151
|
Hepp D, Gonçalves GL, de Freitas TRO. Prediction of the damage-associated non-synonymous single nucleotide polymorphisms in the human MC1R gene. PLoS One 2015; 10:e0121812. [PMID: 25794181 PMCID: PMC4368538 DOI: 10.1371/journal.pone.0121812] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 02/04/2015] [Indexed: 12/13/2022] Open
Abstract
The melanocortin 1 receptor (MC1R) is involved in the control of melanogenesis. Polymorphisms in this gene have been associated with variation in skin and hair color and with elevated risk for the development of melanoma. Here we used 11 computational tools based on different approaches to predict the damage-associated non-synonymous single nucleotide polymorphisms (nsSNPs) in the coding region of the human MC1R gene. Among the 92 nsSNPs arranged according to the predictions 62% were classified as damaging in more than five tools. The classification was significantly correlated with the scores of two consensus programs. Alleles associated with the red hair color (RHC) phenotype and with the risk of melanoma were examined. The R variants D84E, R142H, R151C, I155T, R160W and D294H were classified as damaging by the majority of the tools while the r variants V60L, V92M and R163Q have been predicted as neutral in most of the programs The combination of the prediction tools results in 14 nsSNPs indicated as the most damaging mutations in MC1R (L48P, R67W, H70Y, P72L, S83P, R151H, S172I, L206P, T242I, G255R, P256S, C273Y, C289R and R306H); C273Y showed to be highly damaging in SIFT, Polyphen-2, MutPred, PANTHER and PROVEAN scores. The computational analysis proved capable of identifying the potentially damaging nsSNPs in MC1R, which are candidates for further laboratory studies of the functional and pharmacological significance of the alterations in the receptor and the phenotypic outcomes.
Collapse
Affiliation(s)
- Diego Hepp
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Instituto Federal de Educação, Ciência e Tecnologia do Rio Grande do Sul—Câmpus Porto Alegre, Rio Grande do Sul, Brazil
- * E-mail:
| | - Gislene Lopes Gonçalves
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Instituto de Alta Investigación, Universidad de Tarapacá, Antofagasta, 1520 Arica, Chile
| | | |
Collapse
|
152
|
Barrett JH, Taylor JC, Bright C, Harland M, Dunning AM, Akslen LA, Andresen PA, Avril MF, Azizi E, Bianchi Scarrà G, Brossard M, Brown KM, Dębniak T, Elder DE, Friedman E, Ghiorzo P, Gillanders EM, Gruis NA, Hansson J, Helsing P, Hočevar M, Höiom V, Ingvar C, Landi MT, Lang J, Lathrop GM, Lubiński J, Mackie RM, Molven A, Novaković S, Olsson H, Puig S, Puig-Butille JA, van der Stoep N, van Doorn R, van Workum W, Goldstein AM, Kanetsky PA, Pharoah PDP, Demenais F, Hayward NK, Newton Bishop JA, Bishop DT, Iles MM, GenoMEL Consortium. Fine mapping of genetic susceptibility loci for melanoma reveals a mixture of single variant and multiple variant regions. Int J Cancer 2015; 136:1351-60. [PMID: 25077817 PMCID: PMC4328144 DOI: 10.1002/ijc.29099] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/06/2014] [Indexed: 01/31/2023]
Abstract
At least 17 genomic regions are established as harboring melanoma susceptibility variants, in most instances with genome-wide levels of significance and replication in independent samples. Based on genome-wide single nucleotide polymorphism (SNP) data augmented by imputation to the 1,000 Genomes reference panel, we have fine mapped these regions in over 5,000 individuals with melanoma (mainly from the GenoMEL consortium) and over 7,000 ethnically matched controls. A penalized regression approach was used to discover those SNP markers that most parsimoniously explain the observed association in each genomic region. For the majority of the regions, the signal is best explained by a single SNP, which sometimes, as in the tyrosinase region, is a known functional variant. However in five regions the explanation is more complex. At the CDKN2A locus, for example, there is strong evidence that not only multiple SNPs but also multiple genes are involved. Our results illustrate the variability in the biology underlying genome-wide susceptibility loci and make steps toward accounting for some of the "missing heritability."
Collapse
Affiliation(s)
- Jennifer H Barrett
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Montero I, Requena C, Traves V, García-Casado Z, Kumar R, Nagore E. Age-related characteristics of cutaneous melanoma in a Spanish Mediterranean population. Int J Dermatol 2015; 54:778-84. [PMID: 25771683 DOI: 10.1111/ijd.12496] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 09/06/2013] [Accepted: 09/11/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND Melanoma is considered a heterogeneous tumor with genetic and environmental factors involved in its pathogenesis. The impact of these factors varies depending on age. OBJECTIVE The aim of this study was to characterize the epidemiological, phenotypic, and histological features of patients with melanoma according to three age groups: ≤40, 41-65, and >65 years. METHODS A total of 1122 consecutive patients with invasive melanoma definitively treated in our institution since January 2000 were selected from our melanoma database. Epidemiological, phenotypic, and histological data were retrieved and analyzed as a function of age. RESULTS Female patients predominated in the younger age group. The location of cutaneous malignant melanoma differed with age. In the younger and middle age groups, tumors presented mainly on the trunk, while in the older group they were mainly found on the head/neck. Signs of actinic damage such as actinic keratoses, solar lentigines, or other skin tumors increased with age, while genetic factors such as family history of melanoma or a high number of common melanocytic nevi were more frequent in the younger group. CONCLUSION Our results suggest that melanoma development in younger patients is the result of genetic factors, particularly related to multiple nevi, whereas in older patients environmental factors such as severe chronic sun exposure play a major role.
Collapse
Affiliation(s)
- Iria Montero
- Department of Dermatology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Celia Requena
- Department of Dermatology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Victor Traves
- Department of Pathology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Zaida García-Casado
- Department of Molecular Biology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Rajiv Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Eduardo Nagore
- Department of Dermatology, Instituto Valenciano de Oncología, Valencia, Spain.,Universidad Católica de Valencia, Valencia, Spain
| |
Collapse
|
154
|
MHC class I chain-related gene a diversity in patients with cutaneous malignant melanoma from southeastern Spain. DISEASE MARKERS 2015; 2015:831864. [PMID: 25838620 PMCID: PMC4370202 DOI: 10.1155/2015/831864] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 02/19/2015] [Indexed: 11/17/2022]
Abstract
A limited number of studies have been performed so far on the polymorphism in the transmembrane region (exon 5) of the major histocompatibility complex class I chain-related gene A (MICA) in patients with melanoma. However, the influence of MICA polymorphism in extracellular domains (exons 2, 3, and 4) has not been investigated on melanoma disease. This study aims to characterize the influence of extracellular MICA polymorphism, and its previously described linkage disequilibrium with HLA-B locus, on patients with cutaneous melanoma from southeastern Spain. For this purpose, MICA and HLA-B genotyping was performed in 233 patients and 200 ethnically matched controls by luminex technology. Patients were classified according to the presence of methionine or valine at codon 129 of MICA gene. We found a high frequency of MICA*009 in melanoma patients compared with controls (P = 0.002, Pc = 0.03). Our results also showed an association between MICA*009 and HLA-B*51 alleles in both patients and controls. This association was stronger in patients than controls (P = 0.015). However, a multivariate logistic regression model showed that neither MICA*009 nor the combination MICA*009/HLA-B*51 was associated with melanoma susceptibility. No relationship was observed between MICA-129 dimorphism and melanoma nor when MICA polymorphism was evaluated according to clinical findings at diagnosis.
Collapse
|
155
|
Jacobs LC, Liu F, Pardo LM, Hofman A, Uitterlinden AG, Kayser M, Nijsten T. IRF4, MC1R and TYR genes are risk factors for actinic keratosis independent of skin color. Hum Mol Genet 2015; 24:3296-303. [PMID: 25724930 DOI: 10.1093/hmg/ddv076] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 02/24/2015] [Indexed: 12/13/2022] Open
Abstract
Actinic keratosis (AK) is a pre-malignant skin disease, highly prevalent in elderly Europeans. This study investigates genetic susceptibility to AK with a genome-wide association study (GWAS). A full body skin examination was performed in 3194 elderly individuals from the Rotterdam Study (RS) of exclusive north-western European origin (aged 51-99 years, 45% male). Physicians graded the number of AK into four severity levels: none (76%), 1-3 (14%), 4-9 (6%) and ≥10 (5%), and skin color was quantified using a spectrophotometer on sun-unexposed skin. A GWAS for AK severity was conducted, where promising signals at IRF4 and MC1R (P < 4.2 × 10(-7)) were successfully replicated in an additional cohort of 623 RS individuals (IRF4, rs12203592, Pcombined = 6.5 × 10(-13) and MC1R, rs139810560, Pcombined = 4.1 × 10(-9)). Further, in an analysis of ten additional well-known human pigmentation genes, TYR also showed significant association with AK (rs1393350, P = 5.3 × 10(-4)) after correction for multiple testing. Interestingly, the strength and significance of above-mentioned associations retained largely the same level after skin color adjustment. Overall, our data strongly suggest that IRF4, MC1R and TYR genes likely have pleiotropic effects, a combination of pigmentation and oncogenic functions, resulting in an increased risk of AK.
Collapse
Affiliation(s)
| | - Fan Liu
- Department of Forensic Molecular Biology
| | | | | | - André G Uitterlinden
- Department of Epidemiology and Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | | |
Collapse
|
156
|
A Genome-Wide Association Study Identifies the Skin Color Genes IRF4, MC1R, ASIP, and BNC2 Influencing Facial Pigmented Spots. J Invest Dermatol 2015; 135:1735-1742. [PMID: 25705849 DOI: 10.1038/jid.2015.62] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 01/27/2015] [Accepted: 02/09/2015] [Indexed: 02/02/2023]
Abstract
Facial pigmented spots are a common skin aging feature, but genetic predisposition has yet to be thoroughly investigated. We conducted a genome-wide association study for pigmented spots in 2,844 Dutch Europeans from the Rotterdam Study (mean age: 66.9±8.0 years; 47% male). Using semi-automated image analysis of high-resolution digital facial photographs, facial pigmented spots were quantified as the percentage of affected skin area (mean women: 2.0% ±0.9, men: 0.9% ±0.6). We identified genome-wide significant association with pigmented spots at three genetic loci: IRF4 (rs12203592, P=1.8 × 10(-27)), MC1R (compound heterozygosity score, P=2.3 × 10(-24)), and RALY/ASIP (rs6059655, P=1.9 × 10(-9)). In addition, after adjustment for the other three top-associated loci the BNC2 locus demonstrated significant association (rs62543565, P=2.3 × 10(-8)). The association signals observed at all four loci were successfully replicated (P<0.05) in an independent Dutch cohort (Leiden Longevity Study n=599). Although the four genes have previously been associated with skin color variation and skin cancer risk, all association signals remained highly significant (P<2 × 10(-8)) when conditioning the association analyses on skin color. We conclude that genetic variations in IRF4, MC1R, RALY/ASIP, and BNC2 contribute to the acquired amount of facial pigmented spots during aging, through pathways independent of the basal melanin production.
Collapse
|
157
|
Orlow I, Satagopan JM, Berwick M, Enriquez HL, White KAM, Cheung K, Dusza SW, Oliveria SA, Marchetti MA, Scope A, Marghoob AA, Halpern AC. Genetic factors associated with naevus count and dermoscopic patterns: preliminary results from the Study of Nevi in Children (SONIC). Br J Dermatol 2015; 172:1081-9. [PMID: 25307738 PMCID: PMC4382400 DOI: 10.1111/bjd.13467] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2014] [Indexed: 01/09/2023]
Abstract
BACKGROUND Melanocytic naevi are an important risk factor for melanoma. Naevi with distinct dermoscopic patterns can differ in size, distribution and host pigmentation characteristics. OBJECTIVES We examined MC1R and 85 other candidate loci in a cohort of children to test the hypothesis that the development and dermoscopic type of naevi are modulated by genetic variants. METHODS Buccal DNAs were obtained from a cohort of 353 fifth graders (mean age 10·4 years). Polymorphisms were chosen based on a known or anticipated role in naevi and melanoma. Associations between single-nucleotide polymorphisms (SNPs) and baseline naevus count were determined by multivariate regression adjusting for sex, race/ethnicity and sun sensitivity. Dermoscopic images were available for 853 naevi from 290 children. Associations between SNPs and dermoscopic patterns were determined by polytomous regression. RESULTS Four SNPs were significantly associated with increasing (IRF4) or decreasing (PARP1, CDK6 and PLA2G6) naevus count in multivariate shrinkage analyses with all SNPs included in the model; IRF4 rs12203952 showed the strongest association with log naevus count (relative risk 1·56, P < 0·001). Using homogeneous naevi as the reference, IRF4 rs12203952 and four other SNPs in TERT, CDKN1B, MTAP and PARP1 were associated with either globular or reticular dermoscopic patterns (P < 0·05). CONCLUSIONS Our results provide evidence that subsets of naevi defined by dermoscopic patterns differ in their associations with germline genotypes and support the hypothesis that dermoscopically defined subsets of naevi are biologically distinct. These results require confirmation in larger cohorts. If confirmed, these findings will improve the current knowledge of naevogenesis and assist in the identification of individuals with high-risk phenotypes.
Collapse
Affiliation(s)
- I Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, U.S.A
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
158
|
Sobota RS, Shriner D, Kodaman N, Goodloe R, Zheng W, Gao YT, Edwards TL, Amos CI, Williams SM. Addressing population-specific multiple testing burdens in genetic association studies. Ann Hum Genet 2015; 79:136-47. [PMID: 25644736 DOI: 10.1111/ahg.12095] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 10/06/2014] [Indexed: 01/06/2023]
Abstract
The number of effectively independent tests performed in genome-wide association studies (GWAS) varies by population, making a universal P-value threshold inappropriate. We estimated the number of independent SNPs in Phase 3 HapMap samples by: (1) the LD-pruning function in PLINK, and (2) an autocorrelation-based approach. Autocorrelation was also used to estimate the number of independent SNPs in whole genome sequences from 1000 Genomes. Both approaches yielded consistent estimates of numbers of independent SNPs, which were used to calculate new population-specific thresholds for genome-wide significance. African populations had the most stringent thresholds (1.49 × 10(-7) for YRI at r(2) = 0.3), East Asian populations the least (3.75 × 10(-7) for JPT at r(2) = 0.3). We also assessed how using population-specific significance thresholds compared to using a single multiple testing threshold at the conventional 5 × 10(-8) cutoff. Applied to a previously published GWAS of melanoma in Caucasians, our approach identified two additional genes, both previously associated with the phenotype. In a Chinese breast cancer GWAS, our approach identified 48 additional genes, 19 of which were in or near genes previously associated with the phenotype. We conclude that the conventional genome-wide significance threshold generates an excess of Type 2 errors, particularly in GWAS performed on more recently founded populations.
Collapse
Affiliation(s)
- Rafal S Sobota
- Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Genetics, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire
| | | | | | | | | | | | | | | | | |
Collapse
|
159
|
Aguilo F, Di Cecilia S, Walsh MJ. Long Non-coding RNA ANRIL and Polycomb in Human Cancers and Cardiovascular Disease. Curr Top Microbiol Immunol 2015. [PMID: 26220772 DOI: 10.1007/82_2015_455] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The long non-coding RNA CDKN2B-AS1, commonly referred to as the A ntisense N on-coding R NA in the I NK4 L ocus (ANRIL), is a 3.8-kb-long RNA transcribed from the short arm of human chromosome 9 on p21.3 that overlaps a critical region encompassing three major tumor suppressor loci juxtaposed to the INK4b-ARF-INK4a gene cluster and the methyl-thioadenosine phosphorylase (MTAP) gene. Genome-wide association studies have identified this region with a remarkable and growing number of disease-associated DNA alterations and single nucleotide polymorphisms, which corresponds to increased susceptibility to human disease. Recent attention has been devoted on whether these alterations in the ANRIL sequence affect its expression levels and/or its splicing transcript variation, and in consequence, global cellular homeostasis. Moreover, recent evidence postulates that ANRIL not only can regulate their immediate genomic neighbors in cis, but also has the capacity to regulate additional loci in trans. This action would further increase the complexity for mechanisms imposed through ANRIL and furthering the scope of this lncRNA in disease pathogenesis. In this chapter, we summarize the most recent findings on the investigation of ANRIL and provide a perspective on the biological and clinical significance of ANRIL as a putative biomarker, specifically, its potential role in directing cellular fates leading to cancer and cardiovascular disease.
Collapse
Affiliation(s)
- Francesca Aguilo
- Departments of Structural and Chemical Biology, Genetics and Genomic Sciences and Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Serena Di Cecilia
- Departments of Structural and Chemical Biology, Genetics and Genomic Sciences and Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- European School of Molecular Medicine, CEINGE-Biotecnologie Avanzate, Naples, Italy
| | - Martin J Walsh
- Departments of Structural and Chemical Biology, Genetics and Genomic Sciences and Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
160
|
Olsen CM, Neale RE, Green AC, Webb PM, The QSkin Study, The Epigene Study, Whiteman DC. Independent validation of six melanoma risk prediction models. J Invest Dermatol 2014; 135:1377-1384. [PMID: 25548858 DOI: 10.1038/jid.2014.533] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 11/18/2014] [Accepted: 12/09/2014] [Indexed: 11/10/2022]
Abstract
Identifying people at high risk of melanoma is important for targeted prevention activities and surveillance. Several tools have been developed to classify melanoma risk, but few have been independently validated. We assessed the discriminatory performance of six melanoma prediction tools by applying them to individuals from two independent data sets, one comprising 762 melanoma cases and the second a population-based sample of 42,116 people without melanoma. We compared the model predictions with actual melanoma status to measure sensitivity and specificity. The performance of the models was variable with sensitivity ranging from 97.7 to 10.5% and specificity from 99.6 to 1.3%. The ability of all the models to discriminate between cases and controls, however, was generally high. The model developed by MacKie et al. (1989) had higher sensitivity and specificity for men (0.89 and 0.88) than women (0.79 and 0.72). The tool developed by Cho et al. (2005) was highly specific (men, 0.92; women, 0.99) but considerably less sensitive (men, 0.64; women, 0.37). Other models were either highly specific but lacked sensitivity or had low to very low specificity and higher sensitivity. Poor performance was partly attributable to the use of non-standardized assessment items and various differing interpretations of what constitutes "high risk".
Collapse
Affiliation(s)
- Catherine M Olsen
- Cancer Control Group, Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Rachel E Neale
- Cancer Control Group, Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Adèle C Green
- Cancer Control Group, Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Cancer Research UK Manchester Institute and Institute of Inflammation and Repair, University of Manchester, Manchester, UK
| | - Penelope M Webb
- Cancer Control Group, Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - The QSkin Study
- Cancer Control Group, Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - The Epigene Study
- Cancer Control Group, Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - David C Whiteman
- Cancer Control Group, Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| |
Collapse
|
161
|
Paradisi A, Tabolli S, Didona B, Sobrino L, Russo N, Abeni D. Markedly reduced incidence of melanoma and nonmelanoma skin cancer in a nonconcurrent cohort of 10,040 patients with vitiligo. J Am Acad Dermatol 2014; 71:1110-6. [DOI: 10.1016/j.jaad.2014.07.050] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 07/28/2014] [Accepted: 07/29/2014] [Indexed: 01/25/2023]
|
162
|
Abstract
Melanoma, the deadliest form of skin cancer, is an aggressive disease that is rising in incidence. Although melanoma is a historically treatment-resistant malignancy, in recent years unprecedented breakthroughs in targeted therapies and immunotherapies have revolutionized the standard of care for patients with advanced disease. Here, we provide an overview of recent developments in our understanding of melanoma risk factors, genomics, and molecular pathogenesis and how these insights have driven advances in melanoma treatment. In addition, we review benefits and limitations of current therapies and look ahead to continued progress in melanoma prevention and therapy. Remarkable achievements in the field have already produced a paradigm shift in melanoma treatment: Metastatic melanoma, once considered incurable, can now be treated with potentially curative rather than palliative intent.
Collapse
Affiliation(s)
- Jennifer A Lo
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - David E Fisher
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
163
|
Li DK, Han J, Liu JB, Jin GF, Qu JW, Zhu M, Wang YR, Jiang J, Ma HX. Genetic variants at 6p21.1 and 7p15.3 Identified by GWASs of multiple cancers and ovarian cancer risk: a case-control study in Han Chinese women. Asian Pac J Cancer Prev 2014; 15:123-7. [PMID: 24528012 DOI: 10.7314/apjcp.2014.15.1.123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
A recent study summarized several published genome-wide association studies (GWASs) of cancer and reported two pleiotropic loci at 6p21.1 and 7p15.3 contributing to multiple cancers including lung cancer, noncardia gastric cancer (NCGC), and esophageal squamous-cell carcinoma (ESCC) in Han Chinese. However, it is not known whether such genetic variants have similar effects on the risk of gynecologic cancers, such as ovarian cancer. Hence, we explored associations between genetic variants in 6p21.1 and 7p15.3 and ovarian cancer risk in Han Chinese women. We performed an independent case-control study by genotyping the two loci (rs2494938 A > G at 6p21.1 and rs2285947 A > G at 7p15.3) in a total of 377 ovarian cancer cases and 1,034 cancer-free controls using TaqMan allelic discrimination assay. We found that rs2285947 at 7p15.3 was significantly associated with risk of ovarian cancer with per allele odds ratio (OR) of 1.33 [95% confidence interval (CI): 1.08-1.64, P=0.008]. However, no significant association was observed between rs2494938 and ovarian cancer risk. Our results showed that rs2285947 at 7p15.3 may also contribute to the development of ovarian cancer in Han Chinese women, further suggesting pleiotropy of 7p15.3 in multiple cancers.
Collapse
Affiliation(s)
- Da-Ke Li
- Department of Gynaecology, Jiangsu Provincial Hospital of TCM, Affliated hospital of Nanjing University of TCM, Nanjing, China E-mail :
| | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Athanasiadis EI, Antonopoulou K, Chatzinasiou F, Lill CM, Bourdakou MM, Sakellariou A, Kypreou K, Stefanaki I, Evangelou E, Ioannidis JPA, Bertram L, Stratigos AJ, Spyrou GM. A Web-based database of genetic association studies in cutaneous melanoma enhanced with network-driven data exploration tools. Database (Oxford) 2014; 2014:bau101. [PMID: 25380778 PMCID: PMC4224266 DOI: 10.1093/database/bau101] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 08/21/2014] [Accepted: 09/23/2014] [Indexed: 12/14/2022]
Abstract
The publicly available online database MelGene provides a comprehensive, regularly updated, collection of data from genetic association studies in cutaneous melanoma (CM), including random-effects meta-analysis results of all eligible polymorphisms. The updated database version includes data from 192 publications with information on 1114 significantly associated polymorphisms across 280 genes, along with new front-end and back-end capabilities. Various types of relationships between data are calculated and visualized as networks. We constructed 13 different networks containing the polymorphisms and the genes included in MelGene. We explored the derived network representations under the following questions: (i) are there nodes that deserve consideration regarding their network connectivity characteristics? (ii) What is the relation of either the genome-wide or nominally significant CM polymorphisms/genes with the ones highlighted by the network representation? We show that our network approach using the MelGene data reveals connections between statistically significant genes/ polymorphisms and other genes/polymorphisms acting as 'hubs' in the reconstructed networks. To the best of our knowledge, this is the first database containing data from a comprehensive field synopsis and systematic meta-analyses of genetic polymorphisms in CM that provides user-friendly tools for in-depth molecular network visualization and exploration. The proposed network connections highlight potentially new loci requiring further investigation of their relation to melanoma risk. Database URL: http://www.melgene.org.
Collapse
Affiliation(s)
- Emmanouil I Athanasiadis
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemiology Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, GR, Greece, Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, Norfolk Place, W2 1PG, London, UK, Department of Medicine Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA, Department of Health Research and Policy, Stanford Prevention Research Center, Stanford University School of Medicine, CA, USA, Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA and Department of Medicine, School of Public Health, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London, UK
| | - Kyriaki Antonopoulou
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemiology Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, GR, Greece, Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, Norfolk Place, W2 1PG, London, UK, Department of Medicine Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA, Department of Health Research and Policy, Stanford Prevention Research Center, Stanford University School of Medicine, CA, USA, Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA and Department of Medicine, School of Public Health, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London, UK
| | - Foteini Chatzinasiou
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemiology Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, GR, Greece, Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, Norfolk Place, W2 1PG, London, UK, Department of Medicine Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA, Department of Health Research and Policy, Stanford Prevention Research Center, Stanford University School of Medicine, CA, USA, Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA and Department of Medicine, School of Public Health, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London, UK
| | - Christina M Lill
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemiology Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, GR, Greece, Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, Norfolk Place, W2 1PG, London, UK, Department of Medicine Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA, Department of Health Research and Policy, Stanford Prevention Research Center, Stanford University School of Medicine, CA, USA, Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA and Department of Medicine, School of Public Health, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London, UK Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemio
| | - Marilena M Bourdakou
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemiology Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, GR, Greece, Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, Norfolk Place, W2 1PG, London, UK, Department of Medicine Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA, Department of Health Research and Policy, Stanford Prevention Research Center, Stanford University School of Medicine, CA, USA, Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA and Department of Medicine, School of Public Health, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London, UK
| | - Argiris Sakellariou
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemiology Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, GR, Greece, Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, Norfolk Place, W2 1PG, London, UK, Department of Medicine Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA, Department of Health Research and Policy, Stanford Prevention Research Center, Stanford University School of Medicine, CA, USA, Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA and Department of Medicine, School of Public Health, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London, UK
| | - Katerina Kypreou
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemiology Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, GR, Greece, Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, Norfolk Place, W2 1PG, London, UK, Department of Medicine Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA, Department of Health Research and Policy, Stanford Prevention Research Center, Stanford University School of Medicine, CA, USA, Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA and Department of Medicine, School of Public Health, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London, UK
| | - Irene Stefanaki
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemiology Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, GR, Greece, Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, Norfolk Place, W2 1PG, London, UK, Department of Medicine Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA, Department of Health Research and Policy, Stanford Prevention Research Center, Stanford University School of Medicine, CA, USA, Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA and Department of Medicine, School of Public Health, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London, UK
| | - Evangelos Evangelou
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemiology Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, GR, Greece, Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, Norfolk Place, W2 1PG, London, UK, Department of Medicine Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA, Department of Health Research and Policy, Stanford Prevention Research Center, Stanford University School of Medicine, CA, USA, Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA and Department of Medicine, School of Public Health, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London, UK Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemio
| | - John P A Ioannidis
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemiology Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, GR, Greece, Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, Norfolk Place, W2 1PG, London, UK, Department of Medicine Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA, Department of Health Research and Policy, Stanford Prevention Research Center, Stanford University School of Medicine, CA, USA, Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA and Department of Medicine, School of Public Health, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London, UK Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemio
| | - Lars Bertram
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemiology Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, GR, Greece, Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, Norfolk Place, W2 1PG, London, UK, Department of Medicine Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA, Department of Health Research and Policy, Stanford Prevention Research Center, Stanford University School of Medicine, CA, USA, Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA and Department of Medicine, School of Public Health, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London, UK Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemio
| | - Alexander J Stratigos
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemiology Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, GR, Greece, Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, Norfolk Place, W2 1PG, London, UK, Department of Medicine Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA, Department of Health Research and Policy, Stanford Prevention Research Center, Stanford University School of Medicine, CA, USA, Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA and Department of Medicine, School of Public Health, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London, UK
| | - George M Spyrou
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27 Athens, GR, Greece, Department of Dermatology, University of Athens, School of Medicine, Andreas Sygros Hospital, Ι. Dragoumi 5, 161 21 Athens, GR, Greece, Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, DE, Germany, Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, DE, Germany, Department of Hygiene and Epidemiology, Clinical and Molecular Epidemiology Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, GR, Greece, Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, Norfolk Place, W2 1PG, London, UK, Department of Medicine Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA, Department of Health Research and Policy, Stanford Prevention Research Center, Stanford University School of Medicine, CA, USA, Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA and Department of Medicine, School of Public Health, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London, UK
| |
Collapse
|
165
|
Park SL, Caberto CP, Lin Y, Goodloe RJ, Dumitrescu L, Love SA, Matise TC, Hindorff LA, Fowke JH, Schumacher FR, Beebe-Dimmer J, Chen C, Hou L, Thomas F, Deelman E, Han Y, Peters U, North KE, Heiss G, Crawford DC, Haiman CA, Wilkens LR, Bush WS, Kooperberg C, Cheng I, Le Marchand L. Association of cancer susceptibility variants with risk of multiple primary cancers: The population architecture using genomics and epidemiology study. Cancer Epidemiol Biomarkers Prev 2014; 23:2568-78. [PMID: 25139936 PMCID: PMC4221293 DOI: 10.1158/1055-9965.epi-14-0129] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Multiple primary cancers account for approximately 16% of all incident cancers in the United States. Although genome-wide association studies (GWAS) have identified many common genetic variants associated with various cancer sites, no study has examined the association of these genetic variants with risk of multiple primary cancers (MPC). METHODS As part of the National Human Genome Research Institute (NHGRI) Population Architecture using Genomics and Epidemiology (PAGE) study, we used data from the Multiethnic Cohort (MEC) and Women's Health Initiative (WHI). Incident MPC (IMPC) cases (n = 1,385) were defined as participants diagnosed with more than one incident cancer after cohort entry. Participants diagnosed with only one incident cancer after cohort entry with follow-up equal to or longer than IMPC cases served as controls (single-index cancer controls; n = 9,626). Fixed-effects meta-analyses of unconditional logistic regression analyses were used to evaluate the associations between 188 cancer risk variants and IMPC risk. To account for multiple comparisons, we used the false-positive report probability (FPRP) to determine statistical significance. RESULTS A nicotine dependence-associated and lung cancer variant, CHRNA3 rs578776 [OR, 1.16; 95% confidence interval (CI), 1.05-1.26; P = 0.004], and two breast cancer variants, EMBP1 rs11249433 and TOX3 rs3803662 (OR, 1.16; 95% CI, 1.04-1.28; P = 0.005 and OR, 1.13; 95% CI, 1.03-1.23; P = 0.006), were significantly associated with risk of IMPC. The associations for rs578776 and rs11249433 remained (P < 0.05) after removing subjects who had lung or breast cancers, respectively (P ≤ 0.046). These associations did not show significant heterogeneity by smoking status (Pheterogeneity ≥ 0.53). CONCLUSIONS Our study has identified rs578776 and rs11249433 as risk variants for IMPC. IMPACT These findings may help to identify genetic regions associated with IMPC risk.
Collapse
Affiliation(s)
- S Lani Park
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California.
| | | | - Yi Lin
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Robert J Goodloe
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee
| | - Logan Dumitrescu
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee. Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| | - Shelly-Ann Love
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Tara C Matise
- Department of Statistics and Biostatistics, Rutgers University, Piscataway, New Jersey
| | - Lucia A Hindorff
- Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Jay H Fowke
- Vanderbilt Epidemiology Center, Vanderbilt University, Nashville, Tennessee
| | - Fredrick R Schumacher
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jennifer Beebe-Dimmer
- School of Medicine, Wayne State University, Detroit, Michigan. Karmanos Cancer Institute, Detroit, Michigan
| | - Chu Chen
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Lifang Hou
- Department of Preventative Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Fridtjof Thomas
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Ewa Deelman
- USC Information Sciences Institute, University of Southern California, Marina del Rey, California
| | - Ying Han
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Kari E North
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina. Carolina Center for Genome Sciences, University of North Carolina, Chapel Hill, California
| | - Gerardo Heiss
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Dana C Crawford
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio
| | - Christopher A Haiman
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Lynne R Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - William S Bush
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio
| | - Charles Kooperberg
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Iona Cheng
- Cancer Prevention Institute of California, Fremont, California
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| |
Collapse
|
166
|
Bhatia S. Genetic variation as a modifier of association between therapeutic exposure and subsequent malignant neoplasms in cancer survivors. Cancer 2014; 121:648-63. [PMID: 25355167 DOI: 10.1002/cncr.29096] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/02/2014] [Accepted: 09/05/2014] [Indexed: 11/08/2022]
Abstract
Subsequent malignant neoplasms (SMNs) are associated with significant morbidity and are a major cause of premature mortality among cancer survivors. Several large studies have demonstrated a strong association between the radiation and/or chemotherapy used to treat primary cancer and the risk of developing SMNs. However, for any given therapeutic exposure, the risk of developing an SMN varies between individuals. Genomic variation can potentially modify the association between therapeutic exposures and SMN risk and may explain the observed interindividual variability. In this review, the author provides a brief overview of the current knowledge regarding the role of genomic variation in the development of therapy-related SMNs and discusses the methodological challenges in undertaking an endeavor to develop a deeper understanding of the molecular underpinnings of therapy-related SMNs, such as an appropriate study design, the identification of an adequately sized study population together with a reliable plan for collecting and maintaining high-quality DNA, clinical validation of the phenotype, and the selection of an appropriate approach or platform for genotyping. Understanding the factors that can modify the risk of treatment-related SMNs is critical to developing targeted intervention strategies and optimizing risk-based health care for cancer survivors.
Collapse
Affiliation(s)
- Smita Bhatia
- Department of Population Sciences, City of Hope, Duarte, California
| |
Collapse
|
167
|
Linsley PS, Speake C, Whalen E, Chaussabel D. Copy number loss of the interferon gene cluster in melanomas is linked to reduced T cell infiltrate and poor patient prognosis. PLoS One 2014; 9:e109760. [PMID: 25314013 PMCID: PMC4196925 DOI: 10.1371/journal.pone.0109760] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 09/03/2014] [Indexed: 12/27/2022] Open
Abstract
While immunotherapies are rapidly becoming mainstays of cancer treatment, significant gaps remain in our understanding of how to optimally target them, alone or in combination. Here we describe a novel method to monitor levels of immune cells and pathways in expression data from solid tumors using pre-defined groups or modules of co-regulated immune genes. We show that expression of an interconnected sub-network of type I interferon-stimulated genes (ISGs) in melanomas at the time of diagnosis significantly predicted patient survival, as did, to a lesser extent, sub-networks of T helper/T regulatory and NK/T Cytotoxic cell genes. As a group, poor prognosis tumors with reduced ISG and immune gene levels exhibited significant copy number loss of the interferon gene cluster located at chromosome 9p21.3. Our studies demonstrate a link between type I interferon action and immune cell levels in melanomas, and suggest that therapeutic approaches augmenting both activities may be most beneficial.
Collapse
Affiliation(s)
- Peter S. Linsley
- Department of Systems Immunology, Benaroya Research Institute, Seattle, WA, United States of America
- * E-mail:
| | - Cate Speake
- Department of Systems Immunology, Benaroya Research Institute, Seattle, WA, United States of America
| | - Elizabeth Whalen
- Department of Systems Immunology, Benaroya Research Institute, Seattle, WA, United States of America
| | - Damien Chaussabel
- Department of Systems Immunology, Benaroya Research Institute, Seattle, WA, United States of America
| |
Collapse
|
168
|
Leslie R, O'Donnell CJ, Johnson AD. GRASP: analysis of genotype-phenotype results from 1390 genome-wide association studies and corresponding open access database. Bioinformatics 2014; 30:i185-94. [PMID: 24931982 DOI: 10.1093/bioinformatics/btu273] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
SUMMARY We created a deeply extracted and annotated database of genome-wide association studies (GWAS) results. GRASP v1.0 contains >6.2 million SNP-phenotype association from among 1390 GWAS studies. We re-annotated GWAS results with 16 annotation sources including some rarely compared to GWAS results (e.g. RNAediting sites, lincRNAs, PTMs). MOTIVATION To create a high-quality resource to facilitate further use and interpretation of human GWAS results in order to address important scientific questions. RESULTS GWAS have grown exponentially, with increases in sample sizes and markers tested, and continuing bias toward European ancestry samples. GRASP contains >100 000 phenotypes, roughly: eQTLs (71.5%), metabolite QTLs (21.2%), methylation QTLs (4.4%) and diseases, biomarkers and other traits (2.8%). cis-eQTLs, meQTLs, mQTLs and MHC region SNPs are highly enriched among significant results. After removing these categories, GRASP still contains a greater proportion of studies and results than comparable GWAS catalogs. Cardiovascular disease and related risk factors pre-dominate remaining GWAS results, followed by immunological, neurological and cancer traits. Significant results in GWAS display a highly gene-centric tendency. Sex chromosome X (OR = 0.18[0.16-0.20]) and Y (OR = 0.003[0.001-0.01]) genes are depleted for GWAS results. Gene length is correlated with GWAS results at nominal significance (P ≤ 0.05) levels. We show this gene-length correlation decays at increasingly more stringent P-value thresholds. Potential pleotropic genes and SNPs enriched for multi-phenotype association in GWAS are identified. However, we note possible population stratification at some of these loci. Finally, via re-annotation we identify compelling functional hypotheses at GWAS loci, in some cases unrealized in studies to date. CONCLUSION Pooling summary-level GWAS results and re-annotating with bioinformatics predictions and molecular features provides a good platform for new insights. AVAILABILITY The GRASP database is available at http://apps.nhlbi.nih.gov/grasp.
Collapse
Affiliation(s)
- Richard Leslie
- Cardiovascular Epidemiology and Human Genomics Branch, National Heart, Lung and Blood Institute, The Framingham Heart Study, Framingham, MA 01702, University of Massachusetts Medical School, Worcester, MA 01655 and Division of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USACardiovascular Epidemiology and Human Genomics Branch, National Heart, Lung and Blood Institute, The Framingham Heart Study, Framingham, MA 01702, University of Massachusetts Medical School, Worcester, MA 01655 and Division of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Christopher J O'Donnell
- Cardiovascular Epidemiology and Human Genomics Branch, National Heart, Lung and Blood Institute, The Framingham Heart Study, Framingham, MA 01702, University of Massachusetts Medical School, Worcester, MA 01655 and Division of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USACardiovascular Epidemiology and Human Genomics Branch, National Heart, Lung and Blood Institute, The Framingham Heart Study, Framingham, MA 01702, University of Massachusetts Medical School, Worcester, MA 01655 and Division of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Andrew D Johnson
- Cardiovascular Epidemiology and Human Genomics Branch, National Heart, Lung and Blood Institute, The Framingham Heart Study, Framingham, MA 01702, University of Massachusetts Medical School, Worcester, MA 01655 and Division of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
169
|
Pergoli L, Favero C, Ruth M. P, Tarantini L, Calista D, Cavalleri T, Angelici L, Consonni D, Pier A. B, Angela C. P, Maria T. L, Bollati V. Blood DNA methylation, nevi number, and the risk of melanoma. Melanoma Res 2014; 24:480-7. [PMID: 25026000 PMCID: PMC6857929 DOI: 10.1097/cmr.0000000000000112] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Germline mutations determining increased cutaneous malignant melanoma (CMM) risk have been identified in familial and sporadic CMM cases, but they account only for a small proportion of CMM cases. Recent evidence suggests that germline epimutations (e.g. DNA methylation alterations), which can be inherited similarly to genomic mutations and can be detected in normal body cells (including blood), might increase susceptibility to cancer. The aim of the study was to identify germline epimutations of genes that were found to be mutated in familial CMM (p16, p14, CDK4, MC1R, hTERT), immune and inflammatory genes (ICAM-1, TNFα), DNA mismatch repair gene (MLH1), and repetitive elements (ALU, LINE-1, HERV-w). We measured DNA methylation using bisulfite pyrosequencing in peripheral blood mononuclear cells from 167 CMM cases and 164 sex-matched and age-matched controls. We used multivariable logistic regression models to evaluate the association between methylation levels and CMM status or presence of dysplastic nevi. We found an association between the risk of CMM and peripheral blood mononuclear cell methylation levels of TNFα [odds ratio (OR)=1.11, 95% confidence interval (CI)=1.03-1.18], CDK4 (OR=0.76, 95% CI=0.64-0.91), and MLH1 (OR=1.12, 95% CI=1.02-1.22). In control participants, the risk of developing dysplastic nevi was associated with methylation levels of TNFα (OR=0.81, 95% CI=0.69-0.95), hTERT (OR=0.90, 95% CI=0.82-0.99), and ALU (OR=1.56, 95% CI=1.02-2.39). Epimutations in CMM susceptibility genes and in genes involved in response to oxidative damage are associated with the risk of developing CMM or dysplastic nevi. Further studies measuring methylation levels of these genes in prospectively collected samples are warranted to further elucidate their role in the development and progression of CMM.
Collapse
Affiliation(s)
- Laura Pergoli
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Chiara Favero
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Pfeiffer Ruth M.
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Letizia Tarantini
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | | | - Tommaso Cavalleri
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Laura Angelici
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Dario Consonni
- Epidemiology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Bertazzi Pier A.
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Epidemiology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Pesatori Angela C.
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Epidemiology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Landi Maria T.
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Valentina Bollati
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Epidemiology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
170
|
Andersson U, Wibom C, Cederquist K, Aradottir S, Borg A, Armstrong GN, Shete S, Lau CC, Bainbridge MN, Claus EB, Barnholtz-Sloan J, Lai R, Il'yasova D, Houlston RS, Schildkraut J, Bernstein JL, Olson SH, Jenkins RB, Lachance DH, Wrensch M, Davis FG, Merrell R, Johansen C, Sadetzki S, Bondy ML, Melin BS. Germline rearrangements in families with strong family history of glioma and malignant melanoma, colon, and breast cancer. Neuro Oncol 2014; 16:1333-40. [PMID: 24723567 PMCID: PMC4165415 DOI: 10.1093/neuonc/nou052] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 03/10/2014] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Although familial susceptibility to glioma is known, the genetic basis for this susceptibility remains unidentified in the majority of glioma-specific families. An alternative approach to identifying such genes is to examine cancer pedigrees, which include glioma as one of several cancer phenotypes, to determine whether common chromosomal modifications might account for the familial aggregation of glioma and other cancers. METHODS Germline rearrangements in 146 glioma families (from the Gliogene Consortium; http://www.gliogene.org/) were examined using multiplex ligation-dependent probe amplification. These families all had at least 2 verified glioma cases and a third reported or verified glioma case in the same family or 2 glioma cases in the family with at least one family member affected with melanoma, colon, or breast cancer.The genomic areas covering TP53, CDKN2A, MLH1, and MSH2 were selected because these genes have been previously reported to be associated with cancer pedigrees known to include glioma. RESULTS We detected a single structural rearrangement, a deletion of exons 1-6 in MSH2, in the proband of one family with 3 cases with glioma and one relative with colon cancer. CONCLUSIONS Large deletions and duplications are rare events in familial glioma cases, even in families with a strong family history of cancers that may be involved in known cancer syndromes.
Collapse
Affiliation(s)
- Ulrika Andersson
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Carl Wibom
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Kristina Cederquist
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Steina Aradottir
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Ake Borg
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Georgina N Armstrong
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Sanjay Shete
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Ching C Lau
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Matthew N Bainbridge
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Elizabeth B Claus
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Jill Barnholtz-Sloan
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Rose Lai
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Dora Il'yasova
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Richard S Houlston
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Joellen Schildkraut
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Jonine L Bernstein
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Sara H Olson
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Robert B Jenkins
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Daniel H Lachance
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Margaret Wrensch
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Faith G Davis
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Ryan Merrell
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Christoffer Johansen
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Siegal Sadetzki
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Melissa L Bondy
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| | - Beatrice S Melin
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden (U.A., C.W., B.S.M.); Computational Life Science Cluster (CLiC), Umeå University, Umeå, Sweden (C.W.); Department of Medical Biosciences, Pathology, Umeå University, Umeå Sweden (K.C.); Department of Oncology, Clinical Science, Lund University, Lund, Sweden (S.A., Å.B.); Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas (G.N.A., M.L.B.); Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas (S.S.); Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas (C.C.L.); Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas (M.N.B.); School of Public Health, Yale University, New Haven, Connecticut (E.B.C.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.B.C.); Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio (J.B.-S.); University of Southern California, Los Angeles, California (R.L.); Cancer Control and Prevention Program/Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina (D.I., J.S.); Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK (R.S.H.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center , New York, New York (J.L.B., S.H.O.); Mayo Comprehensive Clinic Cancer, Mayo Clinic, Rochester, Minnesota (R.B.J., D.H.L.); Department of Neurological Surgery, University of California, San Francisco, California (M.W.); School of Public Health, University of Alberta, Edmonton, Canada (F.G.D.); Department of Neurology, NorthShore University Health System, Evanston, Illinois (R.M.); Cancer Late Effects Research, Oncology, Finsencenteret, Rigshospitalet, University of Copenhagen and Head, Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark (C.J.)
| |
Collapse
|
171
|
Gomez-Lira M, Ferronato S, Malerba G, Santinami M, Maurichi A, Sangalli A, Turco A, Perego P, Rodolfo M. Association of promoter polymorphism −765G>C in the PTGS2 gene with malignant melanoma in Italian patients and its correlation to gene expression in dermal fibroblasts. Exp Dermatol 2014; 23:766-8. [DOI: 10.1111/exd.12522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2014] [Indexed: 01/30/2023]
Affiliation(s)
- Macarena Gomez-Lira
- Section of Biology and Genetics; Department of Life and Reproduction Sciences; University of Verona; Verona Italy
| | - Silvia Ferronato
- Section of Biology and Genetics; Department of Life and Reproduction Sciences; University of Verona; Verona Italy
| | - Giovanni Malerba
- Section of Biology and Genetics; Department of Life and Reproduction Sciences; University of Verona; Verona Italy
| | - Mario Santinami
- Unit of Melanoma and Sarcoma; Fondazione IRCCS Istituto Nazionale dei Tumori; Milan Italy
| | - Andrea Maurichi
- Unit of Melanoma and Sarcoma; Fondazione IRCCS Istituto Nazionale dei Tumori; Milan Italy
| | - Antonella Sangalli
- Section of Biology and Genetics; Department of Life and Reproduction Sciences; University of Verona; Verona Italy
| | - Alberto Turco
- Section of Biology and Genetics; Department of Life and Reproduction Sciences; University of Verona; Verona Italy
| | - Paola Perego
- Unit of Molecular Pharmacology; Fondazione IRCCS Istituto Nazionale dei Tumori; Milan Italy
| | - Monica Rodolfo
- Unit of Immunotherapy; Fondazione IRCCS Istituto Nazionale dei Tumori; Milan Italy
| |
Collapse
|
172
|
Li WQ, Pfeiffer RM, Hyland PL, Shi J, Gu F, Wang Z, Bhattacharjee S, Luo J, Xiong X, Yeager M, Deng X, Hu N, Taylor PR, Albanes D, Caporaso NE, Gapstur SM, Amundadottir L, Chanock SJ, Chatterjee N, Landi MT, Tucker MA, Goldstein AM, Yang XR. Genetic polymorphisms in the 9p21 region associated with risk of multiple cancers. Carcinogenesis 2014; 35:2698-705. [PMID: 25239644 DOI: 10.1093/carcin/bgu203] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The chromosome 9p21 region has been implicated in the pathogenesis of multiple cancers. We analyzed 9p21 single nucleotide polymorphisms (SNPs) from eight genome-wide association studies (GWAS) with data deposited in dbGaP, including studies of esophageal squamous cell carcinoma (ESCC), gastric cancer (GC), pancreatic cancer, renal cell carcinoma (RCC), lung cancer (LC), breast cancer (BrC), bladder cancer (BC) and prostate cancer (PrC). The number of subjects ranged from 2252 (PrC) to 7619 (LC). SNP-level analyses for each cancer were conducted by logistic regression or random-effects meta-analysis. A subset-based statistical approach (ASSET) was performed to combine SNP-level P values across multiple cancers. We calculated gene-level P values using the adaptive rank truncated product method. We identified that rs1063192 and rs2157719 in the CDKN2A/2B region were significantly associated with ESCC and rs2764736 (3' of TUSC1) was associated with BC (P ≤ 2.59 × 10(-6)). ASSET analyses identified four SNPs significantly associated with multiple cancers: rs3731239 (CDKN2A intronic) with ESCC, GC and BC (P = 3.96 × 10(-) (4)); rs10811474 (3' of IFNW1) with RCC and BrC (P = 0.001); rs12683422 (LINGO2 intronic) with RCC and BC (P = 5.93 × 10(-) (4)) and rs10511729 (3' of ELAVL2) with LC and BrC (P = 8.63 × 10(-) (4)). At gene level, CDKN2B, CDKN2A and CDKN2B-AS1 were significantly associated with ESCC (P ≤ 4.70 × 10(-) (5)). Rs10511729 and rs10811474 were associated with cis-expression of 9p21 genes in corresponding cancer tissues in the expression quantitative trait loci analysis. In conclusion, we identified several genetic variants in the 9p21 region associated with the risk of multiple cancers, suggesting that this region may contribute to a shared susceptibility across different cancer types.
Collapse
Affiliation(s)
- Wen-Qing Li
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA, Department of Dermatology, Warren Alpert Medical School, Brown University, Providence, RI, USA,
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Paula L Hyland
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Fangyi Gu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Zhaoming Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA, Cancer Genomics Research Laboratory, NCI-Frederick, SAIC-Frederick Inc., Frederick, MD, USA
| | | | - Jun Luo
- Information Management Services, Inc., Calverton, MD, USA and
| | - Xiaoqin Xiong
- Information Management Services, Inc., Calverton, MD, USA and
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA, Cancer Genomics Research Laboratory, NCI-Frederick, SAIC-Frederick Inc., Frederick, MD, USA
| | - Xiang Deng
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA, Cancer Genomics Research Laboratory, NCI-Frederick, SAIC-Frederick Inc., Frederick, MD, USA
| | - Nan Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Philip R Taylor
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Neil E Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Susan M Gapstur
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA
| | - Laufey Amundadottir
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Nilanjan Chatterjee
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Margaret A Tucker
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Alisa M Goldstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Xiaohong R Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| |
Collapse
|
173
|
Genome-wide DNA methylation profile of leukocytes from melanoma patients with and without CDKN2A mutations. Exp Mol Pathol 2014; 97:425-32. [PMID: 25236571 DOI: 10.1016/j.yexmp.2014.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 09/12/2014] [Indexed: 11/22/2022]
Abstract
Melanoma is a highly aggressive cancer, accounting for up to 75% of skin cancer deaths. A small proportion of melanoma cases can be ascribed to the presence of highly penetrant germline mutations, and approximately 40% of hereditary melanoma cases are caused by CDKN2A mutations. The current study sought to investigate whether the presence of germline CDKN2A mutations or the occurrence of cutaneous melanoma would result in constitutive genome-wide DNA methylation changes. The leukocyte methylomes of two groups of melanoma patients (those with germline CDKN2A mutations and those without CDKN2A mutations) were analyzed together with the profile of a control group of individuals. A pattern of DNA hypomethylation was detected in the CDKN2A-negative patients relative to both CDKN2A-mutated patients and controls. Additionally, we delineated a panel of 90 CpG sites that were differentially methylated in CDKN2A-mutated patients relative to controls. Although we identified a possible constitutive epigenetic signature in CDKN2A-mutated patients, the occurrence of reported SNPs at the detected CpG sites complicated the data interpretation. Thus, further studies are required to elucidate the impact of these findings on melanoma predisposition and their possible effect on the penetrance of CDKN2A mutations.
Collapse
|
174
|
Song F, Amos CI, Lee JE, Lian CG, Fang S, Liu H, MacGregor S, Iles MM, Law MH, Lindeman NI, Montgomery GW, Duffy DL, Cust AE, Jenkins MA, Whiteman DC, Kefford RF, Giles GG, Armstrong BK, Aitken JF, Hopper JL, Brown KM, Martin NG, Mann GJ, Bishop DT, Bishop JAN, GenoMEL consortium, Kraft P, Qureshi AA, Kanetsky PA, Hayward NK, Hunter DJ, Wei Q, Han J. Identification of a melanoma susceptibility locus and somatic mutation in TET2. Carcinogenesis 2014; 35:2097-101. [PMID: 24980573 PMCID: PMC4146422 DOI: 10.1093/carcin/bgu140] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 05/16/2014] [Accepted: 06/19/2014] [Indexed: 12/17/2022] Open
Abstract
Although genetic studies have reported a number of loci associated with melanoma risk, the complex genetic architecture of the disease is not yet fully understood. We sought to identify common genetic variants associated with melanoma risk in a genome-wide association study (GWAS) of 2298 cases and 6654 controls. Thirteen of 15 known loci were replicated with nominal significance. A total of 69 single-nucleotide polymorphisms (SNPs) were selected for in silico replication in two independent melanoma GWAS datasets (a total of 5149 cases and 12 795 controls). Seven novel loci were nominally significantly associated with melanoma risk. These seven SNPs were further genotyped in 234 melanoma cases and 238 controls. The SNP rs4698934 was nominally significantly associated with melanoma risk. The combined odds ratio per T allele = 1.18; 95% confidence interval (1.10-1.25); combined P = 7.70 × 10(-) (7). This SNP is located in the intron of the TET2 gene on chromosome 4q24. In addition, a novel somatic mutation of TET2 was identified by next-generation sequencing in 1 of 22 sporadic melanoma cases. TET2 encodes a member of TET family enzymes that oxidizes 5-methylcytosine to 5-hydroxymethylcytosine (5hmC). It is a putative epigenetic biomarker of melanoma as we previously reported, with observation of reduced TET2 transcriptional expression. This study is the first to implicate TET2 genetic variation and mutation in melanoma.
Collapse
Affiliation(s)
- Fengju Song
- Department of Epidemiology and Biostatistics, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Christopher I Amos
- Department of Community and Family Medicine, Center for Genomic Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03755, USA
| | - Jeffrey E Lee
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christine G Lian
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shenying Fang
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Stuart MacGregor
- Queensland Institute of Medical Research, Brisbane, Queensland 4029, Australia
| | - Mark M Iles
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, UK
| | - Matthew H Law
- Queensland Institute of Medical Research, Brisbane, Queensland 4029, Australia
| | - Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Grant W Montgomery
- Queensland Institute of Medical Research, Brisbane, Queensland 4029, Australia
| | - David L Duffy
- Queensland Institute of Medical Research, Brisbane, Queensland 4029, Australia
| | - Anne E Cust
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, School of Population Health, University of Melbourne, Melbourne, Victoria 3052, Australia, Cancer Epidemiology and Services Research, Sydney School of Public Health, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Mark A Jenkins
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, School of Population Health, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - David C Whiteman
- Queensland Institute of Medical Research, Brisbane, Queensland 4029, Australia
| | - Richard F Kefford
- Westmead Institute of Cancer Research, University of Sydney at Westmead Millennium Institute and Melanoma Institute Australia, Westmead, New South Wales 2145, Australia
| | - Graham G Giles
- Cancer Epidemiology Centre, The Cancer Council Victoria, Carlton, Victoria 3053, Australia
| | - Bruce K Armstrong
- Cancer Epidemiology and Services Research, Sydney School of Public Health, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Joanne F Aitken
- Viertel Centre for Research in Cancer Control, Cancer Council Queensland, Brisbane, Queensland 4004, Australia
| | - John L Hopper
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, School of Population Health, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Kevin M Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20852, USA
| | - Nicholas G Martin
- Queensland Institute of Medical Research, Brisbane, Queensland 4029, Australia
| | - Graham J Mann
- Westmead Institute of Cancer Research, University of Sydney at Westmead Millennium Institute and Melanoma Institute Australia, Westmead, New South Wales 2145, Australia
| | - D Timothy Bishop
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, UK
| | | | | | - Peter Kraft
- Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115, USA
| | - Abrar A Qureshi
- Channing Division of Network Medicine and Department of Dermatology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Peter A Kanetsky
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicholas K Hayward
- Oncogenomics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4029, Australia
| | - David J Hunter
- Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115, USA
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA,
| | - Jiali Han
- Department of Epidemiology and Biostatistics, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China, Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115, USA, Channing Division of Network Medicine and Department of Dermatology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA, Department of Epidemiology, Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA, Simon Cancer Center, Indiana University, Indianapolis, IN 46202, USA and Department of Dermatology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| |
Collapse
|
175
|
Wu C, Wang Z, Song X, Feng XS, Abnet CC, He J, Hu N, Zuo XB, Tan W, Zhan Q, Hu Z, He Z, Jia W, Zhou Y, Yu K, Shu XO, Yuan JM, Zheng W, Zhao XK, Gao SG, Yuan ZQ, Zhou FY, Fan ZM, Cui JL, Lin HL, Han XN, Li B, Chen X, Dawsey SM, Liao L, Lee MP, Ding T, Qiao YL, Liu Z, Liu Y, Yu D, Chang J, Wei L, Gao YT, Koh WP, Xiang YB, Tang ZZ, Fan JH, Han JJ, Zhou SL, Zhang P, Zhang DY, Yuan Y, Huang Y, Liu C, Zhai K, Qiao Y, Jin G, Guo C, Fu J, Miao X, Lu C, Yang H, Wang C, Wheeler WA, Gail M, Yeager M, Yuenger J, Guo ET, Li AL, Zhang W, Li XM, Sun LD, Ma BG, Li Y, Tang S, Peng XQ, Liu J, Hutchinson A, Jacobs K, Giffen C, Burdette L, Fraumeni JF, Shen H, Ke Y, Zeng Y, Wu T, Kraft P, Chung CC, Tucker MA, Hou ZC, Liu YL, Hu YL, Liu Y, Wang L, Yuan G, Chen LS, Liu X, Ma T, Meng H, Sun L, Li XM, Li XM, Ku JW, Zhou YF, et alWu C, Wang Z, Song X, Feng XS, Abnet CC, He J, Hu N, Zuo XB, Tan W, Zhan Q, Hu Z, He Z, Jia W, Zhou Y, Yu K, Shu XO, Yuan JM, Zheng W, Zhao XK, Gao SG, Yuan ZQ, Zhou FY, Fan ZM, Cui JL, Lin HL, Han XN, Li B, Chen X, Dawsey SM, Liao L, Lee MP, Ding T, Qiao YL, Liu Z, Liu Y, Yu D, Chang J, Wei L, Gao YT, Koh WP, Xiang YB, Tang ZZ, Fan JH, Han JJ, Zhou SL, Zhang P, Zhang DY, Yuan Y, Huang Y, Liu C, Zhai K, Qiao Y, Jin G, Guo C, Fu J, Miao X, Lu C, Yang H, Wang C, Wheeler WA, Gail M, Yeager M, Yuenger J, Guo ET, Li AL, Zhang W, Li XM, Sun LD, Ma BG, Li Y, Tang S, Peng XQ, Liu J, Hutchinson A, Jacobs K, Giffen C, Burdette L, Fraumeni JF, Shen H, Ke Y, Zeng Y, Wu T, Kraft P, Chung CC, Tucker MA, Hou ZC, Liu YL, Hu YL, Liu Y, Wang L, Yuan G, Chen LS, Liu X, Ma T, Meng H, Sun L, Li XM, Li XM, Ku JW, Zhou YF, Yang LQ, Wang Z, Li Y, Qige Q, Yang WJ, Lei GY, Chen LQ, Li EM, Yuan L, Yue WB, Wang R, Wang LW, Fan XP, Zhu FH, Zhao WX, Mao YM, Zhang M, Xing GL, Li JL, Han M, Ren JL, Liu B, Ren SW, Kong QP, Li F, Sheyhidin I, Wei W, Zhang YR, Feng CW, Wang J, Yang YH, Hao HZ, Bao QD, Liu BC, Wu AQ, Xie D, Yang WC, Wang L, Zhao XH, Chen SQ, Hong JY, Zhang XJ, Freedman ND, Goldstein AM, Lin D, Taylor PR, Wang LD, Chanock SJ. Joint analysis of three genome-wide association studies of esophageal squamous cell carcinoma in Chinese populations. Nat Genet 2014; 46:1001-1006. [PMID: 25129146 PMCID: PMC4212832 DOI: 10.1038/ng.3064] [Show More Authors] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 07/23/2014] [Indexed: 02/05/2023]
Abstract
We conducted a joint (pooled) analysis of three genome-wide association studies (GWAS) of esophageal squamous cell carcinoma (ESCC) in individuals of Chinese ancestry (5,337 ESCC cases and 5,787 controls) with 9,654 ESCC cases and 10,058 controls for follow-up. In a logistic regression model adjusted for age, sex, study and two eigenvectors, two new loci achieved genome-wide significance, marked by rs7447927 at 5q31.2 (per-allele odds ratio (OR) = 0.85, 95% confidence interval (CI) = 0.82-0.88; P = 7.72 × 10(-20)) and rs1642764 at 17p13.1 (per-allele OR = 0.88, 95% CI = 0.85-0.91; P = 3.10 × 10(-13)). rs7447927 is a synonymous SNP in TMEM173, and rs1642764 is an intronic SNP in ATP1B2, near TP53. Furthermore, a locus in the HLA class II region at 6p21.32 (rs35597309) achieved genome-wide significance in the two populations at highest risk for ESSC (OR = 1.33, 95% CI = 1.22-1.46; P = 1.99 × 10(-10)). Our joint analysis identifies new ESCC susceptibility loci overall as well as a new locus unique to the population in the Taihang Mountain region at high risk of ESCC.
Collapse
Affiliation(s)
- Chen Wu
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhaoming Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Xin Song
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiao-Shan Feng
- Department of Oncology, Pathology, Clinical Laboratory and Respiratory Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China
| | - Christian C. Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Jie He
- Department of Thoracic Surgery, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Xian-Bo Zuo
- Key Laboratory of Dermatology, Anhui Medical University, Anhui, Hefei, China
| | - Wen Tan
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qimin Zhan
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhibin Hu
- Department of Epidemiology and Biostatistics, Cancer Center, Nanjing Medical University, Nanjing, China
| | - Zhonghu He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, China
| | - Weihua Jia
- State Key Laboratory of Oncology in Southern China
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yifeng Zhou
- Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, China
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Xiao-Ou Shu
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Jian-Min Yuan
- University of Pittsburgh Cancer Institute, Pittsburg, PA, USA
| | - Wei Zheng
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Xue-Ke Zhao
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - She-Gan Gao
- Department of Oncology, Pathology, Clinical Laboratory and Respiratory Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China
| | - Zhi-Qing Yuan
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
| | | | - Zong-Min Fan
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ji-Li Cui
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Hong-Li Lin
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xue-Na Han
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Bei Li
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xi Chen
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Basic Oncology and Pathology at College of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Sanford M. Dawsey
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Linda Liao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Maxwell P. Lee
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ti Ding
- Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - You-Lin Qiao
- Department of Epidemiology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Liu
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dianke Yu
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiang Chang
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lixuan Wei
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu-Tang Gao
- Shanghai Cancer Institute, Shanghai, People's Republic of China
| | - Woon-Puay Koh
- Duke-National University of Singapore, Graduate Medical School, Singapore
| | - Yong-Bing Xiang
- Shanghai Cancer Institute, Shanghai, People's Republic of China
| | | | - Jin-Hu Fan
- Department of Epidemiology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing-Jing Han
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Basic Oncology and Pathology at College of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Sheng-Li Zhou
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Zhang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Dong-Yun Zhang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuan Yuan
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Huang
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunling Liu
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kan Zhai
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Qiao
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guangfu Jin
- Department of Epidemiology and Biostatistics, Cancer Center, Nanjing Medical University, Nanjing, China
| | - Chuanhai Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, China
| | - Jianhua Fu
- State Key Laboratory of Oncology in Southern China
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xiaoping Miao
- Key Laboratory for Environment and Health (Ministry of Education), School of Public Health, Huazhong University of Sciences and Technology, Wuhan, China
| | | | | | - Chaoyu Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | | | - Mitchell Gail
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Jeff Yuenger
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Er-Tao Guo
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ai-Li Li
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Medical Oncology, Tumor Hospital of Linzhou, Linzhou, Henan, China
| | - Wei Zhang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xue-Min Li
- Department of Pathology and Thoracic Surgery, Centre for Health Screening and Endoscopy, Cixian Hospital, Cixian, Hebei, China
| | - Liang-Dan Sun
- Key Laboratory of Dermatology, Anhui Medical University, Anhui, Hefei, China
| | - Bao-Gen Ma
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Central Laboratory, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yan Li
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Sa Tang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiu-Qing Peng
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, Pathology, Clinical Laboratory and Respiratory Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China
| | - Jing Liu
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Amy Hutchinson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Kevin Jacobs
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Carol Giffen
- Information Management Services Inc., Silver Spring, Maryland, USA
| | - Laurie Burdette
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Joseph F. Fraumeni
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, Cancer Center, Nanjing Medical University, Nanjing, China
| | - Yang Ke
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, China
| | - Yixin Zeng
- State Key Laboratory of Oncology in Southern China
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Tangchun Wu
- Key Laboratory for Environment and Health (Ministry of Education), School of Public Health, Huazhong University of Sciences and Technology, Wuhan, China
| | - Peter Kraft
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Charles C. Chung
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Margaret A. Tucker
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Zhi-Chao Hou
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ya-Li Liu
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yan-Long Hu
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Liu
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Li Wang
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Department of Basic Oncology, Pathology and Gastroenterology, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Guo Yuan
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Li-Sha Chen
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiao Liu
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Teng Ma
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Hui Meng
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Li Sun
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xin-Min Li
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiu-Min Li
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jian-Wei Ku
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The Second Affiliated Hospital of Nanyang Medical College, Nanyang, Henan, China
| | - Ying-Fa Zhou
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Liu-Qin Yang
- Department of Radiotherapy, Pathology and Pediatrics, Central Hospital of Xinxiang, Xinxiang, Henan, China
| | - Zhou Wang
- Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yin Li
- Department of Thoracic Surgery and Radiotherapy, Cancer Hospital of Henan Province, Zhengzhou, Henan, China
| | - Qirenwang Qige
- Department of Internal Mongolia Medicine, The Affiliated Hospital, Inner Mongolia Medical College, Huhhot, Inner Mongolia, China
| | - Wen-Jun Yang
- Department of Biotechnology, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Guang-Yan Lei
- Department of Thoracic Surgery, Tumor Hospital of Shaanxi Province, Xi'an, Shaanxi, China
| | - Long-Qi Chen
- Department of Thoracic and Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - En-Min Li
- Department of Oncologic Pathology, Medical College of Shantou University, Shantou, Guangdong, China
- Department of Biochemistry and Molecular Biology, Medical College of Shantou University, Shantou, Guangdong, China
| | - Ling Yuan
- Department of Thoracic Surgery and Radiotherapy, Cancer Hospital of Henan Province, Zhengzhou, Henan, China
| | - Wen-Bin Yue
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, Puyang City Oil Field General Hospital, Puyang, Henan, China
| | - Ran Wang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Lu-Wen Wang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xue-Ping Fan
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Fang-Heng Zhu
- Department of Radiotherapy, Pathology and Pediatrics, Central Hospital of Xinxiang, Xinxiang, Henan, China
| | - Wei-Xing Zhao
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yi-Min Mao
- Department of Oncology, Pathology, Clinical Laboratory and Respiratory Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China
| | - Mei Zhang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Guo-Lan Xing
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ji-Lin Li
- Department of Pathology and Thoracic Surgery, Linzhou Esophageal Cancer Hospital, Linzhou, Henan, China
| | - Min Han
- Department of Gastroenterology and Thoracic Surgery, The First People's Hospital of Shangqiu, Shangqiu, Henan, China
| | - Jing-Li Ren
- Department of Basic Oncology, Pathology and Gastroenterology, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Bin Liu
- Department of Gastroenterology, Beijing Tongren Hospital Affiliated to Capital Medical University, Beijing, China
| | - Shu-Wei Ren
- Department of Oncology, Xinyang Central Hospital, Xinyang, Henan, China
| | - Qing-Peng Kong
- State Key Laboratory of Genetic Resource and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Feng Li
- Department of Pathology, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Ilyar Sheyhidin
- Department of Thoracic Surgery, Xinjiang Medical University, Urumqi, Xinjiang, China
- Medical Research Center, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Wu Wei
- Institute of Hematologic Disease, Changzhi Medical University, Changzhi, Shanxi, China
- Department of Pathology, Changzhi Medical University, Changzhi, Shanxi, China
| | - Yan-Rui Zhang
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Central Laboratory, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Chang-Wei Feng
- Department of Basic Oncology, Pathology and Gastroenterology, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Jin Wang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yu-Hua Yang
- Department of Surgery, Hebi Dahejian Hospital, Hebi, Henan, China
| | | | - Qi-De Bao
- Anyang District Hospital, Anyang, Henan, China
| | - Bao-Chi Liu
- Surgical Department of Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Ai-Qun Wu
- Department of Anatomy, the Second Military Medical University of Chinese PLA, Shanghai, China
| | - Dong Xie
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wan-Cai Yang
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
| | - Liang Wang
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Medical College of Wisconsin, Cancer Research Center, Milwaukee, WI, USA
| | - Xiao-Hang Zhao
- National Laboratory of Molecular Oncology, Cancer Institute & Hospital, CAMS, Beijing, China
| | - Shu-Qing Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jun-Yan Hong
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- University of Medicine and Dentistry of New Jersey, Newark, NJ, USA
| | - Xue-Jun Zhang
- Key Laboratory of Dermatology, Anhui Medical University, Anhui, Hefei, China
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Alisa M. Goldstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Dongxin Lin
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Philip R. Taylor
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Li-Dong Wang
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
176
|
Current aspects of vitiligo genetics. Postepy Dermatol Alergol 2014; 31:247-55. [PMID: 25254010 PMCID: PMC4171675 DOI: 10.5114/pdia.2014.43497] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 05/17/2014] [Accepted: 05/27/2014] [Indexed: 12/24/2022] Open
Abstract
Vitiligo is a common acquired depigmentation disorder of the skin manifested by the presence of white macules. The disease occurs at a frequency of approximately 1–4% of the world population. Currently, the most popular theory of vitiligo development is a multifactorial hypothesis according to which genetic conditions predispose vitiligo macules to occur as a result of specific environmental factors. According to the genetic hypothesis, vitiligo inheritance is multigenic. Genetic studies conducted so far concern patients with non-segmental vitiligo. There are three basic techniques of genetic studies: candidate gene association studies, genomewide linkage studies and genome-wide association studies (GWAS). The GWAS are the “gold standard” for detecting susceptibility genes. Up to now, approximately 36 convincing non-segmental vitiligo susceptibility loci have been identified. Approximately 90% of them encode immunoregulatory proteins, while approximately 10% encode melanocyte proteins. The existence of various associations between vitiligo and other autoimmune diseases may provide new knowledge on the causes of many disorders. Examples include the inverse relationship between vitiligo and melanoma and association of vitiligo with other autoimmune diseases. The main goal of all researches is to find new, optimal therapeutic strategies for vitiligo and other autoimmune diseases.
Collapse
|
177
|
Hsiao JJ, Fisher DE. The roles of microphthalmia-associated transcription factor and pigmentation in melanoma. Arch Biochem Biophys 2014; 563:28-34. [PMID: 25111671 DOI: 10.1016/j.abb.2014.07.019] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 07/10/2014] [Accepted: 07/15/2014] [Indexed: 01/27/2023]
Abstract
MITF and pigmentation play important roles in both normal melanocyte and transformed melanoma cell biology. MITF is regulated by many pathways and it also regulates many targets, some of which are still being discovered and functionally validated. MITF is involved in a wide range of processes in melanocytes, including pigment synthesis and lineage survival. Pigmentation itself plays an important role as the interface between genetic and environmental factors that contribute to melanoma.
Collapse
Affiliation(s)
- Jennifer J Hsiao
- Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Building 149, 13th Street, Charlestown, MA 02129, USA
| | - David E Fisher
- Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Building 149, 13th Street, Charlestown, MA 02129, USA.
| |
Collapse
|
178
|
Kocarnik JM, Park SL, Han J, Dumitrescu L, Cheng I, Wilkens LR, Schumacher FR, Kolonel L, Carlson CS, Crawford DC, Goodloe RJ, Dilks H, Baker P, Richardson D, Ambite JL, Song F, Quresh AA, Zhang M, Duggan D, Hutter C, Hindorff LA, Bush WS, Kooperberg C, Le Marchand L, Peters U. Replication of associations between GWAS SNPs and melanoma risk in the Population Architecture Using Genomics and Epidemiology (PAGE) Study. J Invest Dermatol 2014; 134:2049-2052. [PMID: 24480881 PMCID: PMC4057959 DOI: 10.1038/jid.2014.53] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Jonathan M Kocarnik
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
| | - Sungshim Lani Park
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Jiali Han
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Epidemiology, Fairbanks School of Public Health, Simon Cancer Center, Indiana University, Indianapolis, Indiana, USA
| | - Logan Dumitrescu
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Iona Cheng
- Cancer Prevention Institute of California, Fremont, California, USA
| | - Lynne R Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Fredrick R Schumacher
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Laurence Kolonel
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Chris S Carlson
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Dana C Crawford
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Robert J Goodloe
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, USA
| | - Holli Dilks
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, USA
| | - Paxton Baker
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, USA
| | - Danielle Richardson
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, USA
| | - José Luis Ambite
- Information Sciences Institute, University of Southern California, Marina del Rey, California, USA
| | - Fengju Song
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Epidemiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Abrar A Quresh
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Mingfeng Zhang
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - David Duggan
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Carolyn Hutter
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, NCI, NIH, Bethesda, Maryland, USA
| | - Lucia A Hindorff
- Division of Genomic Medicine, NHGRI, NIH, Bethesda, Maryland, USA
| | - William S Bush
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, USA; Department of Biomedical Informatics, Vanderbilt University, Nashville, Tennessee, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
| |
Collapse
|
179
|
Lindqvist PG, Epstein E, Landin-Olsson M, Ingvar C, Nielsen K, Stenbeck M, Olsson H. Avoidance of sun exposure is a risk factor for all-cause mortality: results from the Melanoma in Southern Sweden cohort. J Intern Med 2014; 276:77-86. [PMID: 24697969 DOI: 10.1111/joim.12251] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Sunlight exposure and fair skin are major determinants of human vitamin D production, but they are also risk factors for cutaneous malignant melanoma (MM). There is epidemiological evidence that all-cause mortality is related to low vitamin D levels. METHODS We assessed the avoidance of sun exposure as a risk factor for all-cause mortality for 29 518 Swedish women in a prospective 20-year follow-up of the Melanoma in Southern Sweden (MISS) cohort. Women were recruited from 1990 to 1992 and were aged 25 to 64 years at the start of the study. We obtained detailed information at baseline on their sun exposure habits and potential confounders. Multivariable flexible parametric survival analysis was applied to the data. RESULTS There were 2545 deaths amongst the 29 518 women who responded to the initial questionnaire. We found that all-cause mortality was inversely related to sun exposure habits. The mortality rate amongst avoiders of sun exposure was approximately twofold higher compared with the highest sun exposure group, resulting in excess mortality with a population attributable risk of 3%. CONCLUSION The results of this study provide observational evidence that avoiding sun exposure is a risk factor for all-cause mortality. Following sun exposure advice that is very restrictive in countries with low solar intensity might in fact be harmful to women's health.
Collapse
Affiliation(s)
- P G Lindqvist
- Department of Obstetrics and Gynecology, Clintec, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
180
|
Shin YW, Jung KH, Kim JM, Cho YD, Lee ST, Chu K, Kim M, Lee SK, Han MH, Roh JK. Echocardiographic evidence of innate aortopathy in the human intracranial aneurysm. PLoS One 2014; 9:e100569. [PMID: 24964197 PMCID: PMC4070985 DOI: 10.1371/journal.pone.0100569] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/26/2014] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Intracranial aneurysm (IA) is significantly more prevalent in patients with coarctation of the aorta or bicuspid aortic valve than in the general population, suggesting a common pathophysiology connecting IA and aortopathy. Here, we analyzed echocardiographic aortic root dimension (ARD) in patients with IA to confirm this possibility. METHODS From January 2008 to December 2010, 260 consecutive patients with IA who were admitted to our institution for coil embolization or for acute stroke management and who also underwent echocardiography were enrolled. We hypothesized that patients with large, ruptured, or multiple IAs are more likely to harbor co-prevalent aortopathy as measured by ARD compared to patients with small, isolated, unruptured IAs. Eccentric group was defined as patients aged <55 years with at least one ruptured aneurysm, an aneurysm ≥7 mm in size, or multiple aneurysms; the remainder was classified into a non-eccentric group. Clinical, angiographic, and echocardiographic findings of the two groups were compared. RESULTS ARD was significantly larger in the eccentric group than in the non-eccentric group (P = 0.049), and the difference was confirmed by multivariable analysis (P = 0.02). Subgroup analysis of patients aged <55 years showed similar result for ARD (P = 0.03), whereas hypertension was more associated with the non-eccentric group (P = 0.01). In addition, height was inversely related to aneurysm size after adjustment for age, sex, weight, ARD, smoking status, and number of aneurysms (P = 0.004). CONCLUSIONS A certain group of IA patients share a common intrinsic wall defect with aortopathy. Shared neural crest cell origin may give rise to this phenomenon.
Collapse
Affiliation(s)
- Yong-Won Shin
- Department of Neurology, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Keun-Hwa Jung
- Department of Neurology, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
- Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea
| | - Jeong-Min Kim
- Department of Neurology, Chung-Ang University Medical Center, College of Medicine, Chung-Ang University, Seoul, South Korea
| | - Young Dae Cho
- Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | - Soon-Tae Lee
- Department of Neurology, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
- Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea
| | - Kon Chu
- Department of Neurology, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
- Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea
| | - Manho Kim
- Department of Neurology, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
- Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea
| | - Sang Kun Lee
- Department of Neurology, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
- Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea
| | - Moon Hee Han
- Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | - Jae-Kyu Roh
- Department of Neurology, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
- Department of Neurology, Armed Forces Capital Hospital, Seongnam, Gyeunggido, South Korea
| |
Collapse
|
181
|
Field S, Davies J, Bishop DT, Newton-Bishop JA. Vitamin D and melanoma. DERMATO-ENDOCRINOLOGY 2014; 5:121-9. [PMID: 24494045 PMCID: PMC3897580 DOI: 10.4161/derm.25244] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 05/15/2013] [Accepted: 06/01/2013] [Indexed: 01/23/2023]
Abstract
Recreational sun exposure and sunburn are causal for melanoma but the risk is strongly genetically determined. Health promotion advice about sun protection should be aimed at susceptible individuals (pale skin, freckles, large numbers of melanocytic nevi and a family history). We discuss here the evidence that sun-sensitive people have lower vitamin D levels and that, in practice, it is very difficult for such individuals to achieve sufficient levels without supplementation in the UK at least. We conclude that melanoma susceptible sun-avoidant individuals should be advised to avoid insufficiency by supplementation.
Vitamin D is anti-proliferative in vitro for some melanoma cell lines. In a large melanoma cohort we have observed that lower serum 25-hydroxyvitamin D2/D3 levels at diagnosis were associated with thicker tumors and poorer prognosis (study as yet not validated). In the UK, melanoma patients commonly have sub-optimal 25-hydroxyvitamin D2/D3 levels at and post diagnosis; we discuss approaches to management of such patients based on some new data from our group.
Collapse
Affiliation(s)
- Sinead Field
- Section of Epidemiology and Biostatistics; University of Leeds; Leeds, UK
| | - John Davies
- Section of Epidemiology and Biostatistics; University of Leeds; Leeds, UK
| | - D Tim Bishop
- Section of Epidemiology and Biostatistics; University of Leeds; Leeds, UK
| | | |
Collapse
|
182
|
Wang S, Yan Q, Chen P, Zhao P, Gu A. Association of interferon regulatory factor 4 gene polymorphisms rs12203592 and rs872071 with skin cancer and haematological malignancies susceptibility: a meta-analysis of 19 case-control studies. BMC Cancer 2014; 14:410. [PMID: 24906573 PMCID: PMC4059085 DOI: 10.1186/1471-2407-14-410] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 05/23/2014] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Research has indicated that the rs12203592 and rs872071 interferon regulatory factor 4 (IRF4) gene polymorphisms correlate with the risk of cancer, especially skin cancer and haematological malignancies, but the results remain controversial. To understand better the effects of these two polymorphisms on skin cancer and haematological malignancies susceptibility, a cumulative meta-analysis was performed. METHODS We conducted a search using the PubMed and Web of Science databases for relevant case-control studies published before April 2014. Summary odds ratios (ORs) and corresponding 95% confidence intervals (CIs) were estimated using fixed- or random-effects models where appropriate. Heterogeneity test, publication bias test, and sensitivity analysis were also performed. RESULTS In total, 11 articles comprised of 19 case-control studies were identified; five focused on the rs12203592 polymorphism with 7,992 cases and 8,849 controls, and six were on the rs872071 polymorphism with 3108 cases and 8300 controls. As for rs12203592, a significant correlation with overall skin cancer and haematological malignancies risk was found with the homozygote comparison model (OR=1.566, 95% CI 1.087-2.256) and recessive model (OR=1.526, 95% CI 1.107-2.104). For rs872071, a significantly elevated haematological malignancies risk was observed in all genetic models (homozygote comparison: OR=1.805, 95% CI 1.402-2.323; heterozygote comparison: OR=1.427, 95% CI 1.203-1.692; dominant: OR=1.556, 95% CI 1.281-1.891; recessive: OR=1.432, 95% CI 1.293-1.587; additive: OR=1.349, 95% CI 1.201-1.515). Similarly, increased skin cancer and haematological malignancies risk was also identified after stratification of the SNP data by cancer type, ethnicity and source of controls for both polymorphisms. CONCLUSIONS Our meta-analysis indicated that the rs12203592 and rs872071 IRF4 gene polymorphisms are associated with individual susceptibility to skin cancer and haematological malignancies. Moreover, the effect of the rs12203592 polymorphism on skin cancer risk was particularly prominent among Caucasians. Further functional research should be performed to validate the association.
Collapse
Affiliation(s)
| | | | | | - Peng Zhao
- Department of Neurosurgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, China.
| | | |
Collapse
|
183
|
Kvaskoff M, Bijon A, Mesrine S, Vilier A, Baglietto L, Fournier A, Clavel-Chapelon F, Dossus L, Boutron-Ruault MC. Association between melanocytic nevi and risk of breast diseases: The French E3N prospective cohort. PLoS Med 2014; 11:e1001660. [PMID: 24915306 PMCID: PMC4051602 DOI: 10.1371/journal.pmed.1001660] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 04/30/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND While melanocytic nevi have been associated with genetic factors and childhood sun exposure, several observations also suggest a potential hormonal influence on nevi. To test the hypothesis that nevi are associated with breast tumor risk, we explored the relationships between number of nevi and benign and malignant breast disease risk. METHODS AND FINDINGS We prospectively analyzed data from E3N, a cohort of French women aged 40-65 y at inclusion in 1990. Number of nevi was collected at inclusion. Hazard ratios (HRs) for breast cancer and 95% confidence intervals (CIs) were calculated using Cox proportional hazards regression models. Associations of number of nevi with personal history of benign breast disease (BBD) and family history of breast cancer were estimated using logistic regression. Over the period 15 June 1990-15 June 2008, 5,956 incident breast cancer cases (including 5,245 invasive tumors) were ascertained among 89,902 women. In models adjusted for age, education, and known breast cancer risk factors, women with "very many" nevi had a significantly higher breast cancer risk (HR = 1.13, 95% CI = 1.01-1.27 versus "none"; ptrend = 0.04), although significance was lost after adjustment for personal history of BBD or family history of breast cancer. The 10-y absolute risk of invasive breast cancer increased from 3,749 per 100,000 women without nevi to 4,124 (95% CI = 3,674-4,649) per 100,000 women with "very many" nevi. The association was restricted to premenopausal women (HR = 1.40, ptrend = 0.01), even after full adjustment (HR = 1.34, ptrend = 0.03; phomogeneity = 0.04), but did not differ according to breast cancer type or hormone receptor status. In addition, we observed significantly positive dose-response relationships between number of nevi and history of biopsy-confirmed BBD (n = 5,169; ptrend<0.0001) and family history of breast cancer in first-degree relatives (n = 7,472; ptrend = 0.0003). The main limitations of our study include self-report of number of nevi using a qualitative scale, and self-reported history of biopsied BBD. CONCLUSIONS Our findings suggest associations between number of nevi and the risk of premenopausal breast cancer, BBD, and family history of breast cancer. More research is warranted to elucidate these relationships and to understand their underlying mechanisms.
Collapse
Affiliation(s)
- Marina Kvaskoff
- “Nutrition, Hormones and Women's Health” Team, Inserm U1018, Centre for Research in Epidemiology and Population Health (CESP), F-94805, Villejuif, France
- Université Paris Sud 11, UMRS 1018, F-94807, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Cancer Control Group, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- * E-mail:
| | - Anne Bijon
- “Nutrition, Hormones and Women's Health” Team, Inserm U1018, Centre for Research in Epidemiology and Population Health (CESP), F-94805, Villejuif, France
- Université Paris Sud 11, UMRS 1018, F-94807, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
| | - Sylvie Mesrine
- “Nutrition, Hormones and Women's Health” Team, Inserm U1018, Centre for Research in Epidemiology and Population Health (CESP), F-94805, Villejuif, France
- Université Paris Sud 11, UMRS 1018, F-94807, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
| | - Alice Vilier
- “Nutrition, Hormones and Women's Health” Team, Inserm U1018, Centre for Research in Epidemiology and Population Health (CESP), F-94805, Villejuif, France
- Université Paris Sud 11, UMRS 1018, F-94807, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
| | - Laura Baglietto
- Cancer Epidemiology Centre, Cancer Council of Victoria, Melbourne, Victoria, Australia
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, School of Population Health, University of Melbourne, Victoria, Australia
| | - Agnès Fournier
- “Nutrition, Hormones and Women's Health” Team, Inserm U1018, Centre for Research in Epidemiology and Population Health (CESP), F-94805, Villejuif, France
- Université Paris Sud 11, UMRS 1018, F-94807, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
| | - Françoise Clavel-Chapelon
- “Nutrition, Hormones and Women's Health” Team, Inserm U1018, Centre for Research in Epidemiology and Population Health (CESP), F-94805, Villejuif, France
- Université Paris Sud 11, UMRS 1018, F-94807, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
| | - Laure Dossus
- “Nutrition, Hormones and Women's Health” Team, Inserm U1018, Centre for Research in Epidemiology and Population Health (CESP), F-94805, Villejuif, France
- Université Paris Sud 11, UMRS 1018, F-94807, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
| | - Marie-Christine Boutron-Ruault
- “Nutrition, Hormones and Women's Health” Team, Inserm U1018, Centre for Research in Epidemiology and Population Health (CESP), F-94805, Villejuif, France
- Université Paris Sud 11, UMRS 1018, F-94807, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
| |
Collapse
|
184
|
Frigerio S, Disciglio V, Manoukian S, Peissel B, Della Torre G, Maurichi A, Collini P, Pasini B, Gotti G, Ferrari A, Rivoltini L, Massimino M, Rodolfo M. A large de novo 9p21.3 deletion in a girl affected by astrocytoma and multiple melanoma. BMC MEDICAL GENETICS 2014; 15:59. [PMID: 24884915 PMCID: PMC4036080 DOI: 10.1186/1471-2350-15-59] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 05/05/2014] [Indexed: 11/10/2022]
Abstract
Background Association of melanoma, neural system tumors and germ line mutations at the 9p21 region in the CDKN2A, CDKN2B and CDKN2BAS genes has been reported in a small number of families worldwide and described as a discrete syndrome in melanoma families registered as a rare disease, the melanoma–astrocytoma syndrome. Case presentation We here studied two young patients developing melanoma after radiotherapy for astrocytoma, both reporting lack of family history for melanoma or neural system tumors at genetic counselling. Patient A is a girl treated for anaplastic astrocytoma at 10 years and for multiple melanomas on the scalp associated to dysplastic nevi two years later. Her monozygotic twin sister carried dysplastic nevi and a slow growing, untreated cerebral lesion. Direct sequencing analysis showed no alterations in melanoma susceptibility genes including CDKN2A, CDK4, MC1R and MITF or in TP53. By microsatellite analysis, multiplex ligation-dependent probe amplification, and array comparative genomic hybridization a deletion including the CDKN2A, CDKN2B and CDKN2BAS gene cluster was detected in both twin sisters, encompassing a large region at 9p21.3 and occurring de novo after the loss of one paternal allele. Patient B is a boy of 7 years when treated for astrocytoma then developing melanoma associated to congenital nevi on the head 10 years later: sequencing and multiplex ligation-dependent probe amplification revealed a normal profile of the CDKN2A/CDKN2B/CDKN2BAS region. Array comparative genomic hybridization confirmed the absence of deletions at 9p21.3 and failed to reveal known pathogenic copy number variations. Conclusions By comparison with the other germ line deletions at the CDKN2A, CDKN2B and CDKN2BAS gene cluster reported in melanoma susceptible families, the deletion detected in the two sisters is peculiar for its de novo origin and for its extension, as it represents the largest constitutive deletion at 9p21.3 region identified so far. In addition, the two studied cases add to other evidence indicating association of melanoma with exposure to ionizing radiation and with second neoplasm after childhood cancer. Melanoma should be considered in the monitoring of pigmented lesions in young cancer patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Monica Rodolfo
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, Milan 20133, Italy.
| |
Collapse
|
185
|
Cadby G, Ward S, Cole J, Moses E, Millward M, Palmer L. The association of host and genetic melanoma risk factors with Breslow thickness in the Western Australian Melanoma Health Study. Br J Dermatol 2014; 170:851-7. [DOI: 10.1111/bjd.12829] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2014] [Indexed: 01/03/2023]
Affiliation(s)
- G. Cadby
- Centre for Genetic Origins of Health and Disease; The University of Western Australia; M409, 35 Stirling Highway Crawley 6009 WA Australia
| | - S.V. Ward
- Centre for Genetic Origins of Health and Disease; The University of Western Australia; M409, 35 Stirling Highway Crawley 6009 WA Australia
| | - J.M. Cole
- St John of God Dermatology; St John of God Health Care Subiaco; Suite 301, 25 McCourt Street Subiaco 6008 WA Australia
| | - E.K. Moses
- Centre for Genetic Origins of Health and Disease; The University of Western Australia; M409, 35 Stirling Highway Crawley 6009 WA Australia
| | - M. Millward
- School of Medicine and Pharmacology; The University of Western Australia; M409, 35 Stirling Highway Crawley 6009 WA Australia
| | - L.J. Palmer
- Genetic Epidemiology and Biostatistics Platform; Ontario Institute for Cancer Research; MaRS Centre; South Tower, 101 College Street, Suite 800 Toronto M5G 0A3 ON Canada
- Samuel Lunenfeld Research Institute; 60 Murray St Toronto M5T 3L9 ON Canada
| |
Collapse
|
186
|
Tuominen R, Jönsson G, Enerbäck C, Appelqvist F, Olsson H, Ingvar C, Hansson J, Höiom V. Investigation of a putative melanoma susceptibility locus at chromosome 3q29. Cancer Genet 2014; 207:70-4. [PMID: 24721441 DOI: 10.1016/j.cancergen.2014.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 01/14/2014] [Accepted: 02/18/2014] [Indexed: 12/20/2022]
Abstract
Malignant melanoma, the most fatal form of skin cancer, is currently increasing in incidence in many populations. Approximately 10% of all cases occur in families with an inherited predisposition for melanoma. In Sweden, only a minor portion of such melanoma families carry a mutation in the known melanoma gene CDKN2A, and there is a need to identify additional melanoma susceptibility genes. In a recently performed genome-wide linkage screen, novel loci with suggestive evidence of linkage to melanoma were detected. In this study, we have further analyzed one region on chromosome 3q29. In all, 89 affected and 15 nonaffected family members from 42 melanoma-prone families were genotyped for 34 genetic markers. In a pooled linkage analysis of all 42 families, we detected significant evidence of linkage, with a maximum heterogeneity logarithm of odds (HLOD) score of 3.1 with 83% of the families contributing to the linkage score. The minimum critical region of linkage (defined by a 1LOD score support interval) maps to chromosome 3q29, spans 3.5 Mb of genomic sequence, and harbors 44 identified genes. Sequence variants within this region have previously been associated with cancer susceptibility. This study reports the presence of a putative novel melanoma susceptibility locus in the Swedish population, a finding that needs to be replicated in an independent study on other individuals with familial melanoma. Sequencing of genes in the region may identify novel melanoma-associated mutations.
Collapse
Affiliation(s)
- Rainer Tuominen
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Göran Jönsson
- Department of Oncology, Clinical Sciences, Lund University, Lund, Sweden
| | - Charlotta Enerbäck
- Division of Cell Biology and Dermatology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Frida Appelqvist
- Department of Clinical Genetics, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Håkan Olsson
- Department of Oncology, Clinical Sciences, Lund University, Lund, Sweden
| | | | - Johan Hansson
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Veronica Höiom
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
187
|
Chiu CG, Nakamura Y, Chong KK, Huang SK, Kawas NP, Triche T, Elashoff D, Kiyohara E, Irie RF, Morton DL, Hoon DSB. Genome-wide characterization of circulating tumor cells identifies novel prognostic genomic alterations in systemic melanoma metastasis. Clin Chem 2014; 60:873-85. [PMID: 24718909 DOI: 10.1373/clinchem.2013.213611] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Circulating tumor cells (CTC) have been found in patients with metastatic melanoma and are associated with advanced melanoma stage and poor patient outcome. We hypothesize that CTC harbor genomic changes critical in the development of distant systemic metastasis. Here, we present the first genome-wide copy-number aberration (CNA) and loss of heterozygosity (LOH)-based characterization of melanoma CTC. METHODS CTC were isolated from peripheral blood monocytes of 13 melanoma patients with regional metastasis stage IIIB/C using antibodies against melanoma-associated cell surface gangliosides. RESULTS We characterized 251 CNA in CTC. Comparative analysis demonstrated >90% concordance in single-nucleotide polymorphism profiles between paired CTC and tumor metastases. In particular, there were notable recurring CNA across patients. In exploratory studies, the presence of several top CTC-associated CNA was verified in distant metastasis (stage IV) from 27 patients, suggesting that certain genomic changes are propagated from regional metastasis to CTC and to distant systemic metastases. Lastly, an exploratory biomarker panel derived from 5 CTC-associated CNA [CSMD2 (CUB and Sushi multiple domains 2), 1p35.1; CNTNAP5 (contactin associated protein-like 5), 2q14.3; NRDE2 (NRDE-2, necessary for RNA interference, domain containing), 14q32.11; ADAM6 (ADAM metallopeptidase domain 6, pseudogene), 14q32.33; and TRPM2 (transient receptor potential cation channel, subfamily m, member 2), 21q22.3] conferred prognostic utility for melanoma recurrence [hazard ratio (HR), 1.14; CI, 1.00-1.44; P = 0.0471] and death (HR, 2.86; CI, 1.23-14.42; P = 0.0014) in 35 patients with stage IIIB/C melanoma, with a 5-year disease-free survival of 13% vs 69% (P = 0.0006) and overall survival of 28% vs 94% between high-risk and low-risk groups defined by the biomarker panel, respectively. CONCLUSIONS This study provides the first detailed CNA-based profile of melanoma CTC and illustrates how CTC may be used as a novel approach for identification of systemic metastasis.
Collapse
Affiliation(s)
- Connie G Chiu
- Department of Molecular Oncology and Division of Surgical Oncology, John Wayne Cancer Institute, Santa Monica, CA
| | | | | | | | | | - Timothy Triche
- Center for Personalized Medicine, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - David Elashoff
- Department of Medicine Statistics Core, UCLA School of Medicine, Los Angeles, CA
| | | | - Reiko F Irie
- Department of Biotechnology, John Wayne Cancer Institute, Santa Monica, CA
| | - Donald L Morton
- Division of Surgical Oncology, John Wayne Cancer Institute, Santa Monica, CA
| | | |
Collapse
|
188
|
Abstract
In the past decade, major advances have been made in the understanding of melanoma. New predisposition genes have been reported and key somatic events, such as BRAF mutation, directly translated into therapeutic management. Surgery for localised melanoma and regional lymph node metastases is the standard of care. Sentinel-node biopsy provides precise staging, but has not been reported to affect survival. The effect of lymph-node dissection on survival is a topic of investigation. Two distinct approaches have emerged to try to extend survival in patients with metastatic melanoma: immunomodulation with anti-CTLA4 monoclonal antibodies, and targeted therapy with BRAF inhibitors or MEK inhibitors for BRAF-mutated melanoma. The combination of BRAF inhibitors and MEK inhibitors might improve progression-free survival further and, possibly, increase overall survival. Response patterns differ substantially-anti-CTLA4 immunotherapy can induce long-term responses, but only in a few patients, whereas targeted drugs induce responses in most patients, but nearly all of them relapse because of pre-existing or acquired resistance. Thus, the long-term prognosis of metastatic melanoma remains poor. Anti-PD1 and anti-PDL1 antibodies have emerged as breakthrough drugs for melanoma that have high response rates and long durability. Biomarkers that have predictive value remain elusive in melanoma, although emerging data for adjuvant therapy indicate that interferon sensitivity is associated with ulceration of the primary melanoma. Intense investigation continues for clinical and biological markers that predict clinical benefit of immunotherapeutic drugs, such as interferon alfa or anti-CTLA4 antibodies, and the mechanisms that lead to resistance of targeted drugs.
Collapse
Affiliation(s)
- Alexander M M Eggermont
- Melanoma Unit and INSERM U981, Gustave Roussy Cancer Institute, Grand Paris, Villejuif, France; Erasmus University Medical Centre, Rotterdam, Netherlands.
| | - Alan Spatz
- Department of Pathology, McGill University & Lady Davis Institute for Medical Research, Montreal, QC, Canada
| | - Caroline Robert
- Melanoma Unit and INSERM U981, Gustave Roussy Cancer Institute, Grand Paris, Villejuif, France
| |
Collapse
|
189
|
Kvaskoff M, Han J, Qureshi AA, Missmer SA. Pigmentary traits, family history of melanoma and the risk of endometriosis: a cohort study of US women. Int J Epidemiol 2014; 43:255-63. [PMID: 24343850 PMCID: PMC3937978 DOI: 10.1093/ije/dyt235] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2013] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Endometriosis has been associated with a higher risk of cutaneous melanoma, but the mechanisms underlying this association are unknown.Some constitutional factors known to influence melanoma risk have been associated with endometriosis in some retrospective studies. However, prospective data are scarce, and more research is needed to confirm this potentially novel endometriosis risk profile. METHODS To investigate the relationships between pigmentary traits, family history of melanoma and endometriosis risk, we analysed data from the Nurses’ Health Study II, a cohort of 116 430 female US nurses aged 25–42 years at inclusion in 1989. Data were collected every 2 years with 20 years of follow-up for these analyses. We used Cox proportional hazards regression models to compute relative risks(RRs) and 95% confidence intervals (CIs). RESULTS During 1 212 499 woman-years of follow-up, 4763 cases of laparoscopically-confirmed endometriosis were reported among premenopausal Caucasian women. Endometriosis risk was increased with presence of naevi on the lower legs (RR=1.08, 95% CI=1.021.14) and higher level of skin’s burning reaction to sun exposure in childhood/adolescence (‘burn with blisters’: RR=1.20,95% CI=1.061.36) compared with ‘practically none’;P(trend)=0.0006) and family history of melanoma (RR=1.13, 95%CI=1.011.26). CONCLUSION This assessment reports modest associations between several pigmentary traits, family history of melanoma and endometriosis risk,corroborating the results from previous retrospective studies. Our findings call for further research to better understand the mechanisms under lying these associations.
Collapse
Affiliation(s)
- Marina Kvaskoff
- Channing Division of Network Medicine, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA, Inserm U1018, Centre for Research in Epidemiology and Population Health (CESP), Institut Gustave Roussy,Villejuif, France, Université Paris Sud 11, UMRS 1018, Villejuif, France, Cancer Control Group, Queensland Institute of Medical Research, Herston, QLD, Australia, Department of Dermatology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA, Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA and Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Jiali Han
- Channing Division of Network Medicine, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA, Inserm U1018, Centre for Research in Epidemiology and Population Health (CESP), Institut Gustave Roussy,Villejuif, France, Université Paris Sud 11, UMRS 1018, Villejuif, France, Cancer Control Group, Queensland Institute of Medical Research, Herston, QLD, Australia, Department of Dermatology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA, Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA and Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Abrar A Qureshi
- Channing Division of Network Medicine, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA, Inserm U1018, Centre for Research in Epidemiology and Population Health (CESP), Institut Gustave Roussy,Villejuif, France, Université Paris Sud 11, UMRS 1018, Villejuif, France, Cancer Control Group, Queensland Institute of Medical Research, Herston, QLD, Australia, Department of Dermatology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA, Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA and Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Stacey A Missmer
- Channing Division of Network Medicine, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA, Inserm U1018, Centre for Research in Epidemiology and Population Health (CESP), Institut Gustave Roussy,Villejuif, France, Université Paris Sud 11, UMRS 1018, Villejuif, France, Cancer Control Group, Queensland Institute of Medical Research, Herston, QLD, Australia, Department of Dermatology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA, Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA and Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
190
|
Abstract
Familial melanoma accounts for approximately a tenth of all melanoma cases. The most commonly known melanoma susceptibility gene is the highly penetrant CDKN2A (p16INK4a) locus, which is transmitted in an autosomal dominant fashion and accounts for approximately 20-50 % of familial melanoma cases. Mutated p16INK4a shows impaired capacity to inhibit the cyclin D1-CDK4 complex, allowing for unchecked cell cycle progression. Mutations in the second protein coded by CDKN2A, p14ARF, are much less common and result in proteasomal degradation of p53 with subsequent accumulation of DNA damage as the cell progresses through the cell cycle without a functional p53-mediated DNA damage response. Mutations in CDK4 that impair the inhibitory interaction with p16INK4a also increase melanoma risk but these mutations are extremely rare. Genes of the melanin biosynthetic pathway, including MC1R and MITF, have also been implicated in melanomagenesis. MC1R variants were traditionally thought to increase risk for melanoma secondary to intensified UV-mediated DNA damage in the setting of absent photoprotective eumelanin. Accumulation of pheomelanin, which appears to have a carcinogenic effect regardless of UV exposure, may be a more likely mechanism. Impaired SUMOylation of the E318K variant of MITF results in increased transcription of genes that confer melanocytes with a pro-malignant phenotype. Mutations in the tumor suppressor BAP1 enhance the metastatic potential of uveal melanoma and predispose to cutaneous/ocular melanoma, atypical melanocytic tumors, and other internal malignancies (COMMON syndrome). Genome-wide association studies have identified numerous low-risk alleles. Although several melanoma susceptibility genes have been identified, risk assessment tools have been developed only for the most common gene implicated with hereditary melanoma, CDKN2A. MelaPRO, a validated model that relies on Mendelian inheritance and Bayesian probability theories, estimates carrier probability for CDKN2A and future risk of melanoma taking into account a patient's family and past medical history of melanoma. Genetic testing for CDKN2A mutations is currently available but the Melanoma Genetics Consortium recommends offering such testing to patients only in the context of research protocols because clinical utility is uncertain.
Collapse
|
191
|
Nikolaou V, Stratigos AJ. Emerging trends in the epidemiology of melanoma. Br J Dermatol 2014; 170:11-9. [PMID: 23815297 DOI: 10.1111/bjd.12492] [Citation(s) in RCA: 243] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2013] [Indexed: 12/16/2022]
Abstract
Cutaneous melanoma (CM) is one of the most rapidly growing cancers worldwide, with a consistent increase in incidence among white populations over the past four decades. Despite the early detection of primarily thin melanomas and the improved survival rates observed in several countries, the rate of thick melanomas has remained constant or continues to increase, especially in the older age group. Current considerations in the epidemiology of melanoma focus on the observed survival benefit of females vs. males, the contributing role of indoor tanning in melanoma risk and the diverse effect of sun exposure in the development of different types of melanoma with respect to their clinical and mutational profile. Certain well-known risk factors, such as skin, hair and eye pigmentation and melanocytic naevi have been validated in large-scale association studies, while additional lifestyle factors and iatrogenic exposures, such as immunosuppressive agents and nonsteroidal anti-inflammatory drugs are being investigated. In addition, genome-wide association studies have revealed genetic loci that underlie the genetic susceptibility of melanoma, some of which are related to known risk factors. Recently, an interesting association of melanoma with Parkinson disease has been noted, with a higher than expected frequency of melanoma in patients with Parkinson disease and vice versa. This review article provides an update in the epidemiology of cutaneous melanoma and discusses recent developments in the field.
Collapse
Affiliation(s)
- V Nikolaou
- Department of Dermatology, Andreas Sygros Hospital, University of Athens Medical School, 5 Dragoumi Street, Athens, 16121, Greece
| | | |
Collapse
|
192
|
Bastian BC. The molecular pathology of melanoma: an integrated taxonomy of melanocytic neoplasia. ANNUAL REVIEW OF PATHOLOGY 2014; 9:239-71. [PMID: 24460190 PMCID: PMC4831647 DOI: 10.1146/annurev-pathol-012513-104658] [Citation(s) in RCA: 334] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Melanomas comprise multiple biologically distinct categories, which differ in cell of origin, age of onset, clinical and histologic presentation, pattern of metastasis, ethnic distribution, causative role of UV radiation, predisposing germ-line alterations, mutational processes, and patterns of somatic mutations. Neoplasms are initiated by gain-of-function mutations in one of several primary oncogenes, which typically lead to benign melanocytic nevi with characteristic histologic features. The progression of nevi is restrained by multiple tumor-suppressive mechanisms. Secondary genetic alterations override these barriers and promote intermediate or overtly malignant tumors along distinct progression trajectories. The current knowledge about the pathogenesis and clinical, histologic, and genetic features of primary melanocytic neoplasms is reviewed and integrated into a taxonomic framework.
Collapse
Affiliation(s)
- Boris C Bastian
- Departments of Dermatology and Pathology, Cardiovascular Research Institute, University of California, San Francisco, California 94158-9001;
| |
Collapse
|
193
|
Abstract
In this chapter we consider some key elements in conducting a successful genome-wide association study or GWAS. The first step is to design the study well (Subheading 3.1), paying particular attention to case and control selection and achieving adequate sample size to deal with the large burden of multiple testing. Second, we focus on the crucial step of applying stringent quality control (Subheading 3.2) to genotyping results. The most crucial potential confounding factor in GWAS is population stratification, and we describe methods for accounting for this in study design and analysis (Subheading 3.3). The primary association analysis is relatively straightforward, and we describe the main approaches to this, including evaluation of results (Subheading 3.4). More comprehensive coverage of the genome can be achieved by using an external reference panel to estimate genotypes at untyped variants using imputation (Subheading 3.5), which we consider in some detail. We finish with some observations on following up a GWAS (Subheading 3.6).
Collapse
|
194
|
Dong J, Gao J, Nalls M, Gao X, Huang X, Han J, Singleton AB, Chen H. Susceptibility loci for pigmentation and melanoma in relation to Parkinson's disease. Neurobiol Aging 2013; 35:1512.e5-1512.e10. [PMID: 24439955 DOI: 10.1016/j.neurobiolaging.2013.12.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 12/19/2013] [Indexed: 11/16/2022]
Abstract
Growing evidence suggests that Parkinson's disease (PD) patients have a lower risk for most types of cancer except for melanoma, which has a modest positive association with PD. Pigmentation genes have been hypothesized to contribute to this association. We therefore examined whether genetic susceptibility loci for pigmentation or melanoma was associated with PD risk in 2 large independent datasets. In the Parkinson's Genes and Environment (PAGE) study, we examined 11 single-nucleotide polymorphisms (SNPs) identified from previous genome-wide association studies (GWAS) of pigmentation or melanoma in relation to PD among 808 PD cases and 1623 controls; furthermore, we also examined the colors of hair, eye, or skin and melanoma in relation to PD. In the International Parkinson's Disease Genomic Consortium (IPDGC), we examined a broader selection of 360 pigmentation or melanoma GWAS SNPs in relation to PD among 5,333 PD cases and 12,019 controls. All participants were non-Hispanic Whites. As expected, in the PAGE study, most SNPs were associated with 1 or more pigmentation phenotypes. However, neither these SNPs nor pigmentation phenotypes were associated with PD risk after Bonferroni correction with the exception of rs4911414 at the ASIP gene (p = .001). A total of 18 PD cases (2.2%) and 26 controls (1.6%) had a diagnosis of melanoma with an odds ratio of 1.3 (95% confidence interval: 0.7-2.4). In the IPDGC analysis, none of the 360 SNPs, including rs4911414, were associated with PD risk after adjusting for multiple comparisons. In conclusion, we did not find significant associations between GWAS SNPs of pigmentation or melanoma and the risk for PD.
Collapse
Affiliation(s)
- Jing Dong
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina; Dr. Gao is currently at the University of Chicago, Chicago, Illinois
| | - Jianjun Gao
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina; Dr. Gao is currently at the University of Chicago, Chicago, Illinois
| | - Michael Nalls
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland
| | - Xiang Gao
- Departments of Nutrition and Epidemiology, Harvard School of Public Health, Boston, Massachusetts.,Channing Laboratory, Harvard Medical School, Boston, Massachusetts
| | - Xuemei Huang
- Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Jiali Han
- Channing Laboratory, Harvard Medical School, Boston, Massachusetts
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland
| | - Honglei Chen
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina; Dr. Gao is currently at the University of Chicago, Chicago, Illinois
| | | |
Collapse
|
195
|
Genome-wide association studies of cancer predisposition. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
196
|
Turati F, Negri E, Vecchia CL. Family history and the risk of cancer: genetic factors influencing multiple cancer sites. Expert Rev Anticancer Ther 2013; 14:1-4. [DOI: 10.1586/14737140.2014.863713] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
197
|
Xiao F, Ma J, Cai G, Fang S, Lee JE, Wei Q, Amos CI. Natural and orthogonal model for estimating gene-gene interactions applied to cutaneous melanoma. Hum Genet 2013; 133:559-74. [PMID: 24241239 DOI: 10.1007/s00439-013-1392-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 11/03/2013] [Indexed: 01/24/2023]
Abstract
Epistasis, or gene-gene interaction, results from joint effects of genes on a trait; thus, the same alleles of one gene may display different genetic effects in different genetic backgrounds. In this study, we generalized the coding technique of a natural and orthogonal interaction (NOIA) model for association studies along with gene-gene interactions for dichotomous traits and human complex diseases. The NOIA model which has non-correlated estimators for genetic effects is important for estimating influence from multiple loci. We conducted simulations and data analyses to evaluate the performance of the NOIA model. Both simulation and real data analyses revealed that the NOIA statistical model had higher power for detecting main genetic effects and usually had higher power for some interaction effects than the usual model. Although associated genes have been identified for predisposing people to melanoma risk: HERC2 at 15q13.1, MC1R at 16q24.3 and CDKN2A at 9p21.3, no gene-gene interaction study has been fully explored for melanoma. By applying the NOIA statistical model to a genome-wide melanoma dataset, we confirmed the previously identified significantly associated genes and found potential regions at chromosomes 5 and 4 that may interact with the HERC2 and MC1R genes, respectively. Our study not only generalized the orthogonal NOIA model but also provided useful insights for understanding the influence of interactions on melanoma risk.
Collapse
Affiliation(s)
- Feifei Xiao
- Department of Biostatistics, Yale University School of Public Health, New Haven, CT, USA,
| | | | | | | | | | | | | |
Collapse
|
198
|
Nalls MA, Saad M, Noyce AJ, Keller MF, Schrag A, Bestwick JP, Traynor BJ, Gibbs JR, Hernandez DG, Cookson MR, Morris HR, Williams N, Gasser T, Heutink P, Wood N, Hardy J, Martinez M, Singleton AB. Genetic comorbidities in Parkinson's disease. Hum Mol Genet 2013; 23:831-41. [PMID: 24057672 DOI: 10.1093/hmg/ddt465] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Parkinson's disease (PD) has a number of known genetic risk factors. Clinical and epidemiological studies have suggested the existence of intermediate factors that may be associated with additional risk of PD. We construct genetic risk profiles for additional epidemiological and clinical factors using known genome-wide association studies (GWAS) loci related to these specific phenotypes to estimate genetic comorbidity in a systematic review. We identify genetic risk profiles based on GWAS variants associated with schizophrenia and Crohn's disease as significantly associated with risk of PD. Conditional analyses adjusting for SNPs near loci associated with PD and schizophrenia or PD and Crohn's disease suggest that spatially overlapping loci associated with schizophrenia and PD account for most of the shared comorbidity, while variation outside of known proximal loci shared by PD and Crohn's disease accounts for their shared genetic comorbidity. We examine brain methylation and expression signatures proximal to schizophrenia and Crohn's disease loci to infer functional changes in the brain associated with the variants contributing to genetic comorbidity. We compare our results with a systematic review of epidemiological literature, while the findings are dissimilar to a degree; marginal genetic associations corroborate the directionality of associations across genetic and epidemiological data. We show a strong genetically defined level of comorbidity between PD and Crohn's disease as well as between PD and schizophrenia, with likely functional consequences of associated variants occurring in brain.
Collapse
Affiliation(s)
- Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
199
|
Cust AE, Goumas C, Vuong K, Davies JR, Barrett JH, Holland EA, Schmid H, Agha-Hamilton C, Armstrong BK, Kefford RF, Aitken JF, Giles GG, Bishop D, Newton-Bishop JA, Hopper JL, Mann GJ, Jenkins MA. MC1R genotype as a predictor of early-onset melanoma, compared with self-reported and physician-measured traditional risk factors: an Australian case-control-family study. BMC Cancer 2013; 13:406. [PMID: 24134749 PMCID: PMC3766240 DOI: 10.1186/1471-2407-13-406] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 08/29/2013] [Indexed: 11/10/2022] Open
Abstract
Background Melanocortin-1 receptor (MC1R) gene variants are very common and are associated with melanoma risk, but their contribution to melanoma risk prediction compared with traditional risk factors is unknown. We aimed to 1) evaluate the separate and incremental contribution of MC1R genotype to prediction of early-onset melanoma, and compare this with the contributions of physician-measured and self-reported traditional risk factors, and 2) develop risk prediction models that include MC1R, and externally validate these models using an independent dataset from a genetically similar melanoma population. Methods Using data from an Australian population-based, case-control-family study, we included 413 case and 263 control participants with sequenced MC1R genotype, clinical skin examination and detailed questionnaire. We used unconditional logistic regression to estimate predicted probabilities of melanoma. Results were externally validated using data from a similar study in England. Results When added to a base multivariate model containing only demographic factors, MC1R genotype improved the area under the receiver operating characteristic curve (AUC) by 6% (from 0.67 to 0.73; P < 0.001) and improved the quartile classification by a net 26% of participants. In a more extensive multivariate model, the factors that contributed significantly to the AUC were MC1R genotype, number of nevi and previous non-melanoma skin cancer; the AUC was 0.78 (95% CI 0.75-0.82) for the model with self-reported nevi and 0.83 (95% CI 0.80-0.86) for the model with physician-counted nevi. Factors that did not further contribute were sun and sunbed exposure and pigmentation characteristics. Adding MC1R to a model containing pigmentation characteristics and other self-reported risk factors increased the AUC by 2.1% (P = 0.01) and improved the quartile classification by a net 10% (95% CI 1-18%, P = 0.03). Conclusions Although MC1R genotype is strongly associated with skin and hair phenotype, it was a better predictor of early-onset melanoma than was pigmentation characteristics. Physician-measured nevi and previous non-melanoma skin cancer were also strong predictors. There might be modest benefit to measuring MC1R genotype for risk prediction even if information about traditional self-reported or clinically measured pigmentation characteristics and nevi is already available.
Collapse
|
200
|
|