Open Access

Genetics and epigenetics of melanoma (Review)

Retraction in: /10.3892/ol.2020.12045

  • Authors:
    • Xiao-Ying Zhang
    • Pei-Ying Zhang
  • View Affiliations

  • Published online on: September 5, 2016     https://doi.org/10.3892/ol.2016.5093
  • Pages: 3041-3044
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer affects multiple organs in the body Malignant melanoma involves the invasion of skin and occasionally mucosal membrane or eye choroidal tissues. The incidence of cutaneous malignant melanoma is on the increase worldwide and is a major concern in current research. The increase is associated with UV irradiation-induced genetic aberrations that stimulate skin melanocytes to develop unlimited growth. This eventually leads to cell immortality, which in turn causes metastases. The present review examines the genetics and epigenetics of this pathological state together with recent perspectives of the therapeutic management of disease.

Introduction

Cutaneous malignant melanoma (CMM) has been ranked as the 6th most common cancer in men and the 5th most common cancer in women (1,2). MC1R is regarded as a low to median penetrance CMM susceptibility gene (3). Carriage of two or more variants for MC1R was found to increase the risk for CMM. All the variants associated with the appearance of red or blond hair color were significantly associated with an increase in CMM risk. In addition, carriage of MC1R variants without the typical red hair/blond hair and fair skin color was shown to be associated with an increase in CMM risk. This finding suggested that MC1R function exerts CMM-related effects beyond those related to the phenotype (4).

Heredity for CMM includes shared genetic variants associated with the phenotype, as well as germ-line aberrations, as identified in 5–10% of all families with an increased number of CMM cases (5). These mutations are dominated by regionally distinct variants in CDKN2A. Rare germ-line mutations in CDK4 are found in addition to aberrations in MITF, TERF2IP, ACD, POT1 and BAP1 genes, albeit at a low frequency (5). As the prevalence of heredity in CMM is low, the contribution of germ-line mutations for CMM pathology is limited, albeit several of the germ-line altered genes are also somatically altered. In addition, as some of these genes increase the risk for development of other neoplasms [pancreatic cancer for CDKN2A and CDK4; uveal melanoma (UM) for BAP1], screening of the alterations is useful for the identification of subjects with an increased risk for cancer.

Genetic aberrations and melanoma-associated pathways

The major genetic pathways activated in CMM tumors are the mitogen-activated protein kinase (MAPK) (RAS-RAF- MEK-ERK) and PI3K pathways (PIK3CA-AKT-mTOR). The two pathways are known to be directly activated by oncogenic signaling by some receptor tyrosine kinases (RTKs) (EGFR, MET and KIT), whereas any of the two pathways are activated by many RTKs (VEGFR, IGF1R, EPHA2 and FGFR). In addition, the MAPK pathway is activated by oncogenic mutations in RAS (predominantly NRAS in CMM) or BRAF leading to a constitutively active MAPK downstream signaling and unregulated expression of cyclin D1 protein as one of the high impact oncogenic events (6).

Some G-protein coupled receptors (GPCRs) have been shown to carry mutations in the intercellular C-terminal part causing them to signal constitutively downstream, activating the MAPK pathway. GRM3 mutations lead to increased ERK phosphorylation, although the exact downstream signaling details remain to be determined (7). In addition, phenotype-related GPCR MC1R with downstream signaling via the cyclic AMP-mediated pathway may be involved in oncogenic signaling in CMM. Similarly, constitutive MAPK pathway activation occurs in UM. However, the identified alterations associated with this activation are overexpression of RTKs (KIT, IGF1R and MET) and constitutive signaling from GPCRs GNAQ and GNA11, leading to the activation of MAPK, as well as signaling to PLCγ in the PI3K-AKT pathway (8).

In malignant melanoma, a number of mechanisms leading to neoplasia have been previously described (911). CMM development is known to be associated with the sporadic loss of p16INK4A protein translated from one of two reading frames of the CDKN2A gene. Similarly, the association of germ-line mutations in CDKN2A and aggregation of CMM in families has been recognized (12). Loss of this tumor suppressive protein inhibits p53-dependent functions in apoptosis and prevents G2/M cell-cycle checkpoint engagement (13). CMM involves loss of the tumor suppressor gene, CDKN2A, and the derived proteins, as well as constitutive, oncogenic activation of the RAS-RAF-MEK-ERK (MAPK) pathway by oncogenic mutations, primarily in genes for BRAF and NRAS. In addition, CDKN2A is deactivated transcriptionally by promoter methylation in approximately 20% of melanoma metastases (14).

The pattern for the aberrations does not support a simple association to CMM. In particular, the protein expression of GLI1 and GLI3 appears to be elevated in CMM tumors (TCGA database, cbioportal.org/public-portal/).

Epigenetics of melanoma

The epigenetic alterations in DNA and histones have recently become a part of melanoma genetic aberrations. Epigenetic alterations are regarded as being related to transcriptional deregulation leading to loss of tumor suppressor gene expression (transcriptional silencing) and/or upregulation of genes and their proteins, with an enhanced expression in malignant cells compared to normal cells (15). Epigenetic alterations that affect transcription factor binding to DNA may lead to cell adaptation towards increased survival (16).

Transcriptional silencing and histone modifications

Epigenetic regulation of gene expression has been associated with silencing of tumor suppressor genes in melanoma, specifically CDKN2A, RASSF1A and PTEN (1719). Transcriptional silencing by aberrant promoter methylation is associated with dimethylated lysine 9 in histone H3 (me2H3K9), resulting from the activity of H3K9 histone methyltransferases such as SETDB1 and EHMT2, and counteracted by H3K9 histone demethylase LSD1 (20). Transcriptional repression is believed to require binding of methyl-CpG binding protein (MECP2) and methyl-CpG binding domain protein 1 and 2 (MBD1 and MBD2) activity. MBD1 is also a binding partner to SETDB1 (16).

Ectopic overexpression of miR-124a, which regulates EZH2 negatively, has been shown to decrease tumor growth in vivo in CMM and to be associated with tumor aggression. Similar effects have been observed with regard to miR-124a in UM, confirming that EZH2 downregulation is important in melanoma (21).

miRNA-related regulation

Epigenetic regulation of protein expression is derived from miRNA that has an impact on gene expression or protein stability, which affects the oncogenic process. One example of this is miR-125b, a negative regulator of the MAPK downstream targeting c-Jun by altering translation or protein stability. The forced expression of miR-125b was found to suppress cell proliferation and migration, in agreement with downregulation of the MAPK pathway (22,23).

Alterations in epigenetic regulators

Loss of gene regulation in melanoma may be associated with epigenetic regulation and a result of mutations in gene coding for epigenetic regulators of gene expression. Alterations in EZH2 or SETDB1 are found in 37% of CMM tumors, accounting for increased mRNA transcription, gene amplification and mutation (TCGA database, 20). This high frequency together with the appearance of EZH2 mutations in the catalytic domain suggest that these and possibly other epigenetic regulators are associated with the oncogenic potential of melanoma.

Malignant melanoma therapy

Early detection and surgical removal of melanoma does, in ~90% of diagnosed cases, offer successful treatment for the disease (24). However, when surgery is not sufficient to remove the melanoma cells, there is high risk for metastatic spread of the disease, whereby cutaneous melanoma primary tumor thickness (Breslow thickness) and melanoma propensity metastasize to multiple tissues (lymphatic, soft, lung and CNS tissues) (25). For UM, the metastatic spread is often limited to the liver and may allow therapy using liver perfusion and a high local dose of chemotherapy (26).

Classic systemic therapy for disseminated cutaneous melanoma involves alkylating agents including dacarbazine (DTIC), temozolomide (TMZ) as monotherapies or together with platinum compounds (cisplatin or carboplatin), other alkylating drugs (fotemustine, melphalan) or immunostimulatory agents interleukin (IL)-2, and interferon (IFN)-targeted therapies (27,28). However, single-agent therapies nor combinations of different types of agents have been particularly successful in therapy, exhibiting response rates (RRs) of 5–12% and median overall survival (OS) <1–8 months (25). However, in a large phase III trial NCT00091572 conducted on stage IV patients, with 429 patients receiving TMZ (150 mg/m2 orally 7 consecutive days every 14 days) and 430 patients receiving DTIC (1,000 mg/m2 intravenously day 1+/− 3 days every 3 weeks), the median OS was 9.1 and 9.4 months, respectively, with the corresponding objective response rates being 10 and 14% for TMZ and DTIC, respectively (www.clinicaltrials.gov). In a meta-analysis conducted, allowing pooling of data for DTIC and TMZ clinical trial monotherapies, a median OS length of 7.9 months was established for the chemotherapeutic drugs (29). As few patients respond to therapy and the sustained therapeutic efficacy is poor, alternatives to classic chemotherapy have been to treat patients systemically with high-, intermediate- or low-dose IFN in the adjuvant setting. In a meta-analysis focusing on clinical trials, investigating 3,593 treated patients vs. 2,539 control patients (observation), the results showed an overall decreased risk of death with IFN administration, exhibiting a significant, albeit rather weak enhancement of therapeutic efficacy (30).

Future perspectives for melanoma therapy

In general, the high mutational rate observed in CMM cells is a potential source of therapy resistance. It increases the likelihood for the melanoma cells to become heterogenic as clones of melanoma cells develop aberrations in the same pathways but in different components of the pathways (TCGA database; COSMIC database, http://cancer.sanger.ac.uk/cosmic). Heterogeneity inducers other than diversity in genetic aberrations may include hypoxia, tumor microenvironment, selection by therapeutic agents and may be executed by reversible alterations of epigenetic changes in miRNA profiles or histone modifications and be regarded as adaptations. CMM exhibits a relatively small number of tumors activated by mutated RTKs and non-RTKs, whereas the overexpression of the RTKs, often several in the same tumors (including IGF1R, MET, SRC, EPHA2, KIT, ERBB3 and EGRF), is a relatively common feature (TCGA database).

Two additional and closely related concepts are the phenotypic plasticity and stemness observed in melanoma cells. Reasons behind the phenotypic plasticity are largely unknown, although involvement of the melanosomal, lineage-specific transcription factor, MITF, have been suggested (31). In the present model, a low expression of the melanotic lineage-related transcription factor MITF was associated with invasive and stem cell markers and ZEB2 whereas a high MITF expression was associated with differentiation markers, high proliferative capacity and a high ZEB1 expression (32). The transition from general cytotoxic therapies to targeted therapies has led to a change in therapy-associated resistance, from intrinsic resistance to acquired resistance. However, whether the recent advances in immunotherapy are useful in the prevention of acquired therapy resistance remains to be determined.

Conclusions

The abovementioned information and citations indicate that the scientific community is making concerted efforts to combat this pathological state by exploiting genetic and epigenetic approaches. However, considerable research remains to be conducted with regard to the development of improved, highly specific treatment strategy against this lethal disease.

References

1 

Garg K, Maurer M, Griss J, Brüggen MC, Wolf IH, Wagner C, Willi N, Mertz KD and Wagner SN: Tumor associated B cells in cutaneous primary melanoma and improved clinical outcome. Hum Pathol. Apr 20–2016.pii: S0046-8177(16)30043-0. doi: 10.1016/j.humpath.2016.03.022 (Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

2 

Naldi L, Altieri A, Imberti GL, Giordano L, Gallus S and La Vecchia C: Cutaneous malignant melanoma in women. Phenotypic characteristics, sun exposure, and hormonal factors: A case-control study from Italy. Ann Epidemiol. 15:545–550. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Shi CH, Wang H, Mao CY, Yang J, Song B, Liu YT, Yang ZH, Luo HY, Zhang SY, Wu J, et al: MC1R variants in Chinese Han patients with sporadic Parkinsons disease. Neurobiol Aging. 42:217.e5–217.e6. 2016. View Article : Google Scholar

4 

Pasquali E, García-Borrón JC, Fargnoli MC, Gandini S, Maisonneuve P, Bagnardi V, Specchia C, Liu F, Kayser M, Nijsten T, et al: M-SKIP Study Group: MC1R variants increased the risk of sporadic cutaneous melanoma in darker-pigmented Caucasians: a pooled-analysis from the M-SKIP project. Int J Cancer. 136:618–631. 2015.PubMed/NCBI

5 

Aoude LG, Wadt KA, Pritchard AL and Hayward NK: Genetics of familial melanoma: 20 years after CDKN2A. Pigment Cell Melanoma Res. 28:148–160. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Gibney GT, Messina JL, Fedorenko IV, Sondak VK and Smalley KS: Paradoxical oncogenesis - the long-term effects of BRAF inhibition in melanoma. Nat Rev Clin Oncol. 10:390–399. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Prickett TD, Wei X, Cardenas-Navia I, Teer JK, Lin JC, Walia V, Gartner J, Jiang J, Cherukuri PF, Molinolo A, et al: Exon capture analysis of G protein-coupled receptors identifies activating mutations in GRM3 in melanoma. Nat Genet. 43:1119–1126. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Shoushtari AN and Carvajal RD: GNAQ and GNA11 mutations in uveal melanoma. Melanoma Res. 24:525–534. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Geertsen R, Hofbauer G, Kamarashev J, Yue FY and Dummer R: Immune escape mechanisms in malignant melanoma. Int J Mol Med. 3:49–57. 1999.PubMed/NCBI

10 

Serrone L and Hersey P: The chemoresistance of human malignant melanoma: An update. Melanoma Res. 9:51–58. 1999. View Article : Google Scholar : PubMed/NCBI

11 

Luan J, Shattuck-Brandt R, Haghnegahdar H, Owen JD, Strieter R, Burdick M, Nirodi C, Beauchamp D, Johnson KN and Richmond A: Mechanism and biological significance of constitutive expression of MGSA/GRO chemokines in malignant melanoma tumor progression. J Leukoc Biol. 62:588–597. 1997.PubMed/NCBI

12 

Hussussian CJ, Struewing JP, Goldstein AM, Higgins PA, Ally DS, Sheahan MD, Clark WH Jr, Tucker MA and Dracopoli NC: Germline p16 mutations in familial melanoma. Nat Genet. 8:15–21. 1994. View Article : Google Scholar : PubMed/NCBI

13 

Ha L, Ichikawa T, Anver M, Dickins R, Lowe S, Sharpless NE, Krimpenfort P, Depinho RA, Bennett DC, Sviderskaya EV, et al: ARF functions as a melanoma tumor suppressor by inducing p53-independent senescence. Proc Natl Acad Sci USA. 104:10968–10973. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Busch C, Geisler J, Knappskog S, Lillehaug JR and Lønning PE: Alterations in the p53 pathway and p16INK4a expression predict overall survival in metastatic melanoma patients treated with dacarbazine. J Invest Dermatol. 130:2514–2516. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Howell PM Jr, Liu S, Ren S, Behlen C, Fodstad O and Riker AI: Epigenetics in human melanoma. Cancer Control. 16:200–218. 2009.PubMed/NCBI

16 

Klose RJ and Bird AP: Genomic DNA methylation: the mark and its mediators. Trends Biochem Sci. 31:89–97. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Straume O, Smeds J, Kumar R, Hemminki K and Akslen LA: Significant impact of promoter hypermethylation and the 540 C>T polymorphism of CDKN2A in cutaneous melanoma of the vertical growth phase. Am J Pathol. 161:229–237. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Spugnardi M, Tommasi S, Dammann R, Pfeifer GP and Hoon DS: Epigenetic inactivation of RAS association domain family protein 1 (RASSF1A) in malignant cutaneous melanoma. Cancer Res. 63:1639–1643. 2003.PubMed/NCBI

19 

Mirmohammadsadegh A, Marini A, Nambiar S, Hassan M, Tannapfel A, Ruzicka T and Hengge UR: Epigenetic silencing of the PTEN gene in melanoma. Cancer Res. 66:6546–6552. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Miura S, Maesawa C, Shibazaki M, Yasuhira S, Kasai S, Tsunoda K, Maeda F, Takahashi K, Akasaka T and Masuda T: Immunohistochemistry for histone h3 lysine 9 methyltransferase and demethylase proteins in human melanomas. Am J Dermatopathol. 36:211–216. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Tiffen J, Gallagher SJ and Hersey P: EZH2: an emerging role in melanoma biology and strategies for targeted therapy. Pigment Cell Melanoma Res. 28:21–30. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Kappelmann M, Kuphal S, Meister G, Vardimon L and Bosserhoff AK: MicroRNA miR-125b controls melanoma progression by direct regulation of c-Jun protein expression. Oncogene. 32:2984–2991. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Zhang J, Na S, Liu C, Pan S, Cai J and Qiu J: MicroRNA-125b suppresses the epithelial-mesenchymal transition and cell invasion by targeting ITGA9 in melanoma. Tumour Biol. 37:5941–5949. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Rigel DS and Carucci JA: Malignant melanoma: Prevention, early detection, and treatment in the 21st century. CA Cancer J Clin. 50:215–236, 237–240. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Han D, Zager JS, Shyr Y, Chen H, Berry LD, Iyengar S, Djulbegovic M, Weber JL, Marzban SS, Sondak VK, et al: Clinicopathologic predictors of sentinel lymph node metastasis in thin melanoma. J Clin Oncol. 31:4387–4393. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Khalil DN and Carvajal RD: Treatments for noncutaneous melanoma. Hematol Oncol Clin North Am. 28:507–521. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Russo A, Ficili B, Candido S, Pezzino FM, Guarneri C, Biondi A, Travali S, McCubrey JA, Spandidos DA and Libra M: Emerging targeted therapies for melanoma treatment (review). Int J Oncol. 45:516–524. 2014.PubMed/NCBI

28 

Russo AE, Torrisi E, Bevelacqua Y, Perrotta R, Libra M, McCubrey JA, Spandidos DA, Stivala F and Malaponte G: Melanoma: molecular pathogenesis and emerging target therapies (Review). Int J Oncol. 34:1481–1489. 2009.PubMed/NCBI

29 

Garbe C, Eigentler TK, Keilholz U, Hauschild A and Kirkwood JM: Systematic review of medical treatment in melanoma: current status and future prospects. Oncologist. 16:5–24. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Xin Y, Huang Q, Zhang P, Yang M, Hou XY, Tang JQ, Zhang LZ and Jiang G: Meta-analysis of the safety and efficacy of interferon combined with dacarbazine versus dacarbazine alone in cutaneous malignant melanoma. Medicine (Baltimore). 95:e34062016. View Article : Google Scholar : PubMed/NCBI

31 

Hartman ML and Czyz M: MITF in melanoma: Mechanisms behind its expression and activity. Cell Mol Life Sci. 72:1249–1260. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Vandamme N and Berx G: Melanoma cells revive an embryonic transcriptional network to dictate phenotypic heterogeneity. Front Oncol. 4:3522014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 12 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang X and Zhang X: Genetics and epigenetics of melanoma (Review) Retraction in /10.3892/ol.2020.12045. Oncol Lett 12: 3041-3044, 2016
APA
Zhang, X., & Zhang, X. (2016). Genetics and epigenetics of melanoma (Review) Retraction in /10.3892/ol.2020.12045. Oncology Letters, 12, 3041-3044. https://doi.org/10.3892/ol.2016.5093
MLA
Zhang, X., Zhang, P."Genetics and epigenetics of melanoma (Review) Retraction in /10.3892/ol.2020.12045". Oncology Letters 12.5 (2016): 3041-3044.
Chicago
Zhang, X., Zhang, P."Genetics and epigenetics of melanoma (Review) Retraction in /10.3892/ol.2020.12045". Oncology Letters 12, no. 5 (2016): 3041-3044. https://doi.org/10.3892/ol.2016.5093