Research Article

DNA Repair and Cell Cycle Control and the Risk of Young-Onset Lung Cancer

Stefano Landi,1 Federica Gemignani,1,2 Federico Canzian,2,3 Vale´rie Gaborieau,2 Roberto Barale,1 Debora Landi,1 Neonilia Szeszenia-Dabrowska,4 David Zaridze,5 Jolanta Lissowska,6 Peter Rudnai,7 Eleonora Fabianova,8 Dana Mates,9 Lenka Foretova,10 Vladimir Janout,11 Vladimir Bencko,12 Lydie Gioia-Patricola,2 Janet Hall,2 Paolo Boffetta,2 Rayjean J. Hung,2,13 and Paul Brennan2

1Dip. Biologia-Genetics, University of Pisa, Pisa, Italy; 2IARC, Lyon, France; 3German Cancer Research Center, Heidelberg, Germany; 4Department of Epidemiology, Institute of Occupational Medicine, Lodz, Poland; 5Institute of Carcinogenesis, Cancer Research Centre, Moscow, Russia; 6Department of Cancer Epidemiology and Prevention, Cancer Center and Maria Sklodowska-Curie Institute of Oncology, Warsaw, Poland; 7National Institute of Environmental Health, Fodor Jo´zsef National Center for Public Health, Budapest, Hungary; 8Specialized Institute of Hygiene and Epidemiology, Banska Bystrica, Slovakia; 9Institute of Hygiene, Public Health, Bucharest, Romania; 10Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Brno, Czech Republic; 11Department of Preventive Medicine, Faculty of Medicine, Palacky University, Olomouc, Czech Republic; 12Charles University of Prague, First Faculty of Medicine, Institute of Hygiene and Epidemiology, Prague, Czech Republic; and 13School of Public Health, University of California at Berkeley, Berkeley, California

Abstract normal program of cell cycle and growth, favoring the development Exposure to tobacco smoke and to mutagenic xenobiotics can of cancer. Cells counteract the deleterious effects of damaged DNA cause various types of DNA damage in lung cells, which, if not through different repair pathways and strategies, including cell corrected by DNA repair systems, may lead to deregulation of cycle control and apoptosis induction (2). The ‘‘base excision the cell cycle and, ultimately, to cancer. Genetic variation repair’’ (BER) pathway is the primary mechanism to remove could thus be an important factor in determining suscepti- incorrect and damaged bases, including those formed by reactive bility to tobacco-induced lung cancer with genetic suscepti- oxygen species, such as 8-hydroxyguanine, and methylating agents bility playing a larger role in young-onset cases compared with (e.g., 7-methylguanine and 3-methyladenine; ref. 3). The mismatch that in the general population. We have therefore studied 102 repair (MMR) pathway corrects errors of DNA replication that single-nucleotide polymorphisms (SNP) in 34 key DNA repair escape the proofreading functions of DNA polymerases as well as and cell cycle control genes in 299 lung cancer cases heterologies formed during recombination. It has also been linked diagnosed before the age of 50 years and 317 controls from to cell cycle checkpoint activation and cell death, and thus, six countries of Central and Eastern Europe. We have found no alterations in this process can have wide-ranging biological association of lung cancer risk with polymorphisms in genes consequences (4). related to cell cycle control, single-strand/double-strand The ‘‘nucleotide excision repair’’ (NER; either ‘‘global genome’’ break repair, or . Significant associations NER or ‘‘transcription-coupled’’ NER) acts on a variety of helix- (P < 0.05) were found with polymorphisms in genes involved in distorting DNA lesions, such as the pyrimidine dimers caused by DNA damage sensing (ATM) and, interestingly, in four genes UV light, and chemical adducts, such as those caused by benzo(a) encoding proteins involved in mismatch repair (LIG1, LIG3, pyrene (5). Finally, direct reversal of DNA damage, an unusual form MLH1 MSH6 of DNA repair, is also present in human cells. The promutagenic , and ). The strongest associations were observed 6 with heterozygote carriers of LIG1 7C>T [odds ratio (OR), lesion O -methylguanine is repaired by such a process where a 1.73; 95% confidence interval (95% CI), 1.13-2.64] and specific methyltransferase, encoded by the MGMT , transfers homozygote carriers of LIG3 rs1052536 (OR, 2.05; 95% CI, the methyl group from the DNA guanine residue to a cysteine 1.25-3.38). Consideration of the relatively large number of residue located at its in an error-free process (6). markers assessed diminishes the significance of these findings; Cells must also cope with the presence of DNA single-strand and thus, these SNPs should be considered promising candidates double-strand breaks (SSB and DSB). SSBs can arise directly from for further investigation in other independent populations. damage to the DNA or indirectly as intermediates in the BER (Cancer Res 2006; 66(22): 11062-9) pathway. Like many of the other DNA repair pathways in human cells, the processing of such breaks involves a series of coordinated, Introduction sequential reactions in which the damage is detected and processed and any gaps generated are filled and religated. Key Exposure to tobacco smoke and to mutagenic xenobiotics causes proteins in this process are XRCC1, poly(ADP-ribose) polymerase 1 various types of DNA damage to lung cells (1). Such lesions and the (PARP1), and the proliferating cell nuclear antigen (PCNA; ref. 7). mutations that arise from them can lead to alterations in the The repair of DSBs, which are dangerously cytotoxic lesions formed after exposure to ionizing radiation, occurs through two main mechanisms: nonhomologous end joining (NHEJ) and the homol- ogous recombination repair (HRR). These two processes have Note: S. Landi and F. Gemignani contributed equally to this work. Requests for reprints: Paul Brennan, IARC, 150 cours Albert-Thomas, F-69372 different fidelities: HR can be achieved with high fidelity, whereas Lyon Cedex 08, France. Phone: 33-4-72738391; Fax: 33-4-72738342; E-mail: brennan@ NHEJ may result in loss of genetic information (8, 9). iarc.fr. I2006 American Association for Cancer Research. A second arm in the maintenance of genome stability in the doi:10.1158/0008-5472.CAN-06-1039 presence of DNA damage is achieved by the precise regulation of

Cancer Res 2006; 66: (22). November 15, 2006 11062 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2006 American Association for Cancer Research. Young-Onset Lung Cancer the cell cycle. DNA strand breaks and certain base adducts are very Materials and Methods effective in inducing cell cycle blocks. Such damage is detected Study population. This study was conducted in 15 centers in six Central through different mechanisms involving the protein products of and Eastern European countries: the Czech Republic (Prague, Olomouc, and key genes, such as ataxia-telangiectasia mutated (ATM) and ATM Brno), Hungary (Borsod, Heves, Szabolcs, Szolnok, and Budapest), Poland and Rad3-related (ATR), which act upstream of p53 and other (Warsaw and Lodz), Romania (Bucharest), Russia (Moscow), and Slovakia checkpoint proteins to induce cell cycle arrest (10). In addition to (Banska Bystrica, Bratislava, and Nitra). Each center followed an identical these mechanisms, apoptosis, the program of cell suicide, which is protocol and was responsible for recruiting a consecutive group of patients an important escape route to avoid damaged cells progressing to a who were newly diagnosed with lung cancer and a comparable group of malignant phenotype, can be activated in response to DNA hospital-based control subjects without lung cancer from February 1998 to damage. The ATM and p53 proteins are also involved in this October 2002. All cancer diagnoses were confirmed histologically or cytologically. Eligible subjects (case patients and control subjects) must process (11). have resided in the study area for at least 1 year before recruitment. Lung All these pathways contribute to the genome stability of the cell, cancer case patients were identified through an active search of the records and a deficiency in one or more of the relevant genes can lead to of clinical and pathology departments at the participating hospitals. All deregulated cell growth and, ultimately, to cancer development. centers attempted to recruit all eligible patients as soon as possible after the The link between alterations in DNA repair genes and increased patient had received an initial diagnosis of lung cancer. The maximum time cancer risk is well documented in certain inherited disorders, such interval between diagnosis and recruitment was 3 months. All study as xeroderma pigmentosum (deficiencies in NER), Bloom syn- subjects (case patients and control subjects) and their physicians provided drome (deficiency in the DNA helicase RECQ), Fanconi anemia written informed consent. This study was approved by the institutions at all (FANCx genes), ataxia-telangiectasia (ATM), human nonpolyposis study centers, and ethical approval was obtained from the IARC (Lyon, colorectal cancer (HNPCC, MMR genes), retinoblastoma (RB1), France), the coordinating center. At all centers, except the Warsaw center, control subjects were chosen from among inpatients and outpatients familial cutaneous melanoma (CDKN1A), Li-Fraumeni syndrome admitted to the same hospitals as the case patients or hospitals serving the (TP53), familial breast/ovary cancers (BRCA1 or BRCA2), and same population; case patients and control subjects from each hospital Nijmegen breakage syndrome (NBS1). In addition, the importance were frequency matched by sex, age (F3 years), center, and referral (or of the integrity of DNA repair pathways to prevent lung residence) area. Control subjects were eligible for this study if they had been carcinogenesis has been shown in several animal models where diagnosed with non-tobacco-related diseases or had undergone minor individual DNA repair genes are disrupted. For example, Xpc / surgical procedures or had benign disorders, common infections, eye mice show high susceptibility to lung cancer following exposure to conditions (except cataract or diabetic retinopathy), or common orthopedic acetylaminofluorene (12), Mgmt / and Parp1 / mice to diseases (except osteoporosis). At the Warsaw center, control subjects were nitrosamine-induced tumors (13, 14), and Ogg1 / mice to the selected by random sampling of the general population using the Electronic spontaneous development of lung cancers (15). List of Residents. Overall, the average participation rate was 91.0% among case patients and 91.2% among control subjects. Case patients and control Polymorphisms in DNA repair and cell cycle control genes, subjects underwent an identical in-person interview during which they which affect the normal protein activity, may alter the efficiency of completed a detailed questionnaire and provided blood samples. The these processes and lead to genetic instability and increased cancer questionnaire collected information about demographic variables, such as risk. Reduced DNA repair capacity and impaired control of the cell sex, date of birth, and education level, medical history, family history of cycle are phenotypes found in lung cancer patients, and poly- cancer, history of tobacco consumption, including frequency, intensity, morphisms, which have been reported to be associated with the duration, and status, history of alcohol consumption, diet history (using a risk of developing lung cancer, include OGG1 S326C, XRCC1 general food frequency questionnaire), and occupational history. Blood R194W, MLH1 93 G>A, PARP1 V762A, ERCC2 Gln751, RAD23B samples were stored in liquid nitrogen. Of the 2,633 case patients and 2,884 Val249, XPA 5¶-untranslated region (UTR) G>A, CCND1 4 A>G, control subjects who agreed to participate in this study, 2,188 (83%) case and TP53 codon 72, intron 3, and intron 6 (16–21). patients and 2,198 (76%) control subjects provided blood samples during the interview of whom 299 cases and 317 controls were V50 years of age at These results support the hypothesis that genetic variations diagnosis and at recruitment, respectively, and form the study population within DNA repair and cell cycle control pathways might be for this analysis. important in determining an individual risk of developing lung Selection of polymorphisms. The polymorphisms that were included in cancer. Thus, to investigate the role of gene variants in DNA repair the panel to be studied were selected based on the following criteria: (a) and cell cycle control pathways, we have analyzed 102 single- having a biological significance shown through functional studies (e.g., nucleotide polymorphisms (SNPs) in 51 genes in a multicenter OGG1 326Ser has a 7-fold higher activity for repairing 8-oxoG than the case-control study of early-onset lung cancer. Our rationale for 326Cys variant), (b) associated with biological end points in epidemiologic restricting the analysis to subjects diagnosed before age 50 years studies [e.g., XRCC1 399Gln allele was associated with increased levels of was that the role of genetic susceptibility would be expected to be aflatoxin B1-DNA adduct and increased bleomycin sensitivity (27–29)], or more important among subjects with a younger age of diagnosis. (c) associated with the risk of cancer at any site in epidemiologic studies [e.g., the polymorphism at codon 194 within XRCC1 (30)]. However, for Indeed, previous epidemiologic studies have suggested that a large some DNA repair and cell cycle control genes, there were no variants that proportion of lung cancers occurring before age 50 years have a met these criteria. Therefore, for some genes, we added selected SNPs from genetic component. Risks due to genetic factors are further dbSNP,14 which either (a) had a high frequency of the rare allele (to allow amplified by cigarette smoking (22, 23). the highest statistical power to detect associations) or (b) coded for Due to the quite low incidence of the disease occurring at missense changes. Among the missense variants, we focused, in particular, younger ages, there are very few published studies on the genetic on changes having the greatest potential for functional relevance, such as risk factors for early-onset lung cancer. The majority of these involving a proline residue (e.g., PARP P1328T) or the charge/polarity of the investigated the role of xenobiotic metabolism genes (24–26), and none have explored, using a comprehensive panel of variants, the association of variants in DNA repair and cell cycle control genes and lung cancer risk. 14 http://www.ncbi.nlm.nih.gov/SNP/. www.aacrjournals.org 11063 Cancer Res 2006; 66: (22). November 15, 2006

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2006 American Association for Cancer Research. Cancer Research lateral group (e.g., CDKN2A A148T). Although the biological effect of many (V14 pack-years), moderate (>14-38.26 pack-years), or heavy (>38.26 pack- of the polymorphisms selected for study is not known, we expect that these years) smokers based on the tertiles of cumulative tobacco consumption criteria should have maximized the likelihood of having chosen SNPs with among the control group. We tested the Hardy-Weinberg equilibrium of functional significance or associated with cancer. genotype distributions separately among case patients and control subjects. Laboratory techniques. Genomic DNA was extracted from blood The minimum detectable odds ratio (OR) was calculated for each sequence samples using Puregene chemistry (Gentra Systems, Minneapolis, MN). variant based on its genotype frequency, our study sample size, and a DNA concentrations were measured by using PicoGreen dsDNA quantifi- statistical power of 80% as described previously (34). Our study had an 80% cation kits (Molecular Probes, Leiden, the Netherlands). All polymorphisms power to detect a minimum OR of 2.5 for relatively rare variants (5%) and a were analyzed together for a given sample by a microarray technique based minimum OR of 1.6 for common variants (z30%). on the arrayed primer extension (APEX) principle. We used unconditional multivariate logistic regression analysis to APEX consists of a sequencing reaction primed by an oligonucleotide examine associations between genetic polymorphisms and lung cancer anchored with its 5¶-end to a glass slide and terminating just one nucleotide risk by estimating ORs and 95% confidence intervals (95% CI). Genotypes before the polymorphic site. A DNA polymerase extends the oligonucleotide were categorized into three groups (major allele homozygous, heterozygous, by adding one fluorescently labeled dideoxynucleotide triphosphate and homozygous variant) when the allele frequencies allowed or into two (ddNTP) complementary to the variant base. Reading the incorporated groups (major allele homozygous and minor allele carriers) for rare fluorescence identifies the base in the target sequence. This method is polymorphisms. Age, sex, country, and tobacco pack-year were included in suitable not only for SNPs but also for small insertion/deletion poly- all analyses as covariates. morphisms. Because both sense and antisense strands are probed, two We computed false-positive response probabilities (FPRP; ref. 35) for the oligonucleotides were designed for each polymorphism. In general, two nominally significant associations we have observed between SNPs and lung 30-mers, one for each strand, complementary to each side of the cancer risk. Prior probability is likely to be influenced by the biological polymorphism were designed both with their 3¶-end pointing toward the knowledge of the gene, the functional significance of the variants, and the polymorphism. The flanking sequences and their related APEX-oligonucleo- available epidemiologic evidence. It remains a subjective measure that may tides have been previously published (31). Five-prime (C-12) amino-linker vary from one investigator to another based on the importance they assign oligonucleotides were synthesized by Sigma-Genosys (Cambridge, United to the different pieces of evidence. For this reason, we have calculated FPRP Kingdom) and spotted onto silanized slides (32). Genomic DNAs were amplified to enrich the fragments carrying the SNPs Table 1. Demographic characteristics of lung cancer by using specific primer pairs. Then, PCR products were pooled, purified, cases and controls concentrated using Millipore (Bedford, MA) Microcon MY30 columns, and fragmented. For single-base extension reaction, fragmented PCR products Cases, Controls, were incubated onto the slides together with the fluorescently labeled n (%) n (%) ddNTPs (4 50 pmol), 10 buffer, and 4 units ThermoSequenase (GE Healthcare, Little Chalfont, United Kingdom). All the details of the experimental protocol, including primer and probe sequences, were Country reported in previous articles (31, 33). Romania 37 (12.4) 58 (18.3) Slides were imaged by a Genorama-003 four-color detector equipped Hungary 67 (22.4) 51 (16.1) with Genorama image analysis software (Asper Biotech, Tartu, Estonia). Poland 101 (33.8) 108 (34.1) Fluorescence intensities at each position were converted automatically into Russia 42 (14.0) 32 (10.1) base calls by the software under the supervision of trained personnel. In Slovakia 37 (12.4) 25 (7.9) case of more than one signal present on a given position, only the main Czech Republic 15 (5.0) 43 (13.6) signal was considered if the intensity of the weaker signal was <10% of the Gender main signal. APEX gives a very high concordance compared with the Men 199 (66.6) 211 (66.6) standard genotyping or sequencing methods as assessed in previous studies Women 100 (33.4) 106 (33.4) (31, 33). Age (completed), y To ensure quality control, we followed several strategies: DNA samples <30 0 (0.0) 4 (1.3) from case patients and control subjects were randomly distributed, and all 30-34 7 (2.3) 15 (4.7) genotyping was conducted by personnel who were blinded to the case- 35-39 17 (5.7) 26 (8.2) control status of the subjects; each APEX oligonucleotide was spotted in 40-44 82 (27.4) 79 (24.9) replicate; each SNP was analyzed independently by genotyping both the 45-49 193 (64.5) 193 (60.9) sense and the antisense strands of the DNA (in case of disagreement, Education the base call was discarded); internal positive controls allowed to verify that Basic/elementary 21 (7.0) 6 (1.9) the intensities of the four channels (A, C, T, and G) were equilibrated; Apprentice/vocational 108 (36.1) 105 (33.1) base calls were scored by three independent trained operators and dis- Middle schools, ending by graduation 98 (32.8) 107 (33.8) cordant results were rechecked and, in case of disagreement, discarded; Postgradual, not university degree 52 (17.4) 60 (18.9) DNA samples from individuals of known genotypes were added to check University degree 20 (6.7) 35 (11.0) periodically the validity of the genotyping; we randomly selected 10% of the Missing 0 (0.0) 4 (1.3) study subjects (i.e., both case patients and control subjects) and Smoking status regenotyped their DNA samples for each polymorphism. Never smokers 18 (6.0) 90 (28.4) Statistical analysis. The frequency distributions of demographic Former smokers 22 (7.4) 53 (16.7) variables and putative risk factors for lung cancer, including country of Current smokers 259 (86.6) 170 (53.6) residence, age at recruitment (which for case patients was a proxy for age at Missing 0 (0.0) 4 (1.3) diagnosis), sex, highest education level, and smoking status, were examined Histology for case patients and control subjects. Former smokers were defined as Adenocarcinoma 86 (28.8) smokers who stopped smoking at least 2 years before the interview. Small cell carcinoma 61 (20.4) Tobacco consumption included smoking of cigarettes, pipes, and cigars. Squamous cell 85 (28.4) Cumulative tobacco consumption was calculated by multiplying smoking Other 67 (22.4) duration (in years) by smoking intensity (in the equivalent of cigarette Total 299 317 packs) and expressed as pack-years. We categorized the subjects as light

Cancer Res 2006; 66: (22). November 15, 2006 11064 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2006 American Association for Cancer Research. Young-Onset Lung Cancer for a range of prior probabilities from 50% to 0.1%. Following Wacholder Hardy-Weinberg equilibrium in the controls by a m2 test using P = et al., we used a threshold of noteworthiness of FPRP V0.2. All statistical 0.01 as threshold. This threshold was chosen based on anticonser- analyses were conducted using STATA software version 8.0 (Stata Corp. LP, vativeness of this test as noted by Wigginton et al. (36). All SNPs College Station, TX). All statistical tests were two sided. (except for MSH3 235G>A, MSH6 G39E, ERCC1 354T>C, ERCC2 D312N, and XRCC3 17893A>G) were in Hardy-Weinberg equilibri- Results um in this population. The results of the initial and the repeat Table 1 shows the frequency distribution of demographic genotyping analyses were at least 99% concordant. Unfortunately, characteristics among the 299 cases and 317 controls. As expected, because of the nature of the method of genotyping where all the the proportion of current smokers was far higher among the cases SNPs are analyzed on one chip, genotyping that failed for any than controls (86.6% versus 53.6%; P < 0.001). The sex and age individual SNP could not be repeated. Genotyping success rates for distributions were similar among cases and controls, although individual polymorphisms averaged 92%. there was a tendency for cases to have a lower education level than Table 2 shows ORs for polymorphisms in genes involved in the controls (P = 0.01). With respect to the histologic types of the detecting DNA breaks as well as in the negative modulation of cell tumors in the cases, the numbers of adenocarcinoma, squamous, cycle (including apoptosis). Only one SNP (IVS48+238 C>G within and small cell carcinomas were similar. We tested departure from ATM) was found associated with a decreased risk of lung cancer

Table 2. Associations between lung cancer risk and SNPs of genes coding for DNA break sensors and negative regulators of the cell cycle (ATM-, ATR-, and p53-dependent cascades)

SNP name rs no. Homozygotes Heterozygotes Homozygotes Ptrend common allele rarer allele

Ca Co Ca Co OR (95% CI) Ca Co OR (95% CI)

ATM: 5557 G>A -D1853N rs1801516 205 238 73 63 1.21 (0.80-1.85) 7 3 2.06 (0.50-8.49) 0.21 ATM: IVS22-77T>C rs664677 108 85 134 170 0.68 (0.46-1.01) 47 54 0.77 (0.45-1.30) 0.18 ATM: IVS48+238 C>G rs609429 92 70 101 113 0.7 (0.45-1.10) 35 52 0.55 (0.30-0.98) 0.03 ATR: 632C>T -T211M rs2227928 99 111 138 138 1.21 (0.82-1.79) 49 48 1.07 (0.63-1.80) 0.63 CCND1: 687bp 3 of STP G>C rs678653 105 112 105 114 0.84 (0.56-1.27) 34 31 1.04 (0.57-1.88) 0.82 CCND1: 870G>A rs603965 79 81 139 156 0.81 (0.53-1.23) 71 76 0.94 (0.58-1.53) 0.79 CDKN1B: 79C>T rs34330 168 178 111 115 1.09 (0.76-1.57) 17 17 0.96 (0.45-2.07) 0.80 CDKN2A: 29bp 3 of STP C>G rs11515 219 238 69 70 1.09 (0.72-1.66) 3 6 0.47 (0.10-2.19) 0.92 CDKN2A: 69bp 3 of STP C>T rs3088440 245 270 42 42 1.15 (0.69-1.89) 1 0 — 0.45 CDKN2A: A148T rs3731249 240 253 15 10 1.44 (0.60-3.49) 1 0 — 0.16 CDKN2B: C>A intron1 rs2069426 204 199 34 35 0.98 (0.56-1.71) 1 3 0.45 (0.04-4.57) 0.69 CDKN2B: G>A intron1 rs974336 198 201 43 54 0.81 (0.50-1.32) 2 3 0.99 (0.15-6.31) 0.46 GADD45A: 3812T>C rs532446 163 180 97 106 0.98 (0.67-1.43) 22 23 1.28 (0.65-2.50) 0.66 MDM2: 309T>G rs2279744 100 115 147 143 1.27 (0.86-1.87) 35 44 0.88 (0.50-1.55) 0.91 MDM2: E354E; -344 A>T rs769412 200 189 29 40 0.78 (0.44-1.36) 4 2 2.15 (0.37-12.70) 0.79 p21/Cip1/CDKN1A: 20bp 3 of STP C>T rs1059234 251 272 43 36 1.23 (0.73-2.08) 2 2 0.91 (0.12-7.06) 0.52 p21/Cip1/CDKN1A: S31R rs1801270 241 268 36 37 0.96 (0.56-1.65) 3 1 3.05 (0.30-31.35) 0.77 PARP1/ADPRT1: IVS17-12 C>G rs4986819 238 249 48 55 0.92 (0.58-1.45) 6 4 1.49 (0.38-5.87) 1.00 PARP1/ADPRT1: IVS4+12 G>A rs1805403 174 164 86 112 0.68 (0.47-1.01) 19 20 0.84 (0.41-1.72) 0.13 PARP1/ADPRT1: P1328T rs1050112 98 113 100 115 1.16 (0.76-1.76) 32 36 0.98 (0.54-1.78) 0.84 PARP1/ADPRT1: T802 A>T rs4986817 240 250 48 56 0.88 (0.55-1.39) 6 4 1.46 (0.37-5.75) 0.87 PARP1/ADPRT1: V762A rs1136410 207 211 75 84 0.84 (0.57-1.25) 10 12 0.96 (0.38-2.46) 0.50 RAD9A: 730bp 3 of STP G>A rs1064876 240 256 20 22 1.04 (0.53-2.06) 0 2 — 0.59 TP53: 14181 (C>T) rs12947788 211 211 21 31 0.6 (0.31-1.16) 1 3 0.41 (0.04-4.51) 0.09 TP53: R72P -BstUI rs1042522 174 159 100 122 0.78 (0.53-1.13) 19 26 0.62 (0.32-1.23) 0.08 TP53BP1: D353E rs560191 141 154 123 131 0.95 (0.66-1.37) 27 24 1.18 (0.61-2.29) 0.85 TP53BP1: G412S rs689647 205 221 41 41 0.9 (0.54-1.50) 2 3 0.95 (0.13-7.21) 0.70 TP53BP2: rs3738370 C>T 5¶-UTR rs3738370 221 243 53 41 1.31 (0.81-2.12) 4 4 0.9 (0.21-3.95) 0.40 TP53BP2: rs17739 G>A 5¶-UTR rs17739 198 219 78 85 0.99 (0.67-1.47) 12 9 1.22 (0.46-3.19) 0.84 XRCC5: 323bp 3 of STP T>C rs1051677 225 244 56 53 1.29 (0.82-2.03) 4 1 3.45 (0.34-34.69) 0.16 CASP10: 1228G>A -V410I rs5837767 258 283 34 33 1.12 (0.64-1.94) 3 0 — 0.31 CASP3: IVS1-1555 A>C rs3087455 116 119 129 145 0.86 (0.59-1.25) 35 37 0.93 (0.52-1.66) 0.59 CASP8: D302H rs1045485 233 255 60 56 1.15 (0.74-1.79) 3 5 0.47 (0.09-2.51) 0.88 CASP9: Q221R rs1052576 100 99 128 148 0.82 (0.56-1.22) 60 66 0.94 (0.58-1.53) 0.70 XRCC5: 841bp 3 of STP -74582 G>A rs2440 119 105 106 130 0.71 (0.48-1.05) 37 41 0.8 (0.46-1.40) 0.21

NOTE: Statistically significant results (P < 0.05) are reported in bold. Abbreviations: Ca, cases; Co, controls; OR, odds ratios adjusted for age, sex, country, and tobacco smoking.

www.aacrjournals.org 11065 Cancer Res 2006; 66: (22). November 15, 2006

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2006 American Association for Cancer Research. Cancer Research

(Ptrend = 0.03), with the other 34 SNPs (19 genes) showing no cancer cases and controls on the basis that they are likely to be association. Table 3 shows similar results for SNPs within genes enriched for risk-associated genetic variants. The validity of this involved in SSB/DSB repair. These genes can be considered as assumption is supported by the increased report of family history downstream with respect to the first set of genes, being actually of lung and other tobacco-related cancers among the young-onset involved in the NHEJ, HRR, and SSB repair (SSBR). Among these 26 cases as opposed to those who developed lung cancer after the age SNPs (12 genes), we found no overall association among the of 50 years. Among the young-onset cases, there was over a 2-fold homozygote variants. Table 4 shows the results for SNPs within risk of lung cancer in first-degree family members (OR, 2.06; 95% genes in the NER, BER, and MMR pathways and the MGMT gene. CI, 1.16-3.65) and over a 4-fold risk of other tobacco-related cancers We analyzed 40 SNPs (20 genes) and found a borderline association (OR, 4.94; 95% CI, 1.47-16.62). Conversely, among those ages >50 for carriers of Gln751 in ERCC2 (OR, 0.7; 95% CI, 0.49-1.00), an years, the risk of lung cancer among family members was much increased risk for carriers of a SNP in the 3¶-UTR of LIG3 (OR, 1.7; reduced (OR, 1.45; 95% CI, 1.15-1.81) and the risk of other tobacco- 95% CI, 1.13-2.56), a reduced risk for carriers of Val219 in MLH1 related cancers was not apparent (OR, 0.86; 95% CI, 0.58-1.29). (OR, 0.69; 95% CI, 0.48-0.98), a borderline significant association for Although we found several interesting statistically significant the homozygote carriers of the 540T allele in MSH6 (OR, 1.95; 95% associations with modest increases in risk, none were significant CI, 1-3.78), and an increased risk for two SNPs in LIG1 (7C>T and when a prior probability of association of 1% was applied. When IVS9-21), with the strongest OR of 1.89 (95% CI, 1.24-2.87) for interpreting such results, it should be taken into account that, carriers of 7 T allele. Among these associations, it is interesting to among the polymorphisms investigated in the present study, note that LIG1, LIG3, MLH1, and MSH6 are all involved in the information on their effect on expression and protein function is MMR. Calculation of FPRP showed that none of the above available for only few of the SNPs studied. Although haplotype- associations remained noteworthy (FPRP V 0.2) when a prior tagging SNPs are available for all of the gene studies, this probability of association of V1% was considered, and only the information was not available when SNPs were being selected for association of LIG1 7C>T remained noteworthy assuming a prior the current study. We therefore focused on variants with relatively probability of 10% (FPRP = 0.154). high prevalence and those coding for missense changes, as these were more likely to affect the function of the encoded protein. It is interesting to note that most of the genes with a significant Discussion association (i.e., LIG1, LIG3, MLH1, and MSH6) belong to the MMR We have undertaken an exploratory investigation of genes and, to a minor extent, BER pathways. Although lung cancer is not involved in cell cycle and DNA repair among a population of lung considered part of the HNPCC spectrum (37), a significant excess

Table 3. Associations between lung cancer risk and SNPs of genes involved in the DNA break repair pathways (HRR, NHEJ, and SSBR)

SNP name rs no. Homozygotes common allele Heterozygotes Homozygotes rarer allele Ptrend

Ca Co Ca Co OR (95% CI) Ca Co OR (95% CI)

BARD1: 143C>T -P24S rs1048108 97 94 143 154 0.94 (0.63-1.39) 48 59 0.76 (0.45-1.27) 0.32 BARD1: 1592G>A -M507V rs2070094 110 102 134 148 0.77 (0.53-1.14) 42 48 0.80 (0.46-1.37) 0.27 BARD1: H506H C>T rs2070093 204 221 83 80 1.11 (0.75-1.64) 8 9 0.92 (0.32-2.66) 0.75 BRCA1: D693N rs4986850 231 230 27 44 0.66 (0.38-1.15) 1 2 0.59 (0.05-6.82) 0.13 BRCA1: E1038G rs16941 121 136 117 115 1.15 (0.78-1.69) 37 29 1.51 (0.84-2.71) 0.18 BRCA1: P871L rs799917 116 137 137 143 1.26 (0.87-1.82) 41 32 1.59 (0.90-2.81) 0.08 BRCA1: Q356R rs1799950 238 258 38 48 0.99 (0.60-1.62) 1 2 0.92 (0.07-12.13) 0.95 BRCA2: 26G>A rs1799943 138 150 107 114 0.98 (0.67-1.44) 29 32 0.94 (0.51-1.72) 0.83 BRCA2: N372H rs144848 159 188 96 98 1.13 (0.77-1.65) 28 18 1.78 (0.90-3.52) 0.13 BRCA2: T1915M rs4987117 269 290 17 20 1.07 (0.51-2.21) 1 0 — 0.82 LIG4: -176C>T rs1805388 204 206 69 79 1.01 (0.67-1.52) 5 4 1.69 (0.36-8.00) 0.77 LIG4: -194C>T rs1805389 243 263 32 44 0.89 (0.52-1.54) 2 2 0.99 (0.12-8.07) 0.71 NBS1: L34 G>A rs1063045 123 134 117 125 0.90 (0.61-1.32) 32 42 0.82 (0.46-1.45) 0.45 NBS1: Q185E rs1805794 134 140 121 134 0.84 (0.58-1.23) 31 36 0.97 (0.54-1.73) 0.62 RAD51: 135 G>C rs1801320 222 242 65 68 1.01 (0.67-1.54) 7 5 1.33 (0.38-4.65) 0.78 RAD51: 172 G>T rs1801321 76 97 79 80 1.42 (0.88-2.27) 11 17 1.02 (0.42-2.49) 0.37 RAD52: C>T2259 -744bp 3 of STP rs11226 84 101 145 144 1.04 (0.69-1.55) 62 66 0.98 (0.60-1.59) 0.96 RAD54B: N250 T>C rs2291439 116 124 122 138 1.00 (0.68-1.47) 34 39 0.86 (0.48-1.54) 0.70 RECQL: 6bp 3 of STP A>C rs13035 86 99 126 127 1.27 (0.84-1.92) 58 60 1.09 (0.66-1.80) 0.60 XRCC2: 41657C>T rs718282 259 262 37 48 0.70 (0.42-1.16) 0 1 — 0.09 XRCC2: 4234G>C rs3218384 160 161 84 106 0.94 (0.64-1.40) 18 17 1.17 (0.55-2.48) 0.92 XRCC2: R188H rs3218536 217 217 22 27 0.93 (0.48-1.79) 1 2 0.24 (0.02-2.67) 0.44 XRCC3: 17893A>G -IVS5-14 rs1799796 123 119 137 165 0.91 (0.63-1.32) 33 27 1.27 (0.69-2.35) 0.75 XRCC3: 4541A>G rs1799794 176 205 79 74 1.16 (0.77-1.74) 13 17 0.64 (0.28-1.46) 0.81 XRCC3: T241M rs861539 127 129 132 143 1.01 (0.70-1.46) 36 40 0.91 (0.52-1.60) 0.83 XRCC4: N298S -IVS7-1 G>A rs1805377 173 215 48 41 1.47 (0.90-2.41) 3 2 2.98 (0.41-21.85) 0.07

Cancer Res 2006; 66: (22). November 15, 2006 11066 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2006 American Association for Cancer Research. Young-Onset Lung Cancer

Table 4. Associations between lung cancer risk and SNPs of genes mainlyinvolved in NER, BER (GG-NER, TC-NER; single-nucleotide BER, multiple-nucleotide BER), MMR, and MGMT

SNP name rs no. Homozygotes Heterozygotes Homozygotes Ptrend common allele rarer allele

Ca Co Ca Co OR (95% CI) Ca Co OR (95% CI)

ERCC1: 15310G>C rs16979802 207 215 36 40 0.92 (0.54-1.56) 3 1 5.35 (0.53-54.31) 0.69 ERCC1: 17677C>A rs3212961 217 240 61 66 1.14 (0.74-1.75) 9 6 1.87 (0.60-5.79) 0.27 ERCC1: 19716G>C rs3212948 132 131 120 134 0.95 (0.65-1.38) 36 43 0.77 (0.45-1.33) 0.39 ERCC1: 354T>C -19007T>C N118N rs11615 124 119 102 118 0.89 (0.60-1.33) 44 48 0.83 (0.49-1.40) 0.44 ERCC1: 8092C>A rs3212986 162 154 97 101 0.80 (0.54-1.19) 17 18 0.79 (0.37-1.70) 0.28 ERCC2-XPD: D312N rs1799793 118 113 111 126 0.91 (0.61-1.35) 48 66 0.65 (0.40-1.08) 0.12 ERCC2-XPD: L751Q rs13181 132 108 109 135 0.70 (0.47-1.03) 49 58 0.70 (0.42-1.14) 0.08 ERCC2-XPD: R156R C>A rs238406 92 108 137 149 1.05 (0.71-1.55) 65 49 1.42 (0.86-2.35) 0.21 ERCC4-XPF: R415Q rs1800067 258 277 40 34 1.17 (0.68-2.01) 0 3 — 0.95 ERCC5-XPG: 335C>T -H46H rs1047768 92 89 128 140 0.92 (0.61-1.38) 53 60 0.97 (0.58-1.63) 0.86 ERCC5-XPG: 3508G>C H1104D rs17655 167 156 65 83 0.78 (0.51-1.19) 14 17 0.51 (0.22-1.15) 0.07 RAD23B: A249V rs1805329 177 169 89 104 0.86 (0.59-1.26) 13 23 0.49 (0.23-1.04) 0.08 XPC: R939Q rs2228001 93 97 104 122 0.88 (0.57-1.34) 37 43 0.70 (0.39-1.24) 0.23 APEX1/APE1: D148E rs3136820 84 80 140 147 0.91 (0.60-1.38) 69 84 0.90 (0.55-1.46) 0.65 APEX 1/APE1: Q51H rs1048945 276 294 14 19 0.70 (0.32-1.51) 0 0 — 0.36 LIG3: 1508bp 3 of STP C>T rs1052536 62 92 146 153 1.54 (1.00-2.38) 85 68 2.05 (1.25-3.38) <0.01 MLH1: 676A>G -I219V rs1799977 145 129 123 151 0.73 (0.50-1.05) 23 29 0.51 (0.26-0.98) 0.02 MSH2: G322D rs4987188 276 286 15 12 1.22 (0.54-2.76) 0 1 — 0.98 MSH3: 235G>A -V79I -V70I rs1650697 151 155 12 22 0.57 (0.26-1.28) 6 4 1.56 (0.41-5.92) 0.72 MSH3: 2835G>A -R940Q rs184967 212 215 71 83 0.84 (0.56-1.25) 8 9 0.97 (0.34-2.76) 0.48 MSH3: 3124A>G -T1036A T1045A rs26279 153 154 116 123 0.92 (0.64-1.33) 27 35 0.78 (0.43-1.42) 0.41 MSH6: D180 -540T>C rs1800935 158 155 102 133 0.77 (0.53-1.11) 33 19 1.95 (1.00-3.78) 0.55 MSH6: G39E rs1042821 171 162 59 71 0.93 (0.59-1.45) 11 17 0.43 (0.18-1.05) 0.14 MUTYH: Q324H rs3219489 205 201 83 100 0.80 (0.55-1.18) 8 11 0.84 (0.31-2.30) 0.29 MUTYH: V22M rs3219484 253 271 39 44 0.98 (0.59-1.62) 1 1 0.78 (0.05-13.04) 0.89 OGG1: S326C -m6 -Fnu4HI rs1052133 203 209 82 94 1.01 (0.69-1.48) 12 9 1.68 (0.64-4.39) 0.51 PMS2: M622I rs1805324 153 151 10 7 1.47 (0.51-4.26) 0 0 — 0.48 POLB: P242R rs3136797 280 301 16 14 1.52 (0.70-3.33) 1 1 2.32 (0.11-47.78) 0.24 XRCC1: R194W rs1799782 263 262 32 53 0.61 (0.36-1.02) 1 1 2.73 (0.13-58.07) 0.10 XRCC1: R280H rs25489 260 290 32 25 1.45 (0.80-2.65) 1 0 — 0.18 XRCC1: R399Q rs25487 118 123 143 149 1.07 (0.74-1.56) 34 42 0.85 (0.48-1.49) 0.77 LIG1: 7C>T rs20579 206 245 73 61 1.73 (1.13-2.64) 60 — <0.01 LIG1: IVS2+12 C>T rs3730849 114 111 134 142 0.91 (0.62-1.33) 40 56 0.70 (0.42-1.18) 0.20 LIG1: IVS9-21 A>G rs3730931 220 255 64 52 1.73 (1.11-2.72) 5 2 3.19 (0.52-19.42) 0.01 PCNA: 1876A>G rs25405 207 217 52 54 0.94 (0.59-1.49) 6 1 6.85 (0.64-73.88) 0.60 PCNA: 2232C>T rs25406 97 107 135 136 1.19 (0.80-1.77) 44 44 1.29 (0.74-2.23) 0.32 MGMT/AGT: 171C>T -L53L rs1803965 218 223 72 83 0.97 (0.65-1.44) 5 4 1.08 (0.25-4.69) 0.91 MGMT/AGT: 262C>T -L84Frs12917 212 219 76 83 1.04 (0.70-1.55) 5 5 1.01 (0.25-4.12) 0.86 MGMT/AGT: 427A>G -I143V rs2308321 238 241 54 71 0.73 (0.48-1.13) 2 4 0.75 (0.13-4.50) 0.17 MGMT/AGT: 533A>G -K178R rs2308327 236 236 51 70 0.71 (0.46-1.10) 2 4 0.73 (0.12-4.40) 0.13

NOTE: Statistically significant results (P < 0.05) are reported in bold.

of lung, liver, and brain tumors has been detected within HNPCC previous study from Korea, the variation 93 G>A within MLH1 families (38). Moreover, an altered expression, including a complete was found associated with increased risk of squamous cell lung lack of protein, of MLH1, in lung cancer, in conjunction with the carcinoma (42). Moreover, the exposure to chromate seems to presence of microsatellite instability in tumor tissue or allelic promote lung cancer also through the inhibition of hMLH1 (43). imbalance of MSH2, has been reported (39, 40). MLH1 was found Finally, engineered mice deficient in the MMR gene Mutyh are inactivated in lung cancers following promoter methylation, prone to develop lung cancer (44). Our findings are consistent with indicating a role of this gene in lung cancer (27). It has also been the importance of MMR in protecting against the development of suggested that the MLH1 and MSH3 genes could be involved in lung cancer and deserve further investigation. lung tumorigenesis through a gene dosage effect in those tumors One of the variants that was found to be associated with a that are hemizygotes and retaining the wild-type copies of MLH1 protective effect was the IVS48+238 C>G variant in the ATM gene. and MSH3 and not showing microsatellite instability (41). In a This variant, when present in the homozygous state, has been www.aacrjournals.org 11067 Cancer Res 2006; 66: (22). November 15, 2006

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2006 American Association for Cancer Research. Cancer Research previously found to be associated with an increased risk of breast variation in the call rate by country was controlled for in the cancer and, when present in the heterozygous state, with a multivariable models. decreased risk of therapeutic radiation sensitivity in breast cancer Another potential limitation is related to the number of cases patients (45). Exposure to X-rays through occupational examina- and controls, which limits the power of the study to detect tions was associated with an increased risk of lung cancer in a significant associations particularly for rare alleles responsible for larger study population from which the cases and controls weak associations. For example, in our study, the XRCC1 investigated here were selected (46). Clearly, further investigation polymorphism R194W is associated with a reduced risk of cancer, of the role of the variants in the ATM gene and lung cancer risk is in agreement with the results reported in literature for this warranted. polymorphism and a recent meta-analysis (30), but the association The present study has some potential limitations. Potential was not strong enough to reach statistical significance. We also did selection bias can occur when subjects who agree to participate in not observe the association with the OGG1 S326C SNP that we the study have characteristics that differ from those of subjects found when we analyzed the complete study population, regardless who are eligible for the study. The major reason for nonpartici- of age (48). We think that the reason for this is that the OGG1 SNP pation in this study was the refusal of some eligible subjects, which confers risk only to subjects who are homozygous for the minor might represent a form of self-selection. Bias from self-selection allele, a condition too rare to be associated with a detectable risk may affect estimates of exposure to environmental factors; with the sample size of the present study. Despite these limitations, however, it is unlikely that self-selection would be related to a the present study is one of the largest investigations, on the role of subject’s genotype. Results of a simulation study (47) suggested that DNA repair and cell cycle control genes in relation to the risk of selection factors that are related only to environmental factors lung cancer in people with a young age. The findings of this study might still lead to a biased estimate of genetic main effects if the shed a new light on early-onset lung cancer; however, confirmation environmental factors modify the genetic effects. Nevertheless, bias in a second independent study is required. Nonetheless, further in the estimate of the genetic main effect due to the selection studies on the role of LIG1 and LIG3 seem promising. factor of environmental exposures can be adjusted for in the analyses by treating the environmental factors as potential confounders as we have done (47). Another limitation is the Acknowledgments missing data generated in the process of genotyping. These missing Received 3/21/2006; revised 7/20/2006; accepted 9/21/2006. data are likely to be independent from demographic variables Grant support: European Commission (DG-XII; contract IC15-CT96-0313), National Cancer Institute R01 grant (contract CA 092039-01A2), Association for and smoking status, and their consequence would be to reduce International Cancer Research grant (contract 03-281), ‘‘Marie-Curie Reintegration the statistical power of our study. However, the missing data Grant’’ (contract MERG-CT-2004-506373), and Associazione Italiana per la Ricerca sul Cancro principal investigator grant 2005. The Warsaw part of the study was supported were associated with country as the quality of the DNA samples by a local grant from The Polish State Committee for Scientific Research (grant SPUB- varied from one country to another. Nevertheless, the missing data M-COPERNICUS/P-05/DZ-30/99/2000). F. Gemignani is a recipient of a fellowship were not likely to bias the estimates to any meaningful extent from the International Association for the Study on Lung Cancer. The costs of publication of this article were defrayed in part by the payment of page because the call rate was high (with an average of 92%) and charges. This article must therefore be hereby marked advertisement in accordance independent from the case-control status. In addition, any with 18 U.S.C. Section 1734 solely to indicate this fact.

References 12. Friedberg EC, Bond JP, Burns DK, et al. Defective polymorphism with lung cancer risk. Cancer Epidemiol nucleotide excision repair in xpc mutant mice and its Biomarkers Prev 2002;11:409–12. 1. IARC. IARC monographs on the evaluation of association with cancer predisposition. Mutat Res 2000; 21. Shen M, Berndt SI, Rothman N, et al. Polymorphisms carcinogenic risks to humans, vol. 83. Tobacco smoke 459:99–108. in the DNA nucleotide excision repair genes and lung and involuntary smoking. Lyon (France): IARC; 2004. p. 13. Tsutsumi M, Masutani M, Nozaki T, et al. Increased cancer risk in Xuan Wei, China. Int J Cancer 2005;116: 1–1382. susceptibility of poly(ADP-ribose) polymerase-1 knock- 768–73. 2. Friedberg EC. DNA damage and repair. Nature 2003; out mice to nitrosamine carcinogenicity. Carcinogenesis 22. Cote ML, Kardia SL, Wenzlaff AS, Ruckdeschel JC, 421:436–40. 2001;22:1–3. Schwartz AG. Risk of lung cancer among white and 3. Krokan HE, Nilsen H, Skorpen F, Otterlei M, Slupphaug 14. Iwakuma T, Sakumi K, Nakatsuru Y, et al. High black relatives of individuals with early-onset lung G. Base excision repair of DNA in mammalian cells. incidence of nitrosamine-induced tumorigenesis in mice cancer. JAMA 2005;293:3036–42. FEBS Lett 2000;476:73–7. lacking DNA repair methyltransferase. Carcinogenesis 23. Li X, Hemminki K. Inherited predisposition to early 4. Kunkel TA, Erie DA. DNA mismatch repair. Annu Rev 1997;18:1631–5. onset lung cancer according to histological type. Int J Biochem 2005;74:681–710. 15. Sakumi K, Tominaga Y, Furuichi M, et al. Ogg1 Cancer 2004;112:451–7. 5. Friedberg EC. How nucleotide excision repair protects knockout-associated lung tumorigenesis and its sup- 24. Skuladottir H, Autrup H, Autrup J, et al. Poly- against cancer. Nat Rev Cancer 2001;1:22–33. pression by Mth1 gene disruption. Cancer Res 2003;63: morphisms in genes involved in xenobiotic metabolism 6. Sekiguchi M, Nakabeppu Y, Sakumi K, Tuzuki T. DNA- 902–5. and lung cancer risk under the age of 60 years. A pooled repair methyltransferase as a molecular device for 16. Zhang X, Miao X, Liang G, et al. Polymorphisms in study of lung cancer patients in Denmark and Norway. preventing mutation and cancer. J Cancer Res Clin DNA base excision repair genes ADPRT and XRCC1 and Lung Cancer 2005;48:187–99. Oncol 1996;122:199–206. risk of lung cancer. Cancer Res 2005;65:722–6. 25. Cote ML, Kardia SL, Wenzlaff AS, Land SJ, Schwartz 7. Malanga M, Althaus FR. The role of poly(ADP-ribose) 17. Wu X, Zhao H, Amos CI, et al. p53 genotypes and AG. Combinations of glutathione S- geno- in the DNA damage signaling network. Biochem Cell haplotypes associated with lung cancer susceptibility types and risk of early-onset lung cancer in Caucasians Biol 2005;83:354–64. and ethnicity. J Natl Cancer Inst 2002;94:681–90. and African Americans: a population-based study. 8. Weterings E, van Gent DC. The mechanism of non- 18. Sugimura H, Kohno T, Wakai K, et al. hOGG1 Carcinogenesis 2005;26:811–9. homologous end-joining: a synopsis of synapsis. DNA Ser326Cys polymorphism and lung cancer suscepti- 26. Yang P, Bamlet WR, Ebbert JO, Taylor WR, de Repair (Amst) 2004;3:1425–35. bility. Cancer Epidemiol Biomarkers Prev 1999;8: Andrade M. Glutathione pathway genes and lung cancer 9. Johnson RD, Jasin M. Double-strand-break-induced 669–74. risk in young and old populations. Carcinogenesis 2004; homologous recombination in mammalian cells. Bio- 19. Spitz MR, Wei Q, Dong Q, Amos CI, Wu X. Genetic 25:1935–44. chem Soc Trans 2001;29:196–201. susceptibility to lung cancer: the role of DNA damage 27. Wang YC, Lu YP, Tseng RC, et al. Inactivation of 10. Abraham RT. Cell cycle checkpoint signaling through and repair. Cancer Epidemiol Biomarkers Prev 2003;12: hMLH1 and hMSH2 by promoter methylation in the ATM and ATR kinases. Genes Dev 2001;15:2177–96. 689–98. primary non-small cell lung tumors and matched 11. Norbury CJ, Zhivotovsky B. DNA damage-induced 20. Le Marchand L, Donlon T, Lum-Jones A, Seifried A, sputum samples. J Clin Invest 2003;111:887–95. apoptosis. Oncogene 2004;23:2797–808. Wilkens LR. Association of the hOGG1 Ser326Cys 28. Matullo G, Palli D, Peluso M, et al. XRCC1, XRCC3,

Cancer Res 2006; 66: (22). November 15, 2006 11068 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2006 American Association for Cancer Research. Young-Onset Lung Cancer

XPD gene polymorphisms, smoking, and (32)P-DNA 35. Wacholder S, Chanock S, Garcia-Closas M, El hMLH1 and hMSH3 in non-small cell lung cancer. Int J adducts in a sample of healthy subjects. Carcinogenesis Ghormli L, Rothman N. Assessing the probability that Cancer 1998;77:173–80. 2001;22:1437–45. a positive report is false: an approach for molecular 42. Park SH, Lee GY, Jeon HS, et al. 93G!A 29. Lunn RM, Langlois RG, Hsieh LL, Thompson CL, Bell epidemiology studies. J Natl Cancer Inst 2004;96:434–42. polymorphism of hMLH1 and risk of primary lung DA. XRCC1 polymorphisms: effects on aflatoxin B1-DNA 36. Wigginton JE, Cutler DJ, Abecasis GR. A note on cancer. Int J Cancer 2004;112:678–82. adducts and glycophorin A variant frequency. Cancer exact tests of Hardy-Weinberg equilibrium. Am J Hum 43. Takahashi Y, Kondo K, Hirose T, et al. Microsatellite Res 1999;59:2557–61. Genet 2005;76:887–93. instability and protein expression of the DNA mismatch 30. Hu Z, Ma H, Chen F, Wei Q, Shen H. XRCC1 37. Aarnio M, Sankila R, Pukkala E, et al. Cancer risk in repair gene, hMLH1, of lung cancer in chromate- polymorphisms and cancer risk: a meta-analysis of 38 mutation carriers of DNA-mismatch-repair genes. Int J exposed workers. Mol Carcinog 2005;42:150–8. case-control studies. Cancer Epidemiol Biomarkers Prev Cancer 1999;81:214–8. 44. Xie Y, Yang H, Cunanan C, et al. Deficiencies in 2005;14:1810–8. 38. Ponz de Leon M, Benatti P, Pedroni M, Sassatelli R, mouse Myh and Ogg1 result in tumor predisposition 31. Gemignani F, Landi S, Vivant F, Zienolddiny S, Roncucci L. Risk of cancer revealed by follow-up of and G to T mutations in codon 12 of the K-ras oncogene Brennan P, Canzian F. A catalogue of polymorphisms families with hereditary non-polyposis colorectal in lung tumors. Cancer Res 2004;64:3096–102. related to xenobiotic metabolism and cancer suscepti- cancer: a population-based study. Int J Cancer 1993;55: 45. Angele S, Romestaing P, Moullan N, et al. ATM bility. Pharmacogenetics 2002;12:459–63. 202–7. haplotypes and cellular response to DNA damage: 32. Guo Z, Guilfoyle RA, Thiel AJ, Wang R, Smith LM. 39. Chang JW, Chen YC, Chen CY, Chen JT, Chen SK, association with breast cancer risk and clinical radio- Direct fluorescence analysis of genetic polymorphisms Wang YC. Correlation of genetic instability with sensitivity. Cancer Res 2003;63:8717–25. by hybridization with oligonucleotide arrays on glass mismatch repair protein expression and p53 mutations 46. Boffetta P, Mannetje A, Zaridze D, et al. Occupational supports. Nucleic Acids Res 1994;22:5456–65. in non-small cell lung cancer. Clin Cancer Res 2000;6: X-ray examinations and lung cancer risk. Int J Cancer 33. Landi S, Gemignani F, Gioia-Patricola L, Chabrier A, 1639–46. 2005;115:263–7. Canzian F. Evaluation of a microarray for genotyping 40. Xinarianos G, Liloglou T, Prime W, et al. hMLH1 and 47. Morimoto LM, White E, Newcomb PA. Selection bias polymorphisms related to xenobiotic metabolism and hMSH2 expression correlates with allelic imbalance on in the assessment of gene-environment interaction in DNA repair. Biotechniques 2003;35:816–27. 3p in non-small cell lung carcinomas. case-control studies. Am J Epidemiol 2003;158:259–63. 34. Dos Santos Silva I. Size of a study. In: Cancer Cancer Res 2000;60:4216–21. 48. Hung RJ, Brennan P, Canzian F, et al. Large-scale epidemiology: principles and methods. 2nd ed. Barce- 41. Benachenhou N, Guiral S, Gorska-Flipot I, Labuda D, investigation of base excision repair genetic polymor- lona (Spain): International Agency for the Research on Sinnett D. High resolution deletion mapping reveals phisms and lung cancer risk in a multicenter study. J Cancer; 1999. p. 333–53. frequent allelic losses at the DNA mismatch repair loci Natl Cancer Inst 2005;97:567–76.

www.aacrjournals.org 11069 Cancer Res 2006; 66: (22). November 15, 2006

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2006 American Association for Cancer Research. DNA Repair and Cell Cycle Control Genes and the Risk of Young-Onset Lung Cancer

Stefano Landi, Federica Gemignani, Federico Canzian, et al.

Cancer Res 2006;66:11062-11069.

Updated version Access the most recent version of this article at: http://cancerres.aacrjournals.org/content/66/22/11062

Cited articles This article cites 46 articles, 12 of which you can access for free at: http://cancerres.aacrjournals.org/content/66/22/11062.full#ref-list-1

Citing articles This article has been cited by 5 HighWire-hosted articles. Access the articles at: http://cancerres.aacrjournals.org/content/66/22/11062.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/66/22/11062. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2006 American Association for Cancer Research.