1
|
López Riquelme I, Martínez García S, Serrano Ordónez A, Martínez Pilar L. Germline mutations predisposing to melanoma and associated malignancies and syndromes: a narrative review. Int J Dermatol 2024. [PMID: 39651613 DOI: 10.1111/ijd.17602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/17/2024] [Accepted: 11/25/2024] [Indexed: 12/11/2024]
Abstract
The pathogenesis of melanoma is influenced by a complex combination of environmental factors and individual genetic susceptibility. Familial melanoma refers to cases where there are two first-degree relatives with a melanoma diagnosis. Less strict definitions include second-degree relatives or even three or more of any degree from the same family, although this is not clearly defined in the literature. The term hereditary melanoma is reserved for sporadic or familial melanomas linked to high-risk genes with high penetrance. The first genes related to melanoma were CDKN2A and CDK4, but recently, other genes, mostly tumor suppressor genes, have been described. Internal malignancies, particularly pancreatic cancer, have also been associated with melanoma. Recent studies suggest that there could be a link between melanoma and other neoplasms and tumor predisposition syndromes. This review presents an updated overview of familial melanoma criteria and genes involved in melanoma pathogenesis, emphasizing their clinicopathological aspects and other associated malignancies.
Collapse
Affiliation(s)
- Irene López Riquelme
- Dermatology Department, Hospital Regional Universitario de Málaga, Malaga, Spain
| | | | - Ana Serrano Ordónez
- Dermatology Department, Hospital Regional Universitario de Málaga, Malaga, Spain
| | | |
Collapse
|
2
|
Rahimi A, Esmaeili Y, Dana N, Dabiri A, Rahimmanesh I, Jandaghain S, Vaseghi G, Shariati L, Zarrabi A, Javanmard SH, Cordani M. A comprehensive review on novel targeted therapy methods and nanotechnology-based gene delivery systems in melanoma. Eur J Pharm Sci 2023:106476. [PMID: 37236377 DOI: 10.1016/j.ejps.2023.106476] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023]
Abstract
Melanoma, a malignant form of skin cancer, has been swiftly increasing in recent years. Although there have been significant advancements in clinical treatment underlying a well-understanding of melanoma-susceptible genes and the molecular basis of melanoma pathogenesis, the permanency of response to therapy is frequently constrained by the emergence of acquired resistance and systemic toxicity. Conventional therapies, including surgical resection, chemotherapy, radiotherapy, and immunotherapy, have already been used to treat melanoma and are dependent on the cancer stage. Nevertheless, ineffective side effects and the heterogeneity of tumors pose major obstacles to the therapeutic treatment of malignant melanoma through such strategies. In light of this, advanced therapies including nucleic acid therapies (ncRNA, aptamers), suicide gene therapies, and gene therapy using tumor suppressor genes, have lately gained immense attention in the field of cancer treatment. Furthermore, nanomedicine and targeted therapy based on gene editing tools have been applied to the treatment of melanoma as potential cancer treatment approaches nowadays. Indeed, nanovectors enable delivery of the therapeutic agents into the tumor sites by passive or active targeting, improving therapeutic efficiency and minimizing adverse effects. Accordingly, in this review, we summarized the recent findings related to novel targeted therapy methods as well as nanotechnology-based gene systems in melanoma. We also discussed current issues along with potential directions for future research, paving the way for the next-generation of melanoma treatments.
Collapse
Affiliation(s)
- Azadeh Rahimi
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yasaman Esmaeili
- Biosensor Research Center, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Nasim Dana
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arezou Dabiri
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Setareh Jandaghain
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Golnaz Vaseghi
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran; Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8158388994, Iran
| | - Laleh Shariati
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering & Natural Sciences, Istinye University, Istanbul 34396, Turkey
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University, 28040 Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain.
| |
Collapse
|
3
|
Transethosomal Gel for the Topical Delivery of Celecoxib: Formulation and Estimation of Skin Cancer Progression. Pharmaceutics 2022; 15:pharmaceutics15010022. [PMID: 36678651 PMCID: PMC9864437 DOI: 10.3390/pharmaceutics15010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
The topical delivery of therapeutics is a promising strategy for managing skin conditions. Cyclooxygenase-2 (COX-2) inhibitors showed a possible target for chemoprevention and cancer management. Celecoxib (CXB) is a selective COX-2 inhibitor that impedes cell growth and generates apoptosis in different cell tumors. Herein, an investigation proceeded to explore the usefulness of nano lipid vesicles (transethosomes) (TES) of CXB to permit penetration of considerable quantities of the drug for curing skin cancer. The prepared nanovesicles were distinguished for drug encapsulation efficiency, vesicle size, PDI, surface charge, and morphology. In addition, FT-IR and DSC analyses were also conducted to examine the influence of vesicle components. The optimized formulation was dispersed in various hydrogel bases. Furthermore, in vitro CXB release and ex vivo permeability studies were evaluated. A cytotoxicity study proceeded using A431 and BJ1 cell lines. The expression alteration of the cyclin-dependent kinase inhibitor 2A (CDKN2A) gene and DNA damage and fragmentation using qRT-PCR and comet assays were also investigated. Optimized CXB-TES formulation was spherically shaped and displayed a vesicle size of 75.9 ± 11.4 nm, a surface charge of -44.7 ± 1.52 mV, and an entrapment efficiency of 88.8 ± 7.2%. The formulated TES-based hydrogel displayed a sustained in vitro CXB release pattern for 24 h with an enhanced flux and permeation across rat skin compared with the control (free drug-loaded hydrogel). Interestingly, CXB-TES hydrogel has a lower cytotoxic effect on normal skin cells compared with TES suspension and CXB powder. Moreover, the level of expression of the CDKN2A gene was significantly (p ≤ 0.01, ANOVA/Tukey) decreased in skin tumor cell lines compared with normal skin cell lines, indicating that TES are the suitable carrier for topical delivery of CXB to the cancer cells suppressing their progression. In addition, apoptosis demonstrated by comet and DNA fragmentation assays was evident in skin cancer cells exposed to CXB-loaded TES hydrogel formulation. In conclusion, our results illustrate that CXB-TES-loaded hydrogel could be considered a promising carrier and effective chemotherapeutic agent for the management of skin carcinoma.
Collapse
|
4
|
Yeap I, Becker T, Azimi F, Kernohan M. The management of hereditary melanoma, FAMMM syndrome and germline CDKN2A mutations: a narrative review. AUSTRALASIAN JOURNAL OF PLASTIC SURGERY 2022. [DOI: 10.34239/ajops.v5n2.324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Familial atypical multiple mole melanoma (FAMMM) syndrome is a rare autosomal dominant disorder, in which patients present with a large number of melanocytic naevi and a strong history of malignant melanoma, usually at a young age. The most common genetic alteration, implicated in 40 per cent of FAMMM syndrome families, is a mutation of cyclin-dependent kinase inhibitor 2A (CDKN2A).1 CDKN2A encodes the tumour suppressor gene p16INK4a, a critical cell cycle inhibitor.2
The diagnosis and management of patients with FAMMM syndrome is relevant to the plastic surgeon who manages melanoma. However, clear guidelines on its diagnostic criteria and its relationship to associated but distinct syndromes, such as hereditary melanoma and B-K mole syndrome, are lacking in the extant literature.
The aim of this review is to clarify the diagnostic criteria and management principles for FAMMM syndrome. We propose a new system of classifying FAMMM syndrome patients as a subset of all patients with hereditary melanoma. We also present a management algorithm for these distinct patient groups (FAMMM syndrome, hereditary melanoma and germline CDKN2A mutations).
Collapse
|
5
|
Familial Melanoma and Susceptibility Genes: A Review of the Most Common Clinical and Dermoscopic Phenotypic Aspect, Associated Malignancies and Practical Tips for Management. J Clin Med 2021; 10:jcm10163760. [PMID: 34442055 PMCID: PMC8397216 DOI: 10.3390/jcm10163760] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022] Open
Abstract
A family history of melanoma greatly increases the risk of developing cutaneous melanoma, a highly aggressive skin cancer whose incidence has been steadily increasing worldwide. Familial melanomas account for about 10% of all malignant melanomas and display an inheritance pattern consistent with the presence of pathogenic germline mutations, among which those involving CDKN2A are the best characterized. In recent years, a growing number of genes, such as MC1R, MITF, CDK4, POT1, TERT, ACD, TERF2IP, and BAP1, have been implicated in familial melanoma. The fact that individuals harboring these germline mutations along with their close blood relatives have a higher risk of developing multiple primary melanomas as well as other internal organ malignancies, especially pancreatic cancer, makes cascade genetic testing and surveillance of these families of the utmost importance. Unfortunately, due to a polygenic inheritance mechanism involving multiple low-risk alleles, genetic modifiers, and environmental factors, it is still very difficult to predict the presence of these mutations. It is, however, known that germline mutation carriers can sometimes develop specific clinical traits, such as high atypical nevus counts and specific dermoscopic features, which could theoretically help clinicians predict the presence of these mutations in prone families. In this review, we provide a comprehensive overview of the high- and intermediate-penetrance genes primarily linked to familial melanoma, highlighting their most frequently associated non-cutaneous malignancies and clinical/dermoscopic phenotypes.
Collapse
|
6
|
Molecular landscape of Hereditary Melanoma. Crit Rev Oncol Hematol 2021; 164:103425. [PMID: 34245855 DOI: 10.1016/j.critrevonc.2021.103425] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 05/20/2021] [Accepted: 07/04/2021] [Indexed: 12/27/2022] Open
Abstract
Melanoma is considered the most lethal skin cancer and its incidence has increased during the past decades. About 10 % of cases are classified as hereditary melanoma. Genetic predisposition usually manifests itself clinically as early onset and multiple cutaneous melanomas. Several genes have been identified as involved to melanoma susceptibility, some of them still with unknown clinical relevance. Beyond melanoma, the affected families are also more prone to develop other malignancies, such as pancreatic cancer. The identification of risk families and involved genes is of great importance, since different forms of oncological surveillance are recommended. However, well established guidelines to standardize both the selection of individuals and the genetic panel to be requested are still lacking. Given the importance of the genetic counseling and testing in the context of clinical suspicion of hereditary melanoma, this paper aims to review the literature regarding genetic panel indications worldwide.
Collapse
|
7
|
Novel MAPK/AKT-impairing germline NRAS variant identified in a melanoma-prone family. Fam Cancer 2021; 21:347-355. [PMID: 34215961 DOI: 10.1007/s10689-021-00267-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/12/2021] [Indexed: 12/24/2022]
Abstract
While several high-penetrance melanoma risk genes are known, variation in these genes fail to explain melanoma susceptibility in a large proportion of high-risk families. As part of a melanoma family sequencing study, including 435 families from Mediterranean populations we identified a novel NRAS variant (c.170A > C, p.D57A) in an Italian melanoma-prone family. This variant is absent in exomes in gnomAD, ESP, UKBiobank, and the 1000 Genomes Project, as well as in 11,273 Mediterranean individuals and 109 melanoma-prone families from the US and Australia. This variant occurs in the GTP-binding pocket of NRAS. Differently from other RAS activating alterations, NRAS D57A expression is unable to activate MAPK-pathway both constitutively and after stimulation but enhances EGF-induced PI3K-pathway signaling in serum starved conditions in vitro. Consistent with in vitro data demonstrating that NRAS D57A does not enrich GTP binding, molecular modeling suggests that the D57A substitution would be expected to impair Mg2 + binding and decrease nucleotide-binding and GTPase activity of NRAS. While we cannot firmly establish NRAS c.170A > C (p.D57A) as a melanoma susceptibility variant, further investigation of NRAS as a familial melanoma gene is warranted.
Collapse
|
8
|
Scatena C, Murtas D, Tomei S. Cutaneous Melanoma Classification: The Importance of High-Throughput Genomic Technologies. Front Oncol 2021; 11:635488. [PMID: 34123788 PMCID: PMC8193952 DOI: 10.3389/fonc.2021.635488] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Cutaneous melanoma is an aggressive tumor responsible for 90% of mortality related to skin cancer. In the recent years, the discovery of driving mutations in melanoma has led to better treatment approaches. The last decade has seen a genomic revolution in the field of cancer. Such genomic revolution has led to the production of an unprecedented mole of data. High-throughput genomic technologies have facilitated the genomic, transcriptomic and epigenomic profiling of several cancers, including melanoma. Nevertheless, there are a number of newer genomic technologies that have not yet been employed in large studies. In this article we describe the current classification of cutaneous melanoma, we review the current knowledge of the main genetic alterations of cutaneous melanoma and their related impact on targeted therapies, and we describe the most recent high-throughput genomic technologies, highlighting their advantages and disadvantages. We hope that the current review will also help scientists to identify the most suitable technology to address melanoma-related relevant questions. The translation of this knowledge and all actual advancements into the clinical practice will be helpful in better defining the different molecular subsets of melanoma patients and provide new tools to address relevant questions on disease management. Genomic technologies might indeed allow to better predict the biological - and, subsequently, clinical - behavior for each subset of melanoma patients as well as to even identify all molecular changes in tumor cell populations during disease evolution toward a real achievement of a personalized medicine.
Collapse
Affiliation(s)
- Cristian Scatena
- Division of Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Daniela Murtas
- Department of Biomedical Sciences, Section of Cytomorphology, University of Cagliari, Cagliari, Italy
| | - Sara Tomei
- Omics Core, Integrated Genomics Services, Research Department, Sidra Medicine, Doha, Qatar
| |
Collapse
|
9
|
De Summa S, Lasorella A, Strippoli S, Giudice G, Guida G, Elia R, Nacchiero E, Azzariti A, Silvestris N, Guida M, Guida S, Tommasi S, Pinto R. The Genetic Germline Background of Single and Multiple Primary Melanomas. Front Mol Biosci 2021; 7:555630. [PMID: 33748184 PMCID: PMC7973206 DOI: 10.3389/fmolb.2020.555630] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 11/25/2020] [Indexed: 01/01/2023] Open
Abstract
Background: Melanoma has a complex molecular background and multiple genes are involved in its development and progression. The advent of next generation sequencing platforms has enabled the evaluation of multiple genes at a time, thus unraveling new insights into the genetics of melanoma. We investigated a set of germline mutations able to discriminate the development of multiple primary melanomas (MPM) vs. single site primary melanomas (SPM) using a targeted next generation sequencing panel. Materials and Methods: A total of 39 patients, 20 with SPM and 19 with MPM, were enrolled in our study. Next generation analysis was carried out using a custom targeted sequencing panel that included 32 genes known to have a role in several carcinogenic pathways, such as those involved in DNA repair, pigmentation, regulation of kinases, cell cycle control and senescence. Results: We found a significant correlation between PIK3CA:p.I391M and MPMs, compared to SPMs, p = 0.031 and a trend for the association between CYP1B1: p.N453S and SPMs, compared to MPMs (p = 0.096). We also found that both subgroups shared a spectrum of 9 alterations in 8 genes (CYP1B1: p.N453S, BAP1: p.C39fs, PIK3CA: p.I391M, CDKAL1: c.1226_1227TG, POLE: p.V1161fs, OCA2: p.R419Q, OCA2: p.R305W, MC1R: p.V60L, MGMT: p.L115F), which suggested that these genes may play a role in melanoma development. Conclusions: In conclusion, despite the small cohort of patients, we found that germline mutations, such as those of PIK3CAand CYP1B1, might contribute to the differential development of SPM and MPM.
Collapse
Affiliation(s)
- Simona De Summa
- Pharmacogenetics and Molecular Diagnostic Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Antonia Lasorella
- Pharmacogenetics and Molecular Diagnostic Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Sabino Strippoli
- Medical Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Giuseppe Giudice
- Department of Plastic and Reconstructive Surgery, University of Bari, Bari, Italy
| | - Gabriella Guida
- Department of Basic Medical Science and Sense Organs, University of Bari, Bari, Italy
| | - Rossella Elia
- Department of Plastic and Reconstructive Surgery, University of Bari, Bari, Italy
| | - Eleonora Nacchiero
- Department of Plastic and Reconstructive Surgery, University of Bari, Bari, Italy
| | - Amalia Azzariti
- Pharmacology Laboratory IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Nicola Silvestris
- Medical Oncology Unit IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy.,Biomedical Sciences and Human Oncology (DIMO), University of Bari "Aldo Moro", Bari, Italy
| | - Michele Guida
- Medical Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Stefania Guida
- Dermatology Unit, Department of Surgical, Medical, Dental and Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Stefania Tommasi
- Pharmacogenetics and Molecular Diagnostic Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Rosamaria Pinto
- Pharmacogenetics and Molecular Diagnostic Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| |
Collapse
|
10
|
The Melanoma and Breast Cancer Association: An Overview of their 'Second Primary Cancers' and the Epidemiological, Genetic and Biological correlations. Crit Rev Oncol Hematol 2020; 152:102989. [PMID: 32485529 DOI: 10.1016/j.critrevonc.2020.102989] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/22/2020] [Accepted: 05/14/2020] [Indexed: 12/18/2022] Open
Abstract
This study reviews the relevant epidemiological studies associating cutaneous melanoma and breast carcinomas and provides an overview of the possible genetic, biological and bias factors that underpin this relationship. Standardised incidence ratio (SIR) for primary cutaneous melanoma after breast carcinoma ranged from 1.16 to 5.13 and ranged from 1.03 to 4.10 for primary breast carcinoma after cutaneous melanoma. Epidemiological studies highlight age, gender and use of radiotherapy and chemotherapy as potential risk factors for second primary cancers (SPCs). Mutations in BRCA2, CDKN2A, CDK4 and BAP1 may partly underlie any SPC association. The impact of socio-cultural factors and surveillance bias may be attributed to the findings of SPC partially or entirely. In conclusion, this study has highlighted the association between breast carcinoma and melanoma and identified various factors for further research and the optimised management of patients with both cancers.
Collapse
|
11
|
Flint ND, Bishop MD, Smart TC, Strunck JL, Boucher KM, Grossman D, Secrest AM. Low accuracy of self-reported family history of melanoma in high-risk patients. Fam Cancer 2020; 20:41-48. [PMID: 32436000 DOI: 10.1007/s10689-020-00187-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Family history of melanoma is a major melanoma risk factor. However, self-reported family histories for some cancers, including melanoma, are commonly inaccurate. We used a unique database, the Utah Population Database (UPDB), as well as the Utah Cancer Registry to determine the accuracy of self-reported family history of melanoma in a large cohort of high-risk patients. Patient charts were reviewed and compared to records in the UPDB and the UCR to confirm personal and family history of melanoma in 1780 patients enrolled in a total body photography monitoring program. Self-reported family history of melanoma in first-degree relatives had an overall sensitivity of 71%, specificity of 79%, PPV of 31%, and NPV of 95%, with decreased accuracy (PPV) for second-degree relatives. A personal history of melanoma was the only factor significantly associated with accuracy in self-reported family history of melanoma. Patient age, sex, estimated nevus count, and number of prior personal melanomas were not significant predictors. Dermatologists should educate patients on the differences between melanomas, keratinocyte carcinomas, and pre-cancers. Confirming self-reported family history of melanoma may improve risk assessment for patients undergoing screening.
Collapse
Affiliation(s)
| | | | - Tristan C Smart
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | - Kenneth M Boucher
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Douglas Grossman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Dermatology, University of Utah, Salt Lake City, UT, USA
| | - Aaron M Secrest
- Department of Dermatology, University of Utah, Salt Lake City, UT, USA.
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA.
- Department of Dermatology, University of Utah, 30 N 1900 East, 4A330, Salt Lake City, UT, 84132, USA.
| |
Collapse
|
12
|
Pastor-Tomás N, Martínez-Franco A, Bañuls J, Peñalver JC, Traves V, García-Casado Z, Requena C, Kumar R, Nagore E. Risk factors for the development of a second melanoma in patients with cutaneous melanoma. J Eur Acad Dermatol Venereol 2020; 34:2295-2302. [PMID: 32163215 DOI: 10.1111/jdv.16341] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 02/25/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cutaneous melanoma patients have an increased risk of developing other neoplasms, especially cutaneous neoplasms and other melanomas. Identifying factors associated with an increased risk might be useful in the development of melanoma guidelines. OBJECTIVES To identify risk factors related to the development of a second primary melanoma in a series of patients diagnosed with sporadic melanoma and to establish the estimated incidence rate. METHODS A longitudinal study based on prospective follow-up information of patients diagnosed with sporadic cutaneous melanoma at our centre from 2000 to 2015 was performed. Cumulative incidence was estimated based on competing risk models, and the association of characteristics with the risk of a second melanoma was performed by Cox proportional hazard models. RESULTS Out of 1447 patients included in the study, after a median follow-up of 61 months, 55 patients (3.8%) developed a second melanoma. Fair hair colour, more than 100 common melanocytic nevi and the presence of more than 50 cherry angiomas were independently associated with the development of a second melanoma. The site and the histological subtype of the first and second melanomas were not consistent. The second melanomas were thinner than the first ones. CONCLUSIONS Fair-haired and multiple-nevi patients might benefit from more intensive prevention measures. The finding of cherry angiomas as a risk factor suggests that these lesions could be markers of skin sun damage in the setting of certain degree of genetic susceptibility.
Collapse
Affiliation(s)
- N Pastor-Tomás
- Department of Dermatology, Hospital General Universitario de Elda, Alicante, Spain.,Escuela de Doctorado, Universidad Católica de Valencia San Vicente Mártir, València, Spain
| | - A Martínez-Franco
- School of Medicine, Universidad Católica de Valencia San Vicente Mártir, València, Spain
| | - J Bañuls
- Department of Dermatology, Hospital General Universitario de Alicante-ISABIAL, Alicante, Spain.,Departament of Medicina Clínica, Universidad Miguel Hernández. Sant Joan D'Alacant, Alicante, Spain
| | - J C Peñalver
- Department of Thoracic Surgery, Instituto Valenciano de Oncología, València, Spain
| | - V Traves
- Department of Pathology, Instituto Valenciano de Oncología, València, Spain
| | - Z García-Casado
- Laboratory of Molecular Biology, Instituto Valenciano de Oncología, València, Spain
| | - C Requena
- Department of Dermatology, Instituto Valenciano de Oncología, València, Spain
| | - R Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - E Nagore
- School of Medicine, Universidad Católica de Valencia San Vicente Mártir, València, Spain.,Department of Dermatology, Instituto Valenciano de Oncología, València, Spain
| |
Collapse
|
13
|
Sargen MR, Calista D, Elder DE, Massi D, Chu EY, Potrony M, Pfeiffer RM, Carrera C, Aguilera P, Alos L, Puig S, Elenitsas R, Yang XR, Tucker MA, Landi MT, Goldstein AM. Histologic features of melanoma associated with germline mutations of CDKN2A, CDK4, and POT1 in melanoma-prone families from the United States, Italy, and Spain. J Am Acad Dermatol 2020; 83:860-869. [PMID: 32283231 DOI: 10.1016/j.jaad.2020.03.100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND CDKN2A, CDK4, and POT1 are well-established melanoma-susceptibility genes. OBJECTIVE We evaluated melanoma histopathology for individuals with germline mutations of CDKN2A, CDK4, and POT1. METHODS We assessed histopathology for melanomas diagnosed in melanoma-prone families (≥2 individuals with melanoma) from the United States, Italy, and Spain. Comparisons between mutation carriers and noncarriers (no mutation) were adjusted for age, sex, Breslow depth, and correlations among individuals within the same family. RESULTS Histologic slides were evaluated for 290 melanomas (139 from 132 noncarriers, 122 from 68 CDKN2A carriers, 10 from 6 CDK4 carriers, and 19 from 16 POT1 carriers). Superficial spreading was the predominant subtype for all groups. Spitzoid morphology (>25% of tumor) was observed in 10 of 15 invasive melanomas (67%) from POT1 carriers (P < .0001 vs noncarriers). This finding was independently confirmed by 3 expert melanoma dermatopathologists in 9 of 15 invasive melanomas (60%). In situ and invasive melanomas from CDKN2A and CDK4 carriers were histologically similar to melanomas from noncarriers. LIMITATIONS Limited sample sizes for rare melanoma-susceptibility syndromes (CDK4, POT1). CONCLUSION Spitzoid morphology was associated with POT1 mutations suggesting that telomere dysfunction (POT1 mutations) may contribute to spitzoid differentiation in melanocytic tumors.
Collapse
Affiliation(s)
- Michael R Sargen
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland.
| | - Donato Calista
- Department of Dermatology, Maurizio Bufalini Hospital, Cesena, Italy
| | - David E Elder
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniela Massi
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, Florence, Italy
| | - Emily Y Chu
- Department of Dermatology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Míriam Potrony
- Melanoma Unit, Dermatology Department, Hospital Clínic de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Ruth M Pfeiffer
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Cristina Carrera
- Melanoma Unit, Dermatology Department, Hospital Clínic de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Paula Aguilera
- Melanoma Unit, Dermatology Department, Hospital Clínic de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Llucia Alos
- Pathology Department, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Susana Puig
- Melanoma Unit, Dermatology Department, Hospital Clínic de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Rosalie Elenitsas
- Department of Dermatology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xiaohong R Yang
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Margaret A Tucker
- Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Maria Teresa Landi
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Alisa M Goldstein
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| |
Collapse
|
14
|
Nathan V, Johansson PA, Palmer JM, Howlie M, Hamilton HR, Wadt K, Jönsson G, Brooks KM, Pritchard AL, Hayward NK. Germline variants in oculocutaneous albinism genes and predisposition to familial cutaneous melanoma. Pigment Cell Melanoma Res 2019; 32:854-863. [PMID: 31233279 DOI: 10.1111/pcmr.12804] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/28/2019] [Accepted: 06/15/2019] [Indexed: 11/30/2022]
Abstract
Approximately 1%-2% of cutaneous melanoma (CM) is classified as strongly familial. We sought to investigate unexplained CM predisposition in families negative for the known susceptibility genes using next-generation sequencing of affected individuals. Segregation of germline variants of interest within families was assessed by Sanger sequencing. Several heterozygous variants in oculocutaneous albinism (OCA) genes: TYR, OCA2, TYRP1 and SLC45A2, were present in our CM cohort. OCA is a group of autosomal recessive genetic disorders, resulting in pigmentation defects of the eyes, hair and skin. Missense variants classified as pathogenic for OCA were present in multiple families and some fully segregated with CM. The functionally compromised TYR p.T373K variant was present in three unrelated families. In OCA2, known pathogenic variants: p.V443I and p.N489D, were present in three families and one family, respectively. We identified a likely pathogenic SLC45A2 frameshift variant that fully segregated with CM in a family of four cases. Another four-case family harboured cosegregating variants (p.A24T and p.R153C) of uncertain functional significance in TYRP1. We conclude that rare, heterozygous variants in OCA genes confer moderate risk for CM.
Collapse
Affiliation(s)
- Vaishnavi Nathan
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- University of Queensland, Brisbane, Queensland, Australia
| | - Peter A Johansson
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jane M Palmer
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Madeleine Howlie
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Hayley R Hamilton
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Karin Wadt
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
| | - Göran Jönsson
- Department of Oncology, Clinical Sciences, Lund University, Lund, Sweden
| | - Kelly M Brooks
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Antonia L Pritchard
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- University of Highlands and Islands, Inverness, Scotland
| | - Nicholas K Hayward
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
15
|
Wei EX, Li X, Nan H. Having a first-degree relative with melanoma increases lifetime risk of melanoma, squamous cell carcinoma, and basal cell carcinoma. J Am Acad Dermatol 2019; 81:489-499. [DOI: 10.1016/j.jaad.2019.04.044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/01/2019] [Accepted: 04/14/2019] [Indexed: 01/29/2023]
|
16
|
Taylor NJ, Mitra N, Qian L, Avril MF, Bishop DT, Bressac-de Paillerets B, Bruno W, Calista D, Cuellar F, Cust AE, Demenais F, Elder DE, Gerdes AM, Ghiorzo P, Goldstein AM, Grazziotin TC, Gruis NA, Hansson J, Harland M, Hayward NK, Hocevar M, Höiom V, Holland EA, Ingvar C, Landi MT, Landman G, Larre-Borges A, Mann GJ, Nagore E, Olsson H, Palmer JM, Perić B, Pjanova D, Pritchard AL, Puig S, Schmid H, van der Stoep N, Tucker MA, Wadt KAW, Yang XR, Newton-Bishop JA, Kanetsky PA. Estimating CDKN2A mutation carrier probability among global familial melanoma cases using GenoMELPREDICT. J Am Acad Dermatol 2019; 81:386-394. [PMID: 30731170 PMCID: PMC6634996 DOI: 10.1016/j.jaad.2019.01.079] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 01/02/2019] [Accepted: 01/30/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Although rare in the general population, highly penetrant germline mutations in CDKN2A are responsible for 5%-40% of melanoma cases reported in melanoma-prone families. We sought to determine whether MELPREDICT was generalizable to a global series of families with melanoma and whether performance improvements can be achieved. METHODS In total, 2116 familial melanoma cases were ascertained by the international GenoMEL Consortium. We recapitulated the MELPREDICT model within our data (GenoMELPREDICT) to assess performance improvements by adding phenotypic risk factors and history of pancreatic cancer. We report areas under the curve (AUC) with 95% confidence intervals (CIs) along with net reclassification indices (NRIs) as performance metrics. RESULTS MELPREDICT performed well (AUC 0.752, 95% CI 0.730-0.775), and GenoMELPREDICT performance was similar (AUC 0.748, 95% CI 0.726-0.771). Adding a reported history of pancreatic cancer yielded discriminatory improvement (P < .0001) in GenoMELPREDICT (AUC 0.772, 95% CI 0.750-0.793, NRI 0.40). Including phenotypic risk factors did not improve performance. CONCLUSION The MELPREDICT model functioned well in a global data set of familial melanoma cases. Adding pancreatic cancer history improved model prediction. GenoMELPREDICT is a simple tool for predicting CDKN2A mutational status among melanoma patients from melanoma-prone families and can aid in directing these patients to receive genetic testing or cancer risk counseling.
Collapse
Affiliation(s)
- Nicholas J Taylor
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, Texas
| | - Nandita Mitra
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lu Qian
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Marie-Françoise Avril
- Assistance Publique-Hôpitaux de Paris, Hôpital Cochin et Université Paris Descartes, Paris, France
| | - D Timothy Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Medical Research at St. James's, University of Leeds, Leeds, United Kingdom
| | - Brigitte Bressac-de Paillerets
- Gustave Roussy, Université Paris-Saclay, Département de Biopathologie and Institut National de la Santé et de la Recherche Médicale U1186, Villejuif, France
| | - William Bruno
- Department of Internal Medicine and Medical Specialties, University of Genoa and Istituto de Ricovero e Cura a Carattere Scientifico AOU San Martino-IST, Genoa, Italy
| | - Donato Calista
- Dermatology Unit, Maurizio Bufalini Hospital, Cesena, Italy
| | - Francisco Cuellar
- Melanoma Unit, Dermatology Department, Hospital Clinic Barcelona, Institut de Investigacions Biomediques August Pi Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Anne E Cust
- Sydney School of Public Health, The University of Sydney, Sydney, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Florence Demenais
- Institut National de la Santé et de la Recherche Médicale UMR-946, Genetic Variation and Human Disease Unit, Université Paris Diderot, Paris, France
| | - David E Elder
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anne-Marie Gerdes
- Department of Clinical Genetics, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties, University of Genoa and Istituto de Ricovero e Cura a Carattere Scientifico AOU San Martino-IST, Genoa, Italy
| | - Alisa M Goldstein
- Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Thais C Grazziotin
- Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Nelleke A Gruis
- Department of Dermatology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Johan Hansson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Mark Harland
- Section of Epidemiology and Biostatistics, Leeds Institute of Medical Research at St. James's, University of Leeds, Leeds, United Kingdom
| | | | - Marko Hocevar
- Institute of Oncology Ljubljana, Zaloska, Ljubljana, Slovenia
| | - Veronica Höiom
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Elizabeth A Holland
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Westmead Institute for Medical Research, University of Sydney, Sydney, Australia
| | - Christian Ingvar
- Department of Clinical Sciences, Lund University Hospital Lund, Sweden; Department of Surgery, Lund University Hospital, Lund, Sweden
| | - Maria Teresa Landi
- Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Gilles Landman
- Department of Pathology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Alejandra Larre-Borges
- Unidad de Lesiones Pigmentadas, Cátedra de Dermatología, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Graham J Mann
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Westmead Institute for Medical Research, University of Sydney, Sydney, Australia
| | - Eduardo Nagore
- Department of Dermatology, Instituto Valenciano de Oncologia, Valencia, Spain
| | - Håkan Olsson
- Department of Clinical Sciences, Lund University Hospital Lund, Sweden; Department of Surgery, Lund University Hospital, Lund, Sweden
| | - Jane M Palmer
- QIMR Berghofer Medical Research Institute, Herston, Australia
| | - Barbara Perić
- Institute of Oncology Ljubljana, Zaloska, Ljubljana, Slovenia
| | - Dace Pjanova
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | - Susana Puig
- Melanoma Unit, Dermatology Department, Hospital Clinic Barcelona, Institut de Investigacions Biomediques August Pi Sunyer, Universitat de Barcelona, Barcelona, Spain; Centro de Investigacion Biomedica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain
| | - Helen Schmid
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Westmead Institute for Medical Research, University of Sydney, Sydney, Australia
| | - Nienke van der Stoep
- Department of Clinical Genetics, Leiden University Medical Center Leiden, the Netherlands
| | - Margaret A Tucker
- Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Karin A W Wadt
- Department of Clinical Genetics, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Xiaohong R Yang
- Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Julia A Newton-Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Medical Research at St. James's, University of Leeds, Leeds, United Kingdom
| | - Peter A Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| |
Collapse
|
17
|
Sargen MR, Pfeiffer RM, Yang XR, Tucker MA, Goldstein AM. Variation in Cutaneous Patterns of Melanomagenesis According to Germline CDKN2A/CDK4 Status in Melanoma-Prone Families. J Invest Dermatol 2019; 140:174-181.e3. [PMID: 31326397 DOI: 10.1016/j.jid.2019.06.138] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/19/2019] [Accepted: 06/30/2019] [Indexed: 11/26/2022]
Abstract
CDKN2A and CDK4 are well-established melanoma susceptibility genes, but their effect on tumor location and distribution is unknown. We used a case-case study design to assess for differences in tumor location between mutation carriers (CDKN2A = 141 patients, 348 melanomas; CDK4 = 15 patients, 54 melanomas) and noncarriers (104 patients, 157 melanomas) in US melanoma-prone families. Associations between groups were assessed with chi-square tests. Odds ratios (ORs) for tumor location were adjusted for diagnosis age, gender, and superficial spreading subtype. Models included random effects to account for within individual and family correlations. Compared with having a truncal melanoma, CDK4 (vs. noncarriers: lower extremities OR = 14.5, 95% confidence interval [CI] = 5.02-42.0, P < 0.001; upper extremities OR = 6.88, 95% CI = 2.37-19.9, P < 0.001; head and neck OR = 18.6, 95% CI = 4.04-85.2, P < 0.001) and CDKN2A (vs. noncarriers: lower extremities OR = 3.01, 95% CI = 1.56-5.82, P < 0.05; upper extremities OR = 1.91, 95% CI = 1.03-3.52, P < 0.05; head and neck OR = 5.40, 95% CI = 2.10-13.9, P < 0.001) carriers had higher odds of developing melanoma at all other sites. Similar findings were observed for analyses stratified by gender, age, and first versus subsequent melanoma diagnoses. Further studies are needed to understand the biology underlying these genotype-associated patterns of tumor development, which could provide new insights into melanoma treatment and prevention.
Collapse
Affiliation(s)
- Michael R Sargen
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland.
| | - Ruth M Pfeiffer
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Xiaohong R Yang
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Margaret A Tucker
- Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Alisa M Goldstein
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
18
|
Rocca MS, Benna C, Mocellin S, Rossi CR, Msaki A, Di Nisio A, Opocher G, Foresta C. E2F1 germline copy number variations and melanoma susceptibility. J Transl Med 2019; 17:181. [PMID: 31142321 PMCID: PMC6542053 DOI: 10.1186/s12967-019-1933-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/20/2019] [Indexed: 01/29/2023] Open
Abstract
Background Melanoma is an aggressive type of skin cancer whose aetiology remains elusive as both environmental and genetic factors can contribute to its development. Recent studies have demonstrated the existence of multiple copies of E2F1 gene in melanoma specimens which could explain the deregulated E2F1 activity in this type of cancer. This finding suggests a key role for this transcription factor in the malignant transformation of melanocytes. Therefore, E2F1 has been considered as a potential therapeutic target for this form of skin cancer. Since germline copy number variations (CNVs) have been associated with increased susceptibility to different types of cancer, the aim of our study was to assess germline E2F1 CNV in melanoma patients. However, CNVs not necessarily lead to gene dosage imbalance, hence, further factors, in association with CNVs, could contribute to clinical manifestations. Considering that heat stress has been hypothesised as a contributing factor to skin cancer, we also investigated the effect of heat stress on E2F1 expression. Methods E2F1 CNV was measured in genomic DNA isolated from blood of 552 patients diagnosed with melanoma and 520 healthy subjects using TaqMan Copy Number Assays. E2F1 mRNA expression was also evaluated by RT-qPCR in the melanoma cell line, SK MEL 267, before and after exposure to heat stress. Results We found that patients diagnosed with melanoma (1.6%, 9/552) harboured frequently altered germline E2F1 copies compared to healthy subjects (0%, 0/520). Moreover, the difference among the two groups was statistically significant (p = 0.004). Furthermore, we found that heat exposure alone can significantly induce E2F1 expression. Conclusions This is the first study that shows a relation between germline E2F1 CNV and melanoma, suggesting that altered copies of this gene might be a predisposing factor to skin cancer. Our results also suggest that environmental insults, such as heat stress, could contribute to an aberrant E2F1 activity by inducing E2F1 mRNA expression. Therefore, subjects with multiple constitutive copies of E2F1 are at greater risk of developing melanoma when exposed to heat. Altogether our results corroborate with the hypothesis that susceptibility to melanoma depends on both the environment and genetic factors.
Collapse
Affiliation(s)
- Maria Santa Rocca
- Familial Cancer Clinic, Veneto Institute of Oncology (IOV-IRCCS), Padua, Italy
| | - Clara Benna
- Department of Surgery Oncology and Gastroenterology (DISCOG), University of Padua, Padua, Italy.,First Surgical Clinic, Azienda Ospedaliera di Padova, Padua, Italy
| | - Simone Mocellin
- Surgical Oncology Unit, Veneto Institute of Oncology (IOV-IRCCS), Padua, Italy.,Department of Surgery Oncology and Gastroenterology (DISCOG), University of Padua, Padua, Italy
| | - Carlo Riccardo Rossi
- Surgical Oncology Unit, Veneto Institute of Oncology (IOV-IRCCS), Padua, Italy.,Department of Surgery Oncology and Gastroenterology (DISCOG), University of Padua, Padua, Italy
| | - Aichi Msaki
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padua, Via Giustiniani, 2, 35128, Padua, Italy
| | - Andrea Di Nisio
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padua, Via Giustiniani, 2, 35128, Padua, Italy
| | - Giuseppe Opocher
- Familial Cancer Clinic, Veneto Institute of Oncology (IOV-IRCCS), Padua, Italy
| | - Carlo Foresta
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padua, Via Giustiniani, 2, 35128, Padua, Italy.
| |
Collapse
|
19
|
Bourneuf E, Estellé J, Blin A, Créchet F, Schneider MDP, Gilbert H, Brossard M, Vaysse A, Lathrop M, Vincent-Naulleau S, Demenais F. New susceptibility loci for cutaneous melanoma risk and progression revealed using a porcine model. Oncotarget 2018; 9:27682-27697. [PMID: 29963229 PMCID: PMC6021234 DOI: 10.18632/oncotarget.25455] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/05/2018] [Indexed: 01/08/2023] Open
Abstract
Despite major advances, it is estimated that a large part of melanoma predisposing genes remains to be discovered. Animal models of spontaneous diseases are valuable tools and experimental crosses can be used to identify and fine-map new susceptibility loci associated with melanoma. We performed a Genome-Wide Association Study (GWAS) of melanoma occurrence and progression (clinical ulceration and presence of metastasis) in a porcine model of spontaneous melanoma, the MeLiM pig. Five loci on chromosomes 2, 5, 7, 8 and 16 showed genome-wide significant associations (p < 5 × 10–6) with either one of these phenotypes. Suggestive associations (p < 5 × 10–5) were also found at 16 additional loci. Moreover, comparison of the porcine results to those reported by human melanoma GWAS indicated shared association signals notably at CDKAL1 and TERT loci but also nearby CCND1, FTO, PLA2G6 and TMEM38B-RAD23B loci. Extensive search of the literature revealed a potential key role of genes at the identified porcine loci in tumor invasion (DST, PLEKHA5, CBY1, LIMK2 and ETV5) and immune response modulation (ETV5, HERC3 and DICER1) of the progression phenotypes. These biological processes are consistent with the clinico-pathological features of MeLiM tumors and can open new routes for future melanoma research in humans.
Collapse
Affiliation(s)
- Emmanuelle Bourneuf
- CEA, DRF/iRCM/SREIT/LREG, Jouy-en-Josas, France.,GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Jordi Estellé
- CEA, DRF/iRCM/SREIT/LREG, Jouy-en-Josas, France.,GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Amandine Blin
- GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France.,INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Outils et Méthodes de la Systématique Intégrative, OMSI-UMS 2700, CNRS MNHN, Muséum National d'Histoire Naturelle, Paris, France
| | - Françoise Créchet
- CEA, DRF/iRCM/SREIT/LREG, Jouy-en-Josas, France.,GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Maria Del Pilar Schneider
- GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France.,Present address: Ipsen Innovation, Les Ulis, France
| | - Hélène Gilbert
- GenPhyse, INRA, Université de Toulouse, INPT, ENVT, Castanet Tolosan, France
| | - Myriam Brossard
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Amaury Vaysse
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Mark Lathrop
- McGill University and Genome Québec Innovation Centre, Montréal, Québec, Canada
| | - Silvia Vincent-Naulleau
- CEA, DRF/iRCM/SREIT/LREG, Jouy-en-Josas, France.,GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Florence Demenais
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
20
|
Wu YP, Kohlmann W, Curtin K, Yu Z, Hanson HA, Hashibe M, Parsons BG, Wong J, Schiffman JD, Grossman D, Leachman SA. Melanoma risk assessment based on relatives' age at diagnosis. Cancer Causes Control 2017; 29:193-199. [PMID: 29243179 DOI: 10.1007/s10552-017-0994-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 12/08/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE The aim of this study was to determine risk for melanoma among individuals who have a first- or second-degree relative with a history of melanoma, based on the unaffected individual's age and age at diagnosis of the relative. METHODS The study employed a case-control design using a statewide database linked with a Surveillance Epidemiology and End Results cancer registry. A population-based sample of individuals who received at least one diagnosis of first primary, malignant melanoma (n = 14,281), as well as their first- and second-degree relatives, was included. Control individuals with no history of melanoma (n = 70,889) were matched to cases on birth year, gender, race/ethnicity, and county at birth. RESULTS Risk for melanoma among relatives of melanoma patients declined with relative's age and age at diagnosis. Individuals between ages 40 and 49 who are first-degree relatives of melanoma patients diagnosed between ages 40 and 49 had the greatest risk for melanoma compared with individuals without a first-degree relative with a melanoma history (HR 4.89; 95% CI 3.11-7.68). Increased melanoma risk among second-degree relatives of patients was typically lower than that for first-degree relatives. CONCLUSIONS Risk for melanoma, at earlier ages than expected, is increased among relatives of individuals with a history of melanoma, particularly if the melanoma case was diagnosed at a young age. Further research on the relationship between age at diagnosis and relative's melanoma risk could inform melanoma screening recommendations for individuals with a family history of the disease.
Collapse
Affiliation(s)
- Yelena P Wu
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA. .,Departments of Dermatology and Oncological Sciences, University of Utah, Salt Lake City, UT, USA.
| | - Wendy Kohlmann
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Karen Curtin
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Zhe Yu
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Heidi A Hanson
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Surgery, University of Utah, Salt Lake City, UT, USA
| | - Mia Hashibe
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Division of Public Health, Department of Family and Preventive Medicine, University of Utah, Salt Lake City, UT, USA
| | - Bridget G Parsons
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Jathine Wong
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Joshua D Schiffman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Douglas Grossman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Departments of Dermatology and Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Sancy A Leachman
- Department of Dermatology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
21
|
Bourneuf E. The MeLiM Minipig: An Original Spontaneous Model to Explore Cutaneous Melanoma Genetic Basis. Front Genet 2017; 8:146. [PMID: 29081790 PMCID: PMC5645500 DOI: 10.3389/fgene.2017.00146] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/25/2017] [Indexed: 12/19/2022] Open
Abstract
Melanoma is the deadliest skin cancer and is a major public health concern with a growing incidence worldwide. As for other complex diseases, animal models are needed in order to better understand the mechanisms leading to pathology, identify potential biomarkers to be used in the clinics, and eventually molecular targets for therapeutic solutions. Cutaneous melanoma, arising from skin melanocytes, is mainly caused by environmental factors such as UV radiation; however a significant genetic component participates in the etiology of the disease. The pig is a recognized model for spontaneous development of melanoma with features similar to the human ones, followed by a complete regression and a vitiligo-like depigmentation. Three different pig models (MeLiM, Sinclair, and MMS-Troll) have been maintained through the last decades, and different genetic studies have evidenced a complex inheritance of the disease. As in humans, pigmentation seems to play a prominent role, notably through MC1R and MITF signaling. Conversely, cell cycle genes as CDKN2A and CDK4 have been excluded as predisposing for melanoma in MeLiM. So far, only sparse studies have focused on somatic changes occurring during oncogenesis, and have revealed major cytological changes and a potential dysfunction of the telomere maintenance system. Finally, the spontaneous tumor progression and regression occurring in these models could shed light on the interplay between endogenous retroviruses, melanomagenesis, and adaptive immune response.
Collapse
Affiliation(s)
- Emmanuelle Bourneuf
- LREG, CEA, Université Paris-Saclay, Jouy-en-Josas, France.,GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
22
|
Wu YP, Mays D, Kohlmann W, Tercyak KP. Pediatric Predispositional Genetic Risk Communication: Potential Utility for Prevention and Control of Melanoma Risk as an Exemplar. J Genet Couns 2017; 26:887-893. [PMID: 28547663 PMCID: PMC5702278 DOI: 10.1007/s10897-017-0105-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 04/25/2017] [Indexed: 12/20/2022]
Abstract
Predispositional genetic testing among minor children is intensely debated due to the potential benefits and harms of providing this type of genetic information to children and their families. Existing guidelines on pediatric genetic testing state that predispositional testing could be appropriate for minors if preventive services exist that mitigate children's risk for or severity of the health condition in question. We use the example of hereditary melanoma to illustrate the rationale for and potential application of genetic risk communication for an adult-onset cancer to a pediatric population where childhood behaviors may reduce risk of disease later in life. We draw from the adult melanoma genetic risk communication and pediatric health behavior change literatures to suggest ways in which genetic test reporting and complementary education could be delivered to children who carry a hereditary risk for melanoma and their families in order to foster children's engagement in melanoma preventive behaviors. Genetic discoveries will continue to yield new opportunities to provide predispositional genetic risk information to unaffected individuals, including children, and could be delivered within programs that provide personalized and translational approaches to cancer prevention.
Collapse
Affiliation(s)
- Yelena P Wu
- Division of Public Health, Department of Family & Preventive Medicine, University of Utah, 375 Chipeta Way, Suite A, Salt Lake City, UT, 84108, USA.
- Huntsman Cancer Institute, Salt Lake City, UT, USA.
| | - Darren Mays
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D.C, USA
| | | | - Kenneth P Tercyak
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D.C, USA
| |
Collapse
|
23
|
"Better do not touch" and other superstitions concerning melanoma: the cross-sectional web-based survey. Postepy Dermatol Alergol 2016; 33:329-335. [PMID: 27881937 PMCID: PMC5110621 DOI: 10.5114/ada.2016.62837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/01/2016] [Indexed: 11/17/2022] Open
Abstract
Introduction To the authors’ best knowledge, there are no data regarding the prevalence of superstitions concerning melanoma among internet users. Aim To evaluate the prevalence and identify reasons for superstitions associated with excision of pigmented skin lesions as well as to assess the frequency of this procedure. Material and methods Readers of the scientific portal were invited to complete a fully anonymous e-questionnaire. After collection of questionnaires (5,154) and eliminating incomplete ones, 4,919 surveys were analysed. Results A total of 4,104 (83.4%) respondents have been aware that the total surgical excision is the only efficient way of melanoma treatment. This familiarity was related to increased skin cancer awareness but was not linked to regular skin self-examination. Over half of the surveyed agreed that “it is better not to touch naevi”. Moreover, 3,510 (71.3%) individuals believed that naevi located in “harmed places” may turn into melanoma. Conclusions Superstitions associated with surgical treatment of melanoma are widespread. Conducting educational campaigns is necessary, particularly among young people, whose dangerous tanning behaviours are important risk factors for melanoma occurrence in their later life.
Collapse
|
24
|
Menin C, Bojnik E, Del Bianco P, Elefanti L, Gianesin K, Keppel S, Stagni C, Mocellin S, Vecchiato A, De Rossi A. Differences in telomere length between sporadic and familial cutaneous melanoma. Br J Dermatol 2016; 175:937-943. [PMID: 27061190 DOI: 10.1111/bjd.14652] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Several pieces of evidence indicate that a complex relationship exists between constitutional telomere length (TL) and the risk of cutaneous melanoma. Although the general perception is that longer telomeres increase melanoma risk, some studies do not support this association. We hypothesize that discordant data are due to the characteristics of the studied populations. OBJECTIVES To evaluate the association of TL with familial and sporadic melanoma. MATERIALS AND METHODS TL was measured by multiplex quantitative polymerase chain reaction in leukocytes from 310 patients with melanoma according to familial/sporadic and single/multiple cancers and 216 age-matched controls. RESULTS Patients with sporadic melanoma were found to have shorter telomeres compared with those with familial melanoma. In addition, shorter telomeres, while tending to reduce the risk of familial melanoma regardless of single or multiple tumours, nearly trebled the risk of single sporadic melanoma. CONCLUSIONS This is the first time that TL has been correlated to opposite effects on melanoma risk according to the presence or absence of familial predisposition. Individual susceptibility to melanoma should be taken into account when assessing the role of TL as a risk factor.
Collapse
Affiliation(s)
- C Menin
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, via Gattamelata 64, 35128, Padova, Italy.
| | - E Bojnik
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128, Padova, Italy
| | - P Del Bianco
- Clinical Trials and Biostatistics Unit, Veneto Institute of Oncology, IOV-IRCCS, via Gattamelata 64, 35128, Padova, Italy
| | - L Elefanti
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, via Gattamelata 64, 35128, Padova, Italy
| | - K Gianesin
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128, Padova, Italy
| | - S Keppel
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, via Gattamelata 64, 35128, Padova, Italy
| | - C Stagni
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128, Padova, Italy
| | - S Mocellin
- Surgery Branch, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128, Padova, Italy
| | - A Vecchiato
- Oncology Surgery Unit, Veneto Institute of Oncology, IOV-IRCCS, via Gattamelata 64, 35128, Padova, Italy
| | - A De Rossi
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, via Gattamelata 64, 35128, Padova, Italy.,Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128, Padova, Italy
| |
Collapse
|
25
|
Taylor NJ, Handorf EA, Mitra N, Avril MF, Azizi E, Bergman W, Bianchi-Scarrà G, Bishop DT, Bressac-de Paillerets B, Calista D, Cannon-Albright LA, Cuellar F, Cust AE, Demenais F, Elder DE, Friedman E, Gerdes AM, Ghiorzo P, Goldstein AM, Grazziotin TC, Hansson J, Hayward NK, Hocevar M, Höiom V, Holland EA, Ingvar C, Landi MT, Landman G, Larre-Borges A, Leachman SA, Mann GJ, Nagore E, Olsson H, Palmer J, Perić B, Pjanova D, Puig S, Schmid H, van der Stoep N, Tucker MA, Wadt KAW, Whitaker L, Yang XR, Newton Bishop JA, Gruis NA, Kanetsky PA. Phenotypic and Histopathological Tumor Characteristics According to CDKN2A Mutation Status among Affected Members of Melanoma Families. J Invest Dermatol 2016; 136:1066-1069. [PMID: 26827760 PMCID: PMC5287416 DOI: 10.1016/j.jid.2016.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 01/04/2016] [Accepted: 01/05/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Nicholas J Taylor
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Elizabeth A Handorf
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Nandita Mitra
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marie-Françoise Avril
- Assistance Publique-Hôpitaux de Paris, Hôpital Cochin et Université Paris Descartes, Paris, France
| | - Esther Azizi
- Department of Dermatology, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Wilma Bergman
- Department of Dermatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Giovanna Bianchi-Scarrà
- Department of Internal Medicine and Medical Specialties, University of Genoa and IRCCS AOU San Martino-IST Genoa, Italy
| | - D Timothy Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | | | - Donato Calista
- Dermatology Unit, Maurizio Bufalini Hospital, Cesena, Italy
| | - Lisa A Cannon-Albright
- Division of Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Francisco Cuellar
- Melanoma Unit, Dermatology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain and CIBER de Enfermedades Raras, Barcelona, Spain
| | - Anne E Cust
- Sydney School of Public Health, University of Sydney, Sydney, NSW, Australia
| | - Florence Demenais
- INSERM, UMR-946, Genetic Variation and Human Disease Unit, Université Paris Diderot, Paris, France
| | - David E Elder
- Departments of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Eitan Friedman
- The Susanne Levy Gertner Oncogenetics Unit, The Danek Gertner Institute of Human Genetics, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Anne-Marie Gerdes
- Department of Clinical Genetics, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties, University of Genoa and IRCCS AOU San Martino-IST Genoa, Italy
| | - Alisa M Goldstein
- Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Thais C Grazziotin
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre RS, Brazil
| | - Johan Hansson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - Marko Hocevar
- Institute of Oncology Ljubljana, Zaloska, Ljubljana, Slovenia
| | - Veronica Höiom
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Elizabeth A Holland
- Centre for Cancer Research, Westmead Institute for Medical Research and Melanoma Institute Australia, University of Sydney, NSW, Australia
| | | | - Maria Teresa Landi
- Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Gilles Landman
- Department of Pathology, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Alejandra Larre-Borges
- Unidad de Lesiones Pigmentadas, Cátedra de Dermatología, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Sancy A Leachman
- Oregon Health Sciences University School of Medicine, Department of Dermatology, Portland, Oregon, USA
| | - Graham J Mann
- Centre for Cancer Research, Westmead Institute for Medical Research and Melanoma Institute Australia, University of Sydney, NSW, Australia
| | - Eduardo Nagore
- Department of Dermatology, Instituto Valenciano de Oncologia, Valencia, Spain
| | - Håkan Olsson
- Centre for Cancer Research, Westmead Institute for Medical Research and Melanoma Institute Australia, University of Sydney, NSW, Australia
| | - Jane Palmer
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Barbara Perić
- Institute of Oncology Ljubljana, Zaloska, Ljubljana, Slovenia
| | - Dace Pjanova
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Susana Puig
- Melanoma Unit, Dermatology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain and CIBER de Enfermedades Raras, Barcelona, Spain
| | - Helen Schmid
- Centre for Cancer Research, Westmead Institute for Medical Research and Melanoma Institute Australia, University of Sydney, NSW, Australia
| | - Nienke van der Stoep
- Department of Dermatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Margaret A Tucker
- Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Karin A W Wadt
- Department of Clinical Genetics, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Linda Whitaker
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Xiaohong R Yang
- Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Julia A Newton Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Nelleke A Gruis
- Department of Dermatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Peter A Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA.
| |
Collapse
|
26
|
Müller C, Wendt J, Rauscher S, Burgstaller-Muehlbacher S, Sunder-Plassmann R, Scheurecker C, Richtig E, Fae I, Fischer G, Pehamberger H, Okamoto I. Characterization of patients at high risk of melanoma in Austria. Br J Dermatol 2016; 174:1308-17. [PMID: 26800492 DOI: 10.1111/bjd.14407] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Risk of melanoma is determined by genetic and exogenous factors. Only a few studies have included both characteristics in a comprehensive multivariable analysis. OBJECTIVES To find determinants of patients at high risk of melanoma in Austria, including phenotype, genotype and lifestyle characteristics in comprehensive analyses. METHODS In total, 1668 patients with melanoma from the M3 case-control study were studied. Overall, 567 participants were sequenced for CDKN2A, 232 for CDK4, 123 for MITF encoding the variant E318K and 964 for MC1R. RESULTS Patients with melanoma with a positive family history (n = 190, 11·6%), multiple primary melanomas (n = 261, 15·7%) and younger age (< 50 years, n = 675, 40·5%) were defined as being at high risk. All other patients with melanoma were defined as the reference group. We found significant differences between those two groups and between the high-risk subgroups (positive family history, multiple primary melanomas and younger age). Pigmentation phenotype was associated with the high-risk group in general (childhood freckling, odds ratio 1·46, P = 0·007; blond/reddish hair colour, odds ratio 1·43, P = 0·011). Patients with a positive family history and patients with early-onset disease were similar regarding both their phenotypic characteristics and external factors. Established high-risk mutations in CDKN2A were found in cases with a positive family history (n = 12) or multiple melanomas (n = 2). Moreover, we found three patients carrying the MITF p.E318K variant, two with a CDK4 variant and seven with nonsynonymous MC1R variants with undescribed biological significance, of which four were predicted as damaging. CONCLUSIONS Austrian patients could represent a reservoir for novel genetic variants. Further investigation of populations in Central and Eastern Europe might reveal more novel and disease-relevant variants.
Collapse
Affiliation(s)
- C Müller
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - J Wendt
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - S Rauscher
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | - R Sunder-Plassmann
- Clinical Institute for Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - C Scheurecker
- Department of Dermatology and Venereology, General Hospital Linz, Linz, Austria
| | - E Richtig
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
| | - I Fae
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - G Fischer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - H Pehamberger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - I Okamoto
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
27
|
Marzagalli M, Montagnani Marelli M, Casati L, Fontana F, Moretti RM, Limonta P. Estrogen Receptor β in Melanoma: From Molecular Insights to Potential Clinical Utility. Front Endocrinol (Lausanne) 2016; 7:140. [PMID: 27833586 PMCID: PMC5080294 DOI: 10.3389/fendo.2016.00140] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/12/2016] [Indexed: 12/14/2022] Open
Abstract
Cutaneous melanoma is an aggressive tumor; its incidence has been reported to increase fast in the past decades. Melanoma is a heterogeneous tumor, with most patients harboring mutations in the BRAF or NRAS oncogenes, leading to the overactivation of the MAPK/ERK and PI3K/Akt pathways. The current therapeutic approaches are based on therapies targeting mutated BRAF and the downstream pathway, and on monoclonal antibodies against the immune checkpoint blockade. However, treatment resistance and side effects are common events of these therapeutic strategies. Increasing evidence supports that melanoma is a hormone-related cancer. Melanoma incidence is higher in males than in females, and females have a significant survival advantage over men. Estrogens exert their effects through estrogen receptors (ERα and ERβ) that affect cancer growth in an opposite way: ERα is associated with a proliferative action and ERβ with an anticancer effect. ERβ is the predominant ER in melanoma, and its expression decreases in melanoma progression, supporting its role as a tumor suppressor. Thus, ERβ is now considered as an effective molecular target for melanoma treatment. 17β-estradiol was reported to inhibit melanoma cells proliferation; however, clinical trials did not provide the expected survival benefits. In vitro studies demonstrate that ERβ ligands inhibit the proliferation of melanoma cells harboring the NRAS (but not the BRAF) mutation, suggesting that ERβ activation might impair melanoma development through the inhibition of the PI3K/Akt pathway. These data suggest that ERβ agonists might be considered as an effective treatment strategy, in combination with MAPK inhibitors, for NRAS mutant melanomas. In an era of personalized medicine, pretreatment evaluation of the expression of ER isoforms together with the concurrent oncogenic mutations should be considered before selecting the most appropriate therapeutic intervention. Natural compounds that specifically bind to ERβ have been identified. These phytoestrogens decrease the proliferation of melanoma cells. Importantly, these effects are unrelated to the oncogenic mutations of melanomas, suggesting that, in addition to their ERβ activating function, these compounds might impair melanoma development through additional mechanisms. A better identification of the role of ERβ in melanoma development will help increase the therapeutic options for this aggressive pathology.
Collapse
Affiliation(s)
- Monica Marzagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Marina Montagnani Marelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Lavinia Casati
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Milano, Italy
| | - Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Roberta Manuela Moretti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
- *Correspondence: Patrizia Limonta,
| |
Collapse
|
28
|
Wu YP, Aspinwall LG, Michaelis TC, Stump T, Kohlmann WG, Leachman SA. Discussion of photoprotection, screening, and risk behaviors with children and grandchildren after melanoma genetic testing. J Community Genet 2016; 7:21-31. [PMID: 26099287 PMCID: PMC4715817 DOI: 10.1007/s12687-015-0243-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 06/04/2015] [Indexed: 12/20/2022] Open
Abstract
The purpose of the current study was to examine changes in frequency of discussion about melanoma preventive behaviors among adults who received melanoma genetic test reporting and counseling and their children and grandchildren, correspondence of frequency of discussion with intentions, and content of discussions. Participants received CDKN2A/p16 testing and counseling (N = 24, 46 % p16-positive). Discussions about preventive behaviors were assessed before testing and 1 and 6 months post-testing. Intentions to discuss preventive behaviors and perceived preparedness to discuss risk were assessed post-testing. Open-ended questions assessed content of reported discussions. Discussion of preventive behaviors declined following test reporting, with more rapid decline reported by noncarriers. There was a large gap between the percentage of participants who intended to discuss preventive behaviors and who then reported discussions 1 and 6 months after counseling. Participants felt prepared to discuss melanoma risk but also suggested resources to facilitate discussions. Genetic test reporting and counseling alone did not sustain discussions about preventive behaviors for a hereditary cancer with children and grandchildren. The gap between intentions to have discussions and reported discussions has implications for augmentation of counseling to support at-risk families' discussions about preventive behaviors.
Collapse
Affiliation(s)
- Yelena P Wu
- Division of Public Health, Department of Family and Preventive Medicine, University of Utah, 375 Chipeta Way, Suite A, Salt Lake City, UT, 84108, USA.
- Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA.
| | - Lisa G Aspinwall
- Department of Psychology, University of Utah, 380 South 1530 East, Salt Lake City, UT, 84112, USA
| | - Timothy C Michaelis
- School of Medicine, University of Utah, 30 North 1900 East, Salt Lake City, UT, 84132, USA
| | - Tammy Stump
- Department of Psychology, University of Utah, 380 South 1530 East, Salt Lake City, UT, 84112, USA
| | - Wendy G Kohlmann
- Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
| | - Sancy A Leachman
- Department of Dermatology, Oregon Health and Science University, 3303 Southwest Bond Avenue, Portland, OR, 97239, USA
| |
Collapse
|
29
|
Potrony M, Badenas C, Aguilera P, Puig-Butille JA, Carrera C, Malvehy J, Puig S. Update in genetic susceptibility in melanoma. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:210. [PMID: 26488006 DOI: 10.3978/j.issn.2305-5839.2015.08.11] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Melanoma is the most deadly of the common skin cancers and its incidence is rapidly increasing. Approximately 10% of cases occur in a familial context. To date, cyclin-dependent kinase inhibitor 2A (CDKN2A), which was identified as the first melanoma susceptibility gene more than 20 years ago, is the main high-risk gene for melanoma. A few years later cyclin-dependent kinase 4 (CDK4) was also identified as a melanoma susceptibility gene. The technologic advances have allowed the identification of new genes involved in melanoma susceptibility: Breast cancer 1 (BRCA1) associated protein 1 (BAP1), CXC genes, telomerase reverse transcriptase (TERT), protection of telomeres 1 (POT1), ACD and TERF2IP, the latter four being involved in telomere maintenance. Furthermore variants in melanocortin 1 receptor (MC1R) and microphthalmia-associated transcription factor (MITF) give a moderately increased risk to develop melanoma. Melanoma genetic counseling is offered to families in order to better understand the disease and the genetic susceptibility of developing it. Genetic counseling often implies genetic testing, although patients can benefit from genetic counseling even when they do not fulfill the criteria for these tests. Genetic testing for melanoma predisposition mutations can be used in clinical practice under adequate selection criteria and giving a valid test interpretation and genetic counseling to the individual.
Collapse
Affiliation(s)
- Miriam Potrony
- 1 Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain ; 2 Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Valencia, Spain ; 3 Molecular Biology and Genetics Department, Melanoma Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Celia Badenas
- 1 Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain ; 2 Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Valencia, Spain ; 3 Molecular Biology and Genetics Department, Melanoma Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Paula Aguilera
- 1 Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain ; 2 Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Valencia, Spain ; 3 Molecular Biology and Genetics Department, Melanoma Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Joan Anton Puig-Butille
- 1 Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain ; 2 Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Valencia, Spain ; 3 Molecular Biology and Genetics Department, Melanoma Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Cristina Carrera
- 1 Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain ; 2 Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Valencia, Spain ; 3 Molecular Biology and Genetics Department, Melanoma Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Josep Malvehy
- 1 Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain ; 2 Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Valencia, Spain ; 3 Molecular Biology and Genetics Department, Melanoma Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Susana Puig
- 1 Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain ; 2 Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Valencia, Spain ; 3 Molecular Biology and Genetics Department, Melanoma Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
30
|
Aspinwall LG, Stump TK, Taber JM, Kohlmann W, Leaf SL, Leachman SA. Impact of melanoma genetic test reporting on perceived control over melanoma prevention. J Behav Med 2015; 38:754-65. [PMID: 25822116 PMCID: PMC4568125 DOI: 10.1007/s10865-015-9631-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 03/18/2015] [Indexed: 11/29/2022]
Abstract
To determine whether receiving melanoma genetic test results undermines perceived control over melanoma prevention, control-related beliefs were examined among 60 adults from melanoma-prone families receiving CDKN2A/p16 test results (27 unaffected noncarriers, 15 unaffected carriers, 18 affected carriers; response rate at 2 years = 64.9 % of eligible respondents). Multilevel modeling of perceived control ratings over a 2-year period revealed significant variation in individual trajectories: most participants showed increases (45 %) or no change (38.3 %), while 16.7 % showed decreases. At the group level, noncarriers reported sustained increases through the 2-year follow-up (ps < .05); unaffected carriers reported significant short-term increases (ps < .05); and affected carriers reported no change. Participants in all groups continued to rate photoprotection as highly effective in reducing melanoma risk and reported decreased beliefs that carrying the p16 mutation would inevitably lead to the development of melanoma. Qualitative responses immediately following counseling and test reporting corroborated these findings, as 93 % indicated it was possible to either prevent (64.9 %) or decrease the likelihood (28.1 %) of future melanomas. Thus, genetic test reporting does not generally undermine perceived control over melanoma prevention, though variability in response to positive results warrants future study.
Collapse
Affiliation(s)
- Lisa G Aspinwall
- Department of Psychology, University of Utah, 380 South 1530 East, Room 502, Salt Lake City, UT, 84112-0251, USA.
| | - Tammy K Stump
- Department of Psychology, University of Utah, 380 South 1530 East, Room 502, Salt Lake City, UT, 84112-0251, USA
| | - Jennifer M Taber
- Department of Psychology, University of Utah, 380 South 1530 East, Room 502, Salt Lake City, UT, 84112-0251, USA
| | | | - Samantha L Leaf
- Department of Psychology, University of Utah, 380 South 1530 East, Room 502, Salt Lake City, UT, 84112-0251, USA
- ISA Group, Alexandria, VA, USA
| | - Sancy A Leachman
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
31
|
Antonopoulou K, Stefanaki I, Lill CM, Chatzinasiou F, Kypreou KP, Karagianni F, Athanasiadis E, Spyrou GM, Ioannidis JPA, Bertram L, Evangelou E, Stratigos AJ. Updated field synopsis and systematic meta-analyses of genetic association studies in cutaneous melanoma: the MelGene database. J Invest Dermatol 2015; 135:1074-1079. [PMID: 25407435 DOI: 10.1038/jid.2014.491] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 10/09/2014] [Accepted: 10/31/2014] [Indexed: 12/26/2022]
Abstract
We updated a field synopsis of genetic associations of cutaneous melanoma (CM) by systematically retrieving and combining data from all studies in the field published as of August 31, 2013. Data were available from 197 studies, which included 83,343 CM cases and 187,809 controls and reported on 1,126 polymorphisms in 289 different genes. Random-effects meta-analyses of 81 eligible polymorphisms evaluated in >4 data sets confirmed 20 single-nucleotide polymorphisms across 10 loci (TYR, AFG3L1P, CDK10, MYH7B, SLC45A2, MTAP, ATM, CLPTM1L, FTO, and CASP8) that have previously been published with genome-wide significant evidence for association (P<5 × 10(-8)) with CM risk, with certain variants possibly functioning as proxies of already tagged genes. Four other loci (MITF, CCND1, MX2, and PLA2G6) were also significantly associated with 5 × 10(-8)
Collapse
Affiliation(s)
- Kyriaki Antonopoulou
- Department of Dermatology, University of Athens School of Medicine, Andreas Sygros Hospital, Athens, Greece
| | - Irene Stefanaki
- Department of Dermatology, University of Athens School of Medicine, Andreas Sygros Hospital, Athens, Greece
| | - Christina M Lill
- Neuropsychiatric Genetics Group, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany; Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Foteini Chatzinasiou
- Department of Dermatology, University of Athens School of Medicine, Andreas Sygros Hospital, Athens, Greece
| | - Katerina P Kypreou
- Department of Dermatology, University of Athens School of Medicine, Andreas Sygros Hospital, Athens, Greece
| | - Fani Karagianni
- Department of Dermatology, University of Athens School of Medicine, Andreas Sygros Hospital, Athens, Greece
| | - Emmanouil Athanasiadis
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - George M Spyrou
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - John P A Ioannidis
- Department of Medicine, Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, California, USA
| | - Lars Bertram
- Neuropsychiatric Genetics Group, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany; Department of Medicine, School of Public Health, Imperial College London, London, UK
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, Clinical and Molecular Epidemiology Unit, School of Medicine, University of Ioannina, Ioannina, Greece; Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, London, UK
| | - Alexander J Stratigos
- Department of Dermatology, University of Athens School of Medicine, Andreas Sygros Hospital, Athens, Greece.
| |
Collapse
|
32
|
Abstract
The last 30 years has seen a revolution in melanoma. Fundamental elements of the surgical, adjuvant medical, and systemic therapy for the disease have been significantly altered toward improved management and better outcomes. The intent of this article is to reflect on past efforts and research in melanoma and the current landscape of treatment of melanoma. The authors also hope to capture the excitement currently rippling through the field and the hope for a cure. The intent of treatment of advanced melanoma, which was once considered incurable, has changed from palliative to potentially curative.
Collapse
Affiliation(s)
- Vikram C Gorantla
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, University of Pittsburgh, 5150 Centre Avenue, Pittsburgh, PA 15232, USA
| | - John M Kirkwood
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, University of Pittsburgh, 5150 Centre Avenue, Pittsburgh, PA 15232, USA; Melanoma and Skin Cancer Program, University of Pittsburgh Cancer Institute, 5115 Centre Avenue, Suite 1.32, Pittsburgh, PA 15232, USA.
| |
Collapse
|
33
|
Aspinwall LG, Taber JM, Kohlmann W, Leaf SL, Leachman SA. Unaffected family members report improvements in daily routine sun protection 2 years following melanoma genetic testing. Genet Med 2014; 16:846-53. [PMID: 24763292 PMCID: PMC4209010 DOI: 10.1038/gim.2014.37] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/19/2014] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Reducing ultraviolet radiation exposure may decrease melanoma risk in the hereditary melanoma setting. It is unknown whether genetic counseling and test reporting of CDKN2A/p16 mutation status promote long-term compliance with photoprotection recommendations, especially in unaffected mutation carriers. METHODS This study evaluated changes 2 years following melanoma genetic testing in self-reported practice of sun protection (sunscreen, photoprotective clothing, and ultraviolet radiation avoidance) among 37 members of two CDKN2A/p16 kindreds (10 unaffected carriers, 11 affected carriers, and 16 unaffected noncarriers; response rate = 64.9% of eligible participants). RESULTS Multivariate profile analysis indicated that all three participant groups reported increased daily routine practice of sun protection 2 years following melanoma genetic testing (P < 0.02), with 96.9% reporting that at least one sun protection behavior was part of their daily routine, up from 78.1% at baseline (P < 0.015). Unaffected carriers (P < 0.024) and unaffected noncarriers (P < 0.027) reported significantly more frequent use of photoprotective clothing. Affected carriers maintained adherence to all sun protection behaviors. Reported sunburns in the past 6 months decreased significantly (P < 0.018). CONCLUSION Members of high-risk families reported increased daily routine sun protection and decreased sunburns 2 years following melanoma genetic testing, with no net decline in sun protection following negative test results. Thus, genetic testing and counseling may motivate sustained improvements in prevention behaviors.
Collapse
|
34
|
Potrony M, Puig-Butillé JA, Aguilera P, Badenas C, Carrera C, Malvehy J, Puig S. Increased prevalence of lung, breast, and pancreatic cancers in addition to melanoma risk in families bearing the cyclin-dependent kinase inhibitor 2A mutation: implications for genetic counseling. J Am Acad Dermatol 2014; 71:888-95. [PMID: 25064638 DOI: 10.1016/j.jaad.2014.06.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 05/30/2014] [Accepted: 06/22/2014] [Indexed: 11/18/2022]
Abstract
BACKGROUND Cyclin-dependent kinase inhibitor 2A (CDKN2A) is the major high-risk susceptibility gene for melanoma. OBJECTIVE We sought to evaluate the effect of CDKN2A mutations in Spanish patients with a high risk of developing melanoma and the association with clinical and family history features. METHODS A cross-sectional study design was used to analyze the CDKN2A impact in 702 Spanish patients with a high risk of developing melanoma. RESULTS The CDKN2A mutation prevalence was 8.5% in patients with sporadic multiple primary melanoma and 14.1% in familial melanoma. Number of cases in the family, number of primary melanomas, and age of onset were associated with the presence of CDKN2A mutation. Having a CDKN2A mutation in the family increased the prevalence of other cancers (prevalence ratio [PR] 2.99, P=.012) and prevalence of pancreatic (PR 2.97, P=.006), lung (PR 3.04, P<.001), and breast (PR 2.19, P=.018) cancers but not nephrourologic or colon cancer. LIMITATIONS Smoking status was not assessed in the individuals with lung cancer. CONCLUSIONS Melanoma-prone families with mutations in CDKN2A have an increased prevalence of a broad spectrum of cancers including lung, pancreatic, and breast cancer. This information should be included in genetic counseling and cancer prevention programs for CDKN2A mutation carriers.
Collapse
Affiliation(s)
- Miriam Potrony
- Centro de Investigación Biomédica en Red de Enfermedades Raras ISCIII, Barcelona, Spain; Dermatology Department, Melanoma Unit, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Joan Anton Puig-Butillé
- Centro de Investigación Biomédica en Red de Enfermedades Raras ISCIII, Barcelona, Spain; Biochemical and Molecular Genetics Service, Melanoma Unit, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Paula Aguilera
- Centro de Investigación Biomédica en Red de Enfermedades Raras ISCIII, Barcelona, Spain; Dermatology Department, Melanoma Unit, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Celia Badenas
- Centro de Investigación Biomédica en Red de Enfermedades Raras ISCIII, Barcelona, Spain; Biochemical and Molecular Genetics Service, Melanoma Unit, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Cristina Carrera
- Centro de Investigación Biomédica en Red de Enfermedades Raras ISCIII, Barcelona, Spain; Dermatology Department, Melanoma Unit, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Josep Malvehy
- Centro de Investigación Biomédica en Red de Enfermedades Raras ISCIII, Barcelona, Spain; Dermatology Department, Melanoma Unit, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Susana Puig
- Centro de Investigación Biomédica en Red de Enfermedades Raras ISCIII, Barcelona, Spain; Dermatology Department, Melanoma Unit, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain.
| |
Collapse
|
35
|
Abstract
The purpose of this article is to review the common neoplasms, infections, and inflammatory dermatoses that may present around or near the mouth. Dental professionals are well positioned to evaluate perioral skin conditions, further contributing to patients' general health. This article includes a review of seborrheic keratosis, warts, actinic keratoses, actinic cheilitis, and squamous cell carcinoma, among several other perioral cutaneous lesions.
Collapse
Affiliation(s)
- Geoffrey F S Lim
- Department of Dermatology, Drexel University College of Medicine, The Arnold T. Berman, M.D. Building, 219 North Broad Street, Philadelphia, PA 19107, USA.
| | - Carrie Ann R Cusack
- Department of Dermatology, Drexel University College of Medicine, The Arnold T. Berman, M.D. Building, 219 North Broad Street, Philadelphia, PA 19107, USA
| | - Joseph M Kist
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, South Pavilion, 1st Floor, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| |
Collapse
|
36
|
Abstract
Intact miRNAs can be isolated from the circulation in significant quantities despite the presence of extremely high levels of RNase activity. The remarkable stability of circulating miRNAs makes them excellent candidates for biomarkers in diagnostic applications as well as therapeutic targets in a variety of disease states including melanoma. Circulating RNA molecules are resistant to degradation by RNases because they are encapsulated in membrane-bound microvesicles. We describe a convenient method for the use of ExoQuick, a proprietary resin developed by Systems Biosciences (Mountain View, CA), whereby microvesicles can be purified under gentle conditions using readily available laboratory equipment. This protocol allows for isolation all microvesicles, regardless of their origin, and provides a convenient method for identifying potential cancer-specific biomarkers from biological fluids including serum and plasma.
Collapse
|
37
|
Aspinwall LG, Taber JM, Leaf SL, Kohlmann W, Leachman SA. Melanoma genetic counseling and test reporting improve screening adherence among unaffected carriers 2 years later. Cancer Epidemiol Biomarkers Prev 2013; 22:1687-97. [PMID: 23950214 PMCID: PMC3837428 DOI: 10.1158/1055-9965.epi-13-0422] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND A major goal of predictive genetic testing for melanoma is to promote early detection to reduce mortality. This study evaluated the long-term impact of melanoma genetic test reporting and counseling on screening adherence. METHODS This study assessed adherence to recommendations for annual total body skin examinations (TBSE) and monthly skin self-examinations (SSE) among 37 members of Utah CDKN2A/p16 kindreds (10 unaffected carriers, 11 affected carriers, and 16 unaffected noncarriers; response rate = 64.9% of eligible participants). RESULTS Two years following test reporting, adherence to annual TBSE among unaffected carriers increased from 40% to 70%. However, unaffected noncarriers' adherence decreased from 56% to 13%. Affected carriers reported TBSEs at both assessments (91% and 82%, respectively). Monthly SSE frequency remained highly variable in all patient groups: at 2 years, 29.7% reported monthly SSEs, 27.0% reported more frequent self-examinations, and 43.2% reported underscreening. However, SSE quality improved significantly: participants checked more body sites at 2 years than at baseline, especially feet, shoulders, legs, and genitals. Perceived logistic barriers to TBSEs (e.g., expensive, inconvenient) and SSEs (hard to remember, time-consuming) predicted lower adherence. CONCLUSIONS Unaffected carriers reported increased TBSE adherence and thoroughness of SSEs 2 years following melanoma genetic test reporting, suggesting clinical benefit in this modest sample. Unaffected noncarriers reported comparable gains in SSE thoroughness, but decreased TBSEs. IMPACT Melanoma genetic counseling and test reporting may improve adherence among unaffected carrier members of p16 families. Further interventions to reduce logistic barriers and to promote continued screening adherence among unaffected noncarrier family members may be needed.
Collapse
Affiliation(s)
| | | | | | | | - Sancy A. Leachman
- Department of Dermatology, University of Utah
- Huntsman Cancer Institute
| |
Collapse
|
38
|
|
39
|
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
|
40
|
Badenas C, Aguilera P, Puig-Butillé JA, Carrera C, Malvehy J, Puig S. Genetic counseling in melanoma. Dermatol Ther 2013; 25:397-402. [PMID: 23046018 DOI: 10.1111/j.1529-8019.2012.01499.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Genetic counseling may be offered to families with melanoma and to individuals with multiple melanomas to better understand the genetic susceptibility of the disease, the influence of environmental factors, the inheritance of the risk, and behavior that decreases the risk of dying from melanoma, including specific dermatological follow-up such as total body photography and digital dermoscopy. Genetic testing may be offered to those individuals with more than a 10% chance of being a carrier of a mutation. This risk varies according to the incidence of melanoma in the country and sun behavior. In countries with a low-medium incidence of melanoma, genetic testing should be offered to families with two cases of melanoma or an individual with two primary melanomas. In countries with a high incidence, families with three cases of melanoma, with two melanomas and one pancreatic adenocarcinoma, or patients with three primary melanomas, may benefit from genetic testing.
Collapse
Affiliation(s)
- Celia Badenas
- Centro Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Biochemical and Molecular Genetics Service, Melanoma Unit, Hospital Clinic, Villarroel, Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|
41
|
Puntervoll HE, Yang XR, Vetti HH, Bachmann IM, Avril MF, Benfodda M, Catricalà C, Dalle S, Duval-Modeste AB, Ghiorzo P, Grammatico P, Harland M, Hayward NK, Hu HH, Jouary T, Martin-Denavit T, Ozola A, Palmer JM, Pastorino L, Pjanova D, Soufir N, Steine SJ, Stratigos AJ, Thomas L, Tinat J, Tsao H, Veinalde R, Tucker MA, Bressac-de Paillerets B, Newton-Bishop JA, Goldstein AM, Akslen LA, Molven A. Melanoma prone families with CDK4 germline mutation: phenotypic profile and associations with MC1R variants. J Med Genet 2013; 50:264-70. [PMID: 23384855 PMCID: PMC3607098 DOI: 10.1136/jmedgenet-2012-101455] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background CDKN2A and CDK4 are high risk susceptibility genes for cutaneous malignant melanoma. Melanoma families with CDKN2A germline mutations have been extensively characterised, whereas CDK4 families are rare and lack a systematic investigation of their phenotype. Methods All known families with CDK4 germline mutations (n=17) were recruited for the study by contacting the authors of published papers or by requests via the Melanoma Genetics Consortium (GenoMEL). Phenotypic data related to primary melanoma and pigmentation characteristics were collected. The CDK4 exon 2 and the complete coding region of the MC1R gene were sequenced. Results Eleven families carried the CDK4 R24H mutation whereas six families had the R24C mutation. The total number of subjects with verified melanoma was 103, with a median age at first melanoma diagnosis of 39 years. Forty-three (41.7%) subjects had developed multiple primary melanomas (MPM). A CDK4 mutation was found in 89 (including 62 melanoma cases) of 209 tested subjects. CDK4 positive family members (both melanoma cases and unaffected subjects) were more likely to have clinically atypical nevi than CDK4 negative family members (p<0.001). MPM subjects had a higher frequency of MC1R red hair colour variants compared with subjects with one tumour (p=0.010). Conclusion Our study shows that families with CDK4 germline mutations cannot be distinguished phenotypically from CDKN2A melanoma families, which are characterised by early onset of disease, increased occurrence of clinically atypical nevi, and development of MPM. In a clinical setting, the CDK4 gene should therefore always be examined when a melanoma family tests negative for CDKN2A mutation.
Collapse
Affiliation(s)
- Hanne Eknes Puntervoll
- Section for Pathology, The Gade Institute, University of Bergen, Haukeland University Hospital, N-5021 Bergen, Norway
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Vecchiato A, Pasquali S, Menin C, Montesco MC, Alaibac M, Mocellin S, Campana LG, Nitti D, Rossi CR. Histopathological characteristics of subsequent melanomas in patients with multiple primary melanomas. J Eur Acad Dermatol Venereol 2012; 28:58-64. [PMID: 23216522 DOI: 10.1111/jdv.12055] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Multiple primary melanomas (MPM) occur in up to 20% of melanoma patients, and subsequent tumours seem to have a favourable histopathological pattern. OBJECTIVE A prospectively collected cohort of 194 patients with MPM was retrospectively reviewed to investigate clinical and histopathological features of first and subsequent melanomas. METHODS Patients with MPM who were diagnosed at our Department (1985-2011) and who attended at least a follow-up control yearly were identified. RESULTS The number of nevi was <10, 10-50 and >50 in 8.7%, 41% and 50.3% of patients respectively. Histopathological dysplastic nevi have been diagnosed in 105 patients. During a median follow-up of 58 months, 159 (81.9%), 24 (12.3%), 7 (3.6%) and 4 (2%) patients developed 2, 3, 4 and ≥ 5 melanomas, respectively. The median time to second primary melanoma was 45 months. The second primary melanoma was diagnosed within 1-year and after 5-year from the first melanoma in 36.6% and 17.3% of patients respectively. First and second primary melanomas were in situ in 41 (21%) and 104 (54%) patients respectively (P < 0.001). Among patients with ≥ 2 invasive melanomas (N = 80), median tumour thickness and ulceration of first and second primaries were 0.91 and 0.44 mm (P <0.001), and 32% and 7.7% (P = 0.001) respectively. CONCLUSIONS Subsequent melanomas occurred within 1-year from the appearance of the first melanoma in 36% of patients with MPM, while a late melanoma diagnosis was detected in 17% of cases. Second primary melanoma had favourable histopathological features. Our findings support long-term skin surveillance to detect subsequent melanomas at an early stage.
Collapse
Affiliation(s)
- A Vecchiato
- Melanoma and Sarcomas Unit, Veneto Institute of Oncology, Padova, ItalyDepartment of Oncological and Surgical Sciences, University of Padova, Padova, ItalyImmunology and Molecular Oncology Unit, Veneto Institute of Oncology, Padova, ItalyPathology Unit, Veneto Institute of Oncology, Padova, ItalyDermatology Unit, University of Padova, Padova
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Melanoma is an important public health problem in the United States and worldwide. The incidence of melanoma continues to increase at a high rate and deaths from melanoma are also increasing. The endogenous risk factors that are currently recognized are in many cases surrogates for genetic markers yet to be determined. Exogenous risk factors need to be better defined and understood to help develop better public education programs that can change risk behaviors and subsequently lower future incidence and mortality from melanoma.
Collapse
Affiliation(s)
- Julie E Russak
- Department of Dermatology, Mt. Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029-6574, USA.
| | | |
Collapse
|
44
|
Velho TR. Metastatic melanoma - a review of current and future drugs. Drugs Context 2012; 2012:212242. [PMID: 24432031 PMCID: PMC3885142 DOI: 10.7573/dic.212242] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 08/20/2012] [Indexed: 12/21/2022] Open
Abstract
Background: Melanoma is one of the most aggressive cancers, and it is estimated that 76,250 men and women will be diagnosed with melanoma of the skin in the USA in 2012. Over the last few decades many drugs have been developed but only in 2011 have new drugs demonstrated an impact on survival in metastatic melanoma. Methods: A systematic search of literature was conducted, and studies providing data on the effectiveness of current and/or future drugs used in the treatment of metastatic melanoma were selected for review. This review discusses the advantages and limitations of these agents, evaluating past, current and future clinical trials designed to overcome such limitations. Results: To date, there are four drugs approved by the Food and Drug Administration for melanoma (dacarbazine, interleukin-2, ipilimumab and vemurafenib). Despite efforts to develop new drugs, few of them have demonstrated any clinical benefits. Approved in 1975, dacarbazine remains the gold standard in chemotherapy, although ipilimumab and vemurafenib have raised many hopes in the last few years. Combining dacarbazine or other chemotherapy agents with new pharmacological agents may be a new way to achieve better clinical responses in patients with metastatic melanoma. Discussion: Advances in the molecular knowledge of melanoma have led to major improvements in the treatment of patients with metastatic melanoma, providing new targets and insights. However, heterogeneity amongst study populations, different approaches to treatment and the different melanoma types and localisations included in the trials makes their comparison difficult. New studies focusing on drugs developed in recent decades are warranted.
Collapse
|
45
|
Pedace L, De Simone P, Castori M, Sperduti I, Silipo V, Eibenschutz L, De Bernardo C, Buccini P, Moscarella E, Panetta C, Ferrari A, Grammatico P, Catricalà C. Clinical features predicting identification of CDKN2A mutations in Italian patients with familial cutaneous melanoma. Cancer Epidemiol 2011; 35:e116-20. [DOI: 10.1016/j.canep.2011.07.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 07/24/2011] [Accepted: 07/29/2011] [Indexed: 12/12/2022]
|
46
|
Aspinwall LG, Taber JM, Leaf SL, Kohlmann W, Leachman SA. Genetic testing for hereditary melanoma and pancreatic cancer: a longitudinal study of psychological outcome. Psychooncology 2011; 22:276-89. [PMID: 23382133 DOI: 10.1002/pon.2080] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 08/26/2011] [Accepted: 08/27/2011] [Indexed: 11/10/2022]
Abstract
OBJECTIVE CDKN2A/p16 mutations confer 76% lifetime risk of melanoma and up to 17% lifetime risk of pancreatic cancer. Our objective was to determine the short- and long-term impact of CDKN2A/p16 genetic counseling and test reporting on psychological distress, cancer worry, and perceived costs and benefits of testing. METHODS Prospective changes in anxiety, depression, and cancer worry following CDKN2A/p16 counseling and test reporting were evaluated at multiple assessments over 2 years among 60 adult members of melanoma-prone families; 37 participants completed the 2-year follow-up. Quantitative and qualitative assessments of the costs and benefits of testing were carried out. Outcomes were evaluated among unaffected noncarriers (n = 27), unaffected carriers (n = 15), and affected carriers (n = 18). RESULTS Reported anxiety and depression were low. For carriers and noncarriers, anxiety decreased significantly throughout the 2-year period, whereas depression and melanoma worry showed short-term decreases. Worry about pancreatic cancer was low and decreased significantly. In all groups, test-related distress and uncertainty were low, regret was absent, and positive experiences were high. All participants (>93% at each assessment) reported at least one perceived benefit of genetic testing; only 15.9% listed any negative aspect. Carriers reported increased knowledge about melanoma risk and prevention (78.3%) and increased prevention and screening behaviors for self and family (65.2%). Noncarriers reported increased knowledge (95.2%) and emotional benefits (71.4%). CONCLUSION Among US participants familiar with their hereditary melanoma risk through prior epidemiological research participation, CDKN2A/p16 genetic testing provides multiple perceived benefits to both carriers and noncarriers without inducing distress in general or worry about melanoma or pancreatic cancer.
Collapse
Affiliation(s)
- Lisa G Aspinwall
- Department of Psychology, University of Utah, Salt Lake City, UT, USA.
| | | | | | | | | |
Collapse
|
47
|
Brandt A, Sundquist J, Hemminki K. Risk of incident and fatal melanoma in individuals with a family history of incident or fatal melanoma or any cancer. Br J Dermatol 2011; 165:342-8. [DOI: 10.1111/j.1365-2133.2011.10350.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
48
|
Abstract
Malignant melanoma represents a significant and growing public health burden in the US and worldwide. It is estimated that 68, 130 cases of invasive malignant melanoma and at least 48,000 cases of melanoma in-situ will be diagnosed in the US this year. Melanoma is also one of the few remaining cancers with increasing US incidence. In the 1930s, the lifetime risk of an American developing invasive malignant melanoma was 1 in 1,500. Currently, that risk is 1 in 59. Deaths from malignant melanoma are also increasing. The mortality rate from malignant melanoma has risen about 2% annually since 1960. This year, it is estimated that 8,700 Americans will die from this cancer. The identification of individuals at high risk for malignant melanoma is important for the development of focused and efficient prevention efforts. Acute sun exposure resulting in sunburn remains a significant risk factor for the development of melanoma, but numerous other potential risk factors have been cited. Included among these are atypical mole syndrome/dysplastic nevus syndrome, blistering sunburns, immunosuppression, prior therapy with psoralen with ultraviolet A light (UVA) light, UV exposure at tanning salons, elevated socioeconomic status, and history of melanoma in a first-degree relative. With a better understanding of the reasons for the increasing rate of this cancer, and with enhanced early detection approaches, we may be able to decrease the incidence and mortality of malignant melanoma.
Collapse
Affiliation(s)
- Darrell S Rigel
- Department of Dermatology, New York University Medical Center, New York, NY, USA.
| |
Collapse
|
49
|
Chiarugi A, Nardini P, Crocetti E, Carli P, De Giorgi V, Borgognoni L, Brandani P, Pimpinelli N, Manganoni A, Quaglino P. Familial and sporadic melanoma: different clinical and histopathological features in the Italian population - a multicentre epidemiological study - by GIPMe (Italian Multidisciplinary Group on Melanoma). J Eur Acad Dermatol Venereol 2011; 26:194-9. [DOI: 10.1111/j.1468-3083.2011.04035.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
50
|
|