1
|
Gallus R, Rizzo D, Rossi G, Mureddu L, Galli J, Artuso A, Bussu F. p16 Expression in Laryngeal Squamous Cell Carcinoma: A Surrogate or Independent Prognostic Marker? Pathogens 2024; 13:100. [PMID: 38392838 PMCID: PMC10892421 DOI: 10.3390/pathogens13020100] [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: 01/07/2024] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a common malignancy that, despite scientific advancements, has not seen an improvement in its prognosis in the last decades. Few promising predictive markers have been found and none are relevant in clinical practice. p16ink4a, an oncosuppressor protein involved in cell cycle arrest, with a prognostic impact on other cancers, has been widely used in the head and neck region as a surrogate marker of HPV infection. Published papers and recent meta-analyses seem to minimize the biological role of HPV in the context of LSCC's cancerogenesis, and to disprove the reliability of p16ink4a as a surrogate prognostic marker in this context, while still highlighting its potential role as an independent predictor of survival. Unfortunately, the available literature, in particular during the last two decades, is often not focused on its potential role as an independent biomarker and few relevant data are found in papers mainly focused on HPV. The available data suggest that future research should focus specifically on p16ink4a, taking into account both its potential inactivation and overexpression, different patterns of staining, and immunohistochemistry cutoffs, and should focus not on its potential role as a surrogate marker but on its independent role as a predictor of survival.
Collapse
Affiliation(s)
- Roberto Gallus
- Otolaryngology, Mater Olbia Hospital, 07026 Olbia, Italy; (R.G.); (A.A.)
| | - Davide Rizzo
- U.O.C. Otorinolaringoiatria, Azienda Ospedaliero Universitaria di Sassari, Viale San Pietro, 43, 07100 Sassari, Italy; (D.R.); (F.B.)
- Otolaryngology Division, Department of Medicine, Surgery and Pharmacology, University of Sassari, Viale San Pietro, 43, 07100 Sassari, Italy
| | - Giorgia Rossi
- Unit of Otorhinolaryngology and Head-Neck Surgery, “A. Gemelli” Hospital Foundation IRCCS, 00168 Rome, Italy; (G.R.); (J.G.)
| | - Luca Mureddu
- U.O.C. Otorinolaringoiatria, Azienda Ospedaliero Universitaria di Sassari, Viale San Pietro, 43, 07100 Sassari, Italy; (D.R.); (F.B.)
| | - Jacopo Galli
- Unit of Otorhinolaryngology and Head-Neck Surgery, “A. Gemelli” Hospital Foundation IRCCS, 00168 Rome, Italy; (G.R.); (J.G.)
- Department of Head-Neck and Sensory Organs, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Alberto Artuso
- Otolaryngology, Mater Olbia Hospital, 07026 Olbia, Italy; (R.G.); (A.A.)
| | - Francesco Bussu
- U.O.C. Otorinolaringoiatria, Azienda Ospedaliero Universitaria di Sassari, Viale San Pietro, 43, 07100 Sassari, Italy; (D.R.); (F.B.)
- Otolaryngology Division, Department of Medicine, Surgery and Pharmacology, University of Sassari, Viale San Pietro, 43, 07100 Sassari, Italy
| |
Collapse
|
2
|
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
|
3
|
Targeted germline sequencing of patients with three or more primary melanomas reveals high rate of pathogenic variants. Melanoma Res 2021; 30:247-251. [PMID: 31567591 DOI: 10.1097/cmr.0000000000000645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Individuals with multiple primary melanomas have rates of germline CDKN2A pathogenic variants of 3%-18%, and are also frequent carriers of variants in the melanocortin-1 receptor. Few patients with numerous (≥3) primary melanomas have been studied with respect to these or other potential germline pathogenic variants. We investigated 46 patients with ≥3 primary melanomas (3, n = 17; 4, n = 14; 5-14, n = 15) to determine if higher rates of germline pathogenic variants of CDKN2A, MC1R, or other cancer genes could explain their extreme melanoma phenotype. Most (43/46, 93%) patients had variants in MC1R and 11/46 (24%) had CDKN2A pathogenic variants, but only male sex and having two variants in MC1R correlated with increasing number of melanomas. Panel screening of 56 other cancer predisposition genes did not reveal other germline pathogenic variants associated with melanoma (CDK4, BAP1, POT1), although pathogenic variants in TP53, CHEK2, and BRCA2 were present in three separate patients and some patients had variants of uncertain significance. In summary, targeted germline sequencing of patients with ≥3 primary melanomas revealed a high rate of pathogenic variants in CDKN2A and other known cancer genes. Although further investigation of these pathogenic variants and variants of uncertain significance is needed, these results support cancer gene panel testing in individuals diagnosed with ≥3 melanomas.
Collapse
|
4
|
Sawada Y, Nakamura M. Daily Lifestyle and Cutaneous Malignancies. Int J Mol Sci 2021; 22:5227. [PMID: 34069297 PMCID: PMC8156459 DOI: 10.3390/ijms22105227] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/09/2021] [Accepted: 05/13/2021] [Indexed: 12/11/2022] Open
Abstract
Daily lifestyle is a fundamental part of human life and its influence accumulates daily in the human body. We observe that a good daily lifestyle has a beneficial impact on our health; however, the actual effects of individual daily lifestyle factors on human skin diseases, especially skin cancers, have not been summarized. In this review, we focused on the influence of daily lifestyle on the development of skin cancer and described the detailed molecular mechanisms of the development or regulation of cutaneous malignancies. Several daily lifestyle factors, such as circadian rhythm disruption, smoking, alcohol, fatty acids, dietary fiber, obesity, and ultraviolet light, are known to be associated with the risk of cutaneous malignancies, malignant melanoma, squamous cell carcinoma, basal cell carcinoma, and Merkel cell carcinoma. Although the influence of some daily lifestyles on the risk of skin cancers is controversial, this review provides us a better understanding of the relationship between daily lifestyle factors and skin cancers.
Collapse
Affiliation(s)
- Yu Sawada
- Department of Dermatology, University of Occupational and Environmental Health 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka 807-8555, Japan;
| | | |
Collapse
|
5
|
Chan SH, Chiang J, Ngeow J. CDKN2A germline alterations and the relevance of genotype-phenotype associations in cancer predisposition. Hered Cancer Clin Pract 2021; 19:21. [PMID: 33766116 PMCID: PMC7992806 DOI: 10.1186/s13053-021-00178-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/15/2021] [Indexed: 02/08/2023] Open
Abstract
Although CDKN2A is well-known as a susceptibility gene for melanoma and pancreatic cancer, germline variants have also been anecdotally associated with a broader range of neoplasms including neural system tumors, head and neck squamous cell carcinomas, breast carcinomas, as well as sarcomas. The CDKN2A gene encodes for two distinct tumor suppressor proteins, p16INK4A and p14ARF, however, the independent association of germline alterations affecting these two proteins with cancer is under-appreciated. Here, we reviewed CDKN2A germline alterations reported among individuals and families with cancer in the literature, specifically addressing the cancer phenotypes in relation to the molecular consequence on p16INK4A and p14ARF. While melanoma is observed to associate with variants affecting both p16INK4A and p14ARF transcripts, it is noted that variants affecting p14ARF are more frequently observed with a heterogenous range of cancers. Finally, we reflected on the implications of this inferred genotype-phenotype association in clinical practice and proposed that clinical management of CDKN2A germline variant carriers should involve dedicated cancer genetics services, with multidisciplinary input from various healthcare professionals.
Collapse
Affiliation(s)
- Sock Hoai Chan
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Jianbang Chiang
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Joanne Ngeow
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore.
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, 169857, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, 308232, Singapore.
| |
Collapse
|
6
|
Tovar-Parra JD, Gutiérrez-Castañeda LD, Gil-Quiñones SR, Nova JA, Pulido L. CDKN2A Polymorphism in Melanoma Patients in Colombian Population: A Case-Control Study. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7458917. [PMID: 33102592 PMCID: PMC7576359 DOI: 10.1155/2020/7458917] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/06/2020] [Accepted: 10/03/2020] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Melanoma is the most aggressive type of skin cancer, with poor prognosis in advanced stages. The incidence and mortality rates have increased in recent years. Single nucleotide polymorphisms p.R24P, p.M53I, p.G101W, p.V126D, and p.A148T in the CDKN2A (HGNC ID: 1787) gene have been associated with the development of melanoma in different populations; however, this association has not been studied in Colombia. METHODS Cutaneous melanoma patients and healthy controls (85 cases and 166 controls) were included in this study. These subjects were screened through HRM-qPCR assay and detected variants in exon 1 and 2 of CDKN2A gene and confirmed with Sanger sequencing. Chi-square test was used to compare allele and genotype distributions between cases and controls. Odds ratio (OR) with 95% confidence interval (CI) was calculated to determine the association between polymorphisms and haplotypes with melanoma susceptibility. Statistical and haplotype analyses were performed using Stata® and R-Studio®. RESULTS Fifty-four percent of women were identified both in cases and controls. The frequencies of melanoma subtypes were 36,47% lentigo maligna, 24,71% acral lentiginous, 23,53% superficial extension, and 15,29% nodular. Variants in the CDKN2A gene were 11.76% in cases and 8.43% in controls. The most frequent was p.A148T in 5.88% of cases and in 4.82% of controls. GGTTG haplotype showed statistically significant differences between cases and controls (p value = 0.04). CONCLUSION CDKN2A polymorphisms p.G101W, p.R24P, p.M53I, and A148T are not associated with melanoma susceptibility in the Colombian population; further studies regarding genetic interaction and additive effects between more variants are required.
Collapse
Affiliation(s)
- Jose D. Tovar-Parra
- Hospital Universitario-Centro Dermatológico Federico Lleras Acosta, E.S.E., DC, Bogotá, Colombia 111511, Colombia
| | - Luz D. Gutiérrez-Castañeda
- Hospital Universitario-Centro Dermatológico Federico Lleras Acosta, E.S.E., DC, Bogotá, Colombia 111511, Colombia
| | - Sebastián R. Gil-Quiñones
- Hospital Universitario-Centro Dermatológico Federico Lleras Acosta, E.S.E., DC, Bogotá, Colombia 111511, Colombia
| | - Jhon A. Nova
- Hospital Universitario-Centro Dermatológico Federico Lleras Acosta, E.S.E., DC, Bogotá, Colombia 111511, Colombia
| | - Leonardo Pulido
- Hospital Universitario-Centro Dermatológico Federico Lleras Acosta, E.S.E., DC, Bogotá, Colombia 111511, Colombia
| |
Collapse
|
7
|
Gorlov IP, Amos CI, Tsavachidis S, Begg C, Hernando E, Cheng C, Shen R, Orlow I, Luo L, Ernstoff MS, Parker J, Thomas NE, Gorlova OY, Berwick M. Human genes differ by their UV sensitivity estimated through analysis of UV-induced silent mutations in melanoma. Hum Mutat 2020; 41:1751-1760. [PMID: 32643855 PMCID: PMC7794094 DOI: 10.1002/humu.24078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 06/19/2020] [Accepted: 07/02/2020] [Indexed: 11/09/2022]
Abstract
We hypothesized that human genes differ by their sensitivity to ultraviolet (UV) exposure. We used somatic mutations detected by genome-wide screens in melanoma and reported in the Catalog Of Somatic Mutations In Cancer. As a measure of UV sensitivity, we used the number of silent mutations generated by C>T transitions in pyrimidine dimers of a given transcript divided by the number of potential sites for this type of mutations in the transcript. We found that human genes varied by UV sensitivity by two orders of magnitude. We noted that the melanoma-associated tumor suppressor gene CDKN2A was among the top five most UV-sensitive genes in the human genome. Melanoma driver genes have a higher UV-sensitivity compared with other genes in the human genome. The difference was more prominent for tumor suppressors compared with oncogene. The results of this study suggest that differential sensitivity of human transcripts to UV light may explain melanoma specificity of some driver genes. Practical significance of the study relates to the fact that differences in UV sensitivity among human genes need to be taken into consideration whereas predicting melanoma-associated genes by the number of somatic mutations detected in a given gene.
Collapse
Affiliation(s)
- Ivan P Gorlov
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | | | | | - Colin Begg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eva Hernando
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Chao Cheng
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ronglai Shen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Li Luo
- Department of Internal Medicine and Dermatology, University of New Mexico, Albuquerque, New Mexico
| | - Marc S Ernstoff
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Elm, and Carlton, Buffalo, New York
| | - Joel Parker
- Department of Genetics, Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nancy E Thomas
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina
| | - Olga Y Gorlova
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Marianne Berwick
- Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico
| |
Collapse
|
8
|
Li C, Liu T, Liu B, Hernandez R, Facelli JC, Grossman D. A novel CDKN2A variant (p16 L117P ) in a patient with familial and multiple primary melanomas. Pigment Cell Melanoma Res 2019; 32:734-738. [PMID: 31001908 PMCID: PMC6751567 DOI: 10.1111/pcmr.12787] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/18/2019] [Accepted: 04/09/2019] [Indexed: 12/29/2022]
Abstract
Germline mutations in CDKN2A (p16) are commonly found in patients with family history of melanoma or personal history of multiple primary melanomas. The p16 tumor suppressor gene regulates cell cycle progression and senescence through binding of cyclin-dependent kinases (CDK) and also regulates cellular oxidative stress independently of cell cycle control. We identified a germline missense (c.350T>C, p.Leu117Pro) CDKN2A mutation in a patient who had history of four primary melanomas, numerous nevi, and self-reported family history of melanoma. This particular CDKN2A mutation has not been previously reported in prior large studies of melanoma kindreds or patients with multiple primary melanomas. Compared with wild-type p16, the p16L117P mutant largely retained binding capacity for CDK4 and CDK6 but exhibited impaired capacity for repressing cell cycle progression and inducing senescence, while retaining its ability to reduce mitochondrial reactive oxygen species. Structural modeling predicted that the Leu117Pro mutation disrupts a putative adenosine monophosphate (AMP) binding pocket involving residue 117 in the fourth ankyrin domain. Identification of this new likely pathogenic variant extends our understanding of CDKN2A in melanoma susceptibility and implicates AMP as a potential regulator of p16.
Collapse
Affiliation(s)
- Christopher Li
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Tong Liu
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Bin Liu
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Rolando Hernandez
- Department of Biomedical Informatics, University of Utah, Salt Lake City, Utah
| | - Julio C. Facelli
- Department of Biomedical Informatics, University of Utah, Salt Lake City, Utah
| | - Douglas Grossman
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Dermatology, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah
| |
Collapse
|
9
|
Heenatigala Palliyage G, Singh S, Ashby CR, Tiwari AK, Chauhan H. Pharmaceutical Topical Delivery of Poorly Soluble Polyphenols: Potential Role in Prevention and Treatment of Melanoma. AAPS PharmSciTech 2019; 20:250. [PMID: 31297635 DOI: 10.1208/s12249-019-1457-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 06/06/2019] [Indexed: 01/10/2023] Open
Abstract
Melanoma is regarded as the fifth and sixth most common cancer in men and women, respectively, and it is estimated that one person dies from melanoma every hour in the USA. Unfortunately, the treatment of melanoma is difficult because of its aggressive metastasis and resistance to treatment. The treatment of melanoma continues to be a challenging issue due to the limitations of available treatments such as a low response rate, severe adverse reactions, and significant toxicity. Natural polyphenols have attracted considerable attention from the scientific community due to their chemopreventive and chemotherapeutic efficacy. It has been suggested that poorly soluble polyphenols such as curcumin, resveratrol, quercetin, coumarin, and epigallocatechin-3-gallate may have significant benefits in the treatment of melanoma due to their antioxidant, anti-inflammatory, antiproliferative, and chemoprotective efficacies. The major obstacles for the use of polyphenolic compounds are low stability and poor bioavailability. Numerous nanoformulations, including solid lipid nanoparticles, polymeric nanoparticles, micelles, and liposomes, have been formulated to enhance the bioavailability and stability, as well as the therapeutic efficacy of polyphenols. This review will provide an overview of poorly soluble polyphenols that have been reported to have antimetastatic efficacy in melanomas.
Collapse
|
10
|
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
|
11
|
The role of next-generation sequencing in the differential diagnosis of composite neoplasms. Hum Pathol 2018; 81:78-88. [DOI: 10.1016/j.humpath.2018.06.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/02/2018] [Accepted: 06/09/2018] [Indexed: 01/28/2023]
|
12
|
Robert F, Pelletier J. Exploring the Impact of Single-Nucleotide Polymorphisms on Translation. Front Genet 2018; 9:507. [PMID: 30425729 PMCID: PMC6218417 DOI: 10.3389/fgene.2018.00507] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/10/2018] [Indexed: 01/08/2023] Open
Abstract
Over the past 15 years, sequencing of the human genome and The Cancer Genome Atlas (TCGA) project have led to comprehensive lists of single-nucleotide polymorphisms (SNPs) and gene mutations across a large number of human samples. However, our ability to predict the functional impact of SNPs and mutations on gene expression is still in its infancy. Here, we provide key examples to help understand how mutations present in genes can affect translational output.
Collapse
Affiliation(s)
- Francis Robert
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, QC, Canada.,Department of Oncology, McGill University, Montreal, QC, Canada.,Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| |
Collapse
|
13
|
Goldstein AM, Stidd KC, Yang XR, Fraser MC, Tucker MA. Pediatric melanoma in melanoma-prone families. Cancer 2018; 124:3715-3723. [PMID: 30207590 PMCID: PMC6214720 DOI: 10.1002/cncr.31641] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/02/2018] [Accepted: 06/11/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND In the United States, only approximately 0.4% of all melanomas are diagnosed in patients aged <20 years. To the authors' knowledge, melanoma in pediatric members of melanoma-prone families has not been fully investigated to date. The objective of the current study was to evaluate pediatric patients with melanoma with extensive follow-up in melanoma-prone families with and without cyclin-dependent kinase inhibitor 2A (CDKN2A) mutations. METHODS For this non-population-based study, families were followed prospectively for up to 40 years. A total of 60 families with ≥ 3 patients with melanoma were included for analysis: 30 CDKN2A mutation-positive (CDKN2A+) and 30 CDKN2A mutation-negative (CDKN2A-) families. Age at the time of first melanoma and number of melanomas were obtained for each patient and summarized by family or sets (CDKN2A + vs CDKN2A-). For set comparisons and categorical variables (occurrence of melanoma in pediatric patients, number of melanomas, number of patients with single or multiple melanomas), the Pearson chi-square or Fisher exact test was used. RESULTS Regardless of CDKN2A status, melanoma-prone families were found to have 6-fold to 28-fold higher percentages of patients with pediatric melanoma compared with the general population of patients with melanoma in the United States. Within CDKN2A + families, pediatric patients with melanoma were significantly more likely to have multiple melanomas compared with their relatives who were diagnosed at age >20 years (71% vs 38%, respectively; P = .004). CDKN2A + families had significantly higher percentages of pediatric patients with melanoma compared with CDKN2A- families (11.1% vs 2.5%; P = .004). CONCLUSIONS These observations have implications for the prevention of melanoma as well as clinical care for its early detection. Children in melanoma-prone families should have careful sun protection from an early age and skin surveillance to reduce their risk of melanoma.
Collapse
Affiliation(s)
- Alisa M. Goldstein
- Human Genetics Program, Division of Cancer Epidemiology and Genetics,
National Cancer Institute, National Institutes of Health, Department of Health
and Human Services, Bethesda, MD, USA
| | - Kelsey C. Stidd
- Human Genetics Program, Division of Cancer Epidemiology and Genetics,
National Cancer Institute, National Institutes of Health, Department of Health
and Human Services, Bethesda, MD, USA
| | - Xiaohong R. Yang
- Human Genetics Program, Division of Cancer Epidemiology and Genetics,
National Cancer Institute, National Institutes of Health, Department of Health
and Human Services, Bethesda, MD, USA
| | - Mary C. Fraser
- Human Genetics Program, Division of Cancer Epidemiology and Genetics,
National Cancer Institute, National Institutes of Health, Department of Health
and Human Services, Bethesda, MD, USA
| | - Margaret A. Tucker
- Human Genetics Program, Division of Cancer Epidemiology and Genetics,
National Cancer Institute, National Institutes of Health, Department of Health
and Human Services, Bethesda, MD, USA
| |
Collapse
|
14
|
Yuan TA, Yourk V, Farhat A, Ziogas A, Meyskens FL, Anton-Culver H, Liu-Smith F. A Case-Control Study of the Genetic Variability in Reactive Oxygen Species-Metabolizing Enzymes in Melanoma Risk. Int J Mol Sci 2018; 19:ijms19010242. [PMID: 29342889 PMCID: PMC5796190 DOI: 10.3390/ijms19010242] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/09/2018] [Accepted: 01/12/2018] [Indexed: 11/16/2022] Open
Abstract
Recent studies have shown that ultraviolet (UV)-induced chemiexcitation of melanin fragments leads to DNA damage; and chemiexcitation of melanin fragments requires reactive oxygen species (ROS), as ROS excite an electron in the melanin fragments. In addition, ROS also cause DNA damages on their own. We hypothesized that ROS producing and metabolizing enzymes were major contributors in UV-driven melanomas. In this case-control study of 349 participants, we genotyped 23 prioritized single nucleotide polymorphisms (SNPs) in nicotinamide adenine dinucleotide phosphate (NADPH) oxidases 1 and 4 (NOX1 and NOX4, respectively), CYBA, RAC1, superoxide dismutases (SOD1, SOD2, and SOD3) and catalase (CAT), and analyzed their associated melanoma risk. Five SNPs, namely rs1049255 (CYBA), rs4673 (CYBA), rs10951982 (RAC1), rs8031 (SOD2), and rs2536512 (SOD3), exhibited significant genotypic frequency differences between melanoma cases and healthy controls. In simple logistic regression, RAC1 rs10951982 (odds ratio (OR) 8.98, 95% confidence interval (CI): 5.08 to 16.44; p < 0.001) reached universal significance (p = 0.002) and the minor alleles were associated with increased risk of melanoma. In contrast, minor alleles in SOD2 rs8031 (OR 0.16, 95% CI: 0.06 to 0.39; p < 0.001) and SOD3 rs2536512 (OR 0.08, 95% CI: 0.01 to 0.31; p = 0.001) were associated with reduced risk of melanoma. In multivariate logistic regression, RAC1 rs10951982 (OR 6.15, 95% CI: 2.98 to 13.41; p < 0.001) remained significantly associated with increased risk of melanoma. Our results highlighted the importance of RAC1, SOD2, and SOD3 variants in the risk of melanoma.
Collapse
Affiliation(s)
- Tze-An Yuan
- Program in Public Health, University of California Irvine, Irvine, CA 92697, USA.
| | - Vandy Yourk
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA.
| | - Ali Farhat
- Department of Biomedical Engineering, The Henry Samueli School of Engineering, University of California Irvine, Irvine, CA 92697, USA.
| | - Argyrios Ziogas
- Department of Epidemiology, School of Medicine, University of California, Irvine, CA 92697, USA.
| | - Frank L Meyskens
- Program in Public Health, University of California Irvine, Irvine, CA 92697, USA.
- Department of Epidemiology, School of Medicine, University of California, Irvine, CA 92697, USA.
- Chao Family Comprehensive Cancer Center, Irvine, CA 92697, USA.
| | - Hoda Anton-Culver
- Department of Epidemiology, School of Medicine, University of California, Irvine, CA 92697, USA.
| | - Feng Liu-Smith
- Department of Epidemiology, School of Medicine, University of California, Irvine, CA 92697, USA.
- Chao Family Comprehensive Cancer Center, Irvine, CA 92697, USA.
| |
Collapse
|
15
|
Thomas NE, Edmiston SN, Kanetsky PA, Busam KJ, Kricker A, Armstrong BK, Cust AE, Anton-Culver H, Gruber SB, Luo L, Orlow I, Reiner AS, Gallagher RP, Zanetti R, Rosso S, Sacchetto L, Dwyer T, Parrish EA, Hao H, Gibbs DC, Frank JS, Ollila DW, Begg CB, Berwick M, Conway K. Associations of MC1R Genotype and Patient Phenotypes with BRAF and NRAS Mutations in Melanoma. J Invest Dermatol 2017; 137:2588-2598. [PMID: 28842324 PMCID: PMC5701875 DOI: 10.1016/j.jid.2017.07.832] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 07/08/2017] [Accepted: 07/23/2017] [Indexed: 11/21/2022]
Abstract
Associations of MC1R with BRAF mutations in melanoma have been inconsistent between studies. We sought to determine for 1,227 participants in the international population-based Genes, Environment, and Melanoma (GEM) study whether MC1R and phenotypes were associated with melanoma BRAF/NRAS subtypes. We used logistic regression adjusted by age, sex, and study design features and examined effect modifications. BRAF+ were associated with younger age, blond/light brown hair, increased nevi, and less freckling, and NRAS+ with older age relative to the wild type (BRAF-/NRAS-) melanomas (all P < 0.05). Comparing specific BRAF subtypes to the wild type, BRAF V600E was associated with younger age, blond/light brown hair, and increased nevi and V600K with increased nevi and less freckling (all P < 0.05). MC1R was positively associated with BRAF V600E cases but only among individuals with darker phototypes or darker hair (Pinteraction < 0.05) but inversely associated with BRAF V600K (Ptrend = 0.006) with no significant effect modification by phenotypes. These results support distinct etiologies for BRAF V600E, BRAF V600K, NRAS+, and wild-type melanomas. MC1R's associations with BRAF V600E cases limited to individuals with darker phenotypes indicate that MC1R genotypes specifically provide information about BRAF V600E melanoma risk in those not considered high risk based on phenotype. Our results also suggest that melanin pathways deserve further study in BRAF V600E melanomagenesis.
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
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Peter A Kanetsky
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Klaus J Busam
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - 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
| | - Anne E Cust
- Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia; Melanoma Institute Australia, North Sydney, Australia
| | - Hoda Anton-Culver
- Department of Epidemiology, University of California, Irvine, California, USA
| | - Stephen B Gruber
- Univeristy of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, USA
| | - Li Luo
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Anne S Reiner
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, USA
| | | | - 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
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina, USA; Politecnico di Torino, Turin, Italy
| | - Terence Dwyer
- George Institute for Global Health, Nuffield Department of Obstetrics and Gynecology, University of Oxford
| | - Eloise A Parrish
- 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
| | - David C Gibbs
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina, USA; Department of Epidemiology, Emory University, Atlanta, Georgia, USA
| | - Jill S Frank
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - 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
- 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
|
16
|
Burgstaller-Muehlbacher S, Marko M, Müller C, Wendt J, Pehamberger H, Okamoto I. Novel CDKN2A mutations in Austrian melanoma patients. Melanoma Res 2015; 25:412-20. [PMID: 26225579 DOI: 10.1097/cmr.0000000000000179] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
CDKN2A is the most prominent familial melanoma gene, with mutations occurring in up to 40% of the families. Numerous mutations in the gene are known, several of them representing regional founder mutations. We sought to determine, for the first time, germline mutations in CDKN2A in Austria to identify novel mutations. In total, 700 individuals (136 patients with a positive family history and 164 with at least two primary melanomas as the high-risk groups; 200 with single primary melanomas; and 200 healthy individuals as the control groups) were Sanger sequenced for CDKN2A exon 1α, 1β, and 2. The 136 patients with affected relatives were also sequenced for CDK4 exon 2. We found the disease-associated mutations p.R24P (8×), p.N71T (1×), p.G101W (1×), and p.V126D (1×) in the group with affected relatives and p.R24P (2×) in the group with several primary melanomas. Furthermore, we discovered four mutations of unknown significance, two of which were novel: p.A34V and c.151-4 G>C, respectively. Computational effect prediction suggested p.A34V as conferring a high risk for melanoma, whereas c.151-4 G>C, although being predicted as a splice site mutation by MutationTaster, could not functionally be confirmed to alter splicing. Moreover, computational effect prediction confirmed accumulation of high-penetrance mutations in high-risk groups, whereas mutations of unknown significance were distributed across all groups. p.R24P is the most common high-risk mutation in Austria. In addition, we discovered two new mutations in Austrian melanoma patients, p.A34V and c.151-4 G>C, respectively.
Collapse
|
17
|
Thomas NE, Edmiston SN, Alexander A, Groben PA, Parrish E, Kricker A, Armstrong BK, Anton-Culver H, Gruber SB, From L, Busam KJ, Hao H, Orlow I, Kanetsky PA, Luo L, Reiner AS, Paine S, Frank JS, Bramson JI, Marrett LD, Gallagher RP, Zanetti R, Rosso S, Dwyer T, Cust AE, Ollila DW, Begg CB, Berwick M, Conway K. Association Between NRAS and BRAF Mutational Status and Melanoma-Specific Survival Among Patients With Higher-Risk Primary Melanoma. JAMA Oncol 2015; 1:359-68. [PMID: 26146664 PMCID: PMC4486299 DOI: 10.1001/jamaoncol.2015.0493] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
IMPORTANCE NRAS and BRAF mutations in melanoma inform current treatment paradigms, but their role in survival from primary melanoma has not been established. Identification of patients at high risk of melanoma-related death based on their primary melanoma characteristics before evidence of recurrence could inform recommendations for patient follow-up and eligibility for adjuvant trials. OBJECTIVE To determine tumor characteristics and survival from primary melanoma by somatic NRAS and BRAF status. DESIGN, SETTING, AND PARTICIPANTS A population-based study with a median follow-up of 7.6 years (through 2007), including 912 patients from the United States and Australia in the Genes, Environment, and Melanoma (GEM) Study, with first primary cutaneous melanoma diagnosed in the year 2000 and analyzed for NRAS and BRAF mutations. MAIN OUTCOMES AND MEASURES Tumor characteristics and melanoma-specific survival of primary melanoma by NRAS and BRAF mutational status. RESULTS The melanomas were 13% NRAS+, 30% BRAF+, and 57% with neither NRAS nor BRAF mutation (wildtype [WT]). In a multivariable model including clinicopathologic characteristics, relative to WT melanoma (with results reported as odds ratios [95% CIs]), NRAS+ melanoma was associated with presence of mitoses (1.8 [1.0-3.3]), lower tumor-infiltrating lymphocyte (TIL) grade (nonbrisk, 0.5 [0.3-0.8]; and brisk, 0.3 [0.5-0.7] [vs absent TILs]), and anatomic site other than scalp/neck (0.1 [0.01-0.6] for scalp/neck vs trunk/pelvis), and BRAF+ melanoma was associated with younger age (ages 50-69 years, 0.7 [0.5-1.0]; and ages >70 years, 0.5 [0.3-0.8] [vs <50 years]), superficial spreading subtype (nodular, 0.5 [0.2-1.0]; lentigo maligna, 0.4 [0.2-0.7]; and unclassified/other, 0.2 [0.1-0.5] [vs superficial spreading]), and presence of mitoses (1.7 [1.1-2.6]) (P < .05 for all). There was no significant difference in melanoma-specific survival (reported as hazard ratios [95% CIs]) for melanoma harboring mutations in NRAS (1.7 [0.8-3.4]) or BRAF (1.5 [0.8-2.9]) compared with WT melanoma, as adjusted for age, sex, site, American Joint Committee on Cancer (AJCC) tumor stage, TIL grade, and study center. However, melanoma-specific survival was significantly poorer for higher-risk (T2b or higher stage) tumors with NRAS (2.9 [1.1-7.7]) or BRAF (3.1 [1.2-8.5]) mutations (P = .04) but not for lower-risk (T2a or lower) tumors with NRAS (0.9 [0.3-3.0]) or BRAF (0.6 [0.2-1.7]) (P = .65), as adjusted for age, sex, site, AJCC tumor stage, TIL grade, and study center. CONCLUSIONS AND RELEVANCE Lower TIL grade for NRAS+ melanoma suggests it has a more immunosuppressed microenvironment, which may affect its response to immunotherapies. The approximate 3-fold increased risk of death for higher-risk tumors harboring NRAS or BRAF mutations after adjusting for other prognostic factors compared with WT melanomas indicates that the prognostic implication of these mutations deserves further investigation, particularly in higher–AJCC stage primary melanomas.
Collapse
|
18
|
Thomas NE, Kricker A, Waxweiler WT, Dillon PM, Busman KJ, From L, Groben PA, Armstrong BK, Anton-Culver H, Gruber SB, Marrett LD, Gallagher RP, Zanetti R, Rosso S, Dwyer T, Venn A, Kanetsky PA, Orlow I, Paine S, Ollila DW, Reiner AS, Luo L, Hao H, Frank JS, Begg CB, Berwick M. Comparison of clinicopathologic features and survival of histopathologically amelanotic and pigmented melanomas: a population-based study. JAMA Dermatol 2015; 150:1306-314. [PMID: 25162299 DOI: 10.1001/jamadermatol.2014.1348] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
IMPORTANCE Previous studies have reported that histopathologically amelanotic melanoma is associated with poorer survival than pigmented melanoma; however, small numbers of amelanotic melanomas, selected populations, lack of centralized pathologic review, or no adjustment for stage limit the interpretation or generalization of results from prior studies.OBJECTIVE To compare melanoma-specific survival between patients with histopathologically amelanotic and those with pigmented melanoma in a large international population-based study.DESIGN, SETTING, AND PARTICIPANTS Survival analysis with a median follow-up of 7.6 years.The study population comprised 2995 patients with 3486 invasive primary melanomas centrally scored for histologic pigmentation from the Genes, Environment, and Melanoma(GEM) Study, which enrolled incident cases of melanoma diagnosed in 1998 through 2003 from international population-based cancer registries.MAIN OUTCOMES AND MEASURES Clinicopathologic predictors and melanoma-specific survival of histologically amelanotic and pigmented melanoma were compared using generalized estimating equations and Cox regression models, respectively.RESULTS Of 3467 melanomas, 275 (8%) were histopathologically amelanotic. Female sex,nodular and unclassified or other histologic subtypes, increased Breslow thickness, presence of mitoses, severe solar elastosis, and lack of a coexisting nevus were independently associated with amelanotic melanoma (each P < .05). Amelanotic melanoma was generally ofa higher American Joint Committee on Cancer (AJCC) tumor stage at diagnosis (odds ratios[ORs] [95%CIs] between 2.9 [1.8-4.6] and 11.1 [5.8-21.2] for tumor stages between T1b and T3b and ORs [95%CIs] of 24.6 [13.6-44.4] for T4a and 29.1 [15.5-54.9] for T4b relative to T1a;P value for trend, <.001) than pigmented melanoma. Hazard of death from melanoma was higher for amelanotic than for pigmented melanoma (hazard ratio [HR], 2.0; 95%CI, 1.4-3.0)(P < .001), adjusted for age, sex, anatomic site, and study design variables, but survival did not differ once AJCC tumor stage was also taken into account (HR, 0.8; 95%CI, 0.5-1.2)(P = .36).CONCLUSIONS AND RELEVANCE At the population level, survival after diagnosis of amelanotic melanoma is poorer than after pigmented melanoma because of its more advanced stage at diagnosis. It is probable that amelanotic melanomas present at more advanced tumor stages because they are difficult to diagnose. The association of amelanotic melanoma with presence of mitoses independently of Breslow thickness and other clinicopathologic characteristics suggests that amelanotic melanomas might also grow faster than pigmented melanomas. New strategies for early diagnosis and investigation of the biological properties of amelanotic melanoma are warranted.
Collapse
|
19
|
Harland M, Cust AE, Badenas C, Chang YM, Holland EA, Aguilera P, Aitken JF, Armstrong BK, Barrett JH, Carrera C, Chan M, Gascoyne J, Giles GG, Agha-Hamilton C, Hopper JL, Jenkins MA, Kanetsky PA, Kefford RF, Kolm I, Lowery J, Malvehy J, Ogbah Z, Puig-Butille JA, Orihuela-Segalés J, Randerson-Moor JA, Schmid H, Taylor CF, Whitaker L, Bishop DT, Mann GJ, Newton-Bishop JA, Puig S. Prevalence and predictors of germline CDKN2A mutations for melanoma cases from Australia, Spain and the United Kingdom. Hered Cancer Clin Pract 2014; 12:20. [PMID: 25780468 PMCID: PMC4361137 DOI: 10.1186/1897-4287-12-20] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 11/06/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Mutations in the CDKN2A and CDK4 genes predispose to melanoma. From three case-control studies of cutaneous melanoma, we estimated the prevalence and predictors of these mutations for people from regions with widely differing latitudes and melanoma incidence. METHODS Population-based cases and controls from the United Kingdom (1586 cases, 499 controls) and Australia (596 early-onset cases, 476 controls), and a hospital-based series from Spain (747 cases, 109 controls), were screened for variants in all exons of CDKN2A and the p16INK4A binding domain of CDK4. RESULTS The prevalence of mutations for people with melanoma was similar across regions: 2.3%, 2.5% and 2.0% for Australia, Spain and the United Kingdom respectively. The strongest predictors of carrying a mutation were having multiple primaries (odds ratio (OR) = 5.4, 95% confidence interval (CI: 2.5, 11.6) for 2 primaries and OR = 32.4 (95% CI: 14.7, 71.2) for 3 or more compared with 1 primary only); and family history (OR = 3.8; 95% CI:1.89, 7.5) for 1 affected first- or second-degree relative and OR = 23.2 (95% CI: 11.3, 47.6) for 2 or more compared with no affected relatives). Only 1.1% of melanoma cases with neither a family history nor multiple primaries had mutations. CONCLUSIONS There is a low probability (<2%) of detecting a germline CDKN2A mutation in people with melanoma except for those with a strong family history of melanoma (≥2 affected relatives, 25%), three or more primary melanomas (29%), or more than one primary melanoma who also have other affected relatives (27%).
Collapse
Affiliation(s)
- Mark Harland
- />Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology (LICAP), University of Leeds, Leeds, UK
| | - Anne E Cust
- />Cancer Epidemiology and Services Research (CESR), Sydney School of Public Health, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Celia Badenas
- />Dermatology Department and Biochemistry and Molecular Genetics Department, Melanoma Unit, Hospital Clinic, Instituto de Investigaciones Biomédicas August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- />Centro Investigación Biomédica en Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
| | - Yu-Mei Chang
- />Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology (LICAP), University of Leeds, Leeds, UK
| | - Elizabeth A Holland
- />Westmead Institute for Cancer Research and Melanoma Institute, Australia, University of Sydney at Westmead Millennium Institute, Sydney, Australia
| | - Paula Aguilera
- />Dermatology Department and Biochemistry and Molecular Genetics Department, Melanoma Unit, Hospital Clinic, Instituto de Investigaciones Biomédicas August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- />Centro Investigación Biomédica en Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
| | - Joanne F Aitken
- />Viertel Centre for Research in Cancer Control, The Cancer Council Queensland, Spring Hill, Brisbane, Australia
| | - Bruce K Armstrong
- />Cancer Epidemiology and Services Research (CESR), Sydney School of Public Health, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Jennifer H Barrett
- />Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology (LICAP), University of Leeds, Leeds, UK
| | - Cristina Carrera
- />Dermatology Department and Biochemistry and Molecular Genetics Department, Melanoma Unit, Hospital Clinic, Instituto de Investigaciones Biomédicas August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- />Centro Investigación Biomédica en Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
| | - May Chan
- />Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology (LICAP), University of Leeds, Leeds, UK
| | - Joanne Gascoyne
- />Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology (LICAP), University of Leeds, Leeds, UK
| | - Graham G Giles
- />Centre for Epidemiology & Biostatistics, School of Population Health, University of Melbourne, Melbourne, Australia
- />Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
| | - Chantelle Agha-Hamilton
- />Westmead Institute for Cancer Research and Melanoma Institute, Australia, University of Sydney at Westmead Millennium Institute, Sydney, Australia
| | - John L Hopper
- />Centre for Epidemiology & Biostatistics, School of Population Health, University of Melbourne, Melbourne, Australia
| | - Mark A Jenkins
- />Centre for Epidemiology & Biostatistics, School of Population Health, University of Melbourne, Melbourne, Australia
| | - Peter A Kanetsky
- />Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Richard F Kefford
- />Westmead Institute for Cancer Research and Melanoma Institute, Australia, University of Sydney at Westmead Millennium Institute, Sydney, Australia
| | - Isabel Kolm
- />Westmead Institute for Cancer Research and Melanoma Institute, Australia, University of Sydney at Westmead Millennium Institute, Sydney, Australia
| | - Johanna Lowery
- />Genomics Facility, Leeds Cancer Research UK Centre, University of Leeds, Leeds, UK
| | - Josep Malvehy
- />Dermatology Department and Biochemistry and Molecular Genetics Department, Melanoma Unit, Hospital Clinic, Instituto de Investigaciones Biomédicas August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- />Centro Investigación Biomédica en Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
| | - Zighereda Ogbah
- />Dermatology Department and Biochemistry and Molecular Genetics Department, Melanoma Unit, Hospital Clinic, Instituto de Investigaciones Biomédicas August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Joan-Anton Puig-Butille
- />Dermatology Department and Biochemistry and Molecular Genetics Department, Melanoma Unit, Hospital Clinic, Instituto de Investigaciones Biomédicas August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- />Centro Investigación Biomédica en Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
| | | | - Juliette A Randerson-Moor
- />Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology (LICAP), University of Leeds, Leeds, UK
| | - Helen Schmid
- />Westmead Institute for Cancer Research and Melanoma Institute, Australia, University of Sydney at Westmead Millennium Institute, Sydney, Australia
| | - Claire F Taylor
- />Genomics Facility, Leeds Cancer Research UK Centre, University of Leeds, Leeds, UK
| | - Linda Whitaker
- />Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology (LICAP), University of Leeds, Leeds, UK
| | - D Timothy Bishop
- />Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology (LICAP), University of Leeds, Leeds, UK
| | - Graham J Mann
- />Westmead Institute for Cancer Research and Melanoma Institute, Australia, University of Sydney at Westmead Millennium Institute, Sydney, Australia
| | - Julia A Newton-Bishop
- />Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology (LICAP), University of Leeds, Leeds, UK
| | - Susana Puig
- />Dermatology Department and Biochemistry and Molecular Genetics Department, Melanoma Unit, Hospital Clinic, Instituto de Investigaciones Biomédicas August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- />Centro Investigación Biomédica en Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
| |
Collapse
|
20
|
Hemminki K, Fallah M, Hemminki A. Collection and use of family history in oncology clinics. J Clin Oncol 2014; 32:3344-5. [PMID: 25185094 DOI: 10.1200/jco.2014.56.4336] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Kari Hemminki
- German Cancer Research Center, Heidelberg, Germany; Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Mahdi Fallah
- German Cancer Research Center, Heidelberg, Germany
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Haartman Institute, University of Helsinki, Helsinki, Finland
| |
Collapse
|
21
|
Hill VK, Gartner JJ, Samuels Y, Goldstein AM. The genetics of melanoma: recent advances. Annu Rev Genomics Hum Genet 2013; 14:257-79. [PMID: 23875803 DOI: 10.1146/annurev-genom-091212-153429] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cutaneous malignant melanoma results from the interplay of genetic, host, and environmental factors. Genetic factors implicated in melanoma etiology include inherited high-, intermediate-, and low-risk susceptibility genes as well as numerous somatic mutations in melanoma tumors. CDKN2A is the major high-risk melanoma susceptibility gene identified to date. Recent identification of low-risk loci has been accomplished predominantly through genome-wide association studies. Whole-exome and whole-genome studies have identified numerous genes somatically altered in melanoma tumors and highlighted a higher mutation load in melanoma tumors compared with those in other cancers. This higher load is believed to be attributable to the preponderance of cytosine-to-thymine nucleotide substitutions as a result of UV radiation exposure. Technological advances, particularly next-generation sequencing, have increased the opportunities for germline and somatic gene discovery in melanoma and are opening up new avenues for understanding melanoma pathogenesis as well as leading to new opportunities for treatment.
Collapse
Affiliation(s)
- Victoria K Hill
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892
| | | | | | | |
Collapse
|
22
|
Ozola A, Azarjana K, Doniņa S, Proboka G, Mandrika I, Petrovska R, Cēma I, Heisele O, Eņģele L, Streinerte B, Pjanova D. Melanoma risk associated with MC1R gene variants in Latvia and the functional analysis of rare variants. Cancer Genet 2013; 206:81-91. [PMID: 23522749 DOI: 10.1016/j.cancergen.2013.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 01/11/2013] [Accepted: 01/22/2013] [Indexed: 12/30/2022]
Abstract
To evaluate the association of melanocortin 1 receptor gene (MC1R) variants with melanoma risk in a Latvian population, the MC1R gene was sequenced in 200 melanoma patients and 200 control persons. A functional study of previously uncharacterized, rare MC1R variants was also performed. In total, 26 different MC1R variants, including two novel variants Val165Ile and Val188Ile, were detected. The highest risk of melanoma was associated with the Arg151Cys variant (odds ratio (OR) 4.47, 95% confidence interval (CI) 2.19-9.14, P<0.001). A gene dosage effect was observed, with melanoma risk for carriers of two variants being twice (OR 3.98, 95% CI 2.15-7.38, P<0.001) that of carriers of one variant (OR 1.98, 95% CI 1.26-3.11, P=0.003). After stratification according to the pigmentation phenotype, the risk of melanoma remained in groups with otherwise protective phenotypes. Functional analyses of eight previously uncharacterized MC1R variants revealed that a subset of them is functionally relevant. Our results support the contribution of MC1R variants to a genetic predisposition to melanoma in Latvia.
Collapse
Affiliation(s)
- Aija Ozola
- Latvian Biomedical Research and Study Centre, Riga, Latvia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Balogh K, Széll M, Polyánka H, Pagani F, Bussani E, Kemény L, Oláh J. Detection of a rare CDKN2A intronic mutation in a Hungarian melanoma-prone family and its role in splicing regulation. Br J Dermatol 2012; 167:131-3. [PMID: 22292911 DOI: 10.1111/j.1365-2133.2012.10864.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND The major locus for melanoma predisposition is the cell cycle regulatory CDKN2A gene on chromosome 9p21. However, the frequency of germline coding mutations of the CDKN2A gene is lower than expected in melanoma-prone families linked to chromosome 9p21. OBJECTIVES To investigate whether the rare IVS1+37 G/C intronic mutation of the CDKN2A gene, recently identified in a Hungarian melanoma-prone family, influences mRNA splicing regulation. METHODS CDKN2A minigenes containing the wild-type and the mutant intronic sequence were created and transfected into HeLa cells with the aim of studying the mRNA transcripts. RESULTS The results revealed the emergence of a differential splicing pattern from the wild-type and the mutant minigene, suggesting that this mutation may alter the splicing of CDKN2A primary mRNA and therefore might have a pathogenetic role in familial melanoma. CONCLUSIONS We believe that these results confirm the importance of the identification and characterization of CDKN2A intronic mutations with a view to improving our understanding of the pathogenesis, and explain why the frequency of germline coding mutations of the CDKN2A gene is lower than expected in melanoma-prone families linked to chromosome 9p21.
Collapse
Affiliation(s)
- K Balogh
- Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary.
| | | | | | | | | | | | | |
Collapse
|
24
|
Weischer M, Heerfordt IM, Bojesen SE, Eigentler T, Garbe C, Röcken M, Hölmich LR, Schmidt H, Klyver H, Bastholt L, Nordestgaard BG. CHEK2*1100delC and Risk of Malignant Melanoma: Danish and German Studies and Meta-Analysis. J Invest Dermatol 2012; 132:299-303. [DOI: 10.1038/jid.2011.303] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Nikolaou V, Kang X, Stratigos A, Gogas H, Latorre MC, Gabree M, Plaka M, Njauw CN, Kypreou K, Mirmigi I, Stefanaki I, Tsao H. Comprehensive mutational analysis of CDKN2A and CDK4 in Greek patients with cutaneous melanoma. Br J Dermatol 2011; 165:1219-22. [PMID: 21801156 PMCID: PMC3225501 DOI: 10.1111/j.1365-2133.2011.10551.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND The penetrance of CDKN2A mutations is subject to geographical and latitudinal variation and is presumably dictated by ultraviolet radiation exposure and possibly other co-inherited genetic factors. The frequency of mutations increases with the number of family members affected and the number of primary tumours, and also fluctuates with geography. To date, little is known about the prevalence of CDKN2A mutations in patients with melanoma from Greece. OBJECTIVE To characterize the frequency of CDKN2A and CDK4 mutations in a hospital-based population of Greek patients with melanoma. METHODS Three hundred and four consecutive single primary melanoma (SPM), nine familial melanoma (FM) and seven multiple primary melanoma cases (MPM) were assessed for sequence variants in exons 1α, 1β and 2 of CDKN2A and exon 2 of CDK4. RESULTS Germline CDKN2A mutations were detected in 10 of 304 SPM (3·3%), in four of seven MPM (57%) and in two of nine FM (22%) cases. The most common mutation was a Northern European allele (p16 p.R24P) detected in eight individuals. Five previously unreported CDKN2A variants were also identified: -34G>C, c.41_43delins20bp, c.301G>C (p.G101R), c.301G>A (p.G101E) and c.296_297insGACC. We also describe the first report of a CDK4 p.R24H substitution in a Greek family. CONCLUSIONS The Greek population appears to harbour a higher prevalence of the CDKN2A mutation than other reported cohorts. This supports the notion that genetic susceptibility may play a stronger influence in a country with a relatively low incidence of melanoma. Furthermore, the identification of Northern European alleles suggests that gene migration may be responsible, in part, for the observed cases in Greece.
Collapse
Affiliation(s)
- V Nikolaou
- Department of Dermatology, University of Athens Medical School, 'A. Sygros' Hospital, I Dragoumi 5, 16121 Athens, Greece
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Cabanillas R, Astudillo A, Valle M, de la Rosa J, Álvarez R, Durán NS, Cadiñanos J. Novel germline CDKN2A mutation associated with head and neck squamous cell carcinomas and melanomas. Head Neck 2011; 35:E80-4. [PMID: 22083977 DOI: 10.1002/hed.21911] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2011] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The ability to identify individuals at increased risk of cancer is of immediate clinical relevance. Germline mutations in the CDKN2A locus, encoding the key tumor suppressor proteins p16/INK4A and p14/ARF, are frequently present in kindreds with hereditary cutaneous melanoma but have seldom been reported in families with genetic susceptibility to head and neck squamous cell carcinomas (HNSCC). METHODS We report the pedigree of a patient with an unusually high incidence of HNSCC and melanomas. CDKN2A mutation analysis was performed with standard capillary sequencing and multiplex ligation-dependent probe amplification. RESULTS A previously unreported germline CDKN2A mutation affecting only the p16/INK4A open reading frame, c.106delG (p.Ala36ArgfsX17), was detected in the proband. This mutation causes a premature termination codon. CONCLUSIONS Our report emphasizes the need to consider germinal CDKN2A mutations in the differential diagnosis of familial HNSCC and the importance of awareness of these tumors in carriers of CDKN2A mutations.
Collapse
Affiliation(s)
- Rubén Cabanillas
- Instituto de Medicina Oncológica y Molecular de Asturias, Asturias, Spain.
| | | | | | | | | | | | | |
Collapse
|
27
|
Bakos RM, Besch R, Zoratto GG, Godinho JM, Mazzotti NG, Ruzicka T, Bakos L, Santos SE, Ashton-Prolla P, Berking C, Giugliani R. The CDKN2A p.A148T variant is associated with cutaneous melanoma in Southern Brazil. Exp Dermatol 2011; 20:890-3. [PMID: 21895773 DOI: 10.1111/j.1600-0625.2011.01332.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Several germline mutations and sequence variants in cancer predisposition genes have been described. Among these, the CDKN2A p.A148T variant appears to be frequent in patients with melanoma, at least in certain ethnic groups. In this case-control study, we evaluated 127 patients with cutaneous melanoma and 128 controls from Southern Brazil, the region with the highest melanoma incidence rates in the country. Using PCR-RFLP, we demonstrate that CDKN2A p.A148T variant was significantly more frequent in patients with melanoma than in controls (12.6% vs 3.9%; P=0.009). There was no association between presence of the polymorphism and tumor thickness, site of the primary tumor, melanoma subtype, age at diagnosis, quantitative and qualitative number of nevi. Patients with a positive family of history for other cancers were particularly prone to carry the CDKN2A p.A148T allele. All patients with p.A148T-positive melanoma reported European ancestry, especially German, and this was confirmed using a panel of ancestry-informative INDELs. Our data suggest that CDKN2A p.A148T is a melanoma susceptibility allele in Southern Brazil and is particularly common in patients with melanoma of predominantly European ancestry.
Collapse
Affiliation(s)
- Renato M Bakos
- Dermatology Service, Hospital de Clínicas de Porto Alegre (HCPA), Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Interaction of CDKN2A and sun exposure in the etiology of melanoma in the general population. J Invest Dermatol 2011; 131:2500-3. [PMID: 21833009 PMCID: PMC3213249 DOI: 10.1038/jid.2011.235] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
29
|
Carragher LAS, Snell KR, Giblett SM, Aldridge VSS, Patel B, Cook SJ, Winton DJ, Marais R, Pritchard CA. V600EBraf induces gastrointestinal crypt senescence and promotes tumour progression through enhanced CpG methylation of p16INK4a. EMBO Mol Med 2011; 2:458-71. [PMID: 20941790 PMCID: PMC3394506 DOI: 10.1002/emmm.201000099] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The majority of human colorectal cancers (CRCs) are initiated by mutations arising in the adenomatous polyposis coli (APC) tumour suppressor gene. However, a new class of non-APC mutated CRCs has been defined that have a serrated histopathology and carry the V600EBRAF oncogene. Here we have investigated the pathogenesis of serrated CRCs by expressing V600EBraf in the proliferative cells of the mouse gastrointestinal tract. We show that the oncogene drives an initial burst of Mek-dependent proliferation, leading to the formation of hyperplastic crypts. This is associated with β-catenin nuclear localization by a mechanism involving Mapk/Erk kinase (Mek)-dependent, Akt-independent phosphorylation of Gsk3β. However, hyperplastic crypts remain dormant for prolonged periods due to the induction of crypt senescence accompanied by upregulation of senescence-associated β-galactosidase and p16Ink4a. We show that tumour progression is associated with down-regulation of p16Ink4a through enhanced CpG methylation of exon 1 and knockout of Cdkn2a confirms this gene is a barrier to tumour progression. Our studies identify V600EBRAF as an early genetic driver mutation in serrated CRCs and indicate that, unlike APC-mutated cancers, this subtype arises by the bypassing of a V600EBraf driven oncogene-induced senescence programme.
Collapse
|
30
|
Romagosa C, Simonetti S, López-Vicente L, Mazo A, Lleonart ME, Castellvi J, Ramon y Cajal S. p16(Ink4a) overexpression in cancer: a tumor suppressor gene associated with senescence and high-grade tumors. Oncogene 2011; 30:2087-97. [PMID: 21297668 DOI: 10.1038/onc.2010.614] [Citation(s) in RCA: 343] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
p16(Ink4a) is a protein involved in regulation of the cell cycle. Currently, p16(Ink4a) is considered a tumor suppressor protein because of its physiological role and downregulated expression in a large number of tumors. Intriguingly, overexpression of p16(Ink4a) has also been described in several tumors. This review attempts to elucidate when and why p16(Ink4a) overexpression occurs, and to suggest possible implications of p16(Ink4a) in the diagnosis, prognosis and treatment of cancer.
Collapse
Affiliation(s)
- C Romagosa
- Pathology Department, Fundació Institut de Recerca, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
31
|
Lang J, Shennan M, Njauw JCN, Luo S, Bishop JN, Harland M, Hayward NK, Tucker MA, Goldstein AM, Landi MT, Puig S, Gruis NA, Bergman W, Bianchi-Scarra G, Ghiorzo P, Hogg D, Tsao H. A flexible multiplex bead-based assay for detecting germline CDKN2A and CDK4 variants in melanoma-prone kindreds. J Invest Dermatol 2011; 131:480-486. [PMID: 21085193 PMCID: PMC3045700 DOI: 10.1038/jid.2010.331] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The presence of recurrent high-risk mutations in cyclin-dependent kinase inhibitor 2A/cyclin-dependent kinase 4 (CDKN2A/CDK4) among melanoma-prone families suggests that a high-throughput, multiplex assay could serve as an effective initial screening tool. To this end, we have developed a multiplex bead-based assay for high-throughput CDKN2A/CDK4 genotyping in the context of familial melanoma. Genomic DNA from 1,603 subjects (1,005 in training set and 598 in validation set) were amplified by multiplex PCR using five CDKN2A/CDK4 primer sets followed by multiplex allele-specific primer extension for 39 distinct germline variants. The products were then sorted and analyzed using the Luminex xMAP system. Genotypes were compared with previously determined sequence data. In the Toronto training cohort, all 145 samples with known variants were detected by the bead assay (100% concordance). Analysis of the 598 samples from the GenoMEL validation set led to identification of 150/155 expected variants (96.77%). Overall, the bead assay correctly genotyped 1,540/1,603 (96.07%) of all individuals in the study and 1,540/1,545 (99.68%) of individuals whose variants were represented in the probe set. Out of a total of 62,517 allelic calls, 62,512 (99.99%) were correctly assigned. The multiplex bead-based assay is an accurate method for genotyping CDKN2A/CDK4 variants and is potentially useful in genotyping low-to-moderate melanoma risk single-nucleotide polymorphisms.
Collapse
Affiliation(s)
- Julie Lang
- Departments of Medicine and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Michael Shennan
- Departments of Medicine and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jenny Chi-Ni Njauw
- Wellman Center for Photomedicine, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Boston, MA 02114
| | - Su Luo
- Wellman Center for Photomedicine, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Boston, MA 02114
| | - Julia Newton Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Molecular Medicine, University of Leeds, St James’s Hospital, Leeds, UK, LS8 2QG
| | - Mark Harland
- Section of Epidemiology and Biostatistics, Leeds Institute of Molecular Medicine, University of Leeds, St James’s Hospital, Leeds, UK, LS8 2QG
| | - Nicholas K. Hayward
- Oncogenomics Laboratory, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | | | | | | | - Susana Puig
- Melanoma Unit, Dermatology Department, Hospital Clinic, IDIBAPS and CIBER de Enfermedades Raras, Barcelona, Spain
| | - Nelleke A. Gruis
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Wilma Bergman
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Giovanna Bianchi-Scarra
- Department of Oncology, Biology and Genetics, University of Genoa, Italy
- Laboratory of Genetics of Rare HereditaryCancers, San Martino Hospital, Genoa, Italy
| | - Paola Ghiorzo
- Department of Oncology, Biology and Genetics, University of Genoa, Italy
| | - David Hogg
- Departments of Medicine and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Hensin Tsao
- Wellman Center for Photomedicine, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Boston, MA 02114
| |
Collapse
|
32
|
Gerstenblith MR, Goldstein AM, Tucker MA. Hereditary genodermatoses with cancer predisposition. Hematol Oncol Clin North Am 2010; 24:885-906. [PMID: 20816579 PMCID: PMC3276063 DOI: 10.1016/j.hoc.2010.06.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In this article hereditary genodermatoses with cancer predisposition are reviewed, including nevoid basal cell carcinoma syndrome, neurofibromatosis types 1 and 2, tuberous sclerosis complex, xeroderma pigmentosum, and dyskeratosis congenita. Hereditary melanoma is also included, though it differs from the others in several respects. The underlying genetic aberrations causing these syndromes are largely known, allowing novel treatments to be developed for some of these disorders. Early recognition and diagnosis allows for close follow-up and surveillance for associated malignancies.
Collapse
Affiliation(s)
- Meg R. Gerstenblith
- Genetic Epidemiology Branch/Division of Cancer Epidemiology and Genetics, National Cancer Institute/National Institutes of Health, Building EPS/Room 7003, 6120 Executive Boulevard, Rockville, MD 20892-7236, Phone: (301)-435-5164, Fax: (301)-402-4489,
| | - Alisa M. Goldstein
- Genetic Epidemiology Branch/Division of Cancer Epidemiology and Genetics, National Cancer Institute/National Institutes of Health, Building EPS/Room 7004, 6120 Executive Boulevard, Rockville, MD 20892-7236, Phone: (301)-496-4376, Fax: (301)-402-4489,
| | - Margaret A. Tucker
- Genetic Epidemiology Branch/Division of Cancer Epidemiology and Genetics, National Cancer Institute/National Institutes of Health, Building EPS/Room 7003, 6120 Executive Boulevard, Rockville, MD 20892-7236, Phone: (301)-496-4375, Fax: (301)-402-4489,
| |
Collapse
|
33
|
McKenzie HA, Fung C, Becker TM, Irvine M, Mann GJ, Kefford RF, Rizos H. Predicting functional significance of cancer-associated p16(INK4a) mutations in CDKN2A. Hum Mutat 2010; 31:692-701. [PMID: 20340136 DOI: 10.1002/humu.21245] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Inherited mutations affecting the INK4a/ARF locus (CDKN2A) are associated with melanoma susceptibility in 40% of multiple case melanoma families. Over 60 different germline INK4a/ARF mutations have been detected in more than 190 families worldwide. The majority of these alterations are missense mutations affecting p16(INK4a), and only 25% of these have been functionally assessed. There is therefore a need for an accurate and rapid assay to determine the functional significance of p16(INK4a) mutations. We reviewed the performance of several in vivo functional assays that measure critical aspects of p16(INK4a) function, including subcellular location, CDK binding and cell cycle inhibition. In this report the function of 28 p16(INK4a) variants, many associated with melanoma susceptibility were compared. We show that assessment of CDK4 binding and subcellular localization can accurately and rapidly determine the functional significance of melanoma-associated p16(INK4a) mutations. p16(INK4a)-CDK6 binding affinity was unhelpful, as no disease-associated mutation showed reduced CDK6 affinity while maintaining the ability to bind CDK4. Likewise, in silico analyses did not contribute substantially, with only 12 of 25 melanoma-associated missense variants consistently predicted as deleterious. The ability to determine variant functional activity accurately would identify disease-associated mutations and facilitate effective genetic counselling of individuals at high risk of melanoma.
Collapse
Affiliation(s)
- Heather A McKenzie
- Westmead Institute for Cancer Research and Melanoma Institute of Australia, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead NSW 2145, Australia
| | | | | | | | | | | | | |
Collapse
|
34
|
Relationship between germline MC1R variants and BRAF-mutant melanoma in a North Carolina population-based study. J Invest Dermatol 2009; 130:1463-5. [PMID: 20043015 DOI: 10.1038/jid.2009.410] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
35
|
Kannengiesser C, Brookes S, del Arroyo AG, Pham D, Bombled J, Barrois M, Mauffret O, Avril MFM, Chompret A, Lenoir GM, Sarasin A, Peters G, Bressac-de Paillerets B. Functional, structural, and genetic evaluation of 20 CDKN2A germ line mutations identified in melanoma-prone families or patients. Hum Mutat 2009; 30:564-74. [PMID: 19260062 DOI: 10.1002/humu.20845] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Germline mutations of the CDKN2A gene are found in melanoma-prone families and individuals with multiple sporadic melanomas. The encoded protein, p16(INK4A), comprises four ankyrin-type repeats, and the mutations, most of which are missense and occur throughout the entire coding region, can disrupt the conformation of these structural motifs as well as the association of p16(INK4a) with its physiological targets, the cyclin-dependent kinases (CDKs) CDK4 and CDK6. Assessing pathogenicity of nonsynonymous mutations is critical to evaluate melanoma risk in carriers. In the current study, we investigate 20 CDKN2A germline mutations whose effects on p16(INK4A) structure and function have not been previously documented (Thr18_Ala19dup, Gly23Asp, Arg24Gln, Gly35Ala, Gly35Val, Ala57Val, Ala60Val, Ala60Arg, Leu65dup, Gly67Arg, Gly67_Asn71del, Glu69Gly, Asp74Tyr, Thr77Pro, Arg80Pro, Pro81Thr, Arg87Trp, Leu97Arg, Arg99Pro, and [Leu113Leu;Pro114Ser]). By considering genetic information, the predicted impact of each variant on the protein structure, its ability to interact with CDK4 and impede cell proliferation in experimental settings, we conclude that 18 of the 20 CDKN2A variants can be classed as loss of function mutations, whereas the results for two remain ambiguous. Discriminating between mutant and neutral variants of p16(INK4A) not only adds to our understanding of the functionally critical residues in the protein but provides information that can be used for melanoma risk prediction.
Collapse
|
36
|
Cutaneous Melanoma in Childhood and Adolescence Shows Frequent Loss of INK4A and Gain of KIT. J Invest Dermatol 2009; 129:1759-68. [DOI: 10.1038/jid.2008.422] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
37
|
Chaudru V, Lo MT, Lesueur F, Marian C, Mohamdi H, Laud K, Barrois M, Chompret A, Avril MF, Demenais F, Paillerets BBD. Protective effect of copy number polymorphism of glutathione S-transferase T1 gene on melanoma risk in presence of CDKN2A mutations, MC1R variants and host-related phenotypes. Fam Cancer 2009; 8:371-7. [DOI: 10.1007/s10689-009-9249-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Accepted: 05/14/2009] [Indexed: 12/11/2022]
|
38
|
Ibrahim N, Haluska FG. Molecular pathogenesis of cutaneous melanocytic neoplasms. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2009; 4:551-79. [PMID: 19400696 DOI: 10.1146/annurev.pathol.3.121806.151541] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Melanoma is the deadliest form of skin cancer without an effective treatment. An understanding of the genetic basis of melanoma has recently shed light on some of the mechanisms of melanomagenesis. This review explores the major genes involved in familial and sporadic cutaneous melanoma with an emphasis on CDKN2A, CDK4, MC1R, and MAPK pathway targets (e.g., RAS and BRAF), apoptosis regulators (e.g., BCL-2, AKT, and APAF-1), and the tumor-suppressor genes TP53 and PTEN. New directions for therapeutics based on our current knowledge of the genes implicated in melanoma are also discussed.
Collapse
Affiliation(s)
- Nageatte Ibrahim
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA.
| | | |
Collapse
|
39
|
Dhomen N, Reis-Filho JS, da Rocha Dias S, Hayward R, Savage K, Delmas V, Larue L, Pritchard C, Marais R. Oncogenic Braf induces melanocyte senescence and melanoma in mice. Cancer Cell 2009; 15:294-303. [PMID: 19345328 DOI: 10.1016/j.ccr.2009.02.022] [Citation(s) in RCA: 432] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Revised: 01/08/2009] [Accepted: 02/24/2009] [Indexed: 10/20/2022]
Abstract
We show here that inducible expression of Braf(V600E) off the endogenous Braf gene in mouse melanocytes stimulates skin hyperpigmentation and the appearance of nevi harboring senescent melanocytes. Additionally, approximately 70% of Braf(V600E) mice develop melanomas that reproduce many of the cardinal histological and molecular features of human melanoma and whose cells can colonize the lungs of nude mice. We show that the tumor suppressor p16(INK4a) is not required to induce melanocyte senescence and that its loss is not required for tumor progression, although it does regulate tumor penetrance and latency. Thus, we have developed a mouse model of melanoma driven by Braf(V600E) expressed at physiological levels that reflects the genetics and pathology of the human disease.
Collapse
Affiliation(s)
- Nathalie Dhomen
- Signal Transduction Team, Cancer Research UK Centre for Cell and Molecular Biology, The Institute of Cancer Research, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Santillan AA, Cherpelis BS, Glass LF, Sondak VK. Management of familial melanoma and nonmelanoma skin cancer syndromes. Surg Oncol Clin N Am 2009; 18:73-98, viii. [PMID: 19056043 DOI: 10.1016/j.soc.2008.08.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The clinical manifestations of hereditary skin cancer syndromes depend upon the interplay between environmental and genetic factors. Familial melanoma occurs in the setting of hereditary susceptibility, with a complex phenotype of early age of onset, multiple atypical moles, multiple primary melanomas, multiple melanomas in the family, and in some instances pancreatic cancer. Identification of individuals who may have a hereditary susceptibility for the development of melanoma is essential to provide an opportunity for primary prevention, and to target high-risk groups for early diagnosis and treatment. Consequently, the surgeon as one of the primary caregivers should be familiar with hereditary skin cancer syndromes and their pathogenesis, diagnosis, management, and surveillance recommendations. This article discusses a practical approach for some of the issues likely encountered by the surgeon in the management of familial melanoma and non-melanoma skin cancer.
Collapse
Affiliation(s)
- Alfredo A Santillan
- Division of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, 33612, USA
| | | | | | | |
Collapse
|
41
|
Evaluation of the clonal origin of multiple primary melanomas using molecular profiling. J Invest Dermatol 2009; 129:1972-82. [PMID: 19282844 DOI: 10.1038/jid.2009.4] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Numerous investigations have been conducted using molecular profiling to evaluate the possible clonal origin of second malignancies in various cancer types. However, to date no study assessing clonality of multiple primaries has been conducted in melanoma. In this investigation using patients treated at a specialist melanoma treatment center, we compared the somatic mutational profiles of pairs of melanomas designated as independent on the basis of thorough assessment of their clinical and pathologic characteristics. We used a set of highly polymorphic genetic markers selected on the basis of their chromosomal positions and the frequencies of reported allelic losses at these genetic loci. Our statistical testing strategy showed no significant evidence of clonal origin of the two primaries in 17 of the 19 patients examined. The results suggest that most second melanomas designated as independent primary tumors on the basis of their clinicopathologic features are indeed independent occurrences of the disease, supporting the validity of the criteria used by experienced pathologists in distinguishing new primaries from metastases.
Collapse
|
42
|
Capanu M, Orlow I, Berwick M, Hummer AJ, Thomas DC, Begg CB. The use of hierarchical models for estimating relative risks of individual genetic variants: an application to a study of melanoma. Stat Med 2008; 27:1973-92. [PMID: 18335566 DOI: 10.1002/sim.3196] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
For major genes known to influence the risk of cancer, an important task is to determine the risks conferred by individual variants, so that one can appropriately counsel carriers of these mutations. This is a challenging task, since new mutations are continually being identified, and there is typically relatively little empirical evidence available about each individual mutation. Hierarchical modeling offers a natural strategy to leverage the collective evidence from these rare variants with sparse data. This can be accomplished when there are available higher-level covariates that characterize the variants in terms of attributes that could distinguish their association with disease. In this article, we explore the use of hierarchical modeling for this purpose using data from a large population-based study of the risks of melanoma conferred by variants in the CDKN2A gene. We employ both a pseudo-likelihood approach and a Bayesian approach using Gibbs sampling. The results indicate that relative risk estimates tend to be primarily influenced by the individual case-control frequencies when several cases and/or controls are observed with the variant under study, but that relative risk estimates for variants with very sparse data are more influenced by the higher-level covariate values, as one would expect. The analysis offers encouragement that we can draw strength from the aggregating power of hierarchical models to provide guidance to medical geneticists when they offer counseling to patients with rare or even hitherto unobserved variants. However, further research is needed to validate the application of asymptotic methods to such sparse data.
Collapse
Affiliation(s)
- Marinela Capanu
- Memorial Sloan-Kettering Cancer Center, 307 E 63rd Street, New York, NY 10021, U.S.A.
| | | | | | | | | | | |
Collapse
|
43
|
Goldstein AM, Stacey SN, Olafsson JH, Jonsson GF, Helgason A, Sulem P, Sigurgeirsson B, Benediktsdottir KR, Thorisdottir K, Ragnarsson R, Kjartansson J, Kostic J, Masson G, Kristjansson K, Gulcher JR, Kong A, Thorsteinsdottir U, Rafnar T, Tucker MA, Stefansson K. CDKN2A mutations and melanoma risk in the Icelandic population. J Med Genet 2008; 45:284-9. [PMID: 18178632 DOI: 10.1136/jmg.2007.055376] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Germline CDKN2A mutations have been observed in 20-40% of high risk, melanoma prone families; however, little is known about their prevalence in population based series of melanoma cases and controls. METHODS We resequenced the CDKN2A gene, including the p14ARF variant and promoter regions, in approximately 703 registry ascertained melanoma cases and 691 population based controls from Iceland, a country in which the incidence of melanoma has increased rapidly. RESULTS We identified a novel germline variant, G89D, that was strongly associated with increased melanoma risk and appeared to be an Icelandic founder mutation. The G89D variant was present in about 2% of Icelandic invasive cutaneous malignant melanoma cases. Relatives of affected G89D carriers were at significantly increased risk of melanoma, head and neck cancers, and pancreatic carcinoma compared to relatives of other melanoma patients. Nineteen other germline variants were identified, but none conferred an unequivocal risk of melanoma. CONCLUSIONS This population based study of Icelandic melanoma cases and controls showed a frequency of disease related CDKN2A mutant alleles ranging from 0.7% to 1.0%, thus expanding our knowledge about the frequency of CDKN2A mutations in different populations. In contrast to North America and Australia where a broad spectrum of mutations was observed at a similar frequency, in Iceland, functional CDKN2A mutations consist of only one or two different variants. Additional genetic and/or environmental factors are likely critical for explaining the high incidence rates for melanoma in Iceland. This study adds to the geographic regions for which population based estimates of CDKN2A mutation frequencies are available.
Collapse
Affiliation(s)
- A M Goldstein
- Genetic Epidemiology Branch, Division of Cancer Epidemiologyand Genetics/NCI/NIH/DHHS, Executive Plaza South, Room 7004, 6120 Executive Blvd MSC 7236, Bethesda, MD 20892-7236, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
|