1
|
Gibbs DC, Small BM, Autuori I, Leong SF, Ali E, Kenney J, Luo L, Kanetsky PA, Busam KJ, Cust AE, Anton-Culver H, Gallagher RP, Zanetti R, Rosso S, Sacchetto L, Edmiston SN, Conway K, Ollila DW, Begg CB, Berwick M, Orlow I, Thomas NE. Association of Inherited Genetic Variants with Multiple Primary Melanoma. Cancer Epidemiol Biomarkers Prev 2025; 34:805-814. [PMID: 40036058 DOI: 10.1158/1055-9965.epi-24-1442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/24/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Recent genome-wide association studies (GWAS) have identified new susceptibility loci for melanoma, but their associations with multiple primary melanoma (MPM) are unclear. METHODS We investigated the associations of 69 SNPs in 39 GWAS-identified loci with odds of MPM relative to single primary melanoma in the international, population-based Genes, Environment, and Melanoma study. Per-minor-allele ORs and 95% confidence intervals (CI) for individuals with MPM "cases" (n = 1,205) relative to single primary melanoma "controls" (n = 2,458) were estimated using multivariable logistic regression, and polygenic risk scores (PRS) were calculated and weighted based on a 2020 GWAS meta-analysis (57 of the 68 independent GWAS SNPs available). RESULTS Thirteen SNPs in 11 gene regions (PARP1, CYP1B1/RMDN3, TERT, RAPGEF5, TYRP1, MTAP, CDKN2A/CDKN2B, KLF4, TYR, SOX6, and ASIP) were statistically significantly associated (P < 0.05) with MPM adjusting for age, sex, age-by-sex interaction, and study center. The highest versus lowest PRS quintile was associated with a 2.81-fold higher odds of MPM (95% CI, 2.10-3.78; P = 7.5 × 10-13); this association was attenuated but remained statistically significant after excluding SNPs individually associated with MPM (OR = 1.75, 95% CI, 1.32-2.31). CONCLUSIONS Inherited genetic variants spanning 11 gene regions were independently associated with MPM. Nonsignificant SNPs were associated with MPM when aggregated into a PRS, indicating that their cumulative effect may influence MPM risk despite lacking individual statistical significance in our study population. IMPACT Our findings provide additional evidence that these loci are associated with melanoma risk and estimate the magnitude of their genetic effect on subsequent (multiple) primary melanoma risk.
Collapse
Affiliation(s)
- David C Gibbs
- Department of Dermatology, Emory University, Atlanta, Georgia
| | - Brittany M Small
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Isidora Autuori
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Siok F Leong
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Emily Ali
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jessica Kenney
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Li Luo
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, New Mexico
| | - Peter A Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Klaus J Busam
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anne E Cust
- The Daffodil Centre, The University of Sydney, a Joint Venture with Cancer Council NSW, Sydney, Australia
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Hoda Anton-Culver
- Department of Epidemiology, University of California, Irvine, California
| | - Richard P Gallagher
- Cancer Control Research, British Columbia Cancer and Department of Dermatology and Skin Science, University of British Columbia, Vancouver, Canada
| | - Roberto Zanetti
- Piedmont Cancer Registry, Center for Cancer Prevention, Torino, Italy
- Fondo Elena Moroni, Torino, Italy
| | - Stefano Rosso
- Piedmont Cancer Registry, Center for Cancer Prevention, Torino, Italy
- Fondo Elena Moroni, Torino, Italy
| | - Lidia Sacchetto
- Piedmont Cancer Registry, Center for Cancer Prevention, Torino, Italy
- Fondo Elena Moroni, Torino, Italy
| | - Sharon N Edmiston
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Kathleen Conway
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
- Department of Epidemiology, Gillings Global School of Public Health, University of North Carolina, Chapel Hill, North Carolina
| | - David W Ollila
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina
| | - Colin B Begg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marianne Berwick
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, New Mexico
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nancy E Thomas
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
2
|
Zheng C, Sarin KY. Unveiling the genetic landscape of hereditary melanoma: From susceptibility to surveillance. Cancer Treat Res Commun 2024; 40:100837. [PMID: 39137473 DOI: 10.1016/j.ctarc.2024.100837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/15/2024]
Abstract
The multifactorial etiology underlying melanoma development involves an array of genetic, phenotypic, and environmental factors. Genetic predisposition for melanoma is further influenced by the complex interplay between high-, medium-, and low-penetrance genes, each contributing to varying degrees of susceptibility. Within this network, high-penetrance genes, including CDKN2A, CDK4, BAP1, and POT1, are linked to a pronounced risk for disease, whereas medium- and low-penetrance genes, such as MC1R, MITF, and others, contribute only moderately to melanoma risk. Notably, these genetic factors not only heighten the risk of melanoma but may also increase susceptibility towards internal malignancies, such as pancreatic cancer, renal cell cancer, or neural tumors. Genetic testing and counseling hold paramount importance in the clinical context of suspected hereditary melanoma, facilitating risk assessment, personalized surveillance strategies, and informed decision-making. As our understanding of the genomic landscape deepens, this review paper aims to comprehensively summarize the genetic underpinnings of hereditary melanoma, as well as current screening and management strategies for the disease.
Collapse
Affiliation(s)
- Chenming Zheng
- Stanford University Department of Dermatology, Redwood City, CA, USA
| | - Kavita Y Sarin
- Stanford University Department of Dermatology, Redwood City, CA, USA.
| |
Collapse
|
3
|
Wang X, Jia Y, Xu X, Hu Y, Fan G, Jing D, Zhang Z, Wang C, Song C, Qin Y, Peng L. Nuclear receptor coactivator 6 (NCoA6) promotes cell proliferation, migration, and invasion in pancreatic cancer. Cancer Med 2023; 12:18425-18439. [PMID: 37553876 PMCID: PMC10524018 DOI: 10.1002/cam4.6427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Nuclear receptor coactivator 6 (NCoA6) is overexpressed in various cancers and considered a multifunctional coactivator of various transcription factors and nuclear receptors. However, the role of NCoA6 in pancreatic ductal adenocarcinoma (PDAC) remains unclear. METHODS NCoA6 expression data in PDAC were extracted from TCGA and GTEx databases, and their correlation with survival outcomes were analyzed using the Kaplan-Meier plotter database. NCoA6 protein expression in PDAC tissues was evaluated using immunohistochemistry. RNA-sequencing technology was used to sequence the transcriptome of NCoA6-silenced PANC-1 cells, followed by differential expression, GO/KEGG and GSEA analyses. The effects of NCoA6 on cell proliferation, migration, invasion, cell cycle, and apoptosis were determined in two representative cell lines (PANC-1 and SW1990). Western blotting, qPCR, and co-immunoprecipitation were performed to explore the mechanism of action of NCoA6 in PDAC cells. RESULTS NCoA6 expression was markedly increased in PDAC tissues, and high NCoA6 expression was associated with poor survival prognosis. However, there was no significant relationship between NCoA6 expression and metastasis in PDAC patients. Our RNA-sequencing data analysis found 1194 significant differentially expressed genes between the control and NCoA6-silenced PANC-1 cells. GO/KEGG analysis results mainly focused on cytokine production, cytokine activity, and cytokine-cytokine receptor interactions. GSEA results showed that the knockdown of NCoA6 affected the expression of histone deacetylase 1 (HDAC1) targeted genes. NCoA6 knockdown suppressed proliferation, migration, and invasion of PDAC cells. Finally, western blotting, qPCR, and co-immunoprecipitation results showed that NCoA6 interacted with HDAC1 and that NCoA6 expression was negatively correlated with F-box and WD repeat domain-containing 7 (FBW7) and caudal-related homeobox transcription factor 2 (CDX2) expression in pancreatic cancer. CONCLUSIONS NCoA6 has a profound effect on cell proliferation, migration, invasion, and prognosis of PDAC and is potentially related to the expression of HDAC1, FBW7, and CDX2. Our results may provide novel therapeutic strategies for PDAC patients.
Collapse
Affiliation(s)
- Xin Wang
- Department of EmergencyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Yuming Jia
- Department of Hepatobiliary SurgeryThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Xiaowu Xu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer Institute, Fudan UniversityShanghaiChina
| | - Yuheng Hu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer Institute, Fudan UniversityShanghaiChina
| | - Guixiong Fan
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer Institute, Fudan UniversityShanghaiChina
| | - Desheng Jing
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer Institute, Fudan UniversityShanghaiChina
| | - Zhilei Zhang
- Department of Hepatobiliary SurgeryThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Chao Wang
- Department of Hepatobiliary SurgeryThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Changfeng Song
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer Institute, Fudan UniversityShanghaiChina
| | - Yi Qin
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer Institute, Fudan UniversityShanghaiChina
| | - Li Peng
- Department of Hepatobiliary SurgeryThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| |
Collapse
|
4
|
Cai ED, Swetter SM, Sarin KY. Association of multiple primary melanomas with malignancy risk: A population-based analysis of entries from the Surveillance, Epidemiology, and End Results program database during 1973-2014. J Am Acad Dermatol 2023; 88:e211-e219. [PMID: 30287320 PMCID: PMC9132559 DOI: 10.1016/j.jaad.2018.09.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Genetic and environmental risk factors have been associated with the development of multiple primary melanomas (MPMs). We hypothesized that individuals with MPMs might have an increased incidence of internal malignancies. OBJECTIVE To identify the risk for subsequent malignancies in MPM patients. METHODS Multiple primary standardized incidence ratios were analyzed for individuals with ≥1, ≥2 and ≥3 primary melanomas (PMs) recorded in the Surveillance, Epidemiology, and End Results database during 1973-2014. RESULTS We identified 223,799 individuals with ≥1 PM, 19,709 with ≥2 PMs, and 3,995 with ≥3 PMs. Risks of subsequent internal malignancy increased with number of PMs, with observed:expected ratios of 0.99, 1.14, and 1.23 (P < .05) for patients with ≥1 PM, ≥2 PMs, and ≥3 PMs, respectively. Internal malignancy was higher in younger MPM patients and those with superficial spreading melanoma. The most common malignancies among MPM patients included breast, prostate, thyroid, soft tissue, brain, kidney, non-Hodgkin lymphoma, and chronic lymphocytic leukemia. Risk for subsequent cutaneous melanoma increased with observed:expected ratios of 8.09, 22.52, 41.03 (P < .05) for patients with ≥1 PM, ≥2 PMs, and ≥3 PMs, respectively. LIMITATIONS Surveillance, Epidemiology, and End Results records limited information about pigmentation phenotypes, histology, and treatments. CONCLUSION Patients with MPMs have an increased risk for subsequent internal and cutaneous malignancies and might benefit from tight adherence to age-specific cancer screening.
Collapse
Affiliation(s)
- Emily D Cai
- Department of Dermatology, Stanford University Medical Center, Redwood City, California
| | - Susan M Swetter
- Department of Dermatology, Stanford University Medical Center, Redwood City, California; Dermatology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Kavita Y Sarin
- Department of Dermatology, Stanford University Medical Center, Redwood City, California.
| |
Collapse
|
5
|
Bahreyni A, Mohamud Y, Luo H. Recent advancements in immunotherapy of melanoma using nanotechnology-based strategies. Biomed Pharmacother 2023; 159:114243. [PMID: 36641926 DOI: 10.1016/j.biopha.2023.114243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Melanoma is a malignant tumor that accounts for the deadliest form of skin cancers. Despite the significant efforts made recently for development of immunotherapeutic strategies including using immune checkpoint inhibitors and cancer vaccines, the clinical outcomes are unsatisfying. Different factors affect efficient cancer immunotherapy such as side-effects, immunosuppressive tumor microenvironment, and tumor heterogeneity. In the past decades, various nanotechnology-based approaches have been developed to enhance the efficacy of cancer immunotherapy, in addition to diminishing the toxicity associated with it. Several studies have shown that proper application of nanomaterials can revolutionize the outcome of immunotherapy in diverse melanoma models. This review summarizes the recent advancement in the integration of nanotechnology and cancer immunotherapy in melanoma treatment. The importance of nanomaterials and their therapeutic advantages for patients with melanoma are also discussed.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Yasir Mohamud
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada.
| |
Collapse
|
6
|
Olsen CM, Pandeya N, Dusingize JC, Neale RE, MacGregor S, Law MH, Whiteman DC. Risk Factors Associated With First and Second Primary Melanomas in a High-Incidence Population. JAMA Dermatol 2023; 159:37-46. [PMID: 36416830 PMCID: PMC9685542 DOI: 10.1001/jamadermatol.2022.4975] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/23/2022] [Indexed: 11/24/2022]
Abstract
Importance An increasing number of people develop more than 1 primary melanoma, yet to date, no population-based prospective cohort studies have reported on risk factors for developing first vs second primary melanomas. Objective To compare the clinical characteristics of first and second melanomas and then to estimate the relative risks of developing 1 vs multiple melanomas associated with demographic, phenotypic, sun exposure, and genetic factors. Design, Setting, and Participants This population-based prospective cohort study included men and women aged 40 to 69 years recruited in 2011 and followed up until December 2018 in Queensland, Australia. Data analysis was performed from February to July 2022. Exposures Self-reported information about demographic, phenotypic, and sun exposure measures captured using a survey completed at baseline, and polygenic risk score for melanoma. Main Outcomes and Measures Incident first or second primary melanoma diagnosis, and histologic and clinical characteristics thereof. The Wei-Lin-Weissfeld model for recurrent events was used to estimate the association of each factor with the risks of first and second primary melanoma. Results A total of 38 845 patients (mean [SD] age at baseline, 56.1 [8.2] years; 17 775 men and 21 070 women) were included in the study. During a median follow-up period of 7.4 years, 1212 (3.1%) participants had a single primary melanoma diagnosis, and 245 (0.6%) had a second primary melanoma diagnosis. Second melanomas were more likely than first melanomas to be in situ; for invasive tumors, second melanomas were more likely to be thin (ie, ≤1 mm) than first melanomas. Having many moles at age 21 years (self-reported using visual scoring tool) was more strongly associated with second (hazard ratio [HR], 6.36; 95% CI, 3.77-10.75) than first primary melanoma (HR, 3.46; 95% CI, 2.72-4.40) (P value for difference between the HRs = .01). A high genetic predisposition (ie, polygenic risk score in tertile 3) was also more strongly associated with second (HR, 3.28; 95% CI, 2.06-5.23) than first melanoma (HR, 2.06; 95% CI, 1.71-2.49; P = .03). Second melanomas were more strongly associated with a history of multiple skin cancer excisions (HR, 2.63; 95% CI, 1.80-3.83) than first melanomas (HR, 1.86; 95% CI, 1.61-2.16; P = .05). For all other phenotypic characteristics and sun exposure measures, similarly elevated associations with first vs second melanomas were observed. Conclusions and Relevance Findings of this cohort study suggest that within the general population, the presence of many nevi and having a high genetic predisposition to melanoma were associated with the highest risks of developing second primary melanomas.
Collapse
Affiliation(s)
- Catherine M. Olsen
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Queensland, Australia
- Faculty of Medicine, University of Queensland, Queensland, Australia
| | - Nirmala Pandeya
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Queensland, Australia
- Faculty of Medicine, University of Queensland, Queensland, Australia
| | - Jean Claude Dusingize
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Rachel E. Neale
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Queensland, Australia
- Faculty of Medicine, University of Queensland, Queensland, Australia
| | - Stuart MacGregor
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Matthew H. Law
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Queensland, Australia
- Faculty of Health, Queensland University of Technology, Queensland, Australia
- School of Biomedical Sciences, University of Queensland, Queensland, Australia
| | - David C. Whiteman
- Departments of Population Health and Computational Biology, QIMR Berghofer Medical Research Institute, Queensland, Australia
- Faculty of Medicine, University of Queensland, Queensland, Australia
| |
Collapse
|
7
|
Antúnez-Lay A, Podlipnik S, Carrera C, Potrony M, Tell-Martí G, Badenas C, Puig-Butille JA, Espinosa N, Puig S, Malvehy J. Synchronous primary cutaneous melanomas: a descriptive study of their clinical features, histology, genetic background of the patients and clinical outcomes. J Eur Acad Dermatol Venereol 2022; 36:2364-2372. [PMID: 35882522 DOI: 10.1111/jdv.18467] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/30/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Around 0.5% cutaneous melanoma (CM) patients will present with synchronous melanomas when first seen. Moreover, 26%-40% of patients with multiple primary melanomas present with synchronous lesions. OBJECTIVES To assess the prevalence, clinical and histopathological characteristics, germline mutations and outcome in patients with synchronous melanoma. METHODS Clinical and histopathological data from 4703 melanoma patients were included. Clinical, histological and genetic mutational status information was analyzed. Kaplan-Meier curves were used to investigate survival outcomes. RESULTS A total of 144 patients (3.06%) presented simultaneously with 2 or more primary melanomas. During follow-up, 25.7% of patients with synchronous melanoma developed a new primary melanoma compared to 8.6% of patients diagnosed with single melanoma (p < 0.001). Germinal CDKN2A mutations were identified in 10.7% of patients with synchronous melanomas and genetic variants in MC1R in 72%. No significant differences in all survival outcomes between patients with synchronous melanomas and single melanomas were found. CONCLUSION Synchronous melanomas are more frequent than previously reported and are more frequent in older patients compared to single melanomas. Moreover, these patients have a higher risk of developing a new primary melanoma during follow-up and have higher rates of germline susceptibility variants. Nevertheless, these findings were not associated with worse outcomes.
Collapse
Affiliation(s)
- A Antúnez-Lay
- Department of Dermatology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona University, Spain
| | - S Podlipnik
- Department of Dermatology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona University, Spain
| | - C Carrera
- Department of Dermatology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona University, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - M Potrony
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain.,Biochemistry and Molecular Genetics Department, Hospital Clínic de Barcelona, IDIBAPS, University of Barcelona, Spain
| | - G Tell-Martí
- Department of Dermatology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona University, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - C Badenas
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain.,Biochemistry and Molecular Genetics Department, Hospital Clínic de Barcelona, IDIBAPS, University of Barcelona, Spain
| | - J A Puig-Butille
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain.,Molecular Biology CORE. Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona University, Spain
| | - N Espinosa
- Department of Dermatology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona University, Spain
| | - S Puig
- Department of Dermatology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona University, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - J Malvehy
- Department of Dermatology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona University, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| |
Collapse
|
8
|
Davari DR, Orlow I, Kanetsky PA, Luo L, Edmiston SN, Conway K, Parrish EA, Hao H, Busam KJ, Sharma A, Kricker A, Cust AE, Anton-Culver H, Gruber SB, Gallagher RP, Zanetti R, Rosso S, Sacchetto L, Dwyer T, Ollila DW, Begg CB, Berwick M, Thomas NE. Disease-Associated Risk Variants in ANRIL Are Associated with Tumor-Infiltrating Lymphocyte Presence in Primary Melanomas in the Population-Based GEM Study. Cancer Epidemiol Biomarkers Prev 2021; 30:2309-2316. [PMID: 34607836 PMCID: PMC8643342 DOI: 10.1158/1055-9965.epi-21-0686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/19/2021] [Accepted: 09/23/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Genome-wide association studies have reported that genetic variation at ANRIL (CDKN2B-AS1) is associated with risk of several chronic diseases including coronary artery disease, coronary artery calcification, myocardial infarction, and type 2 diabetes mellitus. ANRIL is located at the CDKN2A/B locus, which encodes multiple melanoma tumor suppressors. We investigated the association of these variants with melanoma prognostic characteristics. METHODS The Genes, Environment, and Melanoma Study enrolled 3,285 European origin participants with incident invasive primary melanoma. For each of ten disease-associated SNPs at or near ANRIL, we used linear and logistic regression modeling to estimate, respectively, the per allele mean changes in log of Breslow thickness and ORs for presence of ulceration and tumor-infiltrating lymphocytes (TIL). We also assessed effect modification by tumor NRAS/BRAF mutational status. RESULTS Rs518394, rs10965215, and rs564398 passed false discovery and were each associated (P ≤ 0.005) with TILs, although only rs564398 was independently associated (P = 0.0005) with TILs. Stratified by NRAS/BRAF mutational status, rs564398*A was significantly positively associated with TILs among NRAS/BRAF mutant, but not wild-type, cases. We did not find SNP associations with Breslow thickness or ulceration. CONCLUSIONS ANRIL rs564398 was associated with TIL presence in primary melanomas, and this association may be limited to NRAS/BRAF-mutant cases. IMPACT Pathways related to ANRIL variants warrant exploration in relationship to TILs in melanoma, especially given the impact of TILs on immunotherapy and survival.
Collapse
Affiliation(s)
- Danielle R. Davari
- Department of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Peter A. Kanetsky
- Department of Cancer Epidemiology, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Li Luo
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, New Mexico
| | - Sharon N. Edmiston
- Department of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kathleen Conway
- Department of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Eloise A. Parrish
- Department of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Honglin Hao
- Department of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Klaus J. Busam
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ajay Sharma
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anne Kricker
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
| | - Anne E. Cust
- Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, Australia
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Hoda Anton-Culver
- Department of Epidemiology, University of California, Irvine, Irvine, California
| | | | - Richard P. Gallagher
- BC Cancer and Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Roberto Zanetti
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy
| | - Stefano Rosso
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy
| | - Lidia Sacchetto
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy
| | - Terence Dwyer
- Murdoch Children's Research Institute, Melbourne, Australia
- The Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, United Kingdom
- Department of Pediatrics, University of Melbourne, Melbourne, Australia
- Oxford Martin School, University of Oxford, Oxford, United Kingdom
| | - David W. Ollila
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Colin B. Begg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marianne Berwick
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, New Mexico
| | - Nancy E. Thomas
- Department of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
9
|
Association of Melanoma-Risk Variants with Primary Melanoma Tumor Prognostic Characteristics and Melanoma-Specific Survival in the GEM Study. Curr Oncol 2021; 28:4756-4771. [PMID: 34898573 PMCID: PMC8628692 DOI: 10.3390/curroncol28060401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/11/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
Genome-wide association studies (GWAS) and candidate pathway studies have identified low-penetrant genetic variants associated with cutaneous melanoma. We investigated the association of melanoma-risk variants with primary melanoma tumor prognostic characteristics and melanoma-specific survival. The Genes, Environment, and Melanoma Study enrolled 3285 European origin participants with incident invasive primary melanoma. For each of 47 melanoma-risk single nucleotide polymorphisms (SNPs), we used linear and logistic regression modeling to estimate, respectively, the per allele mean changes in log of Breslow thickness and odds ratios for presence of ulceration, mitoses, and tumor-infiltrating lymphocytes (TILs). We also used Cox proportional hazards regression modeling to estimate the per allele hazard ratios for melanoma-specific survival. Passing the false discovery threshold (p = 0.0026) were associations of IRF4 rs12203592 and CCND1 rs1485993 with log of Breslow thickness, and association of TERT rs2242652 with presence of mitoses. IRF4 rs12203592 also had nominal associations (p < 0.05) with presence of mitoses and melanoma-specific survival, as well as a borderline association (p = 0.07) with ulceration. CCND1 rs1485993 also had a borderline association with presence of mitoses (p = 0.06). MX2 rs45430 had nominal associations with log of Breslow thickness, presence of mitoses, and melanoma-specific survival. Our study indicates that further research investigating the associations of these genetic variants with underlying biologic pathways related to tumor progression is warranted.
Collapse
|
10
|
Aoki MN, Stein A, de Oliveira JC, Chammas R, Uno M, Munhoz FBDA, Marin AM, Canzian F. Susceptibility loci for pancreatic cancer in the Brazilian population. BMC Med Genomics 2021; 14:111. [PMID: 33879152 PMCID: PMC8056496 DOI: 10.1186/s12920-021-00956-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 04/08/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PA) is a very aggressive cancer and has one of the poorest prognoses. Usually, the diagnosis is late and resistant to conventional treatment. Environmental and genetic factors contribute to the etiology, such as tobacco and alcohol consumption, chronic pancreatitis, diabetes and obesity. Somatic mutation in pancreatic cancer cells are known and SNP profile by GWAS could access novel genetic risk factors for this disease in different population context. Here we describe a SNP panel for Brazilian pancreatic cancer, together with clinical and epidemiological data. METHODS 78 pancreatic adenocarcinoma and 256 non-pancreatic cancer subjects had 25 SNPs genotyped by real-time PCR. Unconditional logistic regression methods were used to assess the main effects on PA risk, using allelic, co-dominant and dominant inheritance models. RESULTS 9 SNPs were nominally associated with pancreatic adenocarcinoma risk, with 5 of the minor alleles conferring protective effect while 4 related as risk factor. In epidemiological and clinical data, tobacco smoking, diabetes and pancreatitis history were significantly related to pancreatic adenocarcinoma risk. Polygenic risk scores computed using the SNPs in the study showed strong associations with PA risk. CONCLUSION We could assess for the first time some SNPs related with PA in Brazilian populations, a result that could be used for genetic screening in risk population such as familial pancreatic cancer, smokers, alcohol users and diabetes patients.
Collapse
Affiliation(s)
- Mateus Nóbrega Aoki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, PR, Brazil.
| | - Angelika Stein
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Roger Chammas
- Departamento de Radiologia E Oncologia, Centro de Investigação Translacional Em Oncologia, Instituto Do Câncer Do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brasil
| | - Miyuki Uno
- Departamento de Radiologia E Oncologia, Centro de Investigação Translacional Em Oncologia, Instituto Do Câncer Do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brasil
| | - Francielle Boçon de Araújo Munhoz
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, PR, Brazil
| | - Anelis Maria Marin
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, PR, Brazil
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
Juraleviciute M, Nsengimana J, Newton-Bishop J, Hendriks GJ, Slipicevic A. MX2 mediates establishment of interferon response profile, regulates XAF1, and can sensitize melanoma cells to targeted therapy. Cancer Med 2021; 10:2840-2854. [PMID: 33734579 PMCID: PMC8026919 DOI: 10.1002/cam4.3846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 02/02/2021] [Accepted: 02/23/2021] [Indexed: 01/05/2023] Open
Abstract
MX2 is an interferon inducible gene that is mostly known for its antiviral activity. We have previously demonstrated that MX2 is also associated with the tumorigenesis process in melanoma. However, it remains unknown which molecular mechanisms are regulated by MX2 in response to interferon signaling in this disease. Here, we report that MX2 is necessary for the establishment of an interferon‐induced transcriptional profile partially through regulation of STAT1 phosphorylation and other interferon‐related downstream factors, including proapoptotic tumor suppressor XAF1. MX2 and XAF1 expression tightly correlate in both cultured melanoma cell lines and in patient‐derived primary and metastatic tumors, where they also are significantly related with survival. MX2 mediates IFN growth‐inhibitory signals in both XAF1 dependent and independent ways and in a cell type and context‐dependent manner. Higher MX2 expression renders melanoma cells more sensitive to targeted therapy drugs such as vemurafenib and trametinib; however, this effect is XAF1 independent. In summary, we uncovered a new mechanism in the complex regulation of interferon signaling in melanoma that can influence both survival and response to therapy.
Collapse
Affiliation(s)
- Marina Juraleviciute
- Department of Pathology, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jérémie Nsengimana
- Faculty of Medical Sciences, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Julia Newton-Bishop
- Division of Haematology and Immunology, Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Gert J Hendriks
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ana Slipicevic
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
12
|
Reis LB, Bakos RM, Vianna FSL, Macedo GS, Jacovas VC, Ribeiro-Dos-Santos AM, Santos S, Bakos L, Ashton-Prolla P. Skin pigmentation polymorphisms associated with increased risk of melanoma in a case-control sample from southern Brazil. BMC Cancer 2020; 20:1069. [PMID: 33167923 PMCID: PMC7650158 DOI: 10.1186/s12885-020-07485-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/02/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Melanoma is the most aggressive type of skin cancer and is associated with environmental and genetic risk factors. It originates in melanocytes, the pigment-producing cells. Single nucleotide polymorphisms (SNPs) in pigmentation genes have been described in melanoma risk modulation, but knowledge in the field is still limited. METHODS In a case-control approach (107 cases and 119 controls), we investigated the effect of four pigmentation gene SNPs (TYR rs1126809, HERC2 rs1129038, SLC24A5 rs1426654, and SLC45A2 rs16891982) on melanoma risk in individuals from southern Brazil using a multivariate logistic regression model and multifactor dimensionality reduction (MDR) analysis. RESULTS Two SNPs were associated with an increased risk of melanoma in a dominant model: rs1129038AA and rs1426654AA [OR = 2.094 (95% CI: 1.106-3.966), P = 2.3 10- 2 and OR = 7.126 (95% CI: 1.873-27.110), P = 4.0 10- 3, respectively]. SNP rs16891982CC was associated with a lower risk to melanoma development in a log-additive model when the allele C was inherited [OR = 0.081 (95% CI: 0.008-0.782), P = 3 10- 2]. In addition, MDR analysis showed that the combination of the rs1426654AA and rs16891982GG genotypes was associated with a higher risk for melanoma (P = 3 10- 3), with a redundant effect. CONCLUSIONS These results contribute to the current knowledge and indicate that epistatic interaction of these SNPs, with an additive or correlational effect, may be involved in modulating the risk of melanoma in individuals from a geographic region with a high incidence of the disease.
Collapse
Affiliation(s)
- Larissa B Reis
- Serviço de Pesquisa Experimental, Laboratório de Medicina Genômica, Hospital de Clínicas de Porto Alegre (HCPA), Rua Ramiro Barcelos, Porto Alegre, Rio Grande do Sul, 2350, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Renato M Bakos
- Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Serviço de Dermatologia, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Fernanda S L Vianna
- Serviço de Pesquisa Experimental, Laboratório de Medicina Genômica, Hospital de Clínicas de Porto Alegre (HCPA), Rua Ramiro Barcelos, Porto Alegre, Rio Grande do Sul, 2350, Brazil.,Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Gabriel S Macedo
- Serviço de Pesquisa Experimental, Laboratório de Medicina Genômica, Hospital de Clínicas de Porto Alegre (HCPA), Rua Ramiro Barcelos, Porto Alegre, Rio Grande do Sul, 2350, Brazil
| | - Vanessa C Jacovas
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | | | - Sidney Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará (UFPA), Belém, Pará, Brazil
| | - Lúcio Bakos
- Serviço de Dermatologia, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Patricia Ashton-Prolla
- Serviço de Pesquisa Experimental, Laboratório de Medicina Genômica, Hospital de Clínicas de Porto Alegre (HCPA), Rua Ramiro Barcelos, Porto Alegre, Rio Grande do Sul, 2350, Brazil. .,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil. .,Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.
| |
Collapse
|
13
|
Juraleviciute M, Pozniak J, Nsengimana J, Harland M, Randerson-Moor J, Wernhoff P, Bassarova A, Øy GF, Trøen G, Flørenes VA, Bishop DT, Herlyn M, Newton-Bishop J, Slipicevic A. MX 2 is a novel regulator of cell cycle in melanoma cells. Pigment Cell Melanoma Res 2020; 33:446-457. [PMID: 31660681 PMCID: PMC7180100 DOI: 10.1111/pcmr.12837] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/07/2019] [Accepted: 10/18/2019] [Indexed: 12/14/2022]
Abstract
MX2 protein is a dynamin-like GTPase2 that has recently been identified as an interferon-induced restriction factor of HIV-1 and other primate lentiviruses. A single nucleotide polymorphism (SNP), rs45430, in an intron of the MX2 gene, was previously reported as a novel melanoma susceptibility locus in genome-wide association studies. Functionally, however, it is still unclear whether and how MX2 contributes to melanoma susceptibility and tumorigenesis. Here, we show that MX2 is differentially expressed in melanoma tumors and cell lines, with most metastatic cell lines showing lower MX2 expression than primary melanoma cell lines and melanocytes. Furthermore, high expression of MX2 RNA in primary melanoma tumors is associated with better patient survival. Overexpression of MX2 reduces in vivo proliferation partially through inhibition of AKT activation, suggesting that it can act as a tumor suppressor in melanoma. However, we have also identified a subset of melanoma cell lines with high endogenous MX2 expression where downregulation of MX2 leads to reduced proliferation. In these cells, MX2 downregulation interfered with DNA replication and cell cycle processes. Collectively, our data for the first time show that MX2 is functionally involved in the regulation of melanoma proliferation but that its function is context-dependent.
Collapse
Affiliation(s)
| | - Joanna Pozniak
- Division of Haematology and Immunology, Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jérémie Nsengimana
- Division of Haematology and Immunology, Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Mark Harland
- Division of Haematology and Immunology, Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Juliette Randerson-Moor
- Division of Haematology and Immunology, Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Patrik Wernhoff
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Assia Bassarova
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Geir Frode Øy
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Gunhild Trøen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | | | - David Timothy Bishop
- Division of Haematology and Immunology, Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | | | - Julia Newton-Bishop
- Division of Haematology and Immunology, Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Ana Slipicevic
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
14
|
McMeniman EK, Duffy DL, Jagirdar K, Lee KJ, Peach E, McInerney-Leo AM, De'Ambrosis B, Rayner JE, Smithers BM, Soyer HP, Sturm RA. The interplay of sun damage and genetic risk in Australian multiple and single primary melanoma cases and controls. Br J Dermatol 2020; 183:357-366. [PMID: 31794051 DOI: 10.1111/bjd.18777] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Skin phenotype, host genotype and ultraviolet (UV) damage play a role in the development of melanoma. OBJECTIVES To ascertain whether the level of UV damage at the site of melanomas was associated with genetic polymorphisms. METHODS Deep phenotyping was performed on 1244 individuals; 281 with multiple primary melanomas (MPMs), 304 with single primary melanoma (SPM) and 659 convenience controls. Genotype data was generated using the Illumina CoreExome microarray platform, assaying over 500 000 single-nucleotide polymorphisms. A subset of variants were combined to assess a polygenic risk score (PRS) for melanoma. RESULTS Most MPM cases were diagnosed in patients aged > 40 years, in sites with visible chronic UV damage. Women and those diagnosed at age ≤ 40 years were less likely to have perilesional UV damage. Patients with MPM had higher frequencies of MITF E318K, MC1R R-alleles and the ASIP risk haplotype. Individuals who had melanoma in a visibly UV-damaged site were more likely to carry MC1R rs75570604 [odds ratio (OR) 2·5], 9q31.2 rs10816595 (OR 1·4) and MTAP rs869329 (OR 1·4). These same alleles were more common in patients with MPM who were diagnosed at age ≤ 40 years. The mean PRS was significantly higher in MPM than in SPM and controls. Naevus count was comparable in early-onset MPM cases and those diagnosed at age > 40 years. CONCLUSIONS Our cohort demonstrated higher frequencies of previously reported alleles associated with melanoma. MPM melanomas more commonly occur in UV-damaged areas, and these individuals are more likely to carry MC1R red hair colour alleles. Awareness of the interplay of genetic vulnerability with UV damage can stratify risk and guide recommendations for melanoma screening. What's already known about this topic? Skin phenotype, host genotype and ultraviolet (UV) damage all play a role in melanoma development. One of the main risk factors is a personal history of melanoma; second and subsequent primary melanomas account for over 20% of all melanomas registered in Queensland. Multiple loci are associated with melanoma risk, including many low-penetrance loci, which may have a cumulatively significant risk. Population-wide screening programmes for melanoma are not yet economically viable. What does this study add? Patients diagnosed with melanoma at age ≤ 40 years were more likely than older patients to have melanomas in non-UV-damaged sites. Patients with multiple melanomas had higher frequencies of MITF E318K, MC1R R-alleles, and the ASIP extended risk haplotype than patients with single melanoma. CDKN2A, MC1R and MTAP variants were more frequent in patients who developed melanomas at a younger age, but also in those whose melanomas were all on visibly UV-damaged sites. What is the translational message? Incorporating these genetic findings into the known risk factors of skin phenotype and visible UV damage may allow for a more customized and economically feasible approach to early detection of melanoma, particularly in younger patients. Plain language summary available online.
Collapse
Affiliation(s)
- E K McMeniman
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia.,Dermatology Department, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - D L Duffy
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia.,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - K Jagirdar
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - K J Lee
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - E Peach
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - A M McInerney-Leo
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - B De'Ambrosis
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Dermatology Department, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,South East Dermatology, Annerley Square, Annerley, Brisbane, Queensland, Australia
| | - J E Rayner
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - B M Smithers
- Queensland Melanoma Project, School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - H P Soyer
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia.,Dermatology Department, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - R A Sturm
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| |
Collapse
|
15
|
Aref S, Al Saeed A, El Menshawy N, Abdalla D, El Ashery M. Prognostic relevance of telomere length and telomerase reverse transcriptase variant (rs2242652) on the multiple myeloma patients. J Clin Lab Anal 2019; 34:e23133. [PMID: 31814184 PMCID: PMC7171320 DOI: 10.1002/jcla.23133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND The search for enhancement of multiple myeloma prognostic tools is an area of current research. This study aimed to assess the clinicopathological impact of telomere length and telomerase reverse transcriptase (TERT) polymorphic variant, rs2242652, on multiple myeloma (MM) patients. METHODS Fifty MM patients and 50 healthy controls were included. Relative telomere length (RTL) and rs2242652 genotype polymorphic variants of TERT were analyzed using real-time polymerase chain reaction (PCR). The MM patients' group was categorized into stage I (n = 16); stage II (n = 12), and stage III (n = 22). RESULTS The median telomere length was significantly longer in MM patients' group (0.78) as compared to controls (0.43) (P = .001). Multivariate regression analysis revealed that MM patients with RTL < 0.5 had significant poor response for induction remission therapy with odds ratio 26.45. On the other hand, TERT genotyping analysis of rs2242652 revealed insignificant difference between cases and controls (P = .234), regarding to induction remission response. Survival analysis using Kaplan-Meier curve revealed that patients with shorter telomere length and those with TERT genotype GA had shorter overall survival. CONCLUSION Telomere length and TERT rs2242652 genotype polymorphism could be used for refining risk stratification of MM patients.
Collapse
Affiliation(s)
- Salah Aref
- Hematology Unit, Mansoura University Oncology Center, Mansoura University, Mansoura, Egypt
| | - Alshaimaa Al Saeed
- Hematology Unit, Mansoura University Oncology Center, Mansoura University, Mansoura, Egypt
| | - Nadia El Menshawy
- Hematology Unit, Mansoura University Oncology Center, Mansoura University, Mansoura, Egypt
| | - Doaa Abdalla
- Hematology Unit, Mansoura University Oncology Center, Mansoura University, Mansoura, Egypt
| | | |
Collapse
|
16
|
Inherited Melanoma Risk Variants Associated with Histopathologically Amelanotic Melanoma. J Invest Dermatol 2019; 140:918-922.e7. [PMID: 31568773 DOI: 10.1016/j.jid.2019.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/05/2019] [Accepted: 09/09/2019] [Indexed: 01/26/2023]
|
17
|
Casula M, Paliogiannis P, Ayala F, De Giorgi V, Stanganelli I, Mandalà M, Colombino M, Manca A, Sini MC, Caracò C, Ascierto PA, Satta RR, Lissia A, Cossu A, Palmieri G. Germline and somatic mutations in patients with multiple primary melanomas: a next generation sequencing study. BMC Cancer 2019; 19:772. [PMID: 31382929 PMCID: PMC6683413 DOI: 10.1186/s12885-019-5984-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/26/2019] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Multiple primary melanomas (MPM) occur up to 8% of patients with cutaneous malignant melanoma (CMM). They are often sporadic harbouring several somatic mutations, but also familial cases harbouring a CDKN2A germline mutation have been describe in Caucasian populations. The aim of this study was to investigate the incidence, the distribution patterns and the impact of known and unknown germline and somatic mutations in patients with MPM from Italy. MATERIALS AND METHODS One-hundred and two MPM patients were enrolled for germline mutation analysis, and five patients with at least four MPMs were identified for somatic mutation analysis. The demographic, pathologic and clinical features were retrieved from medical records. Molecular analysis for both germline and somatic mutations was performed in genomic DNA from peripheral blood and tissue samples, respectively, through a next generation sequencing approach, using a specific multiple-gene panel constructed by the Italian Melanoma Intergroup for somatic analysis and a commercial cancer hotspot panel for somatic analysis. RESULTS CDKN2A mutations were detected in 6/16 (37.5%) and 3/86 (3.5%) MPM cases with and without family history for melanoma, respectively. Furthermore, multiple MC1R and, to a lesser extent, ATM variants have been identified. BAP1 variants were found only in MPM patients from southern Italy. The most frequent somatic variants were the pathogenic BRAFV600E and TP53, followed by KIT, PIK3CA, KDR, and NRAS. Single APC, ERBB4, MET, JAK3 and other variants with unknown function were also detected. CONCLUSIONS CDNK2A mutation is the most relevant susceptibility mutation in Italian patients with MPM, especially those with a family history for CMM. The prevalence of this mutation and other sequence variants identified in this study varies among specific sub-populations. Furthermore, some heterogeneity in driver somatic mutations between sporadic MPMs has been observed, as well as in a number of associated sequence variants the clinical impact of which needs to be further elucidated.
Collapse
Affiliation(s)
- Milena Casula
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Traversa La Crucca 3, Baldinca Li Punti, 07100, Sassari, Italy
| | - Panagiotis Paliogiannis
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Fabrizio Ayala
- National Tumor Institute "Fondazione G. Pascale", Napoli, Italy
| | - Vincenzo De Giorgi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | | | - Mario Mandalà
- Unit of Medical Oncology, "Papa Giovanni XXIII" Hospital of Bergamo, Bergamo, Italy
| | - Maria Colombino
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Traversa La Crucca 3, Baldinca Li Punti, 07100, Sassari, Italy
| | - Antonella Manca
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Traversa La Crucca 3, Baldinca Li Punti, 07100, Sassari, Italy
| | - Maria Cristina Sini
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Traversa La Crucca 3, Baldinca Li Punti, 07100, Sassari, Italy
| | - Corrado Caracò
- National Tumor Institute "Fondazione G. Pascale", Napoli, Italy
| | | | - Rosanna Rita Satta
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Amelia Lissia
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Antonio Cossu
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Giuseppe Palmieri
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Traversa La Crucca 3, Baldinca Li Punti, 07100, Sassari, Italy.
| |
Collapse
|
18
|
Immunomodulatory germline variation associated with the development of multiple primary melanoma (MPM). Sci Rep 2019; 9:10173. [PMID: 31308438 PMCID: PMC6629847 DOI: 10.1038/s41598-019-46665-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/28/2019] [Indexed: 12/27/2022] Open
Abstract
Multiple primary melanoma (MPM) has been associated with a higher 10-year mortality risk compared to patients with single primary melanoma (SPM). Given that 3–8% of patients with SPM develop additional primary melanomas, new markers predictive of MPM risk are needed. Based on the evidence that the immune system may regulate melanoma progression, we explored whether germline genetic variants controlling the expression of 41 immunomodulatory genes modulate the risk of MPM compared to patients with SPM or healthy controls. By genotyping these 41 variants in 977 melanoma patients, we found that rs2071304, linked to the expression of SPI1, was strongly associated with MPM risk reduction (OR = 0.60; 95% CI = 0.45–0.81; p = 0.0007) when compared to patients with SPM. Furthermore, we showed that rs6695772, a variant affecting expression of BATF3, is also associated with MPM-specific survival (HR = 3.42; 95% CI = 1.57–7.42; p = 0.0019). These findings provide evidence that the genetic variation in immunomodulatory pathways may contribute to the development of secondary primary melanomas and also associates with MPM survival. The study suggests that inherited host immunity may play an important role in MPM development.
Collapse
|
19
|
Lai M, La Rocca V, Amato R, Freer G, Pistello M. Sphingolipid/Ceramide Pathways and Autophagy in the Onset and Progression of Melanoma: Novel Therapeutic Targets and Opportunities. Int J Mol Sci 2019; 20:ijms20143436. [PMID: 31336922 PMCID: PMC6678284 DOI: 10.3390/ijms20143436] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/04/2019] [Accepted: 07/08/2019] [Indexed: 12/20/2022] Open
Abstract
Melanoma is a malignant tumor deriving from neoplastic transformation of melanocytes. The incidence of melanoma has increased dramatically over the last 50 years. It accounts for most cases of skin cancer deaths. Early diagnosis leads to remission in 90% of cases of melanoma; conversely, for melanoma at more advanced stages, prognosis becomes more unfavorable also because dvanced melanoma is often resistant to pharmacological and radiological therapies due to genetic plasticity, presence of cancer stem cells that regenerate the tumor, and efficient elimination of drugs. This review illustrates the role of autophagy in tumor progression and resistance to therapy, focusing on molecular targets for future drugs.
Collapse
Affiliation(s)
- Michele Lai
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy
| | - Veronica La Rocca
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy
| | - Rachele Amato
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy
| | - Giulia Freer
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy
| | - Mauro Pistello
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy.
- Virology Unit, Pisa University Hospital, 56127 Pisa, Italy.
| |
Collapse
|
20
|
Thomas NE, Edmiston SN, Orlow I, Kanetsky PA, Luo L, Gibbs DC, Parrish EA, Hao H, Busam KJ, Armstrong BK, Kricker A, Cust AE, Anton-Culver H, Gruber SB, Gallagher RP, Zanetti R, Rosso S, Sacchetto L, Dwyer T, Ollila DW, Begg CB, Berwick M, Conway K. Inherited Genetic Variants Associated with Melanoma BRAF/NRAS Subtypes. J Invest Dermatol 2018; 138:2398-2404. [PMID: 29753029 PMCID: PMC6200630 DOI: 10.1016/j.jid.2018.04.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 04/08/2018] [Indexed: 10/16/2022]
Abstract
BRAF and NRAS mutations arise early in melanoma development, but their associations with low-penetrance melanoma susceptibility loci remain unknown. In the Genes, Environment and Melanoma Study, 1,223 European-origin participants had their incident invasive primary melanomas screened for BRAF/NRAS mutations and germline DNA genotyped for 47 single-nucleotide polymorphisms identified as low-penetrant melanoma-risk variants. We used multinomial logistic regression to simultaneously examine each single-nucleotide polymorphism's relationship to BRAF V600E, BRAF V600K, BRAF other, and NRAS+ relative to BRAF-/NRAS- melanoma adjusted for study features. IRF4 rs12203592*T was associated with BRAF V600E (odds ratio [OR] = 0.59, 95% confidence interval [CI] = 0.43-0.79) and V600K (OR = 0.65, 95% CI = 0.41-1.03), but not BRAF other or NRAS+ melanoma. A global test of etiologic heterogeneity (Pglobal = 0.001) passed false discovery (Pglobal = 0.0026). PLA2G6 rs132985*T was associated with BRAF V600E (OR = 1.32, 95% CI = 1.05-1.67) and BRAF other (OR = 1.82, 95% CI = 1.11-2.98), but not BRAF V600K or NRAS+ melanoma. The test for etiologic heterogeneity (Pglobal) was 0.005. The IRF4 rs12203592 associations were slightly attenuated after adjustment for melanoma-risk phenotypes. The PLA2G6 rs132985 associations were independent of phenotypes. IRF4 and PLA2G6 inherited genotypes may influence melanoma BRAF/NRAS subtype development.
Collapse
Affiliation(s)
- Nancy E Thomas
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA.
| | - Sharon N Edmiston
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Peter A Kanetsky
- Department of Cancer Epidemiology, Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Li Luo
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - David C Gibbs
- Department of Epidemiology, Emory University, Atlanta, Georgia, USA
| | - Eloise A Parrish
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Honglin Hao
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Klaus J Busam
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Bruce K Armstrong
- School of Public and Global Health, The University of Western Australia, Perth, Australia
| | - Anne Kricker
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
| | - Anne E Cust
- Sydney School of Public Health, The University of Sydney, Sydney, Australia; Melanoma Institute Australia, The University of Sydney, North Sydney, Australia
| | - Hoda Anton-Culver
- Department of Epidemiology, University of California, Irvine, California, USA
| | - Stephen B Gruber
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, USA
| | - Richard P Gallagher
- British Columbia Cancer and Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Roberto Zanetti
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy
| | - Stefano Rosso
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy
| | - Lidia Sacchetto
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy; Politecnico di Torino, Turin, Italy
| | - Terence Dwyer
- George Institute for Global Health, Nuffield Department of Obstetrics and Gynecology, University of Oxford, Oxford, UK
| | - David W Ollila
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA; Department of Surgery, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Colin B Begg
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Marianne Berwick
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Kathleen Conway
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA; Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
21
|
Yu Y, Hu H, Chen JS, Hu F, Fowler J, Scheet P, Zhao H, Huff CD. Integrated case-control and somatic-germline interaction analyses of melanoma susceptibility genes. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2247-2254. [PMID: 29317335 DOI: 10.1016/j.bbadis.2018.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/20/2017] [Accepted: 01/04/2018] [Indexed: 12/18/2022]
Abstract
While a number of genes have been implicated in melanoma susceptibility, the role of protein-coding variation in melanoma development and progression remains underexplored. To better characterize the role of germline coding variation in melanoma, we conducted a whole-exome case-control and somatic-germline interaction study involving 322 skin cutaneous melanoma cases from The Cancer Genome Atlas and 3607 controls of European ancestry. We controlled for cross-platform technological stratification using XPAT and conducted gene-based association tests using VAAST 2. Four established melanoma susceptibility genes achieved nominal statistical significance, MC1R (p = .0014), MITF (p = .0165) BRCA2 (p = .0206), and MTAP (p = .0393). We also observed a suggestive association for FANCA (p = .002), a gene previously implicated in melanoma survival. The association signal for BRCA2 was driven primarily by likely gene disrupting (LGD) variants, with an Odds Ratio (OR) of 5.62 (95% Confidence Interval (CI) 1.03-30.1). In contrast, the association signals for MC1R and MITF were driven primarily by predicted pathogenic missense variants, with estimated ORs of 1.4 to 3.0 for MC1R and 4.1 for MITF. MTAP exhibited an excess of both LGD and predicted damaging missense variants among cases, with ORs of 5.62 and 3.72, respectively, although neither category was significant. For individuals with known or predicted damaging variants, age of disease onset was significantly lower for two of the four genes, MC1R (p = .005) and MTAP (p = .035). In an analysis of germline carrier status and overlapping copy number alterations, we observed no evidence to support a two-hit model of carcinogenesis in any of the four genes. Although MC1R carriers were represented proportionally among the four molecular tumor subtypes, these individuals accounted for 69% of ultraviolet (UV) radiation mutational signatures among triple-wild type tumors (p = .040), highlighting the increased sensitivity to UV exposure among individuals with loss-of-function variants in MC1R.
Collapse
Affiliation(s)
- Yao Yu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hao Hu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jiun-Sheng Chen
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Fulan Hu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Jerry Fowler
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Scheet
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hua Zhao
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chad D Huff
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
22
|
Adler NR, Kelly JW, Haydon A, McLean CA, Mar VJ. Clinicopathological characteristics and prognosis of patients with multiple primary melanomas. Br J Dermatol 2017; 178:e44-e45. [PMID: 28755438 DOI: 10.1111/bjd.15855] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- N R Adler
- Victorian Melanoma Service, Alfred Hospital, Melbourne, Victoria, Australia.,School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - J W Kelly
- Victorian Melanoma Service, Alfred Hospital, Melbourne, Victoria, Australia
| | - A Haydon
- Victorian Melanoma Service, Alfred Hospital, Melbourne, Victoria, Australia.,Department of Medical Oncology, Alfred Hospital, Melbourne, Victoria, Australia
| | - C A McLean
- Victorian Melanoma Service, Alfred Hospital, Melbourne, Victoria, Australia.,Department of Anatomical Pathology, Alfred Hospital, Melbourne, Victoria, Australia
| | - V J Mar
- Victorian Melanoma Service, Alfred Hospital, Melbourne, Victoria, Australia.,School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia.,Skin and Cancer Foundation, Carlton, Victoria, Australia
| |
Collapse
|
23
|
Tang J, Hu C, Mei H, Peng L, Li H. CLPTM1L gene rs402710 (C > T) and rs401681 (C > T) polymorphisms associate with decreased cancer risk: a meta-analysis. Oncotarget 2017; 8:102446-102457. [PMID: 29254260 PMCID: PMC5731970 DOI: 10.18632/oncotarget.22268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 09/20/2017] [Indexed: 12/15/2022] Open
Abstract
Cleft lip and palate transmembrane 1-like (CLPTM1L) gene rs402710 (C > T) and rs401681 (C > T) polymorphisms have been widely studied for their potential relation to cancer risk, but studies have produced conflicting results. To systematically evaluate the association between these two polymorphisms and overall cancer risk, we conducted a comprehensive meta-analysis on all relevant articles found in the PubMed and EMBASE databases published prior to May 1, 2017. There were 26 articles with 28 studies, including 30,770 cases and 34,089 controls, for the rs402710 polymorphism and 38 articles with 48 studies, including 67,849 cases and 328,226 controls, for the rs401681 polymorphism. The pooled results indicated that both rs402710 and rs401681 polymorphisms are significantly associated with decreased overall cancer risk. In our stratification analysis, a significant association of the rs402710 polymorphism with lung and bladder cancers was identified among Asian and Caucasian populations in both hospital-based and population-based studies. The rs401681 polymorphism was significantly associated with a decreased risk of lung cancer, bladder cancer, and basal cell carcinoma in Asians and in hospital-based studies. CLPTM1L gene rs402710 and rs401681 polymorphisms thus have a protective association with various types of cancer, especially lung cancer among Asians.
Collapse
Affiliation(s)
- Jianzhou Tang
- Department of Biological and Environmental Engineering, Changsha University, Changsha 410003, Hunan, China
| | - Changming Hu
- Department of Molecular Pathology, Guangzhou Kingmed Center for Clinical Laboratory, Guangzhou 510000, Guangdong, China
| | - Hua Mei
- Department of Somatic Stem Cell, Hunan Guangxiu Hospital, Changsha 410002, Hunan, China
| | - Liang Peng
- Department of Biological and Environmental Engineering, Changsha University, Changsha 410003, Hunan, China
| | - Hui Li
- Department of Microbiology and Immunology, Medical School of Jishou University, Jishou 416000, Hunan, China
| |
Collapse
|
24
|
Gibbs DC, Ward SV, Orlow I, Cadby G, Kanetsky PA, Luo L, Busam KJ, Kricker A, Armstrong BK, Cust AE, Anton-Culver H, Gallagher RP, Zanetti R, Rosso S, Sacchetto L, Ollila DW, Begg CB, Berwick M, Thomas NE. Functional melanoma-risk variant IRF4 rs12203592 associated with Breslow thickness: a pooled international study of primary melanomas. Br J Dermatol 2017; 177:e180-e182. [PMID: 28667740 DOI: 10.1111/bjd.15784] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- D C Gibbs
- Department of Epidemiology, Emory University, Atlanta, GA, U.S.A
| | - S V Ward
- Centre for Genetic Origins of Health and Disease, The University of Western Australia, Crawley, Western Australia, Australia.,Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY, U.S.A
| | - I Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY, U.S.A
| | - G Cadby
- Centre for Genetic Origins of Health and Disease, The University of Western Australia, Crawley, Western Australia, Australia
| | - P A Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, U.S.A
| | - L Luo
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM, U.S.A
| | - K J Busam
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY, U.S.A
| | - A Kricker
- Sydney School of Public Health, The University of Sydney, Sydney, New South Wales, Australia
| | - B K Armstrong
- Sydney School of Public Health, The University of Sydney, Sydney, New South Wales, Australia
| | - A E Cust
- Sydney School of Public Health, The University of Sydney, Sydney, New South Wales, Australia.,Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - H Anton-Culver
- Department of Epidemiology, University of California, Irvine, CA, U.S.A
| | - R P Gallagher
- Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - R Zanetti
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy
| | - S Rosso
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy
| | - L Sacchetto
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy.,Politecnico di Torino, Turin, Italy
| | - D W Ollila
- Department of Surgery, University of North Carolina, Chapel Hill, NC, U.S.A.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, U.S.A
| | - C B Begg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY, U.S.A
| | - M Berwick
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM, U.S.A
| | - N E Thomas
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, U.S.A.,Department of Dermatology, University of North Carolina, Chapel Hill, NC, U.S.A
| | | |
Collapse
|
25
|
Vernali S, Waxweiler WT, Dillon PM, Kanetsky PA, Orlow I, Luo L, Busam KJ, Kricker A, Armstrong BK, Anton-Culver H, Gruber SB, Gallagher RP, Zanetti R, Rosso S, Sacchetto L, Dwyer T, Cust AE, Ollila DW, Begg CB, Berwick M, Thomas NE. Association of Incident Amelanotic Melanoma With Phenotypic Characteristics, MC1R Status, and Prior Amelanotic Melanoma. JAMA Dermatol 2017; 153:1026-1031. [PMID: 28746718 PMCID: PMC5650093 DOI: 10.1001/jamadermatol.2017.2444] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Importance We previously reported that survival is poorer from histopathologically amelanotic than pigmented melanoma because of more advanced stage at diagnosis. Identifying patients at risk of amelanotic melanoma might enable earlier diagnosis and improved survival; however, the phenotypic characteristics and underlying genetics associated with amelanotic melanoma are unknown. Objective To determine whether phenotypic characteristics, carriage of MC1R variants, and history of amelanotic melanoma are associated with histopathologically amelanotic melanoma. Design, Setting, and Participants The Genes, Environment, and Melanoma (GEM) study is an international cohort study that enrolled patients with incident primary cutaneous melanomas from population-based and hospital-based cancer registries (1998 to 2003). The GEM participants included here were 2387 patients with data for phenotypes, MC1R genotype, and primary melanomas scored for histopathologic pigmentation. Of these 2387 patients with incident melanomas scored for pigmentation, 527 had prior primary melanomas also scored for pigmentation. Main Outcomes and Measures Associations of phenotypic characteristics (freckles, nevi, phenotypic index) and MC1R status with incident amelanotic melanomas were evaluated using logistic regression models adjusted for age, sex, study center, and primary status (single or multiple primary melanoma); odds ratios (ORs) and 95% CIs are reported. Association of histopathologic pigmentation between incident and prior melanomas was analyzed using an exact logistic regression model. Results This study included 2387 patients (1065 women, 1322 men; mean [SD] age at diagnosis, 58.3 [16.1] years) and 2917 primary melanomas. In a multivariable model including phenotypic characteristics, absence of back nevi, presence of many freckles, and a sun-sensitive phenotypic index were independently associated with amelanotic melanoma. Carriage of MC1R variants was associated with amelanotic melanoma but lost statistical significance in a model with phenotype. Further, patients with incident primary amelanotic melanomas were more likely to have had a prior primary amelanotic melanoma (OR, 4.62; 95% CI, 1.25-14.13) than those with incident primary pigmented melanomas. Conclusions and Relevance Absence of back nevi, presence of many freckles, a sun-sensitive phenotypic index, and prior amelanotic melanoma increase odds for development of amelanotic melanoma. An increased index of suspicion for melanoma in presenting nonpigmented lesions and more careful examination for signs of amelanotic melanoma during periodic skin examination in patients at increased odds of amelanotic melanoma might lead to earlier diagnosis and improved survival.
Collapse
Affiliation(s)
- Steven Vernali
- Department of Dermatology, University of North Carolina, Chapel Hill
| | | | | | - Peter A Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York
| | - Li Luo
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque
| | - Klaus J Busam
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York
| | - Anne Kricker
- Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia
| | - Bruce K Armstrong
- Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia
| | | | - Stephen B Gruber
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles
| | - Richard P Gallagher
- Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Roberto Zanetti
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy
| | - Stefano Rosso
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy
| | - Lidia Sacchetto
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy
- Politecnico di Torino, Turin, Italy
- Universitá degli Studi di Torino, Turin, Italy
| | - Terence Dwyer
- George Institute for Global Health, Nuffield Department of Obstetrics and Gynecology, University of Oxford, England
| | - Anne E Cust
- Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia
| | - David W Ollila
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill
- Department of Surgery, University of North Carolina, Chapel Hill
| | - Colin B Begg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York
| | - Marianne Berwick
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque
| | - Nancy E Thomas
- Department of Dermatology, University of North Carolina, Chapel Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill
| |
Collapse
|
26
|
Wang S, Huo D, Kupfer S, Alleyne D, Ogundiran TO, Ojengbede O, Zheng W, Nathanson KL, Nemesure B, Ambs S, Olopade OI, Zheng Y. Genetic variation in the vitamin D related pathway and breast cancer risk in women of African ancestry in the root consortium. Int J Cancer 2017; 142:36-43. [PMID: 28891071 DOI: 10.1002/ijc.31038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 07/07/2017] [Accepted: 07/12/2017] [Indexed: 01/08/2023]
Abstract
The vitamin D related pathway has been evaluated in carcinogenesis but its genetic contribution remains poorly understood. We examined single-nucleotide polymorphisms (SNPs) in the vitamin D related pathway genes using data from a genome-wide association study (GWAS) of breast cancer in the African Diaspora that included 3,686 participants (1,657 cases). Pathway- and gene-level analyses were conducted using the adaptive rank truncated product test. Odds ratios (OR) and 95% confidence intervals (CI) were estimated at SNP-level. After stringent Bonferroni corrections, we observed no significant association between variants in the vitamin D pathway and breast cancer risk at the pathway-, gene-, or SNP-level. In addition, no association was found for either the reported signals from GWASs of vitamin D related traits, or the SNPs within vitamin D receptor (VDR) binding regions. Furthermore, a decrease in genetically predicted 25(OH)D levels by Mendelian randomization was not associated with breast cancer (p = 0.23). However, an association for breast cancer with the pigment synthesis/metabolism pathway almost approached significance (pathway-level p = 0.08), driven primarily by a nonsense SNP rs41302073 in TYRP1, with an OR of 1.54 (95% CI = 1.24-1.91, padj = 0.007). In conclusion, we found no evidence to support an association between vitamin D status and breast cancer risk in women of African ancestry, suggesting that vitamin D is unlikely to have significant effect on breast carcinogenesis. Interestingly, TYRP1 might be related to breast cancer through a non-vitamin D relevant mechanism but further studies are needed.
Collapse
Affiliation(s)
- Shengfeng Wang
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL
| | - Dezheng Huo
- Department of Public Health Sciences, University of Chicago, Chicago, IL
| | - Sonia Kupfer
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Dereck Alleyne
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Temidayo O Ogundiran
- Department of Surgery, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Oladosu Ojengbede
- Center for Population and Reproductive Health, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN
| | | | - Barbara Nemesure
- Department of Preventive Medicine, State University of New York at Stony Brook, Stony Brook, NY
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, National Cancer Institute, Bethesda, MD
| | - Olufunmilayo I Olopade
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL
| | - Yonglan Zheng
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL
| |
Collapse
|
27
|
Wu D, Zhu G, Zeng J, Song W, Wang K, Wang X, Guo P, He D. Genetic variations in TERC and TERT genes are associated with renal cell carcinoma risk in a Chinese Han population. Oncotarget 2017; 8:76832-76842. [PMID: 29100352 PMCID: PMC5652746 DOI: 10.18632/oncotarget.20163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022] Open
Abstract
Renal cell carcinoma (RCC) is a common malignant tumor of the urinary system, the pathogenesis of RCC is still unclear. It is reported that genetic variations in telomere length related-genes TERT and TERC are involved in the many types of cancers. However, little is known about the association between TERT and TERC polymorphisms and susceptibility to RCC risk. To solve this problem, a total of 293 patients with primary renal cell carcinoma and 459 healthy people were recruited in our study. Six SNPs of TERC and TERT were genotyped, and association analysis was performed. We found TERC-rs35073794 and TERT-rs10069690 were associated with an increased risk of RCC in an allele model. (OR =2.39, 95% CI = 0.99-5.80, p = 0.047; OR =1.39, 95% CI = 1.07-1.81, p = 0.014, respectively). The genotype "TC" of rs10069690 was associated with an increased risk of RCC in the genotype model. (OR =1.52, 95% CI = 1.11-2.08, p = 0.009).TERC-rs35073794 was associated with an increased risk of RCC in the codominant model. (OR =2.61, 95% CI = 1.01-6.76, p = 0.045). Rs10069690 was associated with an increased risk of RCC under the dominant model. (OR=1.44, 95% CI= 1.04-2.01, p = 0.03). Haplotype "CA" was found to be associated with a decreased risk of RCC while haplotype "TA" was associated with an increased risk of RCC without adjustment for gender, age and body mass index (BMI). (OR=0.07; 95% CI= 0.01-0.54; p=0.011; OR= 1.24; 95% CI= 0.92-1.65; p=0.013, respectively). Rs35073794, rs10936599 and rs10069690 were positively correlated with the age older than 55 (OR= 3.27, 95%CI= 1.08-9.93, p=0.031; OR= 1.56, 95%CI= 1.03-2.37, p= 0.034; OR= 4.94, 95%CI= 1.18-20.70, p= 0.022, respectively) with or without history of drinking(OR= 4.47, 95%CI= 0.99-20.25, p= 0.024; OR= 2.62, 95%CI= 1.13-6.08, p= 0.022; OR=2.44, 95%CI=1.03-5.78, p= 0.04, respectively) and clinical stage I/II RCC (OR=2.62, 95%CI=1.02-6.74, p= 0.045; OR= 2.23, 95%CI= 1.08-4.60, p= 0.028; OR= 1.63, 95%CI= 1.17-2.27, p= 0.014, respectively). Our study indicated a significant association between SNPs in the TERC, TERT and RCC risk in a Chinese Han population. It could be used as diagnostic and prognostic markers in clinical studies of renal cell carcinoma patients.
Collapse
Affiliation(s)
- Dapeng Wu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Guodong Zhu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Jin Zeng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Wenbin Song
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Ke Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Xinyang Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Peng Guo
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Dalin He
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| |
Collapse
|
28
|
Lim S, Youn JP, Hong S, Choi D, Moon S, Kim W, Han M, Hwang SY. Customized multiplexing SNP panel for Korean-specific DNA phenotyping in forensic applications. Genes Genomics 2017. [DOI: 10.1007/s13258-016-0509-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
29
|
Mauguen A, Zabor EC, Thomas NE, Berwick M, Seshan VE, Begg CB. Defining Cancer Subtypes With Distinctive Etiologic Profiles: An Application to the Epidemiology of Melanoma. J Am Stat Assoc 2017; 112:54-63. [PMID: 28603323 DOI: 10.1080/01621459.2016.1191499] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
We showcase a novel analytic strategy to identify sub-types of cancer that possess distinctive causal factors, i.e. sub-types that are "etiologically" distinct. The method involves the integrated analysis of two types of study design: an incident series of cases with double primary cancers with detailed information on tumor characteristics that can be used to define the sub-types; a case-series of incident cases with information on known risk factors that can be used to investigate the specific risk factors that distinguish the sub-types. The methods are applied to a rich melanoma dataset with detailed information on pathologic tumor factors, and comprehensive information on known genetic and environmental risk factors for melanoma. Identification of the optimal sub-typing solution is accomplished using a novel clustering analysis that seeks to maximize a measure that characterizes the distinctiveness of the distributions of risk factors across the sub-types and that is a function of the correlations of tumor factors in the case-specific tumor pairs. This analysis is challenged by the presence of extensive missing data. If successful, studies of this nature offer the opportunity for efficient study design to identify unknown risk factors whose effects are concentrated in defined sub-types.
Collapse
Affiliation(s)
- Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Emily C Zabor
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Nancy E Thomas
- Department of Dermatology, University of North Carolina, Chapel Hill, NC.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Marianne Berwick
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Venkatraman E Seshan
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Colin B Begg
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY
| |
Collapse
|
30
|
Stray-Pedersen A, Sorte HS, Samarakoon P, Gambin T, Chinn IK, Coban Akdemir ZH, Erichsen HC, Forbes LR, Gu S, Yuan B, Jhangiani SN, Muzny DM, Rødningen OK, Sheng Y, Nicholas SK, Noroski LM, Seeborg FO, Davis CM, Canter DL, Mace EM, Vece TJ, Allen CE, Abhyankar HA, Boone PM, Beck CR, Wiszniewski W, Fevang B, Aukrust P, Tjønnfjord GE, Gedde-Dahl T, Hjorth-Hansen H, Dybedal I, Nordøy I, Jørgensen SF, Abrahamsen TG, Øverland T, Bechensteen AG, Skogen V, Osnes LTN, Kulseth MA, Prescott TE, Rustad CF, Heimdal KR, Belmont JW, Rider NL, Chinen J, Cao TN, Smith EA, Caldirola MS, Bezrodnik L, Lugo Reyes SO, Espinosa Rosales FJ, Guerrero-Cursaru ND, Pedroza LA, Poli CM, Franco JL, Trujillo Vargas CM, Aldave Becerra JC, Wright N, Issekutz TB, Issekutz AC, Abbott J, Caldwell JW, Bayer DK, Chan AY, Aiuti A, Cancrini C, Holmberg E, West C, Burstedt M, Karaca E, Yesil G, Artac H, Bayram Y, Atik MM, Eldomery MK, Ehlayel MS, Jolles S, Flatø B, Bertuch AA, Hanson IC, Zhang VW, Wong LJ, Hu J, Walkiewicz M, Yang Y, Eng CM, Boerwinkle E, Gibbs RA, Shearer WT, Lyle R, Orange JS, Lupski JR. Primary immunodeficiency diseases: Genomic approaches delineate heterogeneous Mendelian disorders. J Allergy Clin Immunol 2017; 139:232-245. [PMID: 27577878 PMCID: PMC5222743 DOI: 10.1016/j.jaci.2016.05.042] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/10/2016] [Accepted: 05/13/2016] [Indexed: 11/15/2022]
Abstract
BACKGROUND Primary immunodeficiency diseases (PIDDs) are clinically and genetically heterogeneous disorders thus far associated with mutations in more than 300 genes. The clinical phenotypes derived from distinct genotypes can overlap. Genetic etiology can be a prognostic indicator of disease severity and can influence treatment decisions. OBJECTIVE We sought to investigate the ability of whole-exome screening methods to detect disease-causing variants in patients with PIDDs. METHODS Patients with PIDDs from 278 families from 22 countries were investigated by using whole-exome sequencing. Computational copy number variant (CNV) prediction pipelines and an exome-tiling chromosomal microarray were also applied to identify intragenic CNVs. Analytic approaches initially focused on 475 known or candidate PIDD genes but were nonexclusive and further tailored based on clinical data, family history, and immunophenotyping. RESULTS A likely molecular diagnosis was achieved in 110 (40%) unrelated probands. Clinical diagnosis was revised in about half (60/110) and management was directly altered in nearly a quarter (26/110) of families based on molecular findings. Twelve PIDD-causing CNVs were detected, including 7 smaller than 30 Kb that would not have been detected with conventional diagnostic CNV arrays. CONCLUSION This high-throughput genomic approach enabled detection of disease-related variants in unexpected genes; permitted detection of low-grade constitutional, somatic, and revertant mosaicism; and provided evidence of a mutational burden in mixed PIDD immunophenotypes.
Collapse
Affiliation(s)
- Asbjørg Stray-Pedersen
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Center for Human Immunobiology of Texas Children's Hospital/Department of Pediatrics, Baylor College of Medicine, Houston, Tex; Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Norwegian National Unit for Newborn Screening, Oslo University Hospital, Oslo, Norway; Department of Pediatrics, Oslo University Hospital, Oslo, Norway.
| | - Hanne Sørmo Sorte
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Pubudu Samarakoon
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tomasz Gambin
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Institute of Computer Science, Warsaw University of Technology, Warsaw, Poland
| | - Ivan K Chinn
- Center for Human Immunobiology of Texas Children's Hospital/Department of Pediatrics, Baylor College of Medicine, Houston, Tex; Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex
| | - Zeynep H Coban Akdemir
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | | | - Lisa R Forbes
- Center for Human Immunobiology of Texas Children's Hospital/Department of Pediatrics, Baylor College of Medicine, Houston, Tex; Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex
| | - Shen Gu
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Bo Yuan
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Shalini N Jhangiani
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Human Genome Sequencing Center, Baylor College of Medicine, Houston, Tex
| | - Donna M Muzny
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Human Genome Sequencing Center, Baylor College of Medicine, Houston, Tex
| | | | - Ying Sheng
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Sarah K Nicholas
- Center for Human Immunobiology of Texas Children's Hospital/Department of Pediatrics, Baylor College of Medicine, Houston, Tex; Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex
| | - Lenora M Noroski
- Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex
| | - Filiz O Seeborg
- Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex
| | - Carla M Davis
- Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex
| | - Debra L Canter
- Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex
| | - Emily M Mace
- Center for Human Immunobiology of Texas Children's Hospital/Department of Pediatrics, Baylor College of Medicine, Houston, Tex; Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex
| | - Timothy J Vece
- Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex
| | - Carl E Allen
- Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, Tex; Texas Children's Cancer and Hematology Center, Department of Pediatrics, Center for Cell and Gene Therapy, Texas Children's Hospital and Baylor College of Medicine, Houston, Tex
| | - Harshal A Abhyankar
- Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, Tex; Texas Children's Cancer and Hematology Center, Department of Pediatrics, Center for Cell and Gene Therapy, Texas Children's Hospital and Baylor College of Medicine, Houston, Tex
| | - Philip M Boone
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Christine R Beck
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Wojciech Wiszniewski
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Børre Fevang
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Geir E Tjønnfjord
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Hematology, Oslo University Hospital, Oslo, Norway
| | | | - Henrik Hjorth-Hansen
- Department of Hematology, St Olavs Hospital, Trondheim, Norway; Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ingunn Dybedal
- Department of Hematology, Oslo University Hospital, Oslo, Norway
| | - Ingvild Nordøy
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Silje F Jørgensen
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Tore G Abrahamsen
- Department of Pediatrics, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | | | - Vegard Skogen
- Department of Infectious Diseases, Medical Clinic, University Hospital of North-Norway, Tromsø, Norway
| | - Liv T N Osnes
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
| | - Mari Ann Kulseth
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Trine E Prescott
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Cecilie F Rustad
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Ketil R Heimdal
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - John W Belmont
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Nicholas L Rider
- Center for Human Immunobiology of Texas Children's Hospital/Department of Pediatrics, Baylor College of Medicine, Houston, Tex; Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex
| | - Javier Chinen
- Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex
| | - Tram N Cao
- Center for Human Immunobiology of Texas Children's Hospital/Department of Pediatrics, Baylor College of Medicine, Houston, Tex; Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex
| | - Eric A Smith
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Tex
| | - Maria Soledad Caldirola
- Immunology Service, Ricardo Gutierrez Children's Hospital, Ciudad Autonoma de Buenos Aires, Buenos Aires, Argentina
| | - Liliana Bezrodnik
- Immunology Service, Ricardo Gutierrez Children's Hospital, Ciudad Autonoma de Buenos Aires, Buenos Aires, Argentina
| | - Saul Oswaldo Lugo Reyes
- Immunodeficiencies Research Unit, National Institute of Pediatrics, Coyoacan, Mexico City, Mexico
| | | | | | | | - Cecilia M Poli
- Center for Human Immunobiology of Texas Children's Hospital/Department of Pediatrics, Baylor College of Medicine, Houston, Tex; Hospital Roberto del Rio, Universidad de Chile, Santiago, Chile
| | - Jose L Franco
- Grupo de Inmunodeficiencias Primarias, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellin, Colombia
| | - Claudia M Trujillo Vargas
- Grupo de Inmunodeficiencias Primarias, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellin, Colombia
| | | | - Nicola Wright
- Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Thomas B Issekutz
- Department of Pediatrics, Dalhousie University, Izaak Walton Killam Health Centre, Halifax, Nova Scotia, Canada
| | - Andrew C Issekutz
- Department of Pediatrics, Dalhousie University, Izaak Walton Killam Health Centre, Halifax, Nova Scotia, Canada
| | - Jordan Abbott
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Jason W Caldwell
- Section of Pulmonary, Critical Care, Allergic and Immunological Diseases, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston-Salem, NC
| | - Diana K Bayer
- Department of Pediatrics, Division of Pediatric Allergy/Immunology and Pulmonology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Alice Y Chan
- Department of Pediatrics, University of California, San Francisco, Calif
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), and Vita-Salute San Raffaele University, Milan, Italy
| | - Caterina Cancrini
- University Department of Pediatrics, DPUO, Bambino Gesù Children's Hospital, and Tor Vergata University, Rome, Italy
| | - Eva Holmberg
- Department of Clinical Genetics, University Hospital of Umeå, Umeå, Sweden
| | - Christina West
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Magnus Burstedt
- Department of Clinical Genetics, University Hospital of Umeå, Umeå, Sweden
| | - Ender Karaca
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Gözde Yesil
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Department of Medical Genetics, Bezmi Alem Vakif University Faculty of Medicine, Istanbul, Turkey
| | - Hasibe Artac
- Department of Pediatric Immunology and Allergy, Selcuk University Medical Faculty, Alaeddin Keykubat Kampusu, Konya, Turkey
| | - Yavuz Bayram
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Mehmed Musa Atik
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Mohammad K Eldomery
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Mohammad S Ehlayel
- Department of Pediatrics, Section of Pediatric Allergy and Immunology, Hamad Medical Corporation, Doha, Department of Paediatrics, Weill Cornell Medical College, Ar-Rayyan, Qatar
| | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, Wales
| | - Berit Flatø
- Department of Rheumatology, Oslo University Hospital, Oslo, Norway
| | - Alison A Bertuch
- Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, Tex
| | - I Celine Hanson
- Center for Human Immunobiology of Texas Children's Hospital/Department of Pediatrics, Baylor College of Medicine, Houston, Tex; Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex
| | - Victor W Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Lee-Jun Wong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Jianhong Hu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Human Genome Sequencing Center, Baylor College of Medicine, Houston, Tex
| | - Magdalena Walkiewicz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Yaping Yang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Christine M Eng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex
| | - Eric Boerwinkle
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Human Genome Sequencing Center, Baylor College of Medicine, Houston, Tex; Human Genetics Center, University of Texas School of Public Health, Houston, Tex
| | - Richard A Gibbs
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Human Genome Sequencing Center, Baylor College of Medicine, Houston, Tex
| | - William T Shearer
- Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex
| | - Robert Lyle
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Jordan S Orange
- Center for Human Immunobiology of Texas Children's Hospital/Department of Pediatrics, Baylor College of Medicine, Houston, Tex; Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex.
| | - James R Lupski
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, Tex; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tex; Human Genome Sequencing Center, Baylor College of Medicine, Houston, Tex.
| |
Collapse
|
31
|
Jacinto FA, Fisher GH, Espana EM, Leyngold IM, Margo CE. Separate Primary Melanomas of the Bulbar Conjunctiva and Eyelid Skin: Clinical Implications of Multiple Primary Melanomas. Ocul Oncol Pathol 2016; 2:226-229. [PMID: 27843900 DOI: 10.1159/000445543] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 03/14/2016] [Indexed: 12/21/2022] Open
Abstract
PURPOSE We report a patient with previous in situ melanoma of the forehead skin who was referred for treatment of a bulbar conjunctival melanoma and a separate superficially invasive melanoma of the eyelid skin, and we offer a review of the biological and clinical implications of patients who have multiple primary melanomas. METHODS This article offers a clinicopathological correlation with a review of the relevant literature. RESULTS An 80-year-old white man was referred for evaluation of a suspicious conjunctival tumor and a lower-eyelid lesion. Excisional biopsies revealed that both were primary melanomas arising within in situ disease. Over the span of 25 years, the patient had three separate foci of in situ melanoma, two of which spawned invasive melanoma. CONCLUSION Separate melanomas arising from the bulbar conjunctiva and eyelid skin have rarely been reported. Multiple primary melanomas of the skin, however, are not uncommon. Based on studies of persons with multiple cutaneous melanomas, the prognosis is best predicted by the tumor with the greatest depth of invasion. Patients with multiple melanomas should be examined for dysplastic nevi, additional cutaneous melanomas, and screened periodically for future lesions. Ongoing studies enrolling patients with multiple primary melanomas are attempting to generate insights into low-penetrance susceptibility genes.
Collapse
Affiliation(s)
- Frances A Jacinto
- Department of Ophthalmology, Morsani College of Medicine, University of South Florida, Tampa, Fla., USA
| | - George H Fisher
- Department of Ophthalmology, Morsani College of Medicine, University of South Florida, Tampa, Fla., USA
| | - Edgar M Espana
- Department of Ophthalmology, Morsani College of Medicine, University of South Florida, Tampa, Fla., USA
| | - Ilya M Leyngold
- Department of Ophthalmology, Morsani College of Medicine, University of South Florida, Tampa, Fla., USA
| | - Curtis E Margo
- Department of Ophthalmology, Morsani College of Medicine, University of South Florida, Tampa, Fla., USA; Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Fla., USA
| |
Collapse
|
32
|
Hereditary melanoma: Update on syndromes and management: Emerging melanoma cancer complexes and genetic counseling. J Am Acad Dermatol 2016; 74:411-20; quiz 421-2. [PMID: 26892651 DOI: 10.1016/j.jaad.2015.08.037] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 07/29/2015] [Accepted: 08/03/2015] [Indexed: 12/19/2022]
Abstract
Recent advances in cancer genomics have enabled the discovery of many cancer-predisposing genes that are being used to classify new familial melanoma/cancer syndromes. In addition to CDKN2A and CDK4, germline variants in TERT, MITF, and BAP1 have been added to the list of genes harboring melanoma-predisposing mutations. These newer entities may have escaped earlier description in part because of more advanced technologies now being used and in part because of their mixed cancer phenotype as opposed to a melanoma-focused syndrome. Dermatologists should be aware of (and be able to recognize) the clinical signs in high-risk patients in different contexts. Personal and family histories of cancer should always be sought in patients with multiple nevi or a positive history for melanoma, and should be updated annually. Various features that are unique to specific disorders, such as the appearance of melanocytic BAP1-mutated atypical intradermal tumors in cases of BAP1 melanoma syndrome, should also be recognized early. These patients should be offered regular screenings with the use of dermoscopy and total body photography, as needed. More importantly, referral to other specialists may be needed if a risk for internal malignancy is suspected. It is important to have in mind that these patients tend to develop multiple melanomas, along with various internal organ malignancies, often at younger ages; a multidisciplinary approach to their cancer screening and treatment is ideal.
Collapse
|
33
|
Zhang T, Dutton-Regester K, Brown KM, Hayward NK. The genomic landscape of cutaneous melanoma. Pigment Cell Melanoma Res 2016; 29:266-83. [PMID: 26833684 DOI: 10.1111/pcmr.12459] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 01/25/2016] [Indexed: 12/31/2022]
Abstract
Somatic mutation analysis of melanoma has been performed at the single gene level extensively over the past several decades. This has provided considerable insight into the critical pathways controlling melanoma initiation and progression. During the last 5 yr, next-generation sequencing (NGS) has enabled even more comprehensive mutational screening at the level of multigene panels, exomes and genomes. These studies have uncovered many new and unexpected players in melanoma development. The recent landmark study from The Cancer Genome Atlas (TCGA) consortium describing the genomic architecture of 333 cutaneous melanomas provides the largest and broadest analysis to date on the somatic aberrations underlying melanoma genesis. It thus seems timely to review the mutational landscape of melanoma and highlight the key genes and cellular pathways that appear to drive this cancer.
Collapse
Affiliation(s)
- Tongwu Zhang
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Ken Dutton-Regester
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
- Oncogenomics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | - Kevin M Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Nicholas K Hayward
- Oncogenomics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| |
Collapse
|
34
|
Gibbs DC, Orlow I, Bramson JI, Kanetsky PA, Luo L, Kricker A, Armstrong BK, Anton-Culver H, Gruber SB, Marrett LD, Gallagher RP, Zanetti R, Rosso S, Dwyer T, Sharma A, La Pilla E, From L, Busam KJ, Cust AE, Ollila DW, Begg CB, Berwick M, Thomas NE. Association of Interferon Regulatory Factor-4 Polymorphism rs12203592 With Divergent Melanoma Pathways. J Natl Cancer Inst 2016; 108:djw004. [PMID: 26857527 DOI: 10.1093/jnci/djw004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 01/05/2016] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Solar elastosis and neval remnants are histologic markers characteristic of divergent melanoma pathways linked to differences in age at onset, host phenotype, and sun exposure. However, the association between these pathway markers and newly identified low-penetrance melanoma susceptibility loci remains unknown. METHODS In the Genes, Environment and Melanoma (GEM) Study, 2103 Caucasian participants had first primary melanomas that underwent centralized pathology review. For 47 single-nucleotide polymorphisms (SNPs) previously identified as low-penetrant melanoma risk variants, we used multinomial logistic regression to compare melanoma with solar elastosis and melanoma with neval remnants simultaneously to melanoma with neither of these markers, excluding melanomas with both markers. All statistical tests were two-sided. RESULTS IRF4 rs12203592 was the only SNP to pass the false discovery threshold in baseline models adjusted for age, sex, and study center. rs12203592*T was associated positively with melanoma with solar elastosis (odds ratio [OR] = 1.47, 95% confidence interval [CI] = 1.18 to 1.82) and inversely with melanoma with neval remnants (OR = 0.65, 95% CI = 0.48 to 0.87) compared with melanoma with neither marker (P global = 3.78 x 10(-08)). Adjusting for phenotypic characteristics and total sun exposure hours did not materially affect rs12203592's associations. Distinct early- and late-onset age distributions were observed in patients with IRF4 rs12203592 [CC] and [TT] genotypes, respectively. CONCLUSIONS Our findings suggest a role of IRF4 rs12203592 in pathway-specific risk for melanoma development. We hypothesize that IRF4 rs12203592 could underlie in part the bimodal age distribution reported for melanoma and linked to the divergent pathways.
Collapse
Affiliation(s)
- David C Gibbs
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Irene Orlow
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Jennifer I Bramson
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Peter A Kanetsky
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Li Luo
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Anne Kricker
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Bruce K Armstrong
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Hoda Anton-Culver
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Stephen B Gruber
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Loraine D Marrett
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Richard P Gallagher
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Roberto Zanetti
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Stefano Rosso
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Terence Dwyer
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Ajay Sharma
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Emily La Pilla
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Lynn From
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Klaus J Busam
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Anne E Cust
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - David W Ollila
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Colin B Begg
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Marianne Berwick
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF)
| | - Nancy E Thomas
- Department of Dermatology, University of North Carolina, Chapel Hill, NC (DCG, NET); Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC (NET, DWO); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY (IO, AS, ELP, KJB, CBB); Department of Surgery, University of North Carolina, Chapel Hill, NC (JIB, DWO); Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (PAK); Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM (LL, MB); Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia (AEC, AK, BKA); Department of Epidemiology, University of California, Irvine, CA (HAC); USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA (SBG); Department of Population Studies and Surveillance, Cancer Care Ontario, Toronto, Ontario, Canada (LDM); Cancer Control Research, British Columbia Cancer Agency, Vancouver, British Columbia, Canada (RPG); Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy (RZ, SR); The George Institute for Global Health, Oxford Martin School of Public Health, University of Oxford, Oxford, UK (TD); Department of Pathology, Women's College Hospital, Toronto, Ontario, Canada (LF).
| | | |
Collapse
|