risks in case relatives compared to control relatives, adjusting for proband age, proband gender, gender of the relative, smoking status of the proband and smoking status of the relative. SAS (Cary, NC) Version 9.2 was used for all analyses.

Results

Information regarding family history of cancer in first-degree relatives was available for 24,380 cases and 23,399 controls. Of the 57 studies which have contributed data to ILCCO, 24 studies met inclusion criteria for the analysis. Studies from North America accounted for 54% of the datasets analyzed (n=13), 8 were from Europe and 3 were from Asia and Oceania (Table 1). Population-based controls were ascertained by 11 studies, 8 studies employed hospital-based controls, and 5 studies utilized both population and hospital-based control probands (mixed).

Table 1.

Description of the studies included in the pooled analysis of familial aggregation of lung cancer

Principal Control Study Cases, Controls, Total,
Continent Study/Study Center investigator source period Location n n n
North America Samuel Lunenfeld Research
Institute (54)
J. McLaughlin Mixed 1997-2002 Toronto, Ontario, Canada 445 948 1393
University of California at San
Francisco (55)
J.K. Wiencke Population 1999-2002 San Francisco, USA 4314 933 5247
Mayo (56) P. Yang Mixed 1997-2006 USA 3533 1674 5207
*Harvard (57) D.C. Christiani Mixed 1991-
ongoing
Boston, USA 2253 1529 3782
*M.D. Anderson Cancer Center
(58)
M.R. Spitz Hospital 1992-
ongoing
Houston, USA 1841 2000 3841
*Wayne State University and the
Karmanos Cancer Institute (59, 60)
A.G. Schwartz Population 1984-2005 Detroit, USA 1587 1758 3345
*NY Multicenter Study(61) J.E. Muscat Hospital 1969-1999 New York State, USA 1320 1400 2720
University of California at Los
Angeles(62)
Z.F. Zhang
H. Morgenstern
Population 1999-2004 Los Angeles, USA 611 1040 1651
Maryland Lung & Prostate Cancer
Study (63)
C.C. Harris Mixed 1998-2009 Baltimore Metropolitan
Area, USA
551 735 1286
H. Lee Moffitt Cancer Center (64) P. Lazarus Hospital 1999-2003 Florida, USA 498 899 1397
New England Lung Cancer Study
(65)
E. Duell Population 2005-2008 New Hampshire, USA 276 251 527
Memorial Sloan-Kettering (66) I. Orlow Hospital 2005-2008 New York, USA 102 101 203
Europe Central Europe (67) P. Boffetta Hospital 1998-2002 Central/Eastern Europe 2633 2702 5335
LUCAS (68) I. Stucker Hospital 1990-1992 Paris, E. France 310 302 612
ESTHER (69) H. Brenner Population 2001-2003 Germany 206 206 412
CREST- Cancer of the
RESpiratory Tract
biorepository(70)
M. Neri Mixed 1996--
ongoing
N. Italy 413 555 968
The Nijmegen Lung Cancer
Study(71)
L.A.. Kiemeney Population Cases: 2008-
controls:
2002-
The Netherlands 535 2080 2615
Spain (71) J. I. Mayordomo Hospital 2006-- Aragon, Spain 350 1366 1716
Liverpool Lung Project (72) J.K. Field Population 1998-2006 Liverpool, UK 475 954 1429
ReSoLuCENT (73) P.J. Woll Population 2006-
ongoing
N. Trent, UK 214 204 418
Asia and
Oceania
Aichi (74) K. Matsuo Hospital 2001-2005 Japan 716 716 1432
Kyushu (75) C. Kiyohara Population 1994-1996 Japan 190 108 298
*Israel (29) G. Rennert Population 2005-- Israel 372 350 722
Hawaii (76) L. Le Marchand Population 1992-1997 Hawaii, USA 635 588 1223
Total 24380 23399 47779
*

Data from these study sites were included in the generalized estimating equations analysis (Tables 4 and 5). Note that sample sizes reported in early manuscripts for published studies are often smaller than what was included in this pooled analysis.

Table 2 describes characteristics of case and control probands included in the analysis. Men accounted for about half of the study population, and the majority of the population was white (82.3%), followed by Asian (6.8%), and African American (5.4%). Case probands were significantly more likely to be smokers compared to the control probands (p-value<0.0001), and among those who reported smoking, cases had a higher number of pack years compared to controls (p-value<0.0001).

Table 2.

Characteristics of probands included in the familial aggregation analysis

Cases (n=24380) Controls (n=23305) p-value
Gender
Men 13833 (56.7%) 12529 (53.8%) <0.0001
Women 10547 (43.3%) 10776 (46.2%)
Age
<30 73 (0.3%) 476 (2.0%) <0.0001
30≤age<40 551 (2.3%) 1388 (6.0%)
40≤age<50 2551 (10.5%) 3236 (13.9%)
50≤age<60 5458 (22.4%) 5764 (24.7%)
60≤age<70 7813 (32.3%) 7012 (30.1%)
70≤age<80 6480 (26.6%) 4689 (20.1%)
80+ 1454 (5.9%) 740 (3.2%)
Race/Ethnicity
White/Caucasian 20069 (82.3%) 19303 (82.8%) <0.0001
Hispanic/Latino 440 (1.8%) 670 (2.9%)
African American 1304 (5.4%) 1691 (7.3%)
Asian 1656 (6.8%) 1282 (5.5%)
American Indian and Alaskan
Native
185 (0.8%) 33 (0.1%)
Hawaiian/Pacific Islander 398 (1.6%) 150 (0.6%)
Other 169 (0.7%) 152 (0.7%)
Missing/Unknown 159 (0.7%) 24 (0.1%)
Education *
Low 6332 (26.0%) 4550 (19.5%) <0.0001
Medium 9023 (37.0%) 7800 (33.5%)
High 6459 (26.5%) 8513 (36.5%)
Missing 2566 (10.5%) 2442 (10.5%)
Type of Smoker
Never 3301 (13.5%) 8497 (36.5%) <0.0001
Ever** 20961 (86.0%) 14160 (60.8%)
 Former 7940 (32.6%) 7836 (33.6%)
 Current 9877 (40.5%) 5287 (22.7%)
Missing 118 (0.5%) 648 (2.8%)
Mean Pack Years (SD) 44.3 (33.3) 20.4 (25.8) <0.0001
Histology
Small cell carcinoma 2358 (9.7%)
Non-small cell carcinomas 18616 (76.4%)
 Squamous cell carcinomas 5781 (23.7%)
 Large cell carcinomas 1099 (4.5%)
 Adenocarcinoma 9285 (38.1%)
 Bronchioloalveolar carcinoma
  (BAC)
896 (3.7%)
 Non small cell carcinoma, not
 otherwise specified
1555 (6.4%)
Carcinoma§ 1797 (7.4%)
Carcinoid 347 (1.4%)
Others/missing 1262 (5.2%)
*

Low=less than high school, medium=high school diploma or equivalent or some college, high=college degree

**

Includes former smokers, current smokers, and subjects who were either former or current smokers

Pack years of smoking was missing for 9.61% of cases and 14.56% for controls

Non-small cell includes squamous cell, large cell, adenocarcinomas, bronchioloalveolar carcinoma , and non-small cell carcinoma, not otherwise specified

§

No further detail available

Risk of lung cancer associated with having a family history of lung cancer in a first-degree relative (mother, father, sibling) is presented in Table 3. Associations between lung cancer and family history were found in nearly every stratum after adjusting for proband age, proband gender, ethnicity, education, smoking status of the proband, pack years, and study number. Overall, individuals with a first-degree relative with lung cancer had a 1.51-fold increase in risk of lung cancer compared to individuals without a family history, after adjustment (95% CI: 1.39-1.63). There is evidence of interaction between smoking status and family history of lung cancer, with ever smoking individuals with a family history of lung cancer in a first degree relative having a 3.19-fold increase in risk of lung cancer compared to never smokers without a first-degree family history of lung cancer after adjustment (95% CI: 2.03-5.00, data not shown). When stratified by relative type, the association was greatest for those with a history of lung cancer in a sibling (OR=1.82, 95% CI: 1.62, 2.05), compared to lung cancer in a father (OR=1.25, 95% CI: 1.13, 1.39) or mother (OR=1.37 95% CI: 1.17, 1.61). This pattern was similar in male and female probands. There was a significant interaction between race and family history of lung cancer (p=0.002, data not shown). The overall association was strongest among Asians (OR=2.38, 95% CI: 1.50, 3.82), then African Americans (OR=1.67, 95% CI: 1.16, 2.40) and whites (OR=1.46 95% CI: 1.34, 1.58). A family history of lung cancer was associated with lung cancer in every histological type examined, after adjustment. Among never smoking individuals, only a history of lung cancer in a sibling was associated with lung cancer (OR=1.44, 95% CI: 1.07, 1.93). For ever smokers, lung cancer was associated with history of lung cancer in a father (OR=1.27, 95% CI: 1.13, 1.43), mother (OR=1.40, 95% CI: 1.17, 1.66) and sibling (OR=1.91, 95% CI: 1.68, 2.17), after adjustment. The association was stronger in cases diagnosed before the age of 50 than in later onset patients (OR=1.83 and 1.45, respectively). Having a history of lung cancer in siblings among probands <50 years old strengthened the association even further (OR=3.72, 95% CI: 2.00, 6.90), double the odds ratio for the older cases (OR=1.86, 95% CI: 1.65, 2.09). The number of affected relatives among cases and controls, by strata, are available in Supplementary Table 1.

Table 3.

The association between family history of lung cancer and lung cancer, by relative type

Family History of
Lung Cancer in
1st Degree
Relative
Family History of
Lung Cancer in
Father
Family History of
Lung Cancer in
Mother
Family History of
Lung Cancer in
Sibling
OR (95% CI) OR (95% CI) OR (95% CI) OR (95% CI)
Overall 1.51 (1.39,1.63) 1.25 (1.13,1.39) 1.37 (1.17,1.61) 1.82 (1.62,2.05)
Proband Gender
Male 1.53 (1.37,1.70) 1.23 (1.06,1.43) 1.32 (1.05,1.66) 2.07 (1.76,2.45)
Female 1.48 (1.33,1.65) 1.27 (1.09,1.48) 1.43 (1.15,1.77) 1.59 ( 1.35,1.87)
Proband Ethnicity
White 1.46 (1.34,1.58) 1.19 (1.07,1.34) 1.37 (1.16,1.62) 1.77 (1.56,2.00)
Black/African American 1.67 (1.16,2.40) 1.48 (0.87,2.54) 1.13 (0.55,2.30) 2.07 (1.19, 3.60)
Asian 2.38(1.50, 3.82) 2.82(1.54, 5.70) 0.97(0.39,2.38) 4.35(1.83,10.34)
Proband Histology
Small cell carcinoma 1.51 (1.33,1.70) 1.33 (1.13,1.57) 1.31 (1.00,1.73) 1.63 (1.36,1.95)
Non-small cell carcinoma 1.58 (1.44, 1.73) 1.40 (1.23,1.59) 1.28 (1.07,1.54) 1.77 (1.55,2.02)
 Squamous cell
 carcinoma
1.54 (1.39,1.72) 1.32 (1.14,1.53) 1.16 (0.91,1.49) 1.85 (1.59,2.16)
 Large cell carcinoma 1.81(1.60,2.06) 1.55(1.30,1.92) 1.58(1.25,2.10) 1.23(1.78,2.56)
 Adenocarcinoma 1.59 (1.45,1.74) 1.25 (1.10,1.43) 1.61 (1.34,1.93) 1.85 (1.61,2.12)
 BAC 1.56(1.45, 1.69) 1.09(0.90, 1.35) 1.68(1.24,2.12) 1.93(1.64,2.70)
 Non-small cell
 carcinoma, nos Carcinoma
1.78 (1.58,2.01) 1.48 (1.24,1.75) 2.24 (1.78,2.83) 1.73 (1.44,2.08)
Carcinoid 1.28(1.13, 1.45) 0.92(0.69, 1.10) 1.48(1.16, 1.92) 1.78(1.48, 2.42)
Others/missing 1.30(1.17, 1.48) 1.10(0.96, 1.35) 1.20(0.95, 1.62) 1.85(1.55, 2.30)
Proband Smoking Status
Never Smokers 1.25 (1.03,1.52) 1.09 (0.82,1.45) 1.10 (0.74,1.66) 1.44 (1.07,1.93)
Ever Smokers 1.55 (1.42,1.68) 1.27 (1.13,1.43) 1.40 (1.17,1.66) 1.91 (1.68,2.17)
 Former Smokers 1.46 (1.29,1.65) 1.23 (1.03,1.47) 1.26 (0.97,1.62) 1.83 (1.53,2.18)
 Current Smokers 1.57 (1.37,1.79) 1.33 (1.11,1.59) 1.51 (1.15,1.99) 1.85 (1.48,2.32)
Proband Age at Onset
<50 1.83 (1.47,2.28) 1.68 (1.28,2.20) 1.51 (1.04,2.20) 3.72 (2.00,6.90)
50+ 1.45 (1.33,1.57) 1.10 (0.97,1.24) 1.27 (1.06,1.52) 1.86 (1.65,2.09)

All ORs are adjusted for age, gender, ethnicity, education, smoker type, pack years and study site, where appropriate.

The association between lung cancer and family history increased as the number of first-degree relatives with lung cancer increased. The association between family history and lung cancer increased from 1.45 (95% CI: 1.34, 1.58) for those individuals who reported one first-degree relative with lung cancer to 1.97 (95% CI: 1.59, 2.45) for those individuals with 2 or more first-degree affected relatives (data not shown).

Tables 4 and 5 include studies where individual-level information was collected on both affected and unaffected fathers, mothers, and siblings, in addition to the probands. As seen in Table 4, data were available for mothers of 6297 cases and 5907 controls, with 240 case mothers having lung cancer (3.8%) and 171 control mothers having lung cancer (2.9%). Data were also available for 6,263 fathers of cases and 5,886 fathers of controls, with 453 case fathers having lung cancer (7.2%) and 363 control fathers having lung cancer (6.2%). Data were available for 18,948 siblings of cases and 16,625 siblings of controls, with 724 case siblings reported to have lung cancer (3.8%) and 282 control siblings reported to have lung cancer (1.7%). Similar mean ages at diagnosis were reported for case and control fathers (p-value=0.16), mothers (p-value=0.08) and siblings (p-value=0.72).

Table 4.

Characteristics of relatives included in the familial aggregation analysis using generalized estimating equations

Case Relatives Control Relatives P-value
Mothers (n= 12204) n=6297 n=5907
 Ever Smoker 2123 (38.0%) 2270 (40.4%) 0.009
 Never Smoker 3465 (62.0%) 3349 (59.6%)
Number (%) affected w/Lung cancer: 240 (3.8%) 171 (2.9%)
 Median age at diagnosis 68 70
 Mean age at diagnosis (±SD) 66.8±13.0 69.0±10.4 0.08
 Missing age at diagnosis 6 (2.5%) 4 (2.3%) 0.92
Fathers (n= 12149) 6263 5886
 Ever Smoker 3945 (72.7%) 3939 (72.2%) 0.57
 Never Smoker 1483 (27.3%) 1517 (27.8%)
Number (%) affected w/Lung cancer: 453 (7.2%) 363 (6.2%)
 Median age at diagnosis 65 67
 Mean age at diagnosis (±SD) 64.8±10.4 65.9±10.8 0.16
 Missing age at diagnosis 27 (6.0%) 12 (3.3%) 0.08
Siblings (n= 35573) 18948 16625
 Ever Smoker 9869 (59.1%) 8107 (51.5%) <0.0001
 Never Smoker 6819 (40.9%) 7635 (48.5%)
Number (%) affected w/Lung cancer: 724 (3.8%) 282 (1.7%)
 Median age at diagnosis 60 61
 Mean age at diagnosis (±SD) 60.2±11.3 60.0±11.3 0.72
 Missing age at diagnosis 27 (3.7%) 11 (3.9%) 0.90

Table 5.

Odds ratios (95% confidence interval) of risk of lung cancer among first-degree relatives of cases compared to first-degree relatives of controls among subjects included in the generalized estimating equations analysis

# affected
relatives with
lung cancer
# unaffected
relatives
Lung Cancer Risk among First-
Degree Relatives
Adjusted OR (95%CI)
Overall 2233 57693 1.55 (1.39, 1.73)
Proband Gender
 Male 969 26230 1.34 (1.14, 1.58)
 Female 1264 31463 1.74 (1.50, 2.00)
Proband Ethnicity
 White 2093 51842 1.53 (1.37, 1.71)
 Black/African American 122 5306 2.09 (1.28, 3.42)
Proband Histology
 Small cell carcinoma 88 1973 2.45 (1.72, 3.48)
 Non-small cell carcinoma 1167 24738 2.46 (2.02, 2.98)
Proband Smoking Status
 Never Smokers 393 14972 1.38 (1.08, 1.75)
 Ever Smokers 1833 42634 1.57 (1.42, 1.74)
  Ex-Smokers 989 20837 1.62 (1.38, 1.91)
  Current Smokers 841 21670 1.61 (1.34, 1.93)
Proband Age of Onset
  <50 298 15184 1.97 (1.51, 2.58)
  50+ 1935 42509 1.53 (1.36, 1.72)
Relationship to Proband
  Father 816 11333 1.33 (1.13, 1.57)
  Mother 411 11793 1.39 (1.12, 1.75)
  Siblings 1006 34567 1.96 (1.65, 2.34)
Relative Smoking Status
  Never Smoker 177 24091 1.31 (0.96, 1.78)
  Ever Smoker 1750 28503 1.57 (1.41, 1.76)

ORs adjusted for: gender of proband, proband ethnicity, proband histology, proband smoking status, proband age of onset, relationship to proband, gender of relative, smoking status of relative (where appropriate).

Table 5 presents the strength of familial aggregation of lung cancer after adjusting for proband age, proband gender, proband smoking status, relative’s gender, and relative’s smoking status. The association with family history was 1.55-fold greater in first-degree relatives of cases compared to relatives of controls (95% CI: 1.39, 1.73), after adjustment. The association between lung cancer and family history was elevated for case relatives in all subgroups compared to control relatives. Family history was a stronger risk factor for relatives of early-onset (<50 years of age) cases (OR=1.97, 95% CI: 1.51, 2.58) than for those related to someone with later onset disease (OR=1.53, 95% CI: 1.36, 1.72). The association was greater for those who had a sibling with lung cancer (OR=1.96, 95% CI: 1.65, 2.34) compared to a father (OR=1.33, 95% CI: 1.13, 1.57) or mother (OR=1.39, 95% CI: 1.12, 1.75) with the disease. In a sub-analysis of siblings with never smoking parents, the association between having a first-degree relative with lung cancer and lung cancer risk remained (OR=2.18, 95% CI: 1.52-3.11) after adjustment (data not shown). Never smoking relatives of cases were somewhat more likely to have lung cancer compared to never smoking relatives of controls (OR=1.31, 95% CI: 0.96, 1.78), but this association was not statistically significant. Individuals who were ever smokers and related to a case were 1.57-fold more likely to have lung cancer than smokers who were related to a control (95% CI: 1.41, 1.76).

Discussion

The results presented here represent the most comprehensive analysis of the association between family history of lung cancer and lung cancer since the first strong evidence of familial aggregation was reported nearly 50 years ago (17). To date, this is the largest pooled analysis which incorporated a traditional case-control analysis and also used data from individual family members to examine risk adjusted for gender and smoking status of each relative. Individuals with family history in a first-degree relative are at an approximate 50% increased risk of lung cancer compared to those without a family history, and this association remains regardless of gender, race/ethnicity, histological type and after adjusting for other known lung cancer risk factors.

The majority of lung cancers are diagnosed in current or former smokers, and environmental tobacco smoke also increases risk (18, 19). Given the strong evidence linking lung cancer etiology to well-identified occupational or environmental sources, less research has focused on other causes of this disease, including the influence of family history. Twin studies, especially those that can compare concordance between monozygotic and dizygotic twins, can provide information on whether familial aggregation is due to inherited or environmental factors. Evidence of limited heritability of lung cancers has been reported from population-based registry data in Utah, Sweden, Denmark and Finland (20-22). Even in these large, population-based studies, power is limited for most cancers, including lung. The magnitude of risk associated with having a family history of lung cancer are similar to those associated with familial aggregation of colon cancer (23), prostate cancer (24), and breast cancer in non-BRCA families (25). Stronger associations have been reported when the relative had early-onset disease for these cancers (23, 26, 27), as well as for lung cancer (28-30). In this pooled analysis, the association was stronger for individuals who were diagnosed with lung cancer prior to age 50, or who had a family history of lung cancer in a relative under the age 50. The difference was most pronounced among siblings, with the odds in early-onset lung cancer nearly double the odds in later onset probands.

Overall, having a sibling with lung cancer conferred the strongest association, and siblings of cases were at increased risk compared to siblings of controls. Aggregation studies of other cancer types such as prostate (31, 32) and breast (25) also report evidence of a stronger association with affected siblings compared to parents, yet this is less established for colon cancer (33). The association between family history and lung cancer remained for siblings even after adjustment for cigarette smoking in the relative. Similarly, among never smoking probands, the association was greater for those who report lung cancer in a sibling, rather than a parent. The association also remained in siblings from non-smoking households (i.e. both parents were never smokers). Finally, after adjustment for smoking in both the proband and the sibling, there remained strong evidence of an association between family history and lung cancer in the siblings of cases compared to the siblings of controls. Relative type should be considered when assessing risk associated with family history of lung cancer. In general, an elevated risk of disease in siblings signals a recessive effect (34). The elevated risk from affected siblings might be a result from shared environment exposures childhood, or indicate there are recessive genes involved. In the five studies that provided information on all siblings, cases were not more likely to have a sibling compared to controls (82.4% and 83.8%, respectively), and sibships were similar in size, with an average of three siblings for both cases and controls. Adjusting for number of siblings did not change our results. Nevertheless, another factor that needs to be considered when interpreting the strong sibling risk is the age distribution, as affected siblings often have younger ages of onset compared to the affected parents of the case probands. In our study, the mean age of diagnosis for affected siblings and affected parents of the index cases was 60.2 and 66.2, respectively. Therefore, the stronger familial relative risks observed in siblings might be partially associated with an earlier age of onset. These findings may also be the result of the proband having greater recall of the cancer status of their generation (versus their parent’s generation) and better diagnostic techniques in later years, but could also represent evidence of the genetic contribution to lung cancer etiology.

Most studies of lung cancer and familial aggregation included mainly non-small cell lung cancers. While it is difficult to ascertain the histology of lung cancer in the relatives, we were able to ascertain the histological type of lung cancer for the probands and to stratify results according to type. Despite the different etiologies and outcomes associated with various histological types, the odds ratios fell in a fairly narrow range (ORrange 1.28-1.81). For example, the rare (~1%) carcinoid tumors of the lung, which are not considered to be smoking-associated and are characterized by an excellent overall survival (35), showed an association with a positive family history of lung cancer (OR=1.28, 95% CI: 1.13-1.45), confirming in a large dataset early reports from Hassan et al. (2008)(36). The association with more common (15%) and lethal small cell carcinomas (37, 38) is slightly higher (OR=1.51, 95% CI: 1.33, 1.70), as anticipated by a recent meta-analysis (39). Overall, the similar magnitudes of the effect of family history on lung cancer across various histological types suggest that familial aggregation may be independent of the association with cigarette smoking. The hypothesis that familial aggregation of lung cancer may be associated with traits which act independently of cigarette smoking has received support from the results of a follow up study of the chromosome 6q region which reports a lung cancer risk-associated haplotype irrespective of degree of smoking yielded similar risk in carriers (40). The search for other risk factors unrelated to smoking remains crucial, given the number of lung cancer seen among never smokers.

Global estimates suggest that approximately 25% of lung cancer cases world-wide occur among never smoking individuals (41, 42). In this analysis, approximately 14% of all lung cancers were diagnosed in never smokers, and family history of lung cancer was associated with having lung cancer. Various other exposures that were unmeasured in these populations may account for some of this risk, although studies among never smokers with lung cancer, even after adjusting for other known carcinogenic exposures, support family history as an important risk factor (43-45).

The association between having a family history of lung cancer in a first-degree relative and lung cancer varies by ethnicity. The associations were highest among Asians and lowest among whites, with estimates for African Americans falling in between. While some studies have reported differences in risk of lung cancer associated with cigarette smoking by race/ethnicity, (46-48) few report estimates for family history by race/ethnicity, as study populations of individual studies are relatively homogenous. In the current pooled analysis, odds ratios were higher for African Americans, but not statistically different from results in the white populations. Studies investigating Asian populations reported risk estimates for familial aggregation of lung cancer similar to the 2-fold increases found in the present pooled analysis (49, 50). Lower background rates of disease may account for these apparently stronger associations in the Asian population. As smoking patterns, environmental and occupational exposures, diet, and genetics differ by race/ethnicity, it is not unexpected that the strength of the association between family history and lung cancer risk would also vary.

The findings presented are not without limitations. First, all studies relied on self-report of family history and thus may be prone to recall bias. Confirming each lung cancer among relatives would be ideal but time and cost-prohibitive. Studies that have validated the reported family history through another source consistently report high accuracy (sensitivity >0.80) for lung cancers (51-53). Similarly, we were unable to perform a standardized pathologic review to confirm histological type, in the probands or the affected relatives. Next, the methodologies used to ascertain controls differed among the studies. When we limited our analyses to studies using the same control source, findings were essentially the same. Only parents and siblings are included in our analysis, as six studies did not collect information on offspring, and another two studies were focused on early-onset disease, where the majority of the offspring were under 21 years of age. Lastly, pooled analyses are limited by the number of common variables collected by each study. Age, gender, ethnicity, education and smoking history were collected for all study subjects, which are the major known risk factors for lung cancer. It is possible that there remains residual confounding by smoking, as smoking status does not fully assess tobacco smoke exposure. Information on environmental tobacco smoke exposure (in childhood or as an adult) was not collected in a uniform manner, therefore we were not able to adjust for it in a standardize manner, hence our findings may be confounded by this omission. Relative information was more likely to be missing certain variables compared to proband data, and we did not adjust models for family size, as this was not available for the majority of the studies. When we restricted our analyses to those studies with a family size variable, the results were not significantly different.

In summary, family history is a simple proxy for genetic risk, and is influenced by both shared and individual environmental exposures. The association between lung cancer and family history of lung cancer remains even after adjustment for smoking. Unlike germline mutation testing, family history assessment is fairly straightforward and inexpensive to obtain while taking a patient’s history. While the role of genetic variation in the etiology of lung cancer is not yet defined, family history assessment is immediately available and those with a positive history represent a higher risk group. A positive family history of lung cancer should possibly be one of the variables considered in the selection of a population eligible for lung cancer screening.

Supplementary Material

01

Acknowledgements

Drs. Cote, Liu, Etzel and Hung had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

The contributions of the following individuals: Christa Stegmaier, Hartwig Ziegler, Sonja Wolf and Volker Herrmann (ESTHER study) Urvi Mujumdar and Radhai Rastogi (MSKCC), Lynda Forbes, Yvonne Bush, Kelly Montgomery and Angela Wenzlaff (WSU), Dr. Tomas Lynch and Dr. John Wain, (Mass General Hosp), Dr. Kofi Asomaning and Ms. Andrea Shafer (Harvard School of Public Health) are gratefully acknowledged.

They do wish to acknowledge the following support:

ILCCO data management is supported by Canadian Cancer Society (CCSRI no. 020214) and Cancer Care Ontario Research Chair Award.

MLC: NIH K07 CA125203-01A2

AGS: NIH R01CA060691, NIH R01CA87895, NIH N01-PC35145, NIH P30CA22453

DCC: Grants: NIH (NCI) RO1CA74386 and P50 CA090578

GR: Grant number 2003159 from the Bi-national Israel US Science Foundation (BSF)

HB and HM: ESTHER study was supported by a grant from the Baden-Württemberg Ministry of Science Research and Arts.

IO: Steps for Breath and the Labrecque Foundation; and the Society of Memorial Sloan-Kettering Cancer Center

JL: The Warsaw study part of the study was supported by local grant from The Polish State Committee for Scientific Research, grant no. SPUBM-COPERNICUS/P-05/DZ-30/99/2000.

PJW: ReSoLuCENT study funded by the Weston Park Hospital Charitable Trust

Financial support from European Commission (DG-XII), contract no. IC15-CT96-0313 is gratefully acknowledged.

The Nijmegen Ling Cancer Study was funded by an investment grant of the Radboud University Nijmegen Medical Centre.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of interest statement: The authors declare no conflicts of interest exist.

References

  • 1.Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN. Int J Cancer. 2008 Jun 17; doi: 10.1002/ijc.25516. [DOI] [PubMed] [Google Scholar]
  • 2.Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, et al. Cancer statistics, 2008. CA Cancer J Clin. 2008 Mar-Apr;58(2):71–96. doi: 10.3322/CA.2007.0010. [DOI] [PubMed] [Google Scholar]
  • 3.United States Department of Health and Human Services . Department of Health and Human Services Centers for Disease Control and Prevention. National Center for Chronic Disease Prevention and Health Promotion; Atlanta, GA: 2004. The Health Consequences of Smoking: A Report of the Surgeon General. [Google Scholar]
  • 4.Amos CI, Wu X, Broderick P, Gorlov IP, Gu J, Eisen T, et al. Genome-wide association scan of tag SNPs identifies a susceptibility locus for lung cancer at 15q25.1. Nat Genet. 2008 May;40(5):616–22. doi: 10.1038/ng.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hung RJ, McKay JD, Gaborieau V, Boffetta P, Hashibe M, Zaridze D, et al. A susceptibility locus for lung cancer maps to nicotinic acetylcholine receptor subunit genes on 15q25. Nature. 2008 Apr 3;452(7187):633–7. doi: 10.1038/nature06885. [DOI] [PubMed] [Google Scholar]
  • 6.Thorgeirsson TE, Geller F, Sulem P, Rafnar T, Wiste A, Magnusson KP, et al. A variant associated with nicotine dependence, lung cancer and peripheral arterial disease. Nature. 2008 Apr 3;452(7187):638–42. doi: 10.1038/nature06846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Liu P, Vikis HG, Wang D, Lu Y, Wang Y, Schwartz AG, et al. Familial aggregation of common sequence variants on 15q24-25.1 in lung cancer. J Natl Cancer Inst. 2008 Sep 17;100(18):1326–30. doi: 10.1093/jnci/djn268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bailey-Wilson JE, Amos CI, Pinney SM, Petersen GM, de Andrade M, Wiest JS, et al. A major lung cancer susceptibility locus maps to chromosome 6q23-25. Am J Hum Genet. 2004 Sep;75(3):460–74. doi: 10.1086/423857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.D’Amelio AM, Jr., Cassidy A, Asomaning K, Raji OY, Duffy SW, Field JK, et al. Comparison of discriminatory power and accuracy of three lung cancer risk models. Br J Cancer. 2010 Jul 27;103(3):423–9. doi: 10.1038/sj.bjc.6605759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Reduced Lung-Cancer Mortality with Low-Dose Computed Tomographic Screening. N Engl J Med. 2011 Jun 29; doi: 10.1056/NEJMoa1102873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Peres J. Lung cancer screening: ready for prime time? J Natl Cancer Inst. Jan 19;103(2):89–91. doi: 10.1093/jnci/djq555. [DOI] [PubMed] [Google Scholar]
  • 12.National Cancer Institute . PDQ Lung Cancer Prevention. Bethesda, MD: 2010. [Google Scholar]
  • 13.Gu J, Hua F, Zhong D, Chen J, Liu H, Zhou Q. [Systematic review of the relationship between family history of lung cancer and lung cancer risk] Zhongguo Fei Ai Za Zhi. 2010 Mar;13(3):224–9. doi: 10.3779/j.issn.1009-3419.2010.03.07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Matakidou A, Eisen T, Houlston RS. Systematic review of the relationship between family history and lung cancer risk. Br J Cancer. 2005 Oct 3;93(7):825–33. doi: 10.1038/sj.bjc.6602769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hung RJ, Christiani DC, Risch A, Popanda O, Haugen A, Zienolddiny S, et al. International Lung Cancer Consortium: pooled analysis of sequence variants in DNA repair and cell cycle pathways. Cancer Epidemiol Biomarkers Prev. 2008 Nov;17(11):3081–9. doi: 10.1158/1055-9965.EPI-08-0411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zeger SL, Liang KY. Longitudinal data analysis for discrete and continuous outcomes. Biometrics. 1986 Mar;42(1):121–30. [PubMed] [Google Scholar]
  • 17.Tokuhata GK, Lilienfeld AM. Familial aggregation of lung cancer among hospital patients. Public Health Rep. 1963 Apr;78:277–83. [PMC free article] [PubMed] [Google Scholar]
  • 18.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 Jan 8;377(9760):139–46. doi: 10.1016/S0140-6736(10)61388-8. [DOI] [PubMed] [Google Scholar]
  • 19.Tong L, Spitz MR, Fueger JJ, Amos CA. Lung carcinoma in former smokers. Cancer. 1996 Sep 1;78(5):1004–10. doi: 10.1002/(SICI)1097-0142(19960901)78:5<1004::AID-CNCR10>3.0.CO;2-6. [DOI] [PubMed] [Google Scholar]
  • 20.Goldgar DE, Easton DF, Cannon-Albright LA, Skolnick MH. Systematic population-based assessment of cancer risk in first-degree relatives of cancer probands. J Natl Cancer Inst. 1994 Nov 2;86(21):1600–8. doi: 10.1093/jnci/86.21.1600. [DOI] [PubMed] [Google Scholar]
  • 21.Hemminki K, Vaittinen P. Familial cancers in a nationwide family cancer database: age distribution and prevalence. Eur J Cancer. 1999 Jul;35(7):1109–17. doi: 10.1016/s0959-8049(99)00066-0. [DOI] [PubMed] [Google Scholar]
  • 22.Lichtenstein P, Holm NV, Verkasalo PK, Iliadou A, Kaprio J, Koskenvuo M, et al. Environmental and heritable factors in the causation of cancer--analyses of cohorts of twins from Sweden, Denmark, and Finland. N Engl J Med. 2000 Jul 13;343(2):78–85. doi: 10.1056/NEJM200007133430201. [DOI] [PubMed] [Google Scholar]
  • 23.Fuchs CS, Giovannucci EL, Colditz GA, Hunter DJ, Speizer FE, Willett WC. A prospective study of family history and the risk of colorectal cancer. N Engl J Med. 1994 Dec 22;331(25):1669–74. doi: 10.1056/NEJM199412223312501. [DOI] [PubMed] [Google Scholar]
  • 24.Steinberg GD, Carter BS, Beaty TH, Childs B, Walsh PC. Family history and the risk of prostate cancer. Prostate. 1990;17(4):337–47. doi: 10.1002/pros.2990170409. [DOI] [PubMed] [Google Scholar]
  • 25.Pharoah PD, Day NE, Duffy S, Easton DF, Ponder BA. Family history and the risk of breast cancer: a systematic review and meta-analysis. Int J Cancer. 1997 May 29;71(5):800–9. doi: 10.1002/(sici)1097-0215(19970529)71:5<800::aid-ijc18>3.0.co;2-b. [DOI] [PubMed] [Google Scholar]
  • 26.Familial breast cancer: collaborative reanalysis of individual data from 52 epidemiological studies including 58,209 women with breast cancer and 101,986 women without the disease. Lancet. 2001 Oct 27;358(9291):1389–99. doi: 10.1016/S0140-6736(01)06524-2. [DOI] [PubMed] [Google Scholar]
  • 27.Keetch DW, Rice JP, Suarez BK, Catalona WJ. Familial aspects of prostate cancer: a case control study. J Urol. 1995 Dec;154(6):2100–2. [PubMed] [Google Scholar]
  • 28.Cassidy A, Balsan J, Vesin A, Wu X, Liloglou T, Brambilla C, et al. Cancer diagnosis in first-degree relatives and non-small cell lung cancer risk: results from a multi-centre case-control study in Europe. Eur J Cancer. 2009 Nov;45(17):3047–53. doi: 10.1016/j.ejca.2009.05.006. [DOI] [PubMed] [Google Scholar]
  • 29.Etzel CJ, Lu M, Merriman K, Liu M, Vaporciyan A, Spitz MR. An epidemiologic study of early onset lung cancer. Lung Cancer. 2006 May;52(2):129–34. doi: 10.1016/j.lungcan.2005.11.018. [DOI] [PubMed] [Google Scholar]
  • 30.Kreuzer M, Kreienbrock L, Gerken M, Heinrich J, Bruske-Hohlfeld I, Muller KM, et al. Risk factors for lung cancer in young adults. Am J Epidemiol. 1998 Jun 1;147(11):1028–37. doi: 10.1093/oxfordjournals.aje.a009396. [DOI] [PubMed] [Google Scholar]
  • 31.Cerhan JR, Parker AS, Putnam SD, Chiu BC, Lynch CF, Cohen MB, et al. Family history and prostate cancer risk in a population-based cohort of Iowa men. Cancer Epidemiol Biomarkers Prev. 1999 Jan;8(1):53–60. [PubMed] [Google Scholar]
  • 32.Lesko SM, Rosenberg L, Shapiro S. Family history and prostate cancer risk. Am J Epidemiol. 1996 Dec 1;144(11):1041–7. doi: 10.1093/oxfordjournals.aje.a008876. [DOI] [PubMed] [Google Scholar]
  • 33.Johns LE, Houlston RS. A systematic review and meta-analysis of familial colorectal cancer risk. Am J Gastroenterol. 2001 Oct;96(10):2992–3003. doi: 10.1111/j.1572-0241.2001.04677.x. [DOI] [PubMed] [Google Scholar]
  • 34.Ott J. Analysis of human genetic linkage. 3rd ed The Johns Hopkins University Press; Baltimore, MD: 1999. [Google Scholar]
  • 35.Bertino EM, Confer PD, Colonna JE, Ross P, Otterson GA. Pulmonary neuroendocrine/carcinoid tumors: a review article. Cancer. 2009 Oct 1;115(19):4434–41. doi: 10.1002/cncr.24498. [DOI] [PubMed] [Google Scholar]
  • 36.Hassan MM, Phan A, Li D, Dagohoy CG, Leary C, Yao JC. Family history of cancer and associated risk of developing neuroendocrine tumors: a case-control study. Cancer Epidemiol Biomarkers Prev. 2008 Apr;17(4):959–65. doi: 10.1158/1055-9965.EPI-07-0750. [DOI] [PubMed] [Google Scholar]
  • 37.Atlekruse SF, Kosary CL, Krapcho M, et al. [cited based on November 2009];SEER Cancer Statistics Review, 1975-2007. 2010 SEER data submission, posted to the SEER web site, 2010; Available from: http://seer.cancer.gov/csr/1975_2007/
  • 38.Brownson RC, Chang JC, Davis JR. Gender and histologic type variations in smoking-related risk of lung cancer. Epidemiology. 1992 Jan;3(1):61–4. doi: 10.1097/00001648-199201000-00012. [DOI] [PubMed] [Google Scholar]
  • 39.Lissowska J, Foretova L, Dabek J, Zaridze D, Szeszenia-Dabrowska N, Rudnai P, 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 Jul;21(7):1091–104. doi: 10.1007/s10552-010-9537-2. [DOI] [PubMed] [Google Scholar]
  • 40.Amos CI, Pinney SM, Li Y, Kupert E, Lee J, de Andrade MA, et al. A susceptibility locus on chromosome 6q greatly increases lung cancer risk among light and never smokers. Cancer Res. 2010 Mar 15;70(6):2359–67. doi: 10.1158/0008-5472.CAN-09-3096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin. 2005 Mar-Apr;55(2):74–108. doi: 10.3322/canjclin.55.2.74. [DOI] [PubMed] [Google Scholar]
  • 42.Sun S, Schiller JH, Gazdar AF. Lung cancer in never smokers--a different disease. Nat Rev Cancer. 2007 Oct;7(10):778–90. doi: 10.1038/nrc2190. [DOI] [PubMed] [Google Scholar]
  • 43.Schwartz AG, Yang P, Swanson GM. Familial risk of lung cancer among nonsmokers and their relatives. Am J Epidemiol. 1996 Sep 15;144(6):554–62. doi: 10.1093/oxfordjournals.aje.a008965. [DOI] [PubMed] [Google Scholar]
  • 44.Wu AH, Fontham ET, Reynolds P, Greenberg RS, Buffler P, Liff J, et al. Family history of cancer and risk of lung cancer among lifetime nonsmoking women in the United States. Am J Epidemiol. 1996 Mar 15;143(6):535–42. doi: 10.1093/oxfordjournals.aje.a008783. [DOI] [PubMed] [Google Scholar]
  • 45.Wu PF, Lee CH, Wang MJ, Goggins WB, Chiang TA, Huang MS, et al. Cancer aggregation and complex segregation analysis of families with female non-smoking lung cancer probands in Taiwan. Eur J Cancer. 2004 Jan;40(2):260–6. doi: 10.1016/j.ejca.2003.08.021. [DOI] [PubMed] [Google Scholar]
  • 46.Harris RE, Zang EA, Anderson JI, Wynder EL. Race and sex differences in lung cancer risk associated with cigarette smoking. Int J Epidemiol. 1993 Aug;22(4):592–9. doi: 10.1093/ije/22.4.592. [DOI] [PubMed] [Google Scholar]
  • 47.Hinds MW, Stemmermann GN, Yang HY, Kolonel LN, Lee J, Wegner E. Differences in lung cancer risk from smoking among Japanese, Chinese and Hawaiian women in Hawaii. Int J Cancer. 1981 Mar 15;27(3):297–302. doi: 10.1002/ijc.2910270307. [DOI] [PubMed] [Google Scholar]
  • 48.Le Marchand L, Wilkens LR, Kolonel LN. Ethnic differences in the lung cancer risk associated with smoking. Cancer Epidemiol Biomarkers Prev. 1992 Jan-Feb;1(2):103–7. [PubMed] [Google Scholar]
  • 49.Nitadori J, Inoue M, Iwasaki M, Otani T, Sasazuki S, Nagai K, et al. Association between lung cancer incidence and family history of lung cancer: data from a large-scale population-based cohort study, the JPHC study. Chest. 2006 Oct;130(4):968–75. doi: 10.1378/chest.130.4.968. [DOI] [PubMed] [Google Scholar]
  • 50.Wang XR, Yu IT, Chiu YL, Qiu H, Fu Z, Goggins W, et al. Previous pulmonary disease and family cancer history increase the risk of lung cancer among Hong Kong women. Cancer Causes Control. 2009 Jul;20(5):757–63. doi: 10.1007/s10552-008-9289-4. [DOI] [PubMed] [Google Scholar]
  • 51.Chang ET, Smedby KE, Hjalgrim H, Glimelius B, Adami HO. Reliability of self-reported family history of cancer in a large case-control study of lymphoma. J Natl Cancer Inst. 2006 Jan 4;98(1):61–8. doi: 10.1093/jnci/djj005. [DOI] [PubMed] [Google Scholar]
  • 52.Douglas FS, O’Dair LC, Robinson M, Evans DG, Lynch SA. The accuracy of diagnoses as reported in families with cancer: a retrospective study. J Med Genet. 1999 Apr;36(4):309–12. [PMC free article] [PubMed] [Google Scholar]
  • 53.Soegaard M, Jensen A, Frederiksen K, Hogdall E, Hogdall C, Blaakaer J, et al. Accuracy of self-reported family history of cancer in a large case-control study of ovarian cancer. Cancer Causes Control. 2008 Jun;19(5):469–79. doi: 10.1007/s10552-007-9108-3. [DOI] [PubMed] [Google Scholar]
  • 54.Brenner DR, Hung RJ, Tsao MS, Shepherd FA, Johnston MR, Narod S, et al. Lung cancer risk in never-smokers: a population-based case-control study of epidemiologic risk factors. BMC Cancer. 2010 Jun 14;10(1):285. doi: 10.1186/1471-2407-10-285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Aldrich MC, Selvin S, Hansen HM, Barcellos LF, Wrensch MR, Sison JD, et al. CYP1A1/2 haplotypes and lung cancer and assessment of confounding by population stratification. Cancer Res. 2009 Mar 15;69(6):2340–8. doi: 10.1158/0008-5472.CAN-08-2576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Li Y, Sun Z, Wu Y, Babovic-Vuksanovic D, Cunningham JM, Pankratz VS, et al. Cystic fibrosis transmembrane conductance regulator gene mutation and lung cancer risk. Lung Cancer. 2010 Jan 28; doi: 10.1016/j.lungcan.2010.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Zhai R, Liu G, Zhou W, Su L, Heist RS, Lynch TJ, et al. Vascular endothelial growth factor genotypes, haplotypes, gender, and the risk of non-small cell lung cancer. Clin Cancer Res. 2008 Jan 15;14(2):612–7. doi: 10.1158/1078-0432.CCR-07-1655. [DOI] [PubMed] [Google Scholar]
  • 58.Etzel CJ, Amos CI, Spitz MR. Risk for smoking-related cancer among relatives of lung cancer patients. Cancer Res. 2003 Dec 1;63(23):8531–5. [PubMed] [Google Scholar]
  • 59.Cote ML, Kardia SL, Wenzlaff AS, Ruckdeschel JC, Schwartz AG. Risk of lung cancer among white and black relatives of individuals with early-onset lung cancer. JAMA. 2005 Jun 22;293(24):3036–42. doi: 10.1001/jama.293.24.3036. [DOI] [PubMed] [Google Scholar]
  • 60.Cote ML, Yoo W, Wenzlaff AS, Prysak GM, Santer SK, Claeys GB, et al. Tobacco and estrogen metabolic polymorphisms and risk of non-small cell lung cancer in women. Carcinogenesis. 2009 Apr;30(4):626–35. doi: 10.1093/carcin/bgp033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Muscat JE, Chen SQ, Richie JP, Jr., Altorki NK, Citron M, Olson S, et al. Risk of lung carcinoma among users of nonsteroidal antiinflammatory drugs. Cancer. 2003 Apr 1;97(7):1732–6. doi: 10.1002/cncr.11242. [DOI] [PubMed] [Google Scholar]
  • 62.Cui Y, Morgenstern H, Greenland S, Tashkin DP, Mao J, Cao W, et al. Polymorphism of Xeroderma Pigmentosum group G and the risk of lung cancer and squamous cell carcinomas of the oropharynx, larynx and esophagus. Int J Cancer. 2006 Feb 1;118(3):714–20. doi: 10.1002/ijc.21413. [DOI] [PubMed] [Google Scholar]
  • 63.Zheng YL, Loffredo CA, Yu Z, Jones RT, Krasna MJ, Alberg AJ, et al. Bleomycin-induced chromosome breaks as a risk marker for lung cancer: a case-control study with population and hospital controls. Carcinogenesis. 2003 Feb;24(2):269–74. doi: 10.1093/carcin/24.2.269. [DOI] [PubMed] [Google Scholar]
  • 64.Gallagher CJ, Muscat JE, Hicks AN, Zheng Y, Dyer AM, Chase GA, et al. The UDP-glucuronosyltransferase 2B17 gene deletion polymorphism: sex-specific association with urinary 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol glucuronidation phenotype and risk for lung cancer. Cancer Epidemiol Biomarkers Prev. 2007 Apr;16(4):823–8. doi: 10.1158/1055-9965.EPI-06-0823. [DOI] [PubMed] [Google Scholar]
  • 65.Heck JE, Andrew AS, Onega T, Rigas JR, Jackson BP, Karagas MR, et al. Lung cancer in a U.S. population with low to moderate arsenic exposure. Environ Health Perspect. 2009 Nov;117(11):1718–23. doi: 10.1289/ehp.0900566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Orlow I, Park BJ, Mujumdar U, Patel H, Siu-Lau P, Clas BA, et al. DNA damage and repair capacity in patients with lung cancer: prediction of multiple primary tumors. J Clin Oncol. 2008 Jul 20;26(21):3560–6. doi: 10.1200/JCO.2007.13.2654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Richiardi L, Boffetta P, Simonato L, Forastiere F, Zambon P, Fortes C, et al. Occupational risk factors for lung cancer in men and women: a population-based case-control study in Italy. Cancer Causes Control. 2004 Apr;15(3):285–94. doi: 10.1023/B:CACO.0000024223.91059.ed. [DOI] [PubMed] [Google Scholar]
  • 68.Loriot MA, Rebuissou S, Oscarson M, Cenee S, Miyamoto M, Ariyoshi N, et al. Genetic polymorphisms of cytochrome P450 2A6 in a case-control study on lung cancer in a French population. Pharmacogenetics. 2001 Feb;11(1):39–44. doi: 10.1097/00008571-200102000-00005. [DOI] [PubMed] [Google Scholar]
  • 69.Breitling LP, Dahmen N, Illig T, Rujescu D, Nitz B, Raum E, et al. Variants in COMT and spontaneous smoking cessation: retrospective cohort analysis of 925 cessation events. Pharmacogenet Genomics. 2009 Aug;19(8):657–9. doi: 10.1097/FPC.0b013e32832fabf3. [DOI] [PubMed] [Google Scholar]
  • 70.Ugolini D, Neri M, Canessa PA, Casilli C, Catrambone G, Ivaldi GP, et al. The CREST biorepository: a tool for molecular epidemiology and translational studies on malignant mesothelioma, lung cancer, and other respiratory tract diseases. Cancer Epidemiol Biomarkers Prev. 2008 Nov;17(11):3013–9. doi: 10.1158/1055-9965.EPI-08-0524. [DOI] [PubMed] [Google Scholar]
  • 71.Rafnar T, Sulem P, Stacey SN, Geller F, Gudmundsson J, Sigurdsson A, et al. Sequence variants at the TERT-CLPTM1L locus associate with many cancer types. Nat Genet. 2009 Feb;41(2):221–7. doi: 10.1038/ng.296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Field JK, Smith DL, Duffy S, Cassidy A. The Liverpool Lung Project research protocol. Int J Oncol. 2005 Dec;27(6):1633–45. [PubMed] [Google Scholar]
  • 73.Woll P. http://resolucent.group.shef.ac.uk. [cited; Available from:http://resolucent.group.shef.ac.uk.
  • 74.Park JY, Matsuo K, Suzuki T, Ito H, Hosono S, Kawase T, et al. Impact of smoking on lung cancer risk is stronger in those with the homozygous aldehyde dehydrogenase 2 null allele in a Japanese population. Carcinogenesis. 2010 Apr;31(4):660–5. doi: 10.1093/carcin/bgq021. [DOI] [PubMed] [Google Scholar]
  • 75.Kiyohara C, Yamamura KI, Nakanishi Y, Takayama K, Hara N. Polymorphism in GSTM1, GSTT1, and GSTP1 and Susceptibility to Lung Cancer in a Japanese Population. Asian Pac J Cancer Prev. 2000;1(4):293–8. [PubMed] [Google Scholar]
  • 76.Le Marchand L, Donlon T, Lum-Jones A, Seifried A, Wilkens LR. Association of the hOGG1 Ser326Cys polymorphism with lung cancer risk. Cancer Epidemiol Biomarkers Prev. 2002 Apr;11(4):409–12. [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

01

RESOURCES