Open access

An Overview of Important Genetic Aspects in Melanoma

Written By

Rohinton S. Tarapore

Submitted: 19 April 2012 Published: 17 April 2013

DOI: 10.5772/55257

From the Edited Volume

Highlights in Skin Cancer

Edited by Pierre Vereecken

Chapter metrics overview

1,758 Chapter Downloads

View Full Metrics

1. Introduction

Cancer of the skin is the most common form of malignancy in humans and is divided into two categories – non-malignant skin cancer and cutaneous melanoma. Non-melanoma skin cancer (basal cell and small cell carcinoma) make up a vast majority of skin cancers. According to data from National Cancer Institute (NCI) in 2012, more than 2 million new cases of non-melanomas will be identified with less than a 1000 deaths. Despite according for only 4% of all cases, melanoma is the deadliest of skin cancers resulting in over 79% of skin cancer deaths [1]. In the United States, melanoma is the fifth most common cancer in men and the sixth most common in women. In 2011, 70,230 new melanoma cases were identified with 8,790 deaths. The median age of diagnosis is between 45-55; although 25% of melanomas occur in individuals over 40 years.

Advertisement

2. Types of skin cancer

  1. Basal Cell Carcinoma (BCC): This is the most common form of skin cancer and accounts for more than 90% of all skin cancers in the United States. BCC causes damage by growing and invading the surrounding tissue and usually does not metastasize to other parts of the body. Intermittent sun exposure (especially early in life), age and light colored skin are important factors in the development of BCC. Approximately a fifth of BCCs, develop in regions that are not sun-exposed such as chest, arms, neck, back and scalp [2]. Weakening of the immune system on account of the disease or immune-suppressive drugs is known to promote the risk of developing BCCs. Usually BCC begins as a small, dome-shaped bump and is covered by small superficial blood vessels called telangiectases and its texture is often shiny and translucent. Hereditary predisposition to BCC [3,4] occurs among individuals with albinism and Xeroderma Pigmentosum. These disorders can be linked to either instability of the skin or diminished pigmentation.

  2. Squamous Cell Carcinoma (SCC): This cancer begins in the squamous cells that form the surface of the skin, lining of hollow organs of the respiratory and digestive tracts. The earliest form of SCC is called as actinic keratosis (AK) [2] that appear as rough, red bumps on the scalp, face, ears and back of the hands. The rate at which the bumps (keratosis) invade deeper in the skin to become fully developed squamous cell carcinoma is estimated to be 10-20% over a 10 year period. Actinic keratinosis that becomes thicker and more tender could increase the possibility of getting transformed to an invasive squamous cell carcinoma phenotype. The most important risk factor is sun exposure. Lesions appear after years of sun damage in the forehead, cheeks as well as the backs of hands. Other minor factors like exposure to hydrocarbons, arsenic, heat or X-rays could predispose to SCCs. Unlike BCC, SCCs can metastasize to other parts but are easy to treat.

  3. Melanoma: This is the cancer of the melanocytes, the “skin-color producing cells” of the body. An estimated 132,000 new cases of melanoma occur worldwide every year [5,6,7] with approximately 65,161 deaths according to estimates from the World Health Organization (WHO). The high mortality rate of melanoma is remarkably high considering the fact that melanoma is nearly always curable in its early stages; the high mortality rate can be attributed to late diagnosis in which the cancer spreads to other parts of the body [5]. Melanoma incidence has increased more rapidly than that of any other cancer, yet our ability to treat disseminated disease has been lagging [8,9,10]. The predicted 1 year survival for Stage IV melanoma ranges between 41% to 59% [11]. At a very early point in the progression of melanoma, the cancer gains metastatic potential.

Advertisement

3. Risk factors

There are multiple risk factors that contribute to escalating incidence of melanomas in humans (Table 1). Ultraviolet (UV) radiation especially UVA (315-400 nm) and UVB (280-315 nm) from sunlight is an important contributing factor for melanoma progression. A study by Glanz et al [5,12] revealed that 90% of all melanomas are attributed to exposure to ultraviolet radiation.

The damaging effects of UV radiation (UVR) is on account of direct cellular damage and alterations in immunologic functions. UVR causes DNA damage (by formation of pyrimidine dimers), gene mutations, oxidative stress, immunosuppressive and inflammatory responses. All these effects play an important role in photoaging of the skin and predispose to skin cancer [13]. UVR creates mutations in p53, a key tumor suppressor gene that plays an important role in DNA repair and apoptosis. Thus if p53 is mutated, the cells lose the DNA repair process leading to the deregulation of apoptosis, expression of mutated keratinocytes and initiation of skin cancer [13,14]. Darker skinned individuals have lower incidence of cutaneous melanoma primarily as a result of increased epidermal melanin. Studies indicate that epidermal melanin in African-American individuals filters twice as much UVB radiation than in Caucasians. This is on account of the larger, more melanized melanosomes located in the epidermis of dark skin individuals that absorb and scatter more light energy than the smaller, less melanized melanosomes of white skin. The incidence rate of skin cancer (both melanoma and non-melanoma) has increased significantly in the last decade [15]; particularly among young women. For most individuals, exposure to UVR from the sun is the main source of skin cancer. Nonetheless, some individuals are exposed to high UV doses through artificial sources – sunbeds and sunlamps used for tanning purposes. Indoor tanning is widespread in most developed countries in Northern Europe, Australia and the United States [16]. Intense early sunburns and blistering sunburns are closely associated with melanoma development [17,18,19]. Statistics indicate that one severe childhood sunburn is associated with a two-fold increase in melanoma risk [20]. Chronic UV exposure results in increased skin aging, wrinkles, uneven skin pigmentation, loss of elasticity and a distribution in the skin barrier function [21]. Chronic UVR exposure is an important risk factor in the development of actinic keratosis (precursor of SCCs).

Ultraviolet (UV radiation)
UVA
UVB
Genetic syndromes
Xeroderma pigmentosum
Oculocutaneous albinism
Basal cell nevus syndrome
Ionizing radiation
X-rays
Other risk factors
Artificial UV radiation (tanning)
Smoking
Color of skin (having fair skin, especially with blue or hazel eyes)
History of precursor lesions
Chronically injured or non-healing wounds
Working outdoors
Increasing age

Table 1.

Risk factors for skin cancer [1-8]

Candidate Gene Protein Location on chromosome Stage of melanoma References
Gene losses
PTEN
Pten 10q Primary & metastatic [135,136,137]
CDKN2A p16Ink4a/p14Arf 9p Primary & metastatic [135,136]
ITGB3BP Beta-3-endonexin 1p31 Uveal [138]
Gene amplification
BRAF
Braf 7q21.3 Cutaneous [135]
CCND1 cyclinD1 11q13 Acral [136]
CDK4 cdk4 12q14 Acral & mucosal [136]
CDH2 N-cadherin 18q Uveal & metastatic [137,139]
c-MYC c-myc 8q23 Ocular, primary & metastatic [135,137,140,141]
MITF Mitf 3p14 Cutaneous, metastatic [142,143]

Table 2.

Chromosomal aberrations involving important genes found in melanoma

Advertisement

4. Roadway to melanoma

Malignant melanomas arise from epidermal melanocytes or the melanocyte precursor cell which are derived from the neural crest and migrate to the skin and hair follicles [22]. Melanoma initiation and progression is accompanied by a series of histological changes. The five distinct changes are: 1) nevus – benign lesion characterized by an increased number of nested melanocytes; 2) dysplastic nevus – which is characterized by random, discontinuous and atypical melanocytes; 3) radial-growth phase (RGP) melanoma where the cells acquire the ability to proliferate intraepidermally; 4) vertical growth phase (VGP) melanoma – characterized by melanoma cells acquiring the ability to penetrate through the basement membrane (BM) into underlying dermis and subcutaneous tissue; and 5) metastatic melanoma – characterized by the spread of melanoma cells to other areas of the skin and other organs. The most critical event in melanoma progression is the RGP-VGP transition which involves the escape from keratinocyte mediated growth control. This is consistent with tumor thickness being a strong predictor of metastatic disease and adverse clinical outcome [23].

Gene Protein Function Commentary References
Up-regulation
ATM
ATM kinase Cell cycle control, DNA damage response Melanocytic infiltration [135,136,137]
CDH2 N-cadherin Cell-cell adhesion Melanoma invasiveness [144,145]
MMP-1, 2 Metallo proteinases -1, 2 Degradation of ECM Tumor progression [146]
SPP1 Osteopontin Inductor of MMP Risk of metastasis [144,147]
SPARC Osteonectin Angiogenesis Tumor progression [144]
WNT5A Wnt5a Cell signaling Melanoma invasiveness [125,148]
CKS2 Cdc28/cdk1 protein kinase subunit 2 Cell cycle control Poor prognosis [147]
EIF2gamma EIF2γ DNA translation Tumor progression [149]
PCNA PCNA Cofactor DNA polymerase Genome destabilization [150]
Down Regulation
CDH1
E-cadherin Cell-cell adhesion Tumor progression [144,145,147]
CDH3 P-cadherin Cell-cell adhesion Tumor progression [145,151]
CDH10 Cadherin-10 Cell-cell adhesion High risk of metastasis [144]
DSC1 Desmocollin-1 Desmosomal component Loss of cell adhesion [137,139,179]

Table 3.

Genetic expression signatures associated with the progression of melanomas [24]

Acquisition of somatic mutations in key regulatory genes is the driving force behind the initiation and progression of melanoma development. For the past few decades numerous research teams around the world have researched on melanoma genetics leading to an overwhelming body of information.

Advertisement

5. Susceptibility genes

Approximately 8-12% of all melanomas are familial – occurring in individuals with a history of familial melanoma [24]. Two genes have been found to be associated with high penetrance susceptibility – CDKN2A and CDK4. Using linkage analysis of families with high melanoma incidences, the first melanoma incidence susceptibility gene, CDK2N2A was identified at chromosome 9p21 [25,26]. The gene CDKN2A encodes two unrelated proteins – p16Ink4A and p14Arf. These proteins are tumor suppressors involved in cell cycle regulation. Numerous studies indicate that p16Ink4A inhibits G1 cyclin dependent kinase (cdk4/cdkb) mediated phosphorylation of retinoblastoma protein (pRB) resulting in cell cycle progression arrest through G1-S; while p14 favors apoptosis and blocks oncogenic transformation by stabilizing p53 levels through the inhibition of Mdm2-mediated p53 ubiquitination [27,28,29,30]. Loss of p16 promotes hyper-phosphorylation of pRb resulting in its inactivation while the loss of p14 inactivates p53 — leading to unrestricted cell cycle progression. Germline mutations in CDKN2A have been found in 40% of families with 3 or more family members affected by melanoma [31]. Not all individuals carrying germline CDKN2A mutations develop mutations. Individuals with large numbers of pigment lesions or nevi have familial atypical mole-melanoma syndrome (FAMS) are associated with increased risk to developing melanoma [32,33].

The other melanoma susceptibility gene, CDK4 is located at chromosome 12q14 [34,35,36]. Mutations in CDK4 abrogate binding of cdk4 to p16 have been associated with melanoma pathogenesis [32]. This is evidence that links the entire p16Ink4A-cdk4/cdk6-pRb pathway to melanoma indicating that hereditary retinoblastoma patients with germline inactivation of retinoblastoma (Rb1) are predisposed to melanoma [37,38].

Advertisement

6. Acquired genetic alterations in melanoma

Understanding the regulating pathways involved in melanoma development and progression has advanced significantly in recent years. The discovery of genetic alterations that aids in the formation of various cancers has aided in the development of numerous molecularly targeted therapies for individuals with metastatic disease [39,40,41]. These genes are known to be key molecular driver in melanoma; >70% cases harbor activating mutations in these genes. The molecule that is most commonly found to be mutated in melanomas is BRAF (~50% of all cancers) followed by NRAS (20%) and c-kit (1%) [42,43,44]. Melanoma is the result of complex changes in multiple signaling pathways affecting growth, cell mobility, metabolism and the ability to escape cell death progression. The Ras-Raf-Mek-Erk pathway followed by PI3K/Akt pathway is constitutively activated in a significant number of melanoma tumors.

Advertisement

7. The Ras-Raf-Mek-Erk

In 2002, a breakthrough study found that Braf to be mutated in a large percentage of melanomas – triggering new studies that focus on MAPK (mitogen activated protein kinase) signaling in melanomas. Braf is mutated in upto 82% of cutaneous nevi [45,46], 66% of primary melanomas [44] and 40-68% of metastatic melanomas [47,48]. A specific mutation substitution of valine with glutamic acid at residue 600 (BRAF V600E), account for 90%+ BRAF mutation. Raf, a downstream effector of RAS is a serine-threonine specific protein kinase that activates Mek, which inturn activates Erk. Humans have 3 Raf genes: A-raf, B-raf and C-raf. The occurrence of mutation in Nras or Braf is 80-90% of all melanomas suggests that constitutive activation of extracellular signal regulated protein kinase (Ras-Raf-Mek-Erk). Most Ras mutations are present in codon 61 of N-Ras with K-Ras and H-Ras mutations being relatively rare [49,50]. Constitutive activation of Ras-Raf-Mek-Erk cascade has been shown to contribute to tumorigenesis by inhibiting apoptosis and increasing cell proliferation, tumor invasion and metastasis. Activated Erk plays a pivotal role in cell proliferation by controlling the G1- to S-phase transition by negative regulation of p27 inhibition and upregulation of c-myc activity [51,52]. Inhibition of Erk activity is associated with G1 cell cycle arrest by upregulation of p21 and reduced phosphorylation [52]. Activated Erk is also known to stimulate cell proliferation by increasing the transcription and stability of c-Jun which is mediated by CREB (cyclic adenosine monophosphate responsive element-binding) and Gsk-3β (glycogen synthase kinase-3beta) respectively [53]. Erk is also believed to increase proliferation by inhibiting differentiation. Constitutively active Erk limits differentiation in melanoma by targeting MITF (microphthalmia-associated transcription factor) for degradation [54,55,56]. The activated Erk pathway enhances melanoma specific survival by differentially regulating RSK-mediated phosphorylation and inactivation of the pro-apoptotic protein Bad [57] and inhibiting Jak-Stat pathway [58].

Erk signaling also contributes towards tumor invasion and metastasis by regulating the expression of integrin and matrix metalloproteinases (MMPs). Activated Ras-Mek-Erk pathway drives the production of MMP1 [59,60,61].

Advertisement

8. The PI3K/Akt pathway

The PI3K/Akt pathway is activated in various cancers, mostly on accont of mutations in tumor suppressor PTEN (phosphatase and tensin homolog) [62]. In melanoma, los of PTEN on chromosome 10q 23-24 was first reported by Parmiter et al [63]. The PTEN gene encodes a phosphatase that degrades products of PI3K by dephosphorylating phosphatidylinositol 3,4,5-triphosphate and phosphatidylinositol 3,4-biphosphate at 3 positions [64]. Loss of PTEN increases AKT phosphorylation and activity leading to increased mitogenic signaling and decreased apoptosis [65]. Various studies suggest that 30-40% of melanoma cell lines and 5-15% of uncultured melanoma specimens carry inactivating mutations or homozygous deletions of Pten [63,66,67].

Pten encodes a negative regulator of extracellular growth signals that are transcended via PI3K-Akt pathway. Akt/protein kinase B (PKB), a serine-threonine kinase, is a core component of the PI3K signaling cascade and is activated through the phosphorylation of Ser 473/474 and Thr 308/309 [68,69]. Activated Akt regulates a network of factors that control cell proliferation and survival and this pathway is hyperactive in most metastatic melanomas [70,71,72]. Akt activates the transcription of a wide variety of genes involved in a wide range of cellular activities – those involved in immune activation, cell proliferation, apoptosis and cell survival [69]. Several studies have documented Akt activation in melanoma. Dai et al undertook a 292 sample study of pAkt levels using tissue microarray & immunohistochemistry strategies and identified strong pAkt expression in 17%, 43%, 49% and 77% of the biopsies in normal nevi, dysplastic nevi, primary melanoma and melanoma metastasis respectively. An important cell adhesion protein MelCAM that plays critical roles in melanoma development was increased upon active Akt expression [73,74]. PI3K and Akt is known to increase the expression of MMP2 and MMP9 by a mechanism involving Akt activation of NF-kappaB binding to the MMP promoter [75,76]. Akt overexpression led to upregulation of VEGF, increased production of superoxide ROS. Akt can suppress apoptosis by phosphorylating and inactivating many proapoptotic proteins like caspase 9 and Bad [77,78]. PI3K pathway emerges as the central axis that is deregulated in melanoma and along with constitutively active MAPK pathway makes an important role in melanoma development progression. Thus targeting PI3K is expected to be an important therapeutic target modality for melanoma treatment.

Advertisement

9. Wnt/β-catenin pathway

Βeta-catenin (β-catenin) is a key component of the Wnt signaling pathway. Signaling through this pathway controls a wide range of cellular functions and aberrant Wnt/β-catenin signaling can lead to cancer development and progression [79]. Wnts are glycoproteins that act as ligands to stimulate receptor-mediated signal transduction pathways involved in cell survival, proliferation, behavior and fate. Wnt proteins are known to activate 3 different extracellular pathways – Wnt/β-catenin, Wnt/planar-polarity and Wnt/Ca2+ pathways [80]. The Wnt/β-catenin also known as the canonical Wnt pathway plays an important role in melanoma development. In the absence of Wnt ligands, free β-catenin binds to the destructive complex of Axin, adenomatous polyposis coli (APC) and glycogen synthase kinase-3β (GSK-3β). GSK-3β mediates the phosphorylation of β-catenin at specific regulatory sites on the N-terminal side marking β-catenin for ubiquitination and subsequent proteosomal degradation. Upon the binding of Wnt ligand, GSK--catenin for ubiquitination and subsequent proteosomal degradation. Upon the binding of Wnt ligand, GSK-3β activity is inhibited resulting in accumulation of β-catenin in the cytoplasm and shuttles into the nucleus where it serves as an essential co-activator of the Tcf/Lef (T-cell factor / lymphoid enhancer factor) family [81]. Numerous genes implicated in the tumorigenic process like c-myc and cyclinD1 have been identified as targets of the canonical Wnt signaling.

Increased nuclear localization of β-catenin – an important indication of activated Wnt signaling pathway is observed in over a third of melanoma specimens [82,83,84]. Mutations in β-catenin have been observed in about 23% of melanoma cancer cell lines and these mutations affect phosphorylation sites at Ser33, Ser37, Thr41 and Ser45 [85] at the N-terminal domain. These mutations render β-catenin resistant to phosphorylation and subsequent degradation. Low rates of β-catenin mutation have been observed in primary melanomas and metastasis indicating that activating mutations is a rare event in melanoma tumorigenesis [82,83,84,86,87,88]. Mutations in APC were observed sporadically in primary melanomas [82,85,88]. While APC promoter 1A hypermethylation was observed in 17% of melanoma biopsies and 13% of melanoma cell lines. Wnt signaling pathways is activated in tumors through aberration in other genes. ICAT (inhibition of β-catenin & T-cell factor), a gene that negatively regulates the association of β-catenin with TCF4 thus repressing the transactivation of β-catenin-Tcf4 target genes [89]. A study by Reifenberger J et al suggests that loss of ICAT expression may contribute to the progression of melanoma [86]. ICAT mRNA expression analysis in two-third melanoma specimens revealed a 20% or less decrease in ICAT transcription [86]. However the mechanism behind the reduced ICAT mRNA level in melanoma is unclear.

Identification of Wnt target genes is also important towards the study of melanoma progression. Brn1, the POU domain transcription factor is directly controlled by Wnt signaling in transgenic mouse models and melanoma cell lines [90]. Studies indicate that overexpression of Brn2 is associated with increased melanoma progression and tumorigenicity [90,91].MITF (microphthalmia-associated transcription factor), a Wnt target gene, is essential for the development of the melanocyte lineage and has an important role in the control of cell proliferation, survival and differentiation [54,92,93]. The regulation of MITF expression by β-catenin significantly influences the growth and survival behavior of treatment resistant melanoma [94]. A study by Schepsky A et al demonstrated that MITF can directly interact with β-catenin and redirect transcriptional activity away from canonical Wnt signaling-regulated target gene specific for MITF [95]. Induction of Wnt signaling can be blocked by 5 different proteins – Dkk, Wise, sFrp (secreted Frizzled related protein), Wif (wnt inhibitory factor), and Cerebrus that compete for the Wnt ligand or the Lrz-Frp receptor [96]. Interestingly, Dkk1 (Dickkopf 1) expression is negligible in melanomas [97]. Studies by Kuphal et al have demonstrated a downregulated or loss of Dkk-1, -2 and-3 in all melanoma cell lines and most of the melanoma tumor samples that were analyzed [98]. In xenograft mouse model, overexpression of Dkk-1 and Wif-1inhibited melanoma tissue growth [99,100].

Advertisement

10. The JNK/c-Jun pathway

Activation of Jnks is usually in response to diverse stresses. These kinases play an important role in the regulation of cell proliferation, cell survival, cell death, DNA repair and metabolism. A variety of extracellular stimuli by cytokines, growth factors, hormones, UV radiation and tumor promoters are known to activate Jnks [101]. Sequential protein phorphorylation through a MAP-kinase module (MAP3K-MAP2K-MAPK) is responsible for Jnk activation [102]. Depending upon the cellular context, Jnk has been shown to elicit both positive and negative effects on tumor development [103]. Activation of Jnk is required for Ras-mediated transformation and mediate proliferation and tumor growth [104,105]. These observations are consistent with the findings of constitutively active Jnk in tumor samples and cell lines [103,106]. Jnk mediated the phosphorylation at serine 63 & 73 residues enhancing the ability of transcription factor c-Jun, a component of the AP-1 transcription complex [107]. The activation of Jnk leas to the induction of AP-1 dependent target genes that play important roles in cell proliferation, cell death and inflammation. Other members of the AP-1 transcription complex include c-Jun, Jun B, Jun D, c-Fos, Fra1 and Fra 2. The role of Jnk in oncogenesis is emerging; however c-Jun is a well defined oncogene in cancer. c-Jun is amplified and overexpressed in undifferentiated and aggressive sarcomas [108], breast and lung cancer [109,110]. Since the 1990s, the role of Jnk pathway in melanomas was recognized [111,112]. c-Jun, Jun B, c-fos genes play a role in the transformation of melanocytes into malignant melanomas [111].

The possible role Jnk pathway has led research teams to study the clinical relevance of interfering with this pathway. siRNA or chemical inhibitors of Jnk signaling inhibited proliferation in breast and non-small cell lung cancer (NSCLC) [106,113] and also induced apoptosis in prostate cancer cells [114]. A study by Gurzov E et al demonstrated that knockdown of c-Jun and Jun B in B16F10 melanoma cells by siRNA resulted in increased cell cycle arrest and apoptosis also resulting in extended survival of mice inoculated with these modified melanoma cells[115], suggesting that inactivation of c-Jun and Jun B could provide a valuable strategy for antitumor intervention [115].

11. The NFκB (NF-kappaB) pathway

The NFκB family in mammals contains 5 members – p105/p50 (NFκB1), p100/p52 (NFκB2), RelA (p65), RelB and c-Rel (J-206, 207). The canonical activation of NFκB pathway involves TNFα stimulation resulting in the subsequent phosphorylation/activation of IKK (IκB kinase). In turn, IKK-mediated phosphorylation of IκB leading to ubiquitination of IκB and its proteosomal degradation, releasing the NFκB complex which activates a host of target genes [116,117]. The type of genes that get trans-activated depends on the composition of activated NFκB complex. For instance, complexes containing c-Rel activates pro-apoptotic genes (Dr4/Dr5, Bcl-x) and inhibits anti-apoptotic genes (cellular inhibitor of apoptosis (cIAP1, cIAP2), survivin). Complexes containing RelA inhibits the expression of DR4/DR5 and upregulates caspase 8, cIAP1 and cIAP2 [118].

NFκB is activated in various tumors including melanomas and distinct mechanisms have been proposed for the elevated levels of NFκB activity in melanomas. Activation of NFκB in melanomas is also linked to the loss of E-cadherin, a frequent event in melanoma transformation [119]. NIK (NFκB interacting kinase), an activator of IKK is overexpressed in melanoma cells while compared to normal cells. The major contribution of NFκB in melanoma development and progression relates to its function as an important regulator of survival and apoptosis. A study by Meyskens at al demonstrated that in metastatic melanoma cells, an increase in DNA binding activity of NFκB is associated with an increased expression of p50 and RelA resulting in increased expression of anti-apoptotic regulators. Also the expression of c-Rel, the transcriptional activator of pro-apoptotic genes is markedly in melanoma cells compared with normal melanocytes [120]. Strong p50 nuclear staining also correlated with poor prognosis in melanoma patients [121]. Besides eliciting anti-apoptotic activities NFκB mediates the transcription of MMP2 and MMP9 [121,122]. Overexpression of MMPs is associated with tumor invasion, metastasis and angiogenesis.

12. Melanoma stem cells

Stem cells are cells that can self-renew and the ability to differentiate into various cell lineages. These cells are located in the restrictive niche (environment). The interaction between stem cells and their microenvironment is important for the self renewal process. These cells are highly clonogenic and slow cycling (quiescent) in response to proliferation and survival stimuli. Stem cells divide asymmetrically giving rise to a daughter cell that remains a stem cell (capable of self renewal) and another daughter cell that can rapidly divide and differentiate. Melanocytes that are found in the skin and in the choroid layer of the eye is derived from the neural crest (NC). Neural crest cells undergo EMT to migrate along the definite pathways in the embryo. NC cells give rise to a large array of differentiated cells – melanocytes, peripheral neurons & glia, endocrine and cartilage cells [123]. Melanoblasts which are melanocytic precursors – unpigmented cells with the potential to produce melanin, invade the skin areas and differentiate into melanocytes.

The cancer stem theory suggests that cancer originates from a small subpopulation of neoplastic stem cells that have the potential to self renew and are primarily responsible for sustaining the tumor and giving rise to progressively differentiating cells that proliferate rapidly and contribute to the cellular heterogeneity of the tumor (F-194). Cancer stem cells arise either from undifferentiated stem cells or from cells that possess stem cell like characteristics. Evidence suggests that aggressive melanoma cells acquire characteristics of embryonic stem cells having a multipotent plastic phenotype [124]. Studies by Bittner MP et al demonstrated that melanoma cells express genes associated with different cell types like endothelial, epithelial, fibroblastic, neuronal, hematopoietic and progenitor cells [125]. Strangely genes specific for melanocytes are downregulated in metastatic melanomas. Tyrosinase & MLANA (melan A), genes associated with pigmentation are greatly downregulated in aggressive melanomas [124]. Aggressive melanoma cells express endothelial-associated genes and form extravascular fluid-conducting networks which allow melanomas to greatly adapt to the hypoxic microenvironment of rapidly proliferating tumors, a phenomenon called as “vascular mimicry” [124,126]. From different melanoma cell lines, cells with stem cell-like features which have the ability to grow as non-adherent cell aggregates known as spheroids/spheres have been isolated (F-196). These cells have the ability to differentiate into various lineages – adipogenic, osteogenic, chondrogenic and melanogenic. A study by Bittner M et al demonstrated a subset of these spheroid cells express the cell surface marker CD20, a unique molecular signature of aggressive melanomas [125]. For the treatment of non-Hodkin’s lymphoma, CD20 is a standard therapeutic target which raises the possibility that CD20 could be used as a potential target for melanoma treatment [127].

Several studies have demonstrated that aggressive melanoma cells share characteristics with embryonic progenitors. Evidence suggests a major role for stromal components in all stages of tumorigenesis (initiation, progression and metastasis) [128]. Noted scientist Stephen Paget had coined the term “seed & soil” hypothesis predicting that metastatic cells only colonize soils (organs) that are permissive to their growth[129,130]. Studies show embryonic microenvironment has the capacity to reverse the metastatic phenotype of cancer cells. The microenvironment of human embryonic stem cells reprograms aggressive melanoma cells towards a less aggressive phenotype [124]. Nodal, an embryonic morphogen of the TGFβ family is important for sustaining melanoma aggressiveness and plasticity. Nodal is regained in highly aggressive melanoma cell lines, invasive VGP (vertical growth phase)-stage melanoma and metastatic melanoma [131], implicating Nodal as a novel diagnostic marker in melanoma progression and could be a therapeutic target for metastatic melanoma treatment [124].

13. Conclusion

Our understanding of melanoma development and progression has evolved tremendously over the past three decades. Unfortunately our understanding of the molecular biology of melanoma is still far from complete despite extensive research and knowledge gained in chromosomal alterations, mutations in important melanoma-associated genes, epigenetic modifications and melanoma microenvironment. Even to this day, the best prognostic significance of primary melanoma is the thickness of the tumor (i.e. RGP → VGP transition) and the presence/absence of ulcerations. Melanoma still remains as a tumor that is refractory to current chemotherapeutic treatments. A further study of the interaction between various signaling pathways will help researchers decipher the complexity of the genetic and epigenetic changes which eventually would lead to better therapeutic modalities for the treatment of primary and metastatic melanomas.

Glossary

AK - actinic keratosis

BCC - basal cell carcinoma

Cdks - cyclin-dependent kinases

CREB - cyclic adenosine monophosphate responsive element-binding

FAMS - familial atypical mole-melanoma syndrome

GSK3β - glycogen synthase kinase-3beta

IKK - Inhibitor of IκB kinase

MAPK - mitogen activated protein kinase

MITF - microphthalmia-associated transcription factor

MMP - matrix metalloproteinase

NF-κB - nuclear factor kappa-B

NIK - NFκB-interacting kinase

Pten - phosphatase and tensin homolog

Rb - retinoblastoma

RGP - radial growth phase

SCC - Squamous cell carcinoma

Tcf4 - T-cell factor-4

UVR - ultraviolet radiation

VGP - vertical growth phase

WIF1 - Wnt-inhibitory factor 1

References

  1. 1. JemalABrayFCenterM. MFerlayJWardEet al2011Global cancer statistics. CA Cancer J Clin 616990
  2. 2. MarghoobA. A2011Skin cancers and their etiologies. Semin Cutan Med Surg 30: S15
  3. 3. EichbergerTReglGIkramM. SNeillG. WPhilpottM. Pet al2004FOXE1, a new transcriptional target of GLI2 is expressed in human epidermis and basal cell carcinoma. J Invest Dermatol 12211801187
  4. 4. IwasakiJ. KSrivastavaDMoyR. LLinH. JKoubaD. J2012The molecular genetics underlying basal cell carcinoma pathogenesis and links to targeted therapeutics. J Am Acad Dermatol 66: e167178
  5. 5. NarayananD. LSaladiR. NFoxJ. L2010Ultraviolet radiation and skin cancer. Int J Dermatol 49978986
  6. 6. FosterP. JDunnE. AKarlK. ESnirJ. ANyczC. Met al2008Cellular magnetic resonance imaging: in vivo imaging of melanoma cells in lymph nodes of mice. Neoplasia 10207216
  7. 7. MarugameTZhangM. J2010Comparison of time trends in melanoma of skin cancer mortality (1990-2006) between countries based on the WHO mortality database. Jpn J Clin Oncol 40: 710.
  8. 8. KoJ. MVelezN. FTsaoH2010Pathways to melanoma. Semin Cutan Med Surg 29210217
  9. 9. JemalAThunM. JRiesL. AHoweH. LWeirH. Ket al2008Annual report to the nation on the status of cancer, 1975-2005, featuring trends in lung cancer, tobacco use, and tobacco control. J Natl Cancer Inst 10016721694
  10. 10. LinosESwetterS. MCockburnM. GColditzG. AClarkeC. A2009Increasing burden of melanoma in the United States. J Invest Dermatol 12916661674
  11. 11. WargoJ. ATanabeK2009Surgical management of melanoma. Hematol Oncol Clin North Am 23565581x.
  12. 12. GlanzKBullerD. BSaraiyaM2007Reducing ultraviolet radiation exposure among outdoor workers: state of the evidence and recommendations. Environ Health 6: 22.
  13. 13. MeeranS. MPunathilTKatiyarS. K2008IL-12 deficiency exacerbates inflammatory responses in UV-irradiated skin and skin tumors. J Invest Dermatol 12827162727
  14. 14. BenjaminC. LAnanthaswamyH. N200753and the pathogenesis of skin cancer. Toxicol Appl Pharmacol 224: 241-248.
  15. 15. GarbeCLeiterU2009Melanoma epidemiology and trends. Clin Dermatol 2739
  16. 16. GandiniSAutierPBoniolM2011Reviews on sun exposure and artificial light and melanoma. Prog Biophys Mol Biol 107362366
  17. 17. PrestonD. SSternR. S1992Nonmelanoma cancers of the skin. N Engl J Med 32716491662
  18. 18. GilchrestB. AEllerM. SGellerA. CYaarM1999The pathogenesis of melanoma induced by ultraviolet radiation. N Engl J Med 34013411348
  19. 19. GlosterH. MJr., Neal K (2006Skin cancer in skin of color. J Am Acad Dermatol 55741760quiz 761-744.
  20. 20. MaFCollado-mesaFHuSKirsnerR. S2007Skin cancer awareness and sun protection behaviors in white Hispanic and white non-Hispanic high school students in Miami, Florida. Arch Dermatol 143983988
  21. 21. SchroederPHaendelerJKrutmannJ2008The role of near infrared radiation in photoaging of the skin. Exp Gerontol 43629632
  22. 22. BennettD. C1993Genetics, development, and malignancy of melanocytes. Int Rev Cytol 146191260
  23. 23. TsaoHAtkinsM. BSoberA. J2004Management of cutaneous melanoma. N Engl J Med 3519981012
  24. 24. LomasJMartin-duquePPonsMQuintanillaM2008The genetics of malignant melanoma. Front Biosci 1350715093
  25. 25. HussussianC. JStruewingJ. PGoldsteinA. MHigginsP. AAllyD. Set al1994Germline 16mutations in familial melanoma. Nat Genet 8: 15-21.
  26. 26. KambAShattuck-eidensDEelesRLiuQGruisN. Aet al1994Analysis of the 16gene (CDKN2) as a candidate for the chromosome 9p melanoma susceptibility locus. Nat Genet 8: 23-26.
  27. 27. PomerantzJSchreiber-agusNLiegeoisN. JSilvermanAAllandLet al1998The Ink4a tumor suppressor gene product, 19Arfinteracts with MDM2 and neutralizes MDM2’s inhibition of p53. Cell 92: 713-723.
  28. 28. ZhangYXiongYYarbroughW. G1998ARF promotes MDM2 degradation and stabilizes 53ARF-INK4a locus deletion impairs both the Rb and p53 tumor suppression pathways. Cell 92: 725-734.
  29. 29. KamijoTWeberJ. DZambettiGZindyFRousselM. Fet al1998Functional and physical interactions of the ARF tumor suppressor with 53and Mdm2. Proc Natl Acad Sci U S A 95: 8292-8297.
  30. 30. StottF. JBatesSJamesM. CMcconnellB. BStarborgMet al1998The alternative product from the human CDKN2A locus, 14ARF), participates in a regulatory feedback loop with p53 and MDM2. EMBO J 17: 5001-5014.
  31. 31. ChinLGarrawayL. AFisherD. E2006Malignant melanoma: genetics and therapeutics in the genomic era. Genes Dev 2021492182
  32. 32. GandiniSSeraFCattaruzzaM. SPasquiniPAbeniDet al2005Meta-analysis of risk factors for cutaneous melanoma: I. Common and atypical naevi. Eur J Cancer 412844
  33. 33. GandiniSSeraFCattaruzzaM. SPasquiniPPicconiOet al2005Meta-analysis of risk factors for cutaneous melanoma: II. Sun exposure. Eur J Cancer 414560
  34. 34. ZuoLWegerJYangQGoldsteinA. MTuckerM. Aet al1996Germline mutations in the 16INK4abinding domain of CDK4 in familial melanoma. Nat Genet 12: 97-99.
  35. 35. SoufirNAvrilM. FChompretADemenaisFBombledJet al1998Prevalence of 16and CDK4 germline mutations in 48 melanoma-prone families in France. The French Familial Melanoma Study Group. Hum Mol Genet 7: 209-216.
  36. 36. WolfelTHauerMSchneiderJSerranoMWolfelCet al1995A 16INK4aCDK4 mutant targeted by cytolytic T lymphocytes in a human melanoma. Science 269: 1281-1284.
  37. 37. EngCLiF. PAbramsonD. HEllsworthR. MWongF. Let al1993Mortality from second tumors among long-term survivors of retinoblastoma. J Natl Cancer Inst 8511211128
  38. 38. FletcherOEastonDAndersonKGilhamCJayMet al2004Lifetime risks of common cancers among retinoblastoma survivors. J Natl Cancer Inst 96357363
  39. 39. DrukerB. JTalpazMRestaD. JPengBBuchdungerEet al2001Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med 34410311037
  40. 40. DemetriG. DVon MehrenMBlankeC. DVan den Abbeele AD, Eisenberg B, et al. (2002Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N Engl J Med 347472480
  41. 41. LynchT. JBellD. WSordellaRGurubhagavatulaSOkimotoR. Aet al2004Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 35021292139
  42. 42. CurtinJ. ABusamKPinkelDBastianB. C2006Somatic activation of KIT in distinct subtypes of melanoma. J Clin Oncol 2443404346
  43. 43. AlbinoA. PLe Strange R, Oliff AI, Furth ME, Old LJ (1984Transforming ras genes from human melanoma: a manifestation of tumour heterogeneity? Nature 3086972
  44. 44. DaviesHBignellG. RCoxCStephensPEdkinsSet al2002Mutations of the BRAF gene in human cancer. Nature 417949954
  45. 45. Lopez-bergamiPFitchmanBRonaiZ2008Understanding signaling cascades in melanoma. Photochem Photobiol 84289306
  46. 46. PollockP. MHarperU. LHansenK. SYudtL. MStarkMet al2003High frequency of BRAF mutations in nevi. Nat Genet 331920
  47. 47. GordenAOsmanIGaiWHeDHuangWet al2003Analysis of BRAF and N-RAS mutations in metastatic melanoma tissues. Cancer Res 6339553957
  48. 48. KumarRAngeliniSCzeneKSaurojaIHahka-kemppinenMet al2003BRAF mutations in metastatic melanoma: a possible association with clinical outcome. Clin Cancer Res 933623368
  49. 49. CarrJMackieR. M1994Point mutations in the N-ras oncogene in malignant melanoma and congenital naevi. Br J Dermatol 1317277
  50. 50. Van ElsasAZerpSVan Der FlierSKruse-woltersMVaccaAet al1995Analysis of N-ras mutations in human cutaneous melanoma: tumor heterogeneity detected by polymerase chain reaction/single-stranded conformation polymorphism analysis. Recent Results Cancer Res 1395767
  51. 51. KortylewskiMHeinrichP. CKauffmannM. EBohmMMacKiewicz A, et al. (2001Mitogen-activated protein kinases control 27Kip1 expression and growth of human melanoma cells. Biochem J 357: 297-303.
  52. 52. LefevreGCalipelAMouriauxFHecquetCMalecazeFet al2003Opposite long-term regulation of c-Myc and 27Kip1through overactivation of Raf-1 and the MEK/ERK module in proliferating human choroidal melanoma cells. Oncogene 22: 8813-8822.
  53. 53. Lopez-bergamiPHuangCGoydosJ. SYipDBar-eliMet al2007Rewired ERK-JNK signaling pathways in melanoma. Cancer Cell 11447460
  54. 54. GodingC. R2000Mitf from neural crest to melanoma: signal transduction and transcription in the melanocyte lineage. Genes Dev 1417121728
  55. 55. WuMHemesathT. JTakemotoC. MHorstmannM. AWellsA. Get al2000c-Kit triggers dual phosphorylations, which couple activation and degradation of the essential melanocyte factor Mi. Genes Dev 14301312
  56. 56. KimD. SHwangE. SLeeJ. EKimS. YKwonS. Bet al2003Sphingosine-1-phosphate decreases melanin synthesis via sustained ERK activation and subsequent MITF degradation. J Cell Sci 11616991706
  57. 57. EisenmannK. MVanbrocklinM. WStaffendN. AKitchenS. MKooH. M2003Mitogen-activated protein kinase pathway-dependent tumor-specific survival signaling in melanoma cells through inactivation of the proapoptotic protein bad. Cancer Res 6383308337
  58. 58. KrasilnikovMIvanovV. NDongJRonaiZ2003Erkand P. I K negatively regulate STAT-transcriptional activities in human melanoma cells: implications towards sensitization to apoptosis. Oncogene 2240924101
  59. 59. TowerG. BCoonC. CBenbowUVincentiM. PBrinckerhoffC. E2002Erk 1/2 differentially regulates the expression from the 1G/2G single nucleotide polymorphism in the MMP-1 promoter in melanoma cells. Biochim Biophys Acta 1586265274
  60. 60. IshiiYOguraTTatemichiMFujisawaHOtsukaFet al2003Induction of matrix metalloproteinase gene transcription by nitric oxide and mechanisms of MMP-1 gene induction in human melanoma cell lines. Int J Cancer 103161168
  61. 61. RamosM. CSteinbrennerHStuhlmannDSiesHBrenneisenP2004Induction of MMP-10 and MMP-1 in a squamous cell carcinoma cell line by ultraviolet radiation. Biol Chem 3857586
  62. 62. SteelmanL. SBertrandF. EMccubreyJ. A2004The complexity of PTEN: mutation, marker and potential target for therapeutic intervention. Expert Opin Ther Targets 8537550
  63. 63. ParmiterA. HBalabanGClarkW. HJr., Nowell PC (1988Possible involvement of the chromosome region 10q24----q26 in early stages of melanocytic neoplasia. Cancer Genet Cytogenet 30313317
  64. 64. SimpsonLParsonsR2001PTEN: life as a tumor suppressor. Exp Cell Res 2642941
  65. 65. WuHGoelVHaluskaF. G2003PTEN signaling pathways in melanoma. Oncogene 2231133122
  66. 66. HerbstR. AWeissJEhnisACaveneeW. KArdenK. C1994Loss of heterozygosity for 10q22-10qter in malignant melanoma progression. Cancer Res 5431113114
  67. 67. HealyERehmanIAngusBReesJ. L1995Loss of heterozygosity in sporadic primary cutaneous melanoma. Genes Chromosomes Cancer 12152156
  68. 68. HarlanJ. EYoonH. SHajdukP. JFesikS. W1995Structural characterization of the interaction between a pleckstrin homology domain and phosphatidylinositol 4,5-bisphosphate. Biochemistry 3498599864
  69. 69. NicholsonK. MAndersonN. G2002The protein kinase B/Akt signalling pathway in human malignancy. Cell Signal 14381395
  70. 70. StahlJ. MSharmaACheungMZimmermanMChengJ. Qet al2004Deregulated Akt3 activity promotes development of malignant melanoma. Cancer Res 6470027010
  71. 71. RobertsonG. P2005Functional and therapeutic significance of Akt deregulation in malignant melanoma. Cancer Metastasis Rev 24273285
  72. 72. DhawanPSinghA. BEllisD. LRichmondA2002Constitutive activation of Akt/protein kinase B in melanoma leads to up-regulation of nuclear factor-kappaB and tumor progression. Cancer Res 6273357342
  73. 73. LiGKalabisJXuXMeierFOkaMet al2003Reciprocal regulation of MelCAM and AKT in human melanoma. Oncogene 2268916899
  74. 74. JohnsonJ. P1999Cell adhesion molecules in the development and progression of malignant melanoma. Cancer Metastasis Rev 18345357
  75. 75. KimDKimSKohHYoonS. OChungA. Set al2001Akt/PKB promotes cancer cell invasion via increased motility and metalloproteinase production. FASEB J 1519531962
  76. 76. ParkB. KZengXGlazerR. I2001Akt1 induces extracellular matrix invasion and matrix metalloproteinase-2 activity in mouse mammary epithelial cells. Cancer Res 6176477653
  77. 77. CardoneM. HRoyNStennickeH. RSalvesenG. SFrankeT. Fet al1998Regulation of cell death protease caspase-9 by phosphorylation. Science 28213181321
  78. 78. DattaS. RDudekHTaoXMastersSFuHet al1997Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell 91231241
  79. 79. GilesR. HVan EsJ. HCleversH2003Caught up in a Wnt storm: Wnt signaling in cancer. Biochim Biophys Acta 1653124
  80. 80. VeemanM. TAxelrodJ. DMoonR. T2003A second canon. Functions and mechanisms of beta-catenin-independent Wnt signaling. Dev Cell 5367377
  81. 81. ElchevaITaraporeR. SBhatiaNSpiegelmanV. S2008Overexpression of mRNA-binding protein CRD-BP in malignant melanomas. Oncogene 2750695074
  82. 82. RimmD. LCacaKHuGHarrisonF. BFearonE. R1999Frequent nuclear/cytoplasmic localization of beta-catenin without exon 3 mutations in malignant melanoma. Am J Pathol 154325329
  83. 83. DemunterALibbrechtLDegreefHDe Wolf-peetersCvan den Oord JJ (2002Loss of membranous expression of beta-catenin is associated with tumor progression in cutaneous melanoma and rarely caused by exon 3 mutations. Mod Pathol 15454461
  84. 84. OmholtKPlatzARingborgUHanssonJ2001Cytoplasmic and nuclear accumulation of beta-catenin is rarely caused by CTNNB1 exon 3 mutations in cutaneous malignant melanoma. Int J Cancer 92839842
  85. 85. RubinfeldBRobbinsPEl-GamilMAlbertIPorfiriEet al1997Stabilization of beta-catenin by genetic defects in melanoma cell lines. Science 27517901792
  86. 86. ReifenbergerJKnobbeC. BWolterMBlaschkeBSchulteK. Wet al2002Molecular genetic analysis of malignant melanomas for aberrations of the WNT signaling pathway genes CTNNB1, APC, ICAT and BTRC. Int J Cancer 100549556
  87. 87. PollockP. MHaywardN2002Mutations in exon 3 of the beta-catenin gene are rare in melanoma cell lines. Melanoma Res 12183186
  88. 88. WormJChristensenCGronbaekKTulchinskyEGuldbergP2004Genetic and epigenetic alterations of the APC gene in malignant melanoma. Oncogene 2352155226
  89. 89. TagoKNakamuraTNishitaMHyodoJNagaiSet al2000Inhibition of Wnt signaling by ICAT, a novel beta-catenin-interacting protein. Genes Dev 1417411749
  90. 90. GoodallJMartinozziSDexterT. JChampevalDCarreiraSet al2004Brn-2 expression controls melanoma proliferation and is directly regulated by beta-catenin. Mol Cell Biol 2429152922
  91. 91. ThomsonJ. AMurphyKBakerESutherlandG. RParsonsP. Get al1995The brn-2 gene regulates the melanocytic phenotype and tumorigenic potential of human melanoma cells. Oncogene 11691700
  92. 92. HodgkinsonC. AMooreK. JNakayamaASteingrimssonECopelandN. Get al1993Mutations at the mouse microphthalmia locus are associated with defects in a gene encoding a novel basic-helix-loop-helix-zipper protein. Cell 74395404
  93. 93. HughesM. JLingrelJ. BKrakowskyJ. MAndersonK. P1993A Helix-loop helix transcription factor-like gene is located at the mi locus. J Biol Chem 2682068720690
  94. 94. WidlundH. RHorstmannM. APriceE. RCuiJLessnickS. Let al2002Beta-catenin-induced melanoma growth requires the downstream target Microphthalmia-associated transcription factor. J Cell Biol 15810791087
  95. 95. SchepskyABruserKGunnarssonG. JGoodallJHallssonJ. Het al2006The microphthalmia-associated transcription factor Mitf interacts with beta-catenin to determine target gene expression. Mol Cell Biol 2689148927
  96. 96. KawanoYKyptaR2003Secreted antagonists of the Wnt signalling pathway. J Cell Sci 11626272634
  97. 97. ForgetM. ATurcotteSBeauseigleDGodin-ethierJPelletierSet al2007The Wnt pathway regulator DKK1 is preferentially expressed in hormone-resistant breast tumours and in some common cancer types. Br J Cancer 96646653
  98. 98. KuphalSLodermeyerSBatailleFSchuiererMHoangB. Het al2006Expression of Dickkopf genes is strongly reduced in malignant melanoma. Oncogene 2550275036
  99. 99. MikheevA. MMikheevaS. ARostomilyRZarblH2007Dickkopf-1 activates cell death in MDA-MB435 melanoma cells. Biochem Biophys Res Commun 352675680
  100. 100. LinY. CYouLXuZHeBYangC. Tet al2007Wnt inhibitory factor-1 gene transfer inhibits melanoma cell growth. Hum Gene Ther 18379386
  101. 101. WestonC. RDavisR. J2007The JNK signal transduction pathway. Curr Opin Cell Biol 19142149
  102. 102. KarinM1995The regulation of AP-1 activity by mitogen-activated protein kinases. J Biol Chem 2701648316486
  103. 103. KennedyN. JDavisR. J2003Role of JNK in tumor development. Cell Cycle 2199201
  104. 104. YangY. MBostFCharbonoWDeanNMckayRet al2003C-Jun NH(2)-terminal kinase mediates proliferation and tumor growth of human prostate carcinoma. Clin Cancer Res 9391401
  105. 105. CuiJHanS. YWangCSuWHarshyneLet al2006c-Jun NH(2)-terminal kinase 2alpha2 promotes the tumorigenicity of human glioblastoma cells. Cancer Res 661002410031
  106. 106. KhatlaniT. SWislezMSunMSrinivasHIwanagaKet al2007c-Jun N-terminal kinase is activated in non-small-cell lung cancer and promotes neoplastic transformation in human bronchial epithelial cells. Oncogene 2626582666
  107. 107. AdlerVSchafferAKimJDolanLRonaiZ1995UV Irradiation and heat shock mediate JNK activation via alternate pathways. J Biol Chem 2702607126077
  108. 108. MarianiOBrennetotCCoindreJ. MGruelNGanemCet al2007JUN oncogene amplification and overexpression block adipocytic differentiation in highly aggressive sarcomas. Cancer Cell 11361374
  109. 109. VleugelM. MGreijerA. EBosRVan Der WallEVan DiestP. J2006c-Jun activation is associated with proliferation and angiogenesis in invasive breast cancer. Hum Pathol 37668674
  110. 110. MaenoKMasudaAYanagisawaKKonishiHOsadaHet al2006Altered regulation of c-jun and its involvement in anchorage-independent growth of human lung cancers. Oncogene 25271277
  111. 111. YamanishiD. TBuckmeierJ. AMeyskensF. LJr. (1991Expression of c-jun, jun-B, and c-fos proto-oncogenes in human primary melanocytes and metastatic melanomas. J Invest Dermatol 97349353
  112. 112. RutbergS. EGoldsteinI. MYangY. MStackpoleC. WRonaiZ1994Expression and transcriptional activity of AP-1, CRE, and URE binding proteins in B16 mouse melanoma subclones. Mol Carcinog 108287
  113. 113. Mingo-sionA. MMariettaP. MKollerEWolfD. MVan Den Berg CL (2004Inhibition of JNK reduces G2/M transit independent of 53leading to endoreduplication, decreased proliferation, and apoptosis in breast cancer cells. Oncogene 23: 596-604.
  114. 114. UzgareA. RIsaacsJ. T2004Enhanced redundancy in Akt and mitogen-activated protein kinase-induced survival of malignant versus normal prostate epithelial cells. Cancer Res 6461906199
  115. 115. GurzovE. NBakiriLAlfaroJ. MWagnerE. FIzquierdoM2008Targeting c-Jun and JunB proteins as potential anticancer cell therapy. Oncogene 27641652
  116. 116. DixitVMakT. W2002NF-kappaB Signaling Many roads lead to madrid. Cell 111615619
  117. 117. GhoshSKarinM2002Missing pieces in the NF-kappaB puzzle. Cell 109 Suppl: S8196
  118. 118. ChenXKandasamyKSrivastavaR. K2003Differential roles of RelA (65and c-Rel subunits of nuclear factor kappa B in tumor necrosis factor-related apoptosis-inducing ligand signaling. Cancer Res 63: 1059-1066.
  119. 119. PoserIDominguezDDe HerrerosA. GVarnaiABuettnerRet al2001Loss of E-cadherin expression in melanoma cells involves up-regulation of the transcriptional repressor Snail. J Biol Chem 2762466124666
  120. 120. MeyskensF. LJr., Buckmeier JA, McNulty SE, Tohidian NB (1999Activation of nuclear factor-kappa B in human metastatic melanomacells and the effect of oxidative stress. Clin Cancer Res 511971202
  121. 121. BoukercheHSuZ. ZEmdadLSarkarDFisherP. B2007mda-9/Syntenin regulates the metastatic phenotype in human melanoma cells by activating nuclear factor-kappaB. Cancer Res 6718121822
  122. 122. PhilipSKunduG. C2003Osteopontin induces nuclear factor kappa B-mediated promatrix metalloproteinase-2 activation through I kappa B alpha /IKK signaling pathways, and curcumin (diferulolylmethane) down-regulates these pathways. J Biol Chem 2781448714497
  123. 123. DupinELe Douarin NM (2003Development of melanocyte precursors from the vertebrate neural crest. Oncogene 2230163023
  124. 124. HendrixM. JSeftorE. ASeftorR. EKasemeier-kulesaJKulesaP. Met al2007Reprogramming metastatic tumour cells with embryonic microenvironments. Nat Rev Cancer 7246255
  125. 125. BittnerMMeltzerPChenYJiangYSeftorEet al2000Molecular classification of cutaneous malignant melanoma by gene expression profiling. Nature 406536540
  126. 126. HendrixM. JSeftorE. AHessA. RSeftorR. E2003Molecular plasticity of human melanoma cells. Oncogene 2230703075
  127. 127. CoiffierBLepageEBriereJHerbrechtRTillyHet al2002CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med 346235242
  128. 128. LeeJ. THerlynM2007Microenvironmental influences in melanoma progression. J Cell Biochem 101862872
  129. 129. PagetS1989The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev 898101
  130. 130. FidlerI. J2003The pathogenesis of cancer metastasis: the’seed and soil’ hypothesis revisited. Nat Rev Cancer 3453458
  131. 131. TopczewskaJ. MPostovitL. MMargaryanN. VSamAHessA. Ret al2006Embryonic and tumorigenic pathways converge via Nodal signaling: role in melanoma aggressiveness. Nat Med 12925932
  132. 132. MillerS. AHamiltonS. LWesterU. GCyrW. H1998An analysis of UVA emissions from sunlamps and the potential importance for melanoma. Photochem Photobiol 686370
  133. 133. LautenschlagerSWulfH. CPittelkowM. R2007Photoprotection. Lancet 370528537
  134. 134. BrennerMHearingV. J2008The protective role of melanin against UV damage in human skin. Photochem Photobiol 84539549
  135. 135. BastianB. CLeBoit PE, Hamm H, Brocker EB, Pinkel D (1998Chromosomal gains and losses in primary cutaneous melanomas detected by comparative genomic hybridization. Cancer Res 5821702175
  136. 136. CurtinJ. AFridlyandJKageshitaTPatelH. NBusamK. Jet al2005Distinct sets of genetic alterations in melanoma. N Engl J Med 35321352147
  137. 137. BalazsMAdamZTreszlABeganyAHunyadiJet al2001Chromosomal imbalances in primary and metastatic melanomas revealed by comparative genomic hybridization. Cytometry 46222232
  138. 138. HauslerTStangAAnastassiouGJockelK. HMrzykSet al2005Loss of heterozygosity of 1p in uveal melanomas with monosomy 3. Int J Cancer 116909913
  139. 139. WhiteJ. SMcleanI. WBeckerR. LDirector-myskaA. ENathJ2006Correlation of comparative genomic hybridization results of 100 archival uveal melanomas with patient survival. Cancer Genet Cytogenet 1702939
  140. 140. SpeicherM. RPrescherGdu Manoir S, Jauch A, Horsthemke B, et al. (1994Chromosomal gains and losses in uveal melanomas detected by comparative genomic hybridization. Cancer Res 5438173823
  141. 141. VajdicC. MHutchinsA. MKrickerAAitkenJ. FArmstrongB. Ket al2003Chromosomal gains and losses in ocular melanoma detected by comparative genomic hybridization in an Australian population-based study. Cancer Genet Cytogenet 1441217
  142. 142. GarrawayL. AWidlundH. RRubinM. AGetzGBergerA. Jet al2005Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma. Nature 436117122
  143. 143. JonssonGDahlCStaafJSandbergTBendahlP. Oet al2007Genomic profiling of malignant melanoma using tiling-resolution arrayCGH. Oncogene 2647384748
  144. 144. AlonsoS. RTraceyLOrtizPPerez-gomezBPalaciosJet al2007A high-throughput study in melanoma identifies epithelial-mesenchymal transition as a major determinant of metastasis. Cancer Res 6734503460
  145. 145. HoekKRimmD. LWilliamsK. RZhaoHAriyanSet al2004Expression profiling reveals novel pathways in the transformation of melanocytes to melanomas. Cancer Res 6452705282
  146. 146. SeftorR. ESeftorE. AKoshikawaNMeltzerP. SGardnerL. Met al2001Cooperative interactions of laminin 5 gamma2 chain, matrix metalloproteinase-2, and membrane type-1-matrix/metalloproteinase are required for mimicry of embryonic vasculogenesis by aggressive melanoma. Cancer Res 6163226327
  147. 147. JaegerJKoczanDThiesenH. JIbrahimS. MGrossGet al2007Gene expression signatures for tumor progression, tumor subtype, and tumor thickness in laser-microdissected melanoma tissues. Clin Cancer Res 13806815
  148. 148. WeeraratnaA. TJiangYHostetterGRosenblattKDurayPet al2002Wnt5a signaling directly affects cell motility and invasion of metastatic melanoma. Cancer Cell 1279288
  149. 149. BeckerBRoeschAHafnerCStolzWDugasMet al2004Discrimination of melanocytic tumors by cDNA array hybridization of tissues prepared by laser pressure catapulting. J Invest Dermatol 122361368
  150. 150. WinnepenninckxVLazarVMichielsSDessenPStasMet al2006Gene expression profiling of primary cutaneous melanoma and clinical outcome. J Natl Cancer Inst 98472482
  151. 151. HaqqCNosratiMSudilovskyDCrothersJKhodabakhshDet al2005The gene expression signatures of melanoma progression. Proc Natl Acad Sci U S A 10260926097

Written By

Rohinton S. Tarapore

Submitted: 19 April 2012 Published: 17 April 2013