Open access

Melanoma — Epidemiology, Genetics and Risk Factors

Written By

John A. D’Orazio, Stuart Jarrett, Amanda Marsch, James Lagrew and Laura Cleary

Submitted: 25 April 2012 Published: 13 February 2013

DOI: 10.5772/55172

From the Edited Volume

Recent Advances in the Biology, Therapy and Management of Melanoma

Edited by Lester M. Davids

Chapter metrics overview

3,502 Chapter Downloads

View Full Metrics

1. Introduction

1.1. Melanoma a growing problem

The U.S. National Cancer Institute’s Surveillance Epidemiology and End Results (SEER) Cancer Statistics Review estimates over 70,000 people will be diagnosed and 9,000 will die from melanoma in the United States in 2012. Though melanoma can affect persons of essentially any age, it is mainly a disease of adulthood, with median ages of diagnosis and death between 61 and 68 years, respectively (Weinstock, 2012). Nonetheless, melanoma incidence is increasing across age groups, over the past several decades in the United States (Fig. 1) (Ekwueme et al., 2011). In 1935, the average American individual had a 1 in 1,500 lifetime risk of developing melanoma. In 2002, the approximate risk of developing melanoma increased to 1 in 68 individuals (Rigel, 2002). Globally, Australia and New Zealand have the highest incidence rate of melanoma, an abundance of fair-skinned residents living in a UV-rich geography widely believed to be a major factor (Lens and Dawes, 2004). The current melanoma risk for Australian and New Zealander populations may be as high as 1 in 50 (Rigel, 2010). Considering melanoma is being diagnosed more often in young adults, could be prevented by UV-avoiding behaviors, and can be associated with extensive morbidity and mortality, it is truly an emerging public health concern. Part of the apparent increase in melanoma incidence may be due to better surveillance and earlier detection (Erdmann et al., 2012) however, even with heightened melanoma awareness and screening, there seems to have been a real increase in melanoma incidence over the past several decades.

Figure 1.

Increasing incidence of melanoma of the skin, US. Data are reported as lifetime risk and are taken from NCI SEER reports.

1.2. The ultraviolet connection

Historically, humans have been exposed to UV radiation primarily as a consequence of unprotected exposure to sunlight (Holman et al., 1983; Holman et al., 1986;Woodward and Boffetta, 1997). Since the early 20th century, a tanned appearance has been culturally associated with health and well-being in Western civilizations. The desire to sport a dark tan has been matched by increased opportunities for sunbathing outdoors as well as proliferation of indoor tanning salons. UV radiation has many deleterious effects on cells (Zaidi et al., 2012), producing both direct and indirect DNA damage, resulting in mutations that can contributed to carcinogenesis in skin cells. Direct damage occurs when DNA absorbs UV photons and undergoes cleavage of the 5-6 double bond of pyrimidines. When two adjacent pyrimidines undergo this 5-6 double bond opening, a covalent ring structure referred to as a cyclobutane pyrimidine dimer (thymine dimer) can be formed. Alternatively, a pyrimidine 6-4 pyrimidone (6,4)- photoproduct can result when a 5-6 double bond in a pyrimidine opens and reacts with the exocyclic moiety of the adjacent 3' pyrimidine to form a covalent 6-4 linkage (Kadekaro et al., 2003; Pfeifer et al., 2005; Maddodi and Setaluri, 2008). Both (6,4)-photoproducts and cyclobutane dimers can result in characteristic transition mutations between adjacent pyrimidines. “UV signature mutations” involving T-to-C or C-to-T changes are a common feature of UV-induced malignancies such as skin cancers (Kanjilal et al., 1993; Nataraj et al., 1996; Soehnge et al., 1997; Sarasin, 1999). UV radiation also damages cellular macromolecules indirectly, through production of oxidative free radicals [20]. Several DNA modifications can result from oxidative injury, including 7,8-dihydro-8-oxoguanine (8-oxoguanine; 8-OH-dG), which promotes mutagenesis (specifically GC-TA transversion mutations) (Kino and Sugiyama, 2005). Both direct and indirect DNA changes interfere with transcription and replication, and render skin cells susceptible to mutagenesis. It is estimated that one day’s worth of sun exposure can cause up to 100,000 potentially mutagenic UV-induced photolesions in each skin cell (Nakabeppu et al., 2006).

Much of solar UV energy is absorbed by stratospheric ozone, and gradual depletion of stratospheric ozone over the last several decades has resulted in higher levels of solar UV radiation striking Earth’s surface (van der Leun et al., 2008). Increased ambient UV radiation from global climate change may be an important factor to explain the burgeoning prevalence of melanoma (Schmalwieser et al., 2005). Increased exposure to ambient UV radiation is a feature of global climate change because of thinning of atmospheric ozone and increased outdoor occupational and recreational activities associated with a warming climate (de Gruijl et al., 2003; van der Leun et al., 2008; Andrady et al., 2010; Makin, 2011; McKenzie et al., 2011; Norval et al., 2011). UV exposure in youth seems particularly important, affording the longest amount of time for the gradual accumulation of mutagenic UV lesions. Thus, high UV exposures in childhood, adolescence and young adulthood are strongly linked to risk of skin cancer later in life. For example, first exposure to indoor tanning before the age of 35 years raises lifetime risk of melanoma by seventy five percent (Schulman and Fisher, 2009).

1.3. Geographic location

UV radiation varies with altitude and with proximity to the equator. Since UV radiation can be absorbed, reflected back into space or scattered by particles in the atmosphere, ambient UV doses on the surface of the Earth vary according to the amount of atmosphere solar radiation must pass through. The more atmosphere solar radiation passing through, the weaker the corresponding UV content of the sunlight realized on the surface of the Earth. Sunlight strikes Earth most directly at the equator and more tangentially toward the poles. The more direct the sunlight’s path, the less atmosphere radiation has to traverse and the more powerful the UV component will be (Fig. 2). Thus, UV content of sunlight is most powerful in equatorial locations and weakest in polar extremes. Equatorial locations are also typically the hottest environments, therefore people living in such places tend to wear lesser amounts of clothing. Fabrics and other materials used for clothing typically block large amounts of UV radiation, as evidenced by the pattern of “farmer tans” among people who wear short sleeve tee shirts. Persons living in cold, polar climates would be expected to realize far less UV radiation from sunlight both because the UV dosage in ambient sunlight is weaker in such locations and because people living there probably are covered with more clothing. Thus in general, individuals living in equatorial locations typically receive much higher ambient UV doses than persons inhabiting temperate climates (Lee and Scotto, 1993). In the United States, risk of melanoma is higher in the South than in the North (Crombie, 1979). Worldwide, melanoma rates are highest in UV-rich environments such as Australia (Franceschi and Cristofolini, 1992; Elwood and Koh, 1994; Marks, 1995). One study examining the low rates of melanoma in Scandinavia pointed to data showing that ambient UV levels in Norway were significantly lower than most of the world because of its high latitude (Moan et al., 2009). Altitude and the amount of particulate matter in the atmosphere also influence the amount of UV found in a particular geographic location. The higher the altitude, the nearer the location to the sun and the more powerful the sunlight’s UV dose will be. Similarly, the more particles in the atmosphere, the higher the likelihood of interference with UV and the weaker the UV energy at the earth’s surface (Atkinson et al., 2011).

Figure 2.

Strength of ambient UV varies with geographic location. UV radiation in sunlight can be blocked by the atmosphere. Consequently, the longer the distance sunlight must travel, the weaker the UV component hitting Earth will be. The highest UV doses in sunlight are found at the equator, where the sun hits the Earth at a direct angle.

Advertisement

2. Risk factors

2.1. Older age

Melanoma incidence increases markedly with advancing age (Fig. 3), presumably because of the time it takes to accumulate mutations in melanocyte-relevant genes that drive carcinogenesis (Gilchrest et al., 1999). However other factors may also be relevant, including a more permissive environment for tumors to develop because of the natural age-related decline in cellular immunity (Weiskopf et al., 2009; Malaguarnera et al., 2010). According to the SEER data, from 2005-2009, the median age of melanoma diagnosis was 61 years. Nonetheless, although older adults are more at risk for melanoma, the incidence of melanoma in young adults, especially in young adult women, is increasing at a faster rate (Reed et al., 2012). For women and men between the ages of 20-29, melanoma is the second and third most commonly diagnosed cancer respectively (Siegel et al., 2012).

2.2. Solar UV exposure

Decreasing UV radiation exposure, from both sun exposure and artificial UV light, may be the single best preventable factor for decreasing the incidence rate of melanoma (Lucas et al., 2008). The ultraviolet portion of sunlight is divided into UVC (<280 nm), UVB (280-315 nm) and UVA (315-400 nm), with wavelengths below 290 nm being absorbed by stratospheric ozone (Fig. 4). UVB constitutes 5 -10% of solar UV irradiation and is mainly absorbed by the epidermal layer of the skin. The most frequent form of DNA damage induced by UVB are molecular rearrangements resulting in the dimerization of pyrimidines, generating 2 classes of mutagenic lesions, cyclobutane pyrimidine dimers (CPDs) and pyrimidine (6-4) pyrimidone photoproducts (6-4 PP) through direct absorption by DNA. CPDs are formed through a ring structure involving C5 and C6 of neighboring bases whereas 6-4 PP are formed with a non-cyclic bond between C6 and C4 (Budiyanto et al., 2002). These photoproducts promote cytosines (C)- thymines (T) and CC-TT transitions, with regions of DNA containing 5-methylocytosine being hot spots for UVB-induced mutations. Radiation in UVA range is associated with lower energy but has the ability to penetrate deeper into the dermis. In contrast to UVB, UVA is poorly absorbed by DNA, but excites numerous endogenous chromophores, generating reactive oxygen species (ROS) e.g. singlet oxygen and hydroxyl radicals. The predominant ROS-induced lesions formed are oxidized bases, such as 8-oxo-dG with DNA single and double strand breaks (Mouret et al., 2006). Both ultraviolet A radiation (320 to 400 nm) and ultraviolet B radiation (290 to 320 nm) contribute to the development of melanoma (Gilchrest et al., 1999).

Figure 3.

Melanoma incidence by age, US. Incidence rates (per 100,000 individuals) are based on NCI SEER data. Note the marked increase in melanoma incidence with increasing age. Also evident is the tremendous discrepancy in melanoma incidence between persons of fair- and dark-skinned complexions.

Figure 4.

Electromagnetic spectrum of visible and UV radiation and biologic effects on the skin. The diagram shows the subdivision of the solar UV spectrum with the shorter UV wavelengths (i.e. UVC) being entirely absorbed by stratospheric oxygen, and the majority of UVB (> 90 %) being absorbed by ozone. UV light penetrates the skin and is absorbed by different layers in a wavelength- dependent manner. The visible and UVA components of solar radiation penetrates deeply into the dermis reaching the dermal stratum papillare. In contrast, UVB is almost completely absorbed by the epidermis, with only ~20 % reaching the epidermal stratum basale. UVA and visible light make up the majority of the total terrestrial solar energy and are able to generate reactive oxygen species that can damage DNA via indirect photosensitizing reactions. UVB is directly absorbed by DNA which causes molecular rearrangements forming the specific photoproducts CPD and 6-4 PP. Mutations and cancer can result from a variety of modifications to DNA.

2.3. Sunburns

While squamous cell carcinoma of the skin has been closely associated with long term occupational exposure to the sun, risk of developing melanoma seems to be more associated with intermittent, high intensity sun exposure (MacKie and Aitchison, 1982; Lew et al., 1983). Prevalence of sunburns among children is high, with one study finding that approximately 69% of adolescents experienced sunburn the previous summer and only 40% used sun protection methods (Buller et al., 2011). Positive association between severe, painful sunburn and the development of melanoma and a negative association between Early found a positive melanoma and long-term recreational/occupational sun exposure (MacKie and Aitchison, 1982; Lew et al., 1983). Sunburn represents an inflammatory response of the skin to a significant amount of acute UV damage. It is mediated by a complex series of cellular and hormonal events, including the generation of cytokines and mediators of vasodilatation. Risk of sunburn is related not only to UV exposure, but also to innate melanin content of the skin. Thus, sunburn mostly occurs in fair skinned people without sun protection exposed to high intensities of UV radiation, for example in equatorial or high altitude locations. Various epidemiologic studies support the finding that the number of severe sunburns and total childhood sun exposure are positively associated with the development of melanoma (Holman et al., 1986; Scotto and Fears, 1987; Cust et al., 2011; Newton-Bishop et al., 2011; Volkovova et al., 2012). The carcinogenic effects of sunburn have also been demonstrated experimentally using transgenic mice forcing overexpression of the hepatocyte growth/scatter factor (HGF/SF) in melanocytes. In these mice, HGF over-expression altered the distribution of melanocytes to create a “humanized” model, which mimics human skin with melanocytes located in the basal layer of the epidermis, rendering them more susceptible to DNA damaging effects of UVR. Remarkably, a single erythemal UV dose to neonatal mice caused the development of melanomas in roughly half of animals at one year of age (Noonan et al., 2001). This animal model has been used by several laboratories to study a variety of melanoma susceptibility genes in context of UV-induced childhood sunburn and melanoma initiation and metastasis (Recio et al., 2002).

2.4. Indoor tanning

Whereas only one percent of Americans ever used a tanning bed in 1988, now more than twenty five percent have participated in indoor tanning. With more than 25,000 facilities in the US alone, indoor tanning represents a multi-billion dollar industry. Employing over 160,000 people, the tanning industry has a customer base of nearly thirty million people and exerts political influence through powerful lobbying efforts. Nonetheless, there are clear health risks associated with indoor tanning. UV radiation emitted by tanning lamps is typically more powerful than direct ambient sunlight. It is estimated that half an hour in a tanning booth yields the same UV damage to skin as much as 300 minutes in unprotected sun. Levels of UVA/UVB emitted by tanning beds are unpredictable, widely unregulated, and much higher than environmental exposure. A study of 62 tanning salons in North Carolina found that the average UVA output of a tanning bed was 192.1 W/m2 (vs. average UVA summer solar output at noon in Washington D.C. of 48 W/m2) and the average UVB output of a tanning bed was 0.35 W/m2 (vs. average UVB summer solar output at noon in Washington D.C. of 0.18 W/m2) (Hornung et al., 2003). Tanning bed use is clearly associated with skin cancers of all varieties. Persons who have ever used a tanning bed have a 50% increased risk of basal cell carcinoma and more than a 100% increased risk of squamous cell carcinoma (Karagas et al., 2002).

The risk association between melanoma development and indoor tanning has been well substantiated (Autier, 2004; Rados, 2005; Han et al., 2006). Data accumulated from several studies suggest that the use of a tanning salon before the age of 35 is associated with a 75% increased lifetime risk of melanoma, while over-use of tanning salons was associated with a 15% increased risk of melanoma (Fig. 5) (Schulman and Fisher, 2009). Risk of carcinogenesis is enhanced for all types of tanning beds (UVA, UVB and mixed output) and increases with years of use, number of sessions, and hours exposed (Lazovich et al., 2010). There currently is no “safe” way to tan by UV without the inherent risk of photodamage and malignancy. The use of tanning salons despite the established risks, however, remains popular, especially in female young adults and adolescents. A recent survey found that 18.1% of female and 6.3% of male Caucasian adults reported using a tanning salon in the past 12 months (Choi et al., 2010). Among 10,000 adolescents across the 50 states, 24.6% of girls under 18 reported tanning, with prevalence of use steadily increasing from age 12 to 18 years (Geller et al., 2002). California and Vermont have recently banned (January 2012 and July 2012 respectively) use of indoor tanning beds for minors, while many other states require parental permission or have proposed legislation for restricting the use of tanning beds for minors. The use of tanning salons by adolescents did not decline from 1998 to 2004, even though more states restricted use by minors (Cokkinides et al., 2009), which suggests that these partial restrictions may not be effective. Predictors of using tanning salons for women were residing in the Midwest and the South and using spray tan products, while men who lived in metropolitan areas were more likely to visit tanning salons.

Figure 5.

Relative risk of melanoma associated with exposure to indoor tanning. Results of seven studies and overall estimate. Values higher than 1.0 indicate heightened risk of melanoma. Modified from (Schulman and Fisher, 2009).

2.5. PUVA therapy

Ultraviolet A radiation therapy (PUVA) is a common and effective treatment for psoriasis that was first introduced in the 1970s. Since UVA exposure from the sun and artificial sources like tanning beds is a clear risk for melanoma, there is concern that PUVA therapy may predispose to malignancies including melanoma. One large cohort study that followed patients for 20 years found that there was a 10-fold increase in the incidence of invasive melanoma in patients who had received PUVA therapy versus age matched controls (Stern, 2001). Increased risk began at 15 years post-PUVA therapy exposure, and there was a stronger association with patients exposed to higher doses of PUVA therapy, more treatments (greater than 250), and in patients with fair skin. Thus, limiting exposure to PUVA to minimal doses and carefully selecting appropriate patients for the treatment can maximize the effectiveness of this treatment and minimize the risks. Patients who receive PUVA therapy should be carefully followed to facilitate early detection of melanoma and other skin cancers.

2.6. Skin pigmentation

Although individuals from any race or skin pigmentation group can be affected by melanoma, risk of disease is much higher in fair-skinned persons (Fig. 6) (Beral et al., 1983; Rees and Healy, 1997; Sturm, 2002). Created by Dr. Thomas Fitzpatrick in 1975, the Fitzpatrick scale is commonly used to describe skin tone and resultant UV sensitivity (Draelos, 2011). Skin complexion is mainly determined by the amount of black melanin in the epidermis. This pigment, called eumelanin, is a potent blocker of UV radiation. Thus the more eumelanin in the skin, the less UV penetrates into the deep layers of the epidermis, and the less UV-mediated mutagenesis will occur. Risk of sunburn is also heavily influenced by epidermal eumelanin content. In fair-skinned individuals with low Fitzpatrick skin types, it takes a much lower dose of UV to induce inflammation. The amount of UV needed to cause a sunburn is termed the “minimal erythematous dose” (MED), and a lower MED correlates with low levels of epidermal eumelanin and a higher risk of melanoma (Ravnbak et al., 2010) (Fig. 7). Thus, risk of melanoma for Caucasian males and females is 31.6 and 19.9 per 100,000 respectively, while risk for African

American males and females is only 1.1 and 0.9 per 100,000 in comparison (Ekwueme et al., 2011; Park et al., 2012).

Figure 6.

Racial Disparity in Melanoma Incidence (US). Incidence rates based on NCI SEER data. Note that in general, the darker a race’s average skin tone, the lower their incidence of melanoma, irrespective of gender.

Figure 7.

Melanoma risk varies according to skin complexion. Skin complexion can be estimated by the Fitzpatrick scale wherein the higher the number, the more deeply melanized and pigmented the skin is. Fair-skinned individuals are much more UV sensitive and tend to burn rather than tan after UV exposure. Melanoma risk is highest in fair-skinned individuals.

2.7. Nevi

The majority of melanomas arise out of pre-existing moles (nevi), therefore the more nevi a person has, the higher the likelihood that a melanoma may develop (Grob et al., 1990; Newton-Bishop et al., 2010). One study found a seven-fold increased relative risk for melanoma if a patient has more than one hundred nevi (Gandini et al., 2005). Most patients, however, do a poor job in estimating their own mole counts (Melia et al., 2001), and a patient’s self assessment of nevus count should not be relied upon in lieu of a full skin exam for melanoma screening (Psaty et al., 2010). Despite the link between nevi and melanoma, risk of any given mole progressing to malignancy is very low (Metcalf and Maize, 1985; Halpern et al., 1993). One study estimated the 60 year risk of malignant transformation to be 1:11,000 for an individual nevus on a 20 year-old woman (Tsao et al., 2003).

A molecular link between benign nevi and malignant melanoma was established in 2003 when Pollock and coworkers reported that a gain of function mutation in the BRAF gene was common to the majority of benign nevi and melanomas (Pollock et al., 2003). Specifically, the V599E amino-acid substitution in BRAF results in enhanced MAPkinase signaling which stimulates melanocytes to proliferate. Clearly other genetic and/or epigenetic cellular events, such as loss of the tumor suppressor p16, are required for full malignant transformation, as BRAF mutation is sufficient for nevi formation but not melanomagenesis.

Congenital melanocytic nevi are pigmented lesions found on individuals at birth (Zaal et al., 2005; Krengel et al., 2006). Those that are particularly large (>20 cm in diameter) seem particularly prone to malignant transformation, and are associated with a lifetime melanoma risk of approximately 10% (Krengel et al., 2006). Smaller congenital melanocytic nevi have a significantly lower risk of malignant degeneration. Given their relatively high malignant potential, large congenital melanocytic nevi warrant consideration for prophylactic excision (Psaty et al., 2010) preferably during childhood, since up to 70% of melanomas associated with congenital melanocytic nevi occur by the individual’s tenth year (Marghoob et al., 2006).

Atypical Mole Syndrome (also referred to as Dysplastic Nevus Syndrome or Familial Atypical Multiple-Mole Melanoma Syndrome) has emerged as one of the most significant risk factors for the development of melanoma (Carey et al., 1994; Slade et al., 1995; Seykora and Elder, 1996). In the general population, dysplastic nevi are relatively common: found on 2-8% of Caucasians especially among those under 30 (Naeyaert and Brochez, 2003). A combination of both UV exposure and genetic susceptibility is believed to contribute to dysplastic nevi formation (Naeyaert and Brochez, 2003). Atypical Mole Syndrome is an important melanoma risk factor (Halpern et al., 1993); individual melanoma risk approaches 82% in affected individuals by the age of 72 (Tucker et al., 1993).

2.8. Chemical exposure and occupational risk

Geographic discrepancy in melanoma incidence may be influenced by factors other than UV exposure and skin pigmentation (Fortes and de Vries, 2008). A number of environmental and occupational substances have been linked to development of malignant melanoma including heavy metals, polycyclic aromatic hydrocarbons (PAHs) and benzene (Ingram, 1992; Vinceti et al., 2005; Meyskens and Yang, 2011 ). As a result of working around many of these chemicals, petroleum workers, for example, have been reported to have up to an eight-fold increased risk of melanoma (Magnani et al., 1987). Polyvinyl chloride (PVC), a substance used widely in the clothing and chemical industries, is also linked to increased risk of melanoma (Lundberg et al., 1993; Langard et al., 2000). Printers and lithographers, through their exposure to PAH and benzene solvents, have up to a 4.6-fold increased risk of disease (McLaughlin et al., 1988). Ionizing radiation exposure, as might occur from medical radiation exposure or atomic energy occupational exposure has also been linked to melanoma risk (Fink and Bates, 2005; Lie et al., 2008; Korcum et al., 2009). Pesticide exposure was reported to almost triple melanoma risk (Burkhart and Burkhart, 2000). Clearly a better understanding of occupational risk factors, especially when coupled to UV risk, is needed to guide more targeted public health efforts for the prevention of melanoma (Fortes and de Vries, 2008).

2.9. Immunodeficiency

Immunodeficiency, either from inherited defects in cell-mediated immunity or from infection-associated immunosuppression (e.g. AIDS) clearly predisposes individuals for the development of melanoma (Silverberg et al., 2011). Furthermore, with the increasing prevalence of autoimmune disorders and solid organ transplantation requiring medical restraint of native immune function, iatrogenic immunosupression is becoming an increasingly important risk factor for malignancy (DePry et al., 2011). The number of individuals in the US living with solid organ transplant has more than doubled since 1998 to more than 225,000 individuals (Sullivan et al., 2012). Although immunosuppressive agents expose patients to increased risk for a large number of malignancies, cutaneous cancer risk is particularly affected (Engels et al., 2011), and skin cancers in immunsuppressed patients may behave more aggressively than those in immunocompetent persons (Brewer et al., 2011). Cancer patients treated with chemotherapy also have a higher incidence of melanoma, presumably either because of the mutagenic effects of chemotherapy on melanocytes or perhaps through immunosuppression (Smith et al., 1993). Therefore, solid tumor transplant patients, persons with inborn or acquired deficiencies of T cell function and anyone with a current or past pharmacologic history of chemotherapy or immunosuppressive medications should be advised to practice UV-avoiding strategies and be regularly screened for cutaneous malignancies.

Advertisement

3. Genetic factors

While UV exposure is the most significant environmental risk factor for melanoma, there are several genes that when mutated clearly influence melanoma risk (Meyle and Guldberg, 2009; Nelson and Tsao, 2009; Ward et al., 2012). These genes have been identified largely through studying melanoma-prone families or individuals with extraordinary UV sensitivity or melanoma predisposition. Some of these genetic defects cause bone fide familial cancer syndromes, characterized by heritable predisposition to one or more types of malignancy. Each cancer syndrome is associated with unique cancer risk. Clinical “clues” to melanoma familial cancer syndromes include: melanomas diagnosed at a young age (e.g. below forty years of age), multiple primary melanomas diagnosed in the same person over time, multiple family members affected by melanoma, and extreme UV sensitivity (D'Orazio 2010). It is estimated that up to twelve percent of patients diagnosed with melanoma will have a positive family history of melanoma, yet even among this group, there is often no identifiable melanoma susceptibility gene (Gandini et al., 2005). Many of these melanoma susceptibility genes can portend risk vastly exceeding that of the general population (Udayakumar and Tsao, 2009).

3.1. Cyclin-Dependent Kinase Inhibitor 2A (CDKN2A)

The familial atypical multiple mole and melanoma (FAMMM) syndrome was first described in two families in which affected individuals harbored more than one hundred dysplastic nevi and had a lifetime cumulative incidence of melanoma approaching one hundred percent (Clark et al., 1978; Lynch et al., 1978). This syndrome, also called “dysplastic nevus syndrome” was associated with many of the features of a familial cancer syndrome, including melanomas at young ages (median age of 33 years in one study) (Goldstein et al., 1994). Heterozygous loss of CDKN2A function is associated with roughly 40% of cases of familial melanoma (Kamb et al., 1994; Holland et al., 1995)

Linkage studies performed in melanoma pedigrees identified loss of heterozygosity in the chromosome 9p21 region (Fountain et al., 1992). Later, the cyclin-dependent kinase inhibitor 2A gene was identified through positional cloning to be the tumor suppressor on 9p21 that was mutated in many melanoma-prone families (Kamb et al., 1994; Weaver-Feldhaus et al., 1994). Interestingly, affected individuals were not only at higher risk of malignant melanoma of the skin, but also for central nervous system gliomas, lung cancers and leukemias (Nobori et al., 1994). CDKN2A actually encodes two distinct tumor suppressor proteins- p16 and p14ARF that are transcribed in alternate reading frames directed through the use of alternative first exons (Chin et al., 1998; Sharpless and DePinho, 1999; Sharpless and Chin, 2003). p16/INK4a is produced from a transcript generated from exons 1α, 2 and 3, and p14/Arf is generated in an alternative reading frame, from a transcript of exons 1β, 2 and 3 (Udayakumar and Tsao, 2009). The majority of melanoma-associated mutations impacting exon 1β, which is specific for p16INK4a. Most germline mutations in CDKN2A found to contribute to melanoma susceptibility are loss-of-function missense or nonsense mutations of p16 (Goldstein et al., 2006; Goldstein et al., 2007).

The p16 tumor suppressor protein acts to regulate cell proliferation at the G1/S cell cycle checkpoint by inhibiting the cyclin-dependent kinases CDK4 and CDK6 to prevent entry into S-phase of the cell cycle (Serrano et al., 1993; Ohtani et al., 2001). Cyclin-dependent kinases in complex with cyclin D function jointly to inactivate the retinoblastoma (RB1) by phosphorylation. Once phosphorylated, RB1 is released from the transcription factor E2F-1, thereby permitting E2F-dependent transcription of genes that propel cells into proliferation. By binding to and inhibiting CDK4, p16 acts to prevent cell cycle progression, and when p16 function is lost, cells lose regulatory control over CDK/cyclin activity and proceed into unregulated cell division (Bartkova et al., 1996; Chin et al., 1998; Liggett and Sidransky, 1998). As with many other tumor suppressor genes, it is thought that inactivation or underexpression of both alleles of CDKN2A may be required for a cancer phenotype to emerge (the “two-hit” hypothesis) (Knudson, 1996; Tomlinson et al., 2001; Payne and Kemp, 2005). Thus individuals with inherited loss of one copy of p16 are at risk for p16-dependent malignancies such as melanoma (Ranade et al., 1995), with cancers developing only if the remaining p16 allele is inactivated to a sufficient extent either through mutation or epigenetic inactivation (Berger et al., 2011).

3.2. Cyclin-Dependent Kinase 4 (CDK4)

Several melanoma-prone kindreds have been discovered who carry mutations not in CDKN2A, but in its target- cyclin-dependent kinase 4 (CDK4) (Zuo et al., 1996; Soufir et al., 1998; Goldstein et al., 2000). Unlike CDKN2A whose p16 protein product acts as a tumor suppressor by negatively regulating melanocyte proliferation, CDK4 is an oncogene whose activity enhances cell division. The gain-of-function mutations in CDK4 melanoma-prone families typically result in amino acid changes that render the CDK4 enzyme insensitive to p16 inhibition, thereby resulting in a functional p16 null phenotype (Zuo et al., 1996; Newton Bishop et al., 1999; Goldstein et al., 2000).

3.3. Xeroderma Pigmentosum (XP) genes

Xeroderma pigmentosum (XP) is a rare autosomal recessive disorder of the nucleotide excision DNA repair (NER) pathway caused by homozygous deficiency of any one of at least eight genes (XPA, XPB, XPC, XPD, XPE, XPF, XPG and XPV) that work in complex to repair bulky DNA lesions such as mutagenic DNA photoproducts caused by UV radiation (Leibeling et al., 2006) (Fig. 8). NER functions by recruiting a protein complex known as XPC-hHR23B to UV-induced photoproducts in the DNA, with XPE aiding lesion verification. TFIIH, a transcription factor containing multiple enzymes including XPA, XPB and XPD then unwind the DNA in the vicinity of the damaged bases, and then two endonucleases XPF-ERCC1 and XPG incise the lesion on either side of the photodamage to release the damaged DNA section. Finally, using the undamaged strand as a template to ensure fidelity, DNA polymerase, PCNA, RFC and DNA ligase I act in concert to synthesize and ligate the new DNA fragment. In this manner, the NER pathway is the cell’s major defense against DNA damage and if defective, UV-induced mutations accumulate in the genome.

Figure 8.

UV-induced cyclobutane dimers- structure (A) and repair by the Nucleotide Excision DNA Repair (NER) pathway (B). The NER pathway is mediated by at least eight enzymes that work together to identify bulky DNA lesions that distort the structure of the double helix, excise the damaged portion and replace the excised region by DNA synthesis directed by the complementary strand. Homozygous deficiency in any one of the NER enzymes leads to the clinical condition known as Xeroderma Pigmentosum (XP). Please note that although not shown, NER can also be initiated in actively transcribed regions of the genome by involvement of the Cockayne syndrome proteins A and B.

As a result of the inability of the skin to recover after UV exposure, intense sun sensitivity is one of the first manifestations of XP. Estimated incidences vary from 1 in 20,000 in Japan to 1 in 250,000 in the US (Robbins et al., 1974). Beginning in the first or second year of life, UV-exposed skin (e.g. on the face and arms) develops areas of hypo- or hyper-pigmented macules, telangiectasias and atrophy, all signs of chronic sun exposure that normally take decades to develop. Premalignant lesions such as actinic keratoses develop, and typically malignancies such as basal cell carcinomas, squamous cell carcinomas and melanomas start appearing by the age of ten years. XP patients have more than a thousand-fold increased risk of skin cancer and develop malignancies decades earlier than unaffected patients (Van Patter and Drummond, 1953; Lynch et al., 1981; Cleaver, 2005; Jen et al., 2009; Rao et al., 2009; Wang et al., 2009). Melanomas isolated from XP patients clearly bear evidence of “UV signature mutations”, lending support to the concept that defective repair of UV-induced photodimers underlies carcinogenesis of melanocytes (Takebe et al., 1989; Sato et al., 1993). Beside skin cancer, XP patients suffer a 20-fold increased risk of other malignancies including lung cancer, gastric carcinoma and brain cancer, perhaps reflecting the importance of NER in the repair of damage produced by agents other than UV. Overall, approximately 70% of persons with XP die by the age of 40 years from cancer. Currently there is no accepted therapy for treating XP other than avoidance of sunlight and careful surveillance and local control of pre-malignant or malignant lesions as they appear. The use of topical DNA repair enzymes such asT4 endonuclease V which cleaves UV-induced photolesions(Cafardi and Elmets, 2008) and novel UV-protective strategies such as the pharmacologic induction of cutaneous melanin levels which block penetration of UV radiation (D'Orazio et al., 2006) are being developed and may hold great promise for these exceptionally UV-sensitive individuals.

Intriguingly, although the homozygous condition known as XP reveals much about the importance of the NER pathway in melanoma resistance and ability of UV radiation to fuel melanomagenesis, evidence is accumulating that polymorphisms in NER enzymes in the general (non-XP) population may influence melanoma risk. For example, several studies have found an association between polymorphisms in certain NER enzymes and melanoma including XPD (Tomescu et al., 2001; Mocellin et al., 2009), XPC and XPF (Winsey et al., 2000; Baccarelli et al., 2004; Blankenburg et al., 2005; Debniak et al., 2006). Some groups have posited that multiple NER variants in a single individual may be required to influence melanoma susceptibility (Li et al., 2006).

3.4. Melanocortin 1 Receptor (MC1R)

The MC1R is a seven transmembrane Gs-coupled protein that, when bound by melanocyte stimulating hormone (MSH), activates adenylyl cyclase and cAMP generation (Fig. 9). This cAMP second messenger signaling leads to activation of the protein kinase A (PKA) cascade which, in turn, leads to up-regulation of the MITF and CREB transcription factors that together induce expression of melanin biosynthetic enzymes such as tyrosinase and dopachrome tautomerase (Yasumoto et al., 1994; Bertolotto et al., 1996; Fang and Setaluri, 1999). In this manner, MC1R signaling enhances the production and export of melanin by melanocytes to maturing epidermal keratinocytes, thereby controlling the melanin levels and innate UV resistance of the skin (Fig. 9). When MC1R signaling is defective, then melanocytes alter the type and amount of melanin they manufacture. Specifically, a red/blonde sulfated pigment known as pheomelanin is produced rather than the brown/black eumelanin species. Pheomelanin is a much poorer blocker of UV photons and may even contribute to oxidative damage within melanocytes, itself a possible mutagenic mechanism.

Figure 9.

The central role of the melanocortin 1 receptor (MC1R) in the epidermal response to UV radiation. UV-induced cellular and DNA damage to epidermal keratinocytes induces activation of the global damage response protein p53, which mediates transcriptional activation of the pro-opiomelanocortin (POMC) gene. The POMC gene encodes a propeptide that is cleaved into melanocyte stimulating hormone (MSH) along with β-endorphin and adrenocorticotropic hormone (ACTH). MSH secreted from UV-exposed keratinocytes then is hypothesized to bind melanocortin 1 receptors (MC1R) on melanocytes in the basal epidermis. MSH binding induces generation of the second messenger cAMP via MC1R-mediated activation of adenylate cyclase in melanocytes. Generation of cAMP triggers a number of downstream events including activation of the protein kinase A signaling pathway and up-regulation of the cAMP responsive binding element (CREB) and microphthalmia (Mitf) transcription factors. CREB and Mitf induce melanin production through transcriptional up-regulation of melanin biosynthetic enzymes. Thus, MSH-MC1R signaling leads to enhanced pigment synthesis and subsequent transfer of melanin to epidermal keratinocytes. In this manner, the skin is more protected against subsequent UV exposure. MSH-MC1R signaling may also enhance nucleotide excision repair (NER) in melanocytes, which would enhance recovery from UV damage.

Loss-of-function polymorphisms have been identified in MC1R, with the vast majority of allelic variation occurring in European and Asian populations. The most prevalent MC1R mutations (D84E, R142H, R151C, R160W, and D294H) are known as the “RHC” (red hair color) alleles because of the association with a blonde/red hair color, freckling and tendency to burn rather than tan after UV exposure (Scherer and Kumar, 2010). RHC MC1R alleles are also associated with a relatively high lifetime risk of melanoma (increased odds ratio of 2.40 in one study) (Williams et al., 2011). MC1R variants may also modify other melanoma-relevant alleles (van der Velden et al., 2001; Demenais et al., 2010; Kanetsky et al., 2010; Kricker et al., 2010). In a Australian cohort, for example, co-inheritance of either the MC1R variants R151C, R160W or D294H with CDKN2A mutations and decreased latency for melanoma by approximately 20 years (Box et al., 2001). A more recent study found that MC1R variants significantly increased penetrance and lowered the age of melanoma diagnosis in people with CDKN2A mutations, (Fargnoli et al., 2010).

In addition to its role in skin melanization, MC1R may influence melanoma development by non-pigmentary pathways as well (Matichard et al., 2004; Goldstein et al., 2005). Specifically, we and others have found that MC1R signaling influences the ability of melanocytes to recover from UV-induced DNA damage (Hauser et al., 2006; Abdel-Malek et al., 2009; Song et al., 2009). MC1R signaling directly enhances NER in melanocytes, and studies are underway to discover the molecular mechanisms linking MC1R signaling to the NER DNA damage repair pathway. Overall, there is much evidence placing MC1R as a “master regulator” of melanocyte UV physiologic responses.

3.5. Microphthalmia (MITF)

Mitf is a myc-like transcription factor that is critical to melanocyte development and survival (Levy et al., 2006). Defective Mitf leads to disorders of melanocyte function and pigmentation (Fisher, 2000; Goding, 2000; Widlund and Fisher, 2003; Steingrimsson et al., 2004). In humans, for example, Waardenburg syndrome type 2 is caused by inactivating mutations of Mitf, and is characterized by pigmentary defects due to the congenital absence of melanocytes in distinct anatomic locations (hair, skin eyes) (Hughes et al., 1994; Tassabehji et al., 1994). Mitf Immunohistochemical staining has long been used to identify surgical tumor isolates as melanomas (King et al., 1999; Salti et al., 2000), but its oncogenic contribution to melanoma wasn’t realized until Garraway and colleagues reported Mitf to be amplified in a subset of melanomas, particularly in aggressive disease (Garraway et al., 2005). Mitf may be amplified in up to 20% of metastatic melanomas and is associated with activation of the hypoxia inducible factor (HIF1A) pathway (Busca et al., 2005; Cheli et al., 2012) and reduced patient survival (Ugurel et al., 2007).

More recently, melanoma predisposition due to point mutations of Mitf (rather than gene amplification) were described. The E318K Mitf variant correlated with a positive melanoma family history, multiple primary melanomas or risk of melanoma and renal cell carcinoma in the same patient. Mechanistically, it is thought that the E318K Mitf variant leads to gain-of-function in Mitf by impairing its SUMO-mediated clearance (Bertolotto et al., 2011; Yokoyama et al., 2011). Thus whether by increased gene doseage or increased protein stability, Mitf seems to be a relevant melanoma oncogene, and current research efforts are attempting to devise pharmacologic targeting of MITF (Flaherty et al., 2010).

Advertisement

4. Conclusions

An explosion of information regarding the molecular pathways involved in melanoma development has been witnessed in the last several years. The MAPkinase cascade, for example, has emerged as a critical oncogenic pathway that drives the majority of cases of melanoma. Gain of function mutations in BRAF, most notably the V600E point mutation that results in unregulated BRAF signaling, have been described in at least half of all cutaneous melanomas (Davies et al., 2002; Pollock and Meltzer, 2002; Pollock et al., 2003). Oncogenic BRAF mutations lead to constitutive activation of kinase activity of BRAF, providing continuous growth signals in the absence of extracellular stimuli (Nikolaou et al., 2012). In many melanomas in which BRAF has not been mutated or in melanomas treated with BRAF inhibitors, N-Ras oncogene upregulation has been observed, leading again to increased signaling through the MAPkinase cascade (Padua et al., 1984; van 't Veer et al., 1989; Ball et al., 1994; Carr and Mackie, 1994; Wagner et al., 1995; Goydos et al., 2005; Nazarian et al., 2010). Though targeted inhibition of the MAPkinase signaling pathway has led to significant advances in the treatment of melanoma (Flaherty et al., 2010; Falchook et al., 2012), to date, somatic inheritance of MAPkinase activating mutations leading to melanoma predisposition, have not been reported.

The incidence of melanoma has risen at an alarming rate over the last several decades. The reasons for this increase are unclear, but probably represent the confluence of a variety of environmental and inherited risk factors. Though significant progress has been made over the last several years in immunotherapy (Hodi et al., 2010; Kaplan, 2011; Wilson, 2011) and targeted kinase inhibition against melanoma (Flaherty et al., 2010; Chapman et al., 2011; Flaherty et al., 2012; Sosman et al., 2012), clearly it would be better to prevent development of disease in the first place. As our understanding of the molecular mechanisms that underlie the malignant degeneration of melanocytes expands, so hopefully will our ability to develop rational interventions to prevent the development of melanoma.

References

  1. 1. Abdel-malekZ. AARuweRKavanagh-starnerA. LKadekaroVSwopeet al2009alpha-MSH tripeptide analogs activate the melanocortin 1 receptor and reduce UV-induced DNA damage in human melanocytes." Pigment Cell Melanoma Res.
  2. 2. AndradyAP. JAucampA. FBaisC. LBallareL. OBjornet al2010Environmental effects of ozone depletion and its interactions with climate change: progress report, 2009." Photochem Photobiol Sci 93275294
  3. 3. AtkinsonS. KV. LMarlattL. EKimpeD. RLeanV. LTrudeauet al2011Environmental factors affecting ultraviolet photodegradation rates and estrogenicity of estrone and ethinylestradiol in natural waters." Arch Environ Contam Toxicol 60117
  4. 4. AutierP2004Perspectives in melanoma prevention: the case of sunbeds." Eur J Cancer 401623672376
  5. 5. BaccarelliADCalistaPMinghettiBMarinelliBAlbettiet al2004XPD gene polymorphism and host characteristics in the association with cutaneous malignant melanoma risk." Br J Cancer 902497502
  6. 6. BallN. JJ. JYohnJ. GMorelliD. ANorrisL. EGolitzet al1994Ras mutations in human melanoma: a marker of malignant progression." J Invest Dermatol 1023285290
  7. 7. BartkovaJJLukasPGuldbergJAlsnerA. FKirkinet al1996The p16–cyclin D/Cdk4-pRb pathway as a functional unit frequently altered in melanoma pathogenesis." Cancer Res 56(23): 5475-5483.
  8. 8. BeralVSEvansHShawand GMilton1983Cutaneous factors related to the risk of malignant melanoma." Br J Dermatol 1092165172
  9. 9. BergerA. HA. GKnudsonand P. PPandolfi2011A continuum model for tumour suppression." Nature 4767359163169
  10. 10. BertolottoCKBilleJ. POrtonneand RBallotti1996Regulation of tyrosinase gene expression by cAMP in B16 melanoma cells involves two CATGTG motifs surrounding the TATA box: implication of the microphthalmia gene product." J Cell Biol 1343747755
  11. 11. BertolottoCFLesueurSGiulianoTStrubMDe Lichyet al2011A SUMOylation-defective MITF germline mutation predisposes to melanoma and renal carcinoma." Nature 48073759498
  12. 12. BlankenburgSI. RKonigRMoessnerPLaspeK. MThomset al2005Assessment of 3 xeroderma pigmentosum group C gene polymorphisms and risk of cutaneous melanoma: a case-control study." Carcinogenesis 26610851090
  13. 13. BohmMIWolffT. EScholzenS. JRobinsonEHealyet al2005alpha-Melanocyte-stimulating hormone protects from ultraviolet radiation-induced apoptosis and DNA damage." J Biol Chem 280757955802
  14. 14. BoxN. FD. LDuffyWChenMStarkN. GMartinet al2001MC1R genotype modifies risk of melanoma in families segregating CDKN2A mutations." Am J Hum Genet 694765773
  15. 15. BrewerJ. DL. JChristensonA. LWeaverD. CDapprichR. HWeeniget al2011Malignant melanoma in solid transplant recipients: collection of database cases and comparison with surveillance, epidemiology, and end results data for outcome analysis." Arch Dermatol 1477790796
  16. 16. BudiyantoAMUedaTUedaand MIchihashi2002Formation of cyclobutane pyrimidine dimers and 8-oxo-7,8-dihydro-2’-deoxyguanosine in mouse and organ-cultured human skin by irradiation with broadband or with narrowband UVB." Photochem Photobiol 764397400
  17. 17. BullerD. BVCokkinidesH. IHallA. MHartmanMSaraiyaet al2011Prevalence of sunburn, sun protection, and indoor tanning behaviors among Americans: review from national surveys and case studies of 3 states." J Am Acad Dermatol 65(5 Suppl 1): S114123
  18. 18. BurkhartC. Gand C. NBurkhart2000Melanoma and insecticides: is there a connection?" J Am Acad Dermatol 42(2 Pt 1): 302-303.
  19. 19. BuscaREBerraCGaggioliMKhaledKBilleet al2005Hypoxia-inducible factor 1{alpha} is a new target of microphthalmia-associated transcription factor (MITF) in melanoma cells." J Cell Biol 17014959
  20. 20. CafardiJ. Aand C. AElmets2008T4 Endonuclease V: Review and application to dermatology." Expert Opin Biol Ther 86829838
  21. 21. CareyW. PJrC. JThompsonMSynnestvedtD. tGuerryAHalpern, et al1994Dysplastic nevi as a melanoma risk factor in patients with familial melanoma." Cancer 741231183125
  22. 22. CarrJand R. MMackie1994Point mutations in the N-ras oncogene in malignant melanoma and congenital naevi." Br J Dermatol 13117277
  23. 23. ChapmanP. BAHauschildCRobertJ. BHaanenPAsciertoet al2011Improved survival with vemurafenib in melanoma with BRAF 600Emutation." N Engl J Med 364(26): 2507-2516.
  24. 24. CheliYSGiulianoNFenouilleMAllegraVHofmanet al2012Hypoxia and MITF control metastatic behaviour in mouse and human melanoma cells." Oncogene 311924612470
  25. 25. ChinLJPomerantzand R. ADepinho1998The INK4a/ARF tumor suppressor: one gene--two products--two pathways." Trends Biochem Sci 238291296
  26. 26. ChoiKDLazovichBSouthwellJForsterS. JRolnicket al2010Prevalence and characteristics of indoor tanning use among men and women in the United States." Arch Dermatol 1461213561361
  27. 27. ClarkW. HJrR. RReimerMGreeneA. MAinsworthand M. J. Mastrangelo(1978Origin of familial malignant melanomas from heritable melanocytic lesions.’The B-K mole syndrome’." Arch Dermatol 1145732738
  28. 28. CleaverJ. E2005Cancer in xeroderma pigmentosum and related disorders of DNA repair." Nat Rev Cancer 57564573
  29. 29. CokkinidesVMWeinstockDLazovichEWardand MThun2009Indoor tanning use among adolescents in the US, 1998 to 2004." Cancer 1151190198
  30. 30. CrombieI. K1979Variation of melanoma incidence with latitude in North America and Europe." Br J Cancer 405774781
  31. 31. CustA. EM. AJenkinsCGoumasB. KArmstrongHSchmidet al2011Early-life sun exposure and risk of melanoma before age 40 years." Cancer Causes Control 226885897
  32. 32. DOrazioJ. A. , TNobuhisaRCuiMAryaMSpryet al2006Topical drug rescue strategy and skin protection based on the role of Mc1r in UV-induced tanning." Nature 4437109340344
  33. 33. DaviesHG. RBignellCCoxPStephensSEdkinset al2002Mutations of the BRAF gene in human cancer." Nature 4176892949954
  34. 34. De GruijlF. RJLongstrethMNorvalA. PCullenHSlaperet al2003Health effects from stratospheric ozone depletion and interactions with climate change." Photochem Photobiol Sci 211628
  35. 35. DebniakTR. JScottTHuzarskiTByrskiBMasojcet al2006XPD common variants and their association with melanoma and breast cancer risk." Breast Cancer Res Treat 982209215
  36. 36. DemenaisFHMohamdiVChaudruA. MGoldsteinJ. ANewton Bishop, et al. (2010Association of MC1R variants and host phenotypes with melanoma risk in CDKN2A mutation carriers: a GenoMEL study." J Natl Cancer Inst 1022015681583
  37. 37. DepryJ. LK. BReedR. HCook-norrisand J. DBrewer2011Iatrogenic immunosuppression and cutaneous malignancy." Clin Dermatol 296602613
  38. 38. DraelosZ. D2011Genetic mosaicism and Fitzpatrick skin type." J Cosmet Dermatol 103169170
  39. 39. EkwuemeD. UG. PGuyJrCLiS. HRimPParelkar, et al2011The health burden and economic costs of cutaneous melanoma mortality by race/ethnicity-United States, 2000 to 2006." J Am Acad Dermatol 65(5 Suppl 1): S133143
  40. 40. ElwoodJ. Mand H. KKoh1994Etiology, epidemiology, risk factors, and public health issues of melanoma." Curr Opin Oncol 62179187
  41. 41. EngelsE. AR. MPfeifferJ. FFraumeniJrB. LKasiskeA. KIsrani, et al2011Spectrum of cancer risk among US solid organ transplant recipients." JAMA 3061718911901
  42. 42. ErdmannFJLortet-tieulentJSchuzHZeebRGreinertet al2012International trends in the incidence of malignant melanoma 1953-2008-are recent generations at higher or lower risk?" Int J Cancer.
  43. 43. FalchookG. SK. DLewisJ. RInfanteM. SGordonN. JVogelzanget al2012Activity of the oral MEK inhibitor trametinib in patients with advanced melanoma: a phase 1 dose-escalation trial." Lancet Oncol 138782789
  44. 44. FangDand VSetaluri1999Role of microphthalmia transcription factor in regulation of melanocyte differentiation marker TRP-1." Biochem Biophys Res Commun 2563657663
  45. 45. FargnoliM. CSGandiniKPerisPMaisonneuveand SRaimondi2010MC1R variants increase melanoma risk in families with CDKN2A mutations: a meta-analysis." Eur J Cancer 46814131420
  46. 46. FinkC. Aand M. NBates2005Melanoma and ionizing radiation: is there a causal relationship?" Radiat Res 1645701710
  47. 47. FisherD. E2000Microphthalmia: a signal responsive transcriptional regulator in development." Pigment Cell Res 13 Suppl 8145149
  48. 48. FlahertyK. TF. SHodiand B. CBastian2010Mutation-driven drug development in melanoma." Curr Opin Oncol 223178183
  49. 49. FlahertyK. TIPuzanovK. BKimARibasG. AMcarthuret al2010Inhibition of mutated, activated BRAF in metastatic melanoma." N Engl J Med 3639809819
  50. 50. FlahertyK. TCRobertPHerseyPNathanCGarbeet al2012Improved survival with MEK inhibition in BRAF-mutated melanoma." N Engl J Med 3672107114
  51. 51. FortesCand EDe Vries2008Nonsolar occupational risk factors for cutaneous melanoma." Int J Dermatol 474319328
  52. 52. FountainJ. WMKarayiorgouM. SErnstoffJ. MKirkwoodD. RVlocket al1992Homozygous deletions within human chromosome band 921in melanoma." Proc Natl Acad Sci U S A 89(21): 10557-10561.
  53. 53. FranceschiSand MCristofolini1992Cutaneous malignant melanoma: epidemiological considerations." Semin Surg Oncol 86345352
  54. 54. GandiniSFSeraM. SCattaruzzaPPasquiniDAbeniet al2005Meta-analysis of risk factors for cutaneous melanoma: I. Common and atypical naevi." Eur J Cancer 4112844
  55. 55. GandiniSFSeraM. SCattaruzzaPPasquiniRZanettiet al2005Meta-analysis of risk factors for cutaneous melanoma: III. Family history, actinic damage and phenotypic factors." Eur J Cancer 411420402059
  56. 56. GarrawayL. AH. RWidlundM. ARubinGGetzA. JBergeret al2005Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma." Nature 4367047117122
  57. 57. GellerA. CGColditzSOliveriaKEmmonsCJorgensenet al2002Use of sunscreen, sunburning rates, and tanning bed use among more than 10 000 US children and adolescents." Pediatrics 109610091014
  58. 58. GilchrestB. AM. SEllerA. CGellerand MYaar1999The pathogenesis of melanoma induced by ultraviolet radiation." N Engl J Med 3401713411348
  59. 59. GodingC. R2000Mitf from neural crest to melanoma: signal transduction and transcription in the melanocyte lineage." Genes Dev 141417121728
  60. 60. GoldsteinA. MMChanMHarlandE. MGillandersN. KHaywardet al2006High-risk melanoma susceptibility genes and pancreatic cancer, neural system tumors, and uveal melanoma across GenoMEL." Cancer Res 662098189828
  61. 61. GoldsteinA. MMChanMHarlandN. KHaywardFDemenaiset al2007Features associated with germline CDKN2A mutations: a GenoMEL study of melanoma-prone families from three continents." J Med Genet 44299106
  62. 62. GoldsteinA. MM. CFraserW. HClarkJrand M. A. Tucker (1994Age at diagnosis and transmission of invasive melanoma in 23 families with cutaneous malignant melanoma/dysplastic nevi." J Natl Cancer Inst 861813851390
  63. 63. GoldsteinA. MM. TLandiSTsangM. CFraserD. JMunroeet al2005Association of MC1R variants and risk of melanoma in melanoma-prone families with CDKN2A mutations." Cancer Epidemiol Biomarkers Prev 14922082212
  64. 64. GoldsteinA. MJ. PStruewingAChidambaramM. CFraserand M. ATucker2000Genotype-phenotype relationships in U.S. melanoma-prone families with CDKN2A and CDK4 mutations." J Natl Cancer Inst 921210061010
  65. 65. GoydosJ. SBMannH. JKimE. MGabrielJAlsinaet al2005Detection of B-RAF and N-RAS mutations in human melanoma." J Am Coll Surg 2003362370
  66. 66. GrobJ. JJGouvernetDAymarAMostaqueM. HRomanoet al1990Count of benign melanocytic nevi as a major indicator of risk for nonfamilial nodular and superficial spreading melanoma." Cancer 662387395
  67. 67. HalpernA. CD. tGuerryD. EElderBTrockMSynnestvedtet al1993Natural history of dysplastic nevi." J Am Acad Dermatol 2915157
  68. 68. HanJG. AColditzand D. JHunter2006Risk factors for skin cancers: a nested case-control study within the Nurses’ Health Study." Int J Epidemiol 35615141521
  69. 69. HauserJ. EA. LKadekaroR. JKavanaghKWakamatsuSTerzievaet al2006Melanin content and MC1R function independently affect UVR-induced DNA damage in cultured human melanocytes." Pigment Cell Res 194303314
  70. 70. HealyENFlannaganARayCToddI. JJacksonet al2000Melanocortin-1-receptor gene and sun sensitivity in individuals without red hair." Lancet 355920910721073
  71. 71. HodiF. SS. J. ODayD. FMcdermottR. WWeberJ. ASosmanet al2010Improved survival with ipilimumab in patients with metastatic melanoma." N Engl J Med 3638711723
  72. 72. HollandE. AS. CBeatonT. MBeckerO. MGruletB. APeterset al1995Analysis of the 16gene, CDKN2, in 17 Australian melanoma kindreds." Oncogene 11(11): 2289-2294.
  73. 73. HolmanC. DB. KArmstrongand P. JHeenan1986Relationship of cutaneous malignant melanoma to individual sunlight-exposure habits." J Natl Cancer Inst 763403414
  74. 74. HolmanC. DI. MGibsonMStephensonand B. KArmstrong1983Ultraviolet irradiation of human body sites in relation to occupation and outdoor activity: field studies using personal UVR dosimeters." Clin Exp Dermatol 83269277
  75. 75. HornungR. LK. HMageeW. JLeeL. AHansenand Y. CHsieh2003Tanning facility use: are we exceeding Food and Drug Administration limits?" J Am Acad Dermatol 494655661
  76. 76. HughesA. EV. ENewtonX. ZLiuand A. PRead1994A gene for Waardenburg syndrome type 2 maps close to the human homologue of the microphthalmia gene at chromosome 312p14Nat Genet 7(4): 509-512.
  77. 77. IngramA. J1992Review of chemical and UV light-induced melanomas in experimental animals in relation to human melanoma incidence." J Appl Toxicol 1213943
  78. 78. JenMMMurphyand J. MGrant-kels2009Childhood melanoma." Clin Dermatol 276529536
  79. 79. KadekaroA. LR. JKavanaghKWakamatsuSItoM. APipitoneet al2003Cutaneous photobiology. The melanocyte vs. the sun: who will win the final round?" Pigment Cell Res 165434447
  80. 80. KambAN. AGruisJWeaver-feldhausQLiuKHarshmanet al1994A cell cycle regulator potentially involved in genesis of many tumor types." Science 2645157436440
  81. 81. KambADShattuck-eidensREelesQLiuN. AGruiset al1994Analysis of the 16gene (CDKN2) as a candidate for the chromosome 9p melanoma susceptibility locus." Nat Genet 8(1): 23-26.
  82. 82. KanetskyP. ASPanossianD. EElderDGuerryM. EMinget al2010Does MC1R genotype convey information about melanoma risk beyond risk phenotypes?" Cancer 1161024162428
  83. 83. KanjilalSW. EPierceallK. KCummingsM. LKripkeand H. NAnanthaswamy1993High frequency of 53mutations in ultraviolet radiation-induced murine skin tumors: evidence for strand bias and tumor heterogeneity." Cancer Res 53(13): 2961-2964.
  84. 84. KaplanM. G2011Ipilimumab plus dacarbazine in melanoma." N Engl J Med 3651312561257author reply 1257-1258.
  85. 85. KaragasM. RV. AStannardL. AMottM. JSlatteryS. KSpenceret al2002Use of tanning devices and risk of basal cell and squamous cell skin cancers." J Natl Cancer Inst 943224226
  86. 86. KingRK. NWeilbaecherGMcgillECooleyMMihmet al1999Microphthalmia transcription factor. A sensitive and specific melanocyte marker for MelanomaDiagnosis." Am J Pathol 1553731738
  87. 87. KinoKand HSugiyama2005UVR-induced G-C to C-G transversions from oxidative DNA damage." Mutat Res 571(1-2): 33-42.
  88. 88. KnudsonA. G1996Hereditary cancer: two hits revisited." J Cancer Res Clin Oncol 1223135140
  89. 89. KorcumA. FSSanliogluGAksuNTunceland NErin2009Radiotherapy-induced decreases in substance P levels may potentiate melanoma growth." Mol Med Report 22319326
  90. 90. KrengelSAHauschildand TSchafer2006Melanoma risk in congenital melanocytic naevi: a systematic review." Br J Dermatol 155118
  91. 91. KrickerAB. KArmstrongCGoumasPKanetskyR. PGallagheret al2010MC1R genotype may modify the effect of sun exposure on melanoma risk in the GEM study." Cancer Causes Control.
  92. 92. LangardSJRosenbergAAndersenand S. SHeldaas2000Incidence of cancer among workers exposed to vinyl chloride in polyvinyl chloride manufacture." Occup Environ Med 5716568
  93. 93. LazovichDR. IVogelMBerwickM. AWeinstockK. EAndersonet al2010Indoor tanning and risk of melanoma: a case-control study in a highly exposed population." Cancer Epidemiol Biomarkers Prev 19615571568
  94. 94. LeeJ. Aand JScotto1993Melanoma: linked temporal and latitude changes in the United States." Cancer Causes Control 45413418
  95. 95. LeibelingDPLaspeand SEmmert2006Nucleotide excision repair and cancer." J Mol Histol 37(5-7): 225-238.
  96. 96. LensM. Band MDawes2004Global perspectives of contemporary epidemiological trends of cutaneous malignant melanoma." Br J Dermatol 1502179185
  97. 97. LevyCMKhaledand D. EFisher2006MITF: master regulator of melanocyte development and melanoma oncogene." Trends Mol Med 129406414
  98. 98. LewR. AA. JSoberNCookRMarvelland T. BFitzpatrick1983Sun exposure habits in patients with cutaneous melanoma: a case control study." J Dermatol Surg Oncol 912981986
  99. 99. LiCZHuZLiuL. EWangS. SStromet al2006Polymorphisms in the DNA repair genes XPC, XPD, and XPG and risk of cutaneous melanoma: a case-control analysis." Cancer Epidemiol Biomarkers Prev 151225262532
  100. 100. LieJ. AKKjaerheimand TTynes2008Ionizing radiation exposure and cancer risk among Norwegian nurses." Eur J Cancer Prev 174369375
  101. 101. LiggettW. HJrand D. Sidransky (1998Role of the 16tumor suppressor gene in cancer." J Clin Oncol 16(3): 1197-1206.
  102. 102. LucasR. MA. JMcmichaelB. KArmstrongand W. TSmith2008Estimating the global disease burden due to ultraviolet radiation exposure." Int J Epidemiol 373654667
  103. 103. LundbergIAGustavssonBHolmbergGMolinaand PWesterholm1993Mortality and cancer incidence among PVC-processing workers in Sweden." Am J Ind Med 232313319
  104. 104. LynchH. TB. CFrichotRdand J. F. Lynch (1978Familial atypical multiple mole-melanoma syndrome." J Med Genet 155352356
  105. 105. LynchH. TRFusaroJEdlundWAlbanoand JLynch1981Skin cancer developing in xeroderma pigmentosum patient relaxing sunlight avoidance." Lancet 2(8257): 1230.
  106. 106. MacKie R. M. and T. Aitchison (1982Severe sunburn and subsequent risk of primary cutaneous malignant melanoma in scotland." Br J Cancer 466955960
  107. 107. MaddodiNand VSetaluri2008Role of UV in cutaneous melanoma." Photochem Photobiol 842528536
  108. 108. MagnaniCDCoggonCOsmondand E. DAcheson1987Occupation and five cancers: a case-control study using death certificates." Br J Ind Med 4411769776
  109. 109. MakinJ2011Implications of climate change for skin cancer prevention in Australia." Health Promot J Austr 22 Spec S39-413941
  110. 110. MalaguarneraLECristaldiand MMalaguarnera2010The role of immunity in elderly cancer." Crit Rev Oncol Hematol 7414060
  111. 111. MarghoobA. AA. LAgeroCBenvenuto-andradeand S. WDusza2006Large congenital melanocytic nevi, risk of cutaneous melanoma, and prophylactic surgery." J Am Acad Dermatol 545868870discussion 871-863.
  112. 112. MarksR1995The epidemiology of non-melanoma skin cancer: who, why and what can we do about it." J Dermatol 2211853857
  113. 113. MatichardEPVerpillatRMezianiBGerardVDescampset al2004Melanocortin 1 receptor (MC1R) gene variants may increase the risk of melanoma in France independently of clinical risk factors and UV exposure." J Med Genet 41(2): e13.
  114. 114. MckenzieR. LP. JAucampA. FBaisL. OBjornMIlyaset al2011Ozone depletion and climate change: impacts on UV radiation." Photochem Photobiol Sci 102182198
  115. 115. MclaughlinJ. KH. SMalkerW. JBlotJ. LEricssonGGemneet al1988Malignant melanoma in the printing industry." Am J Ind Med 132301304
  116. 116. MeliaJSMossDColemanTFrostRGraham-brownet al2001The relation between mortality from malignant melanoma and early detection in the Cancer Research Campaign Mole Watcher Study." Br J Cancer 856803807
  117. 117. MetcalfJ. Sand J. CMaize1985Melanocytic nevi and malignant melanoma." Dermatol Clin 32217224
  118. 118. MeyleK. Dand PGuldberg2009Genetic risk factors for melanoma." Hum Genet 1264499510
  119. 119. MeyskensF. Land SYang2011Thinking about the role (largely ignored) of heavy metals in cancer prevention: hexavalent chromium and melanoma as a case in point." Recent Results Cancer Res 1886574
  120. 120. MoanJADahlbackZLagunovaECicarmaand A. CPorojnicu2009Solar radiation, vitamin D and cancer incidence and mortality in Norway." Anticancer Res 29935013509
  121. 121. MocellinSDVerdiand DNitti2009DNA repair gene polymorphisms and risk of cutaneous melanoma: a systematic review and meta-analysis." Carcinogenesis 301017351743
  122. 122. MouretSCBaudouinMCharveronAFavierJCadetet al2006Cyclobutane pyrimidine dimers are predominant DNA lesions in whole human skin exposed to UVA radiation." Proc Natl Acad Sci U S A 103371376513770
  123. 123. NaeyaertJ. Mand LBrochez2003Clinical practice. Dysplastic nevi." N Engl J Med 3492322332240
  124. 124. NakabeppuYKSakumiKSakamotoDTsuchimotoTTsuzukiet al2006Mutagenesis and carcinogenesis caused by the oxidation of nucleic acids." Biol Chem 3874373379
  125. 125. NatarajA. JH. SBlackand H. NAnanthaswamy1996Signature p53 mutation at DNA cross-linking sites in 8-methoxypsoralen and ultraviolet A (PUVA)-induced murine skin cancers." Proc Natl Acad Sci U S A 931579617965
  126. 126. NazarianRHShiQWangXKongR. CKoyaet al2010Melanomas acquire resistance to B-RAF(600Einhibition by RTK or N-RAS upregulation." Nature 468(7326): 973-977.
  127. 127. NelsonA. Aand HTsao2009Melanoma and genetics." Clin Dermatol 2714652
  128. 128. Newton-bishopJ. AY. MChangFElliottMChanSLeakeet al2011Relationship between sun exposure and melanoma risk for tumours in different body sites in a large case-control study in a temperate climate." Eur J Cancer 475732741
  129. 129. Newton-bishopJ. AY. MChangM. MIlesJ. CTaylorBBakkeret al2010Melanocytic nevi, nevus genes, and melanoma risk in a large case-control study in the United Kingdom." Cancer Epidemiol Biomarkers Prev 19820432054
  130. 130. Newton BishopJ. A., M. Harland, D. C. Bennett, V. Bataille, A. M. Goldstein, et al. (1999Mutation testing in melanoma families: INK4A, CDK4 and INK4D." Br J Cancer 80(1-2): 295-300.
  131. 131. NikolaouV. AA. JStratigosK. TFlahertyand HTsao2012Melanoma: new insights and new therapies." J Invest Dermatol 132(3 Pt 2): 854-863.
  132. 132. NoboriTKMiuraD. JWuALoisKTakabayashiet al1994Deletions of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers." Nature 3686473753756
  133. 133. NoonanF. PJ. ARecioHTakayamaPDurayM. RAnveret al2001Neonatal sunburn and melanoma in mice." Nature 4136853271272
  134. 134. NorvalMR. MLucasA. PCullenF. RDe GruijlJLongstrethet al2011The human health effects of ozone depletion and interactions with climate change." Photochem Photobiol Sci 102199225
  135. 135. OhtaniNZZebedeeT. JHuotJ. AStinsonMSugimotoet al2001Opposing effects of Ets and Id proteins on 16INK4aexpression during cellular senescence." Nature 409(6823): 1067-1070.
  136. 136. PaduaR. ANBarrassand G. ACurrie1984A novel transforming gene in a human malignant melanoma cell line." Nature 3115987671673
  137. 137. PalmerJ. SD. LDuffyN. FBoxJ. FAitkenL. E. OGormanet al2000Melanocortin-1 receptor polymorphisms and risk of melanoma: is the association explained solely by pigmentation phenotype?" Am J Hum Genet 661176186
  138. 138. ParkS. LLLeMarchandL. RWilkensL. NKolonelB. E. Henderson, et al. (2012Risk factors for malignant melanoma in white and non-white/non-African American populations: the multiethnic cohort." Cancer Prev Res (Phila) 53423434
  139. 139. PayneS. Rand C. JKemp2005Tumor suppressor genetics." Carcinogenesis 261220312045
  140. 140. PfeiferG. PY. HYouand ABesaratinia2005Mutations induced by ultraviolet light." Mutat Res 571(1-2): 19-31.
  141. 141. PollockP. MU. LHarperK. SHansenL. MYudtMStarket al2003High frequency of BRAF mutations in nevi." Nat Genet 3311920
  142. 142. PollockP. Mand P. SMeltzer2002A Genome-based strategy uncovers frequent BRAF mutations in melanoma." Cancer Cell 2157
  143. 143. PsatyE. LAScopeA. CHalpernand A. AMarghoob2010Defining the patient at high risk for melanoma." Int J Dermatol 494362376
  144. 144. RadosC2005Teen tanning hazards." FDA Consum 39289
  145. 145. RanadeKC. JHussussianR. SSikorskiH. EVarmusA. MGoldsteinet al1995Mutations associated with familial melanoma impair 16INK4function." Nat Genet 10(1): 114-116.
  146. 146. RaoT. NABhagyalaxmiKAhmedT. SMohana Rao and K. Venkatachalam (2009A case of melanoma in xeroderma pigmentosum." Indian J Pathol Microbiol 524524526
  147. 147. RavnbakM. HP. APhilipsenand H. CWulf2010The minimal melanogenesis dose/minimal erythema dose ratio declines with increasing skin pigmentation using solar simulator and narrowband ultraviolet B exposure." Photodermatol Photoimmunol Photomed 263133137
  148. 148. RecioJ. AF. PNoonanHTakayamaM. RAnverPDurayet al2002Ink4a/arf deficiency promotes ultraviolet radiation-induced melanomagenesis." Cancer Res 622267246730
  149. 149. ReedK. BJ. DBrewerC. MLohseK. EBringeC. NPruittet al2012Increasing incidence of melanoma among young adults: an epidemiological study in Olmsted County, Minnesota." Mayo Clin Proc 874328334
  150. 150. ReesJ. L2000The melanocortin 1 receptor (MC1R): more than just red hair." Pigment Cell Res 133135140
  151. 151. ReesJ. Land EHealy1997Melanocortin receptors, red hair, and skin cancer." J Investig Dermatol Symp Proc 219498
  152. 152. RigelD. S2002The effect of sunscreen on melanoma risk." Dermatol Clin 204601606
  153. 153. RigelD. S2010Trends in dermatology: melanoma incidence." Arch Dermatol 146(3): 318.
  154. 154. RobbinsJ. HK. HKraemerM. ALutznerB. WFestoffand H. GCoon1974Xeroderma pigmentosum. An inherited diseases with sun sensitivity, multiple cutaneous neoplasms, and abnormal DNA repair." Ann Intern Med 802221248
  155. 155. RobinsonSSDixonSAugustBDiffeyKWakamatsuet al2010Protection against UVR involves MC1R-mediated non-pigmentary and pigmentary mechanisms in vivo." J Invest Dermatol 130719041913
  156. 156. SaltiG. ITManougianMFarolanAShilkaitisDMajumdaret al2000Micropthalmia transcription factor: a new prognostic marker in intermediate-thickness cutaneous malignant melanoma." Cancer Res 601850125016
  157. 157. SarasinA1999The molecular pathways of ultraviolet-induced carcinogenesis." Mutat Res 428(1-2): 5-10.
  158. 158. SatoMCNishigoriMZghalTYagiand HTakebe1993Ultraviolet-specific mutations in 53gene in skin tumors in xeroderma pigmentosum patients." Cancer Res 53(13): 2944-2946.
  159. 159. SchererDand RKumar2010Genetics of pigmentation in skin cancer--a review." Mutat Res 7052141153
  160. 160. SchmalwieserA. WGSchaubergerMJanouchMNunezTKoskelaet al2005Global forecast model to predict the daily dose of the solar erythemally effective UV radiation." Photochem Photobiol 811154162
  161. 161. SchulmanJ. Mand D. EFisher2009Indoor ultraviolet tanning and skin cancer: health risks and opportunities." Curr Opin Oncol 212144149
  162. 162. ScottoJand T. RFears1987The association of solar ultraviolet and skin melanoma incidence among caucasians in the United States." Cancer Invest 54275283
  163. 163. SerranoMG. JHannonand DBeach1993A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4." Nature 3666456704707
  164. 164. SeykoraJand DElder1996Dysplastic nevi and other risk markers for melanoma." Semin Oncol 236682687
  165. 165. SharplessEand LChin2003The INK4a/ARF locus and melanoma." Oncogene 222030923098
  166. 166. SharplessN. Eand R. ADepinho1999The INK4A/ARF locus and its two gene products." Curr Opin Genet Dev 912230
  167. 167. SiegelRCDesantisKVirgoKSteinAMariottoet al2012Cancer treatment and survivorship statistics, 2012." CA Cancer J Clin 624220241
  168. 168. SilverbergM. JCChaoW. ALeydenLXuM. AHorberget al2011HIV infection, immunodeficiency, viral replication, and the risk of cancer." Cancer Epidemiol Biomarkers Prev 201225512559
  169. 169. SladeJA. AMarghoobT. GSalopekD. SRigelA. WKopfet al1995Atypical mole syndrome: risk factor for cutaneous malignant melanoma and implications for management." J Am Acad Dermatol 323479494
  170. 170. SmithM. BHXueLStrongHTakahashiNJaffeet al1993Forty-year experience with second malignancies after treatment of childhood cancer: analysis of outcome following the development of the second malignancy." J Pediatr Surg 281013421348discussion 1348-1349.
  171. 171. SoehngeHAOuhtitand O. NAnanthaswamy1997Mechanisms of induction of skin cancer by UV radiation." Front Biosci 2: d538551
  172. 172. SongXNMosbyJYangAXuZAbdel-maleket al2009alpha-MSH activates immediate defense responses to UV-induced oxidative stress in human melanocytes." Pigment Cell Melanoma Res 226809818
  173. 173. SosmanJ. AK. BKimLSchuchterRGonzalezA. CPavlicket al2012Survival in BRAF 600-mutantadvanced melanoma treated with vemurafenib." N Engl J Med 366(8): 707-714.
  174. 174. SoufirNM. FAvrilAChompretFDemenaisJBombledet al1998Prevalence of 16and CDK4 germline mutations in 48 melanoma-prone families in France. The French Familial Melanoma Study Group." Hum Mol Genet 7(2): 209-216.
  175. 175. SteingrimssonEN. GCopelandand N. AJenkins2004Melanocytes and the microphthalmia transcription factor network." Annu Rev Genet 38365411
  176. 176. SternR. S2001The risk of melanoma in association with long-term exposure to PUVA." J Am Acad Dermatol 445755761
  177. 177. SturmR. A2002Skin colour and skin cancer- MC1R, the genetic link." Melanoma Res 125405416
  178. 178. SullivanA. NE. ABryantand L. AMark2012Malignant melanoma in transplant patients: a case report and review of the literature." Cutis 893133136
  179. 179. TakebeHCNishigoriand KTatsumi1989Melanoma and other skin cancers in xeroderma pigmentosum patients and mutation in their cells." J Invest Dermatol 92(5 Suppl): 236S-238S.
  180. 180. TassabehjiMV. ENewtonand A. PRead1994Waardenburg syndrome type 2 caused by mutations in the human microphthalmia (MITF) gene." Nat Genet 83251255
  181. 181. TomescuDGKavanaghTHaHCampbelland D. WMelton2001Nucleotide excision repair gene XPD polymorphisms and genetic predisposition to melanoma." Carcinogenesis 223403408
  182. 182. TomlinsonI. PRRoylanceand R. SHoulston2001Two hits revisited again." J Med Genet 3828185
  183. 183. TsaoHCBevonaWGogginsand TQuinn2003The transformation rate of moles (melanocytic nevi) into cutaneous melanoma: a population-based estimate." Arch Dermatol 1393282288
  184. 184. TuckerM. AW. ACrutcherPHartgeand R. WSagebiel1993Familial and cutaneous features of dysplastic nevi: a case-control study." J Am Acad Dermatol 284558564
  185. 185. UdayakumarDand HTsao2009Melanoma genetics: an update on risk-associated genes." Hematol Oncol Clin North Am 233415429vii.
  186. 186. UgurelSRHoubenDSchramaHVoigtMZapatkaet al2007Microphthalmia-associated transcription factor gene amplification in metastatic melanoma is a prognostic marker for patient survival, but not a predictive marker for chemosensitivity and chemotherapy response." Clin Cancer Res 132163446350
  187. 187. ValverdePEHealySSikkinkFHaldaneA. JThodyet al1996The Asp84Glu variant of the melanocortin 1 receptor (MC1R) is associated with melanoma." Hum Mol Genet 51016631666
  188. 188. van’tVeerL. J. , B. MBurgeringRVersteegA. JBootD. JRuiteret al1989N-ras mutations in human cutaneous melanoma from sun-exposed body sites." Mol Cell Biol 9731143116
  189. 189. Van Der LeunJ. CR. DPiacentiniand F. RDe Gruijl2008Climate change and human skin cancer." Photochem Photobiol Sci 76730733
  190. 190. Van Der VeldenP. AL. ASandkuijlWBergmanSPavelLVan Mouriket al2001Melanocortin-1 receptor variant R151C modifies melanoma risk in Dutch families with melanoma." Am J Hum Genet 694774779
  191. 191. Van PatterH. Tand J. ADrummond1953Malignant melanoma occurring in xeroderma pigmentosum; report of a case." Cancer 65942947
  192. 192. VincetiMSBassissiCMalagoliGPellacaniDAlberet al2005Environmental exposure to trace elements and risk of cutaneous melanoma." J Expo Anal Environ Epidemiol 155458462
  193. 193. VolkovovaKDBilanicovaABartonovaSLetasiovaand MDusinska2012Associations between environmental factors and incidence of cutaneous melanoma. Review." Environ Health 11 Suppl 1: S12.
  194. 194. WagnerS. NH. MOckenfelsCWagnerHHoflerand MGoos1995Ras gene mutations: a rare event in nonmetastatic primary malignant melanoma." J Invest Dermatol 1045868871
  195. 195. WangYJ. JDigiovannaJ. BSternT. JHornyakMRaffeldet al2009Evidence of ultraviolet type mutations in xeroderma pigmentosum melanomas." Proc Natl Acad Sci U S A 1061562796284
  196. 196. WardK. ADLazovichand M. KHordinsky2012Germline melanoma susceptibility and prognostic genes: A review of the literature." J Am Acad Dermatol.
  197. 197. Weaver-feldhausJN. AGruisSNeuhausenDLePaslierEStockert, et al. (1994Localization of a putative tumor suppressor gene by using homozygous deletions in melanomas." Proc Natl Acad Sci U S A 911675637567
  198. 198. WeinstockM. A2012Reducing death from melanoma and standards of evidence." J Invest Dermatol 132513111312
  199. 199. WeiskopfDBWeinbergerand BGrubeck-loebenstein2009The aging of the immune system." Transpl Int 221110411050
  200. 200. WidlundH. Rand D. EFisher2003Microphthalamia-associated transcription factor: a critical regulator of pigment cell development and survival." Oncogene 222030353041
  201. 201. WilliamsP. FC. MOlsenN. KHaywardand D. CWhiteman2011Melanocortin 1 receptor and risk of cutaneous melanoma: a meta-analysis and estimates of population burden." Int J Cancer 129717301740
  202. 202. WilsonK. S2011Ipilimumab plus dacarbazine in melanoma." N Engl J Med 365(13): 1256; author reply 12571258
  203. 203. WinseyS. LN. AHaldarH. PMarshMBunceS. EMarshallet al2000A variant within the DNA repair gene XRCC3 is associated with the development of melanoma skin cancer." Cancer Res 602056125616
  204. 204. WoodwardAand PBoffetta1997Environmental exposure, social class, and cancer risk." IARC Sci Publ(138): 361-367.
  205. 205. YasumotoKKYokoyamaKShibataYTomitaand SShibahara1994Microphthalmia-associated transcription factor as a regulator for melanocyte-specific transcription of the human tyrosinase gene." Mol Cell Biol 141280588070
  206. 206. YokoyamaSS. LWoodsG. MBoyleL. GAoudeSMacGregor, et al. (2011A novel recurrent mutation in MITF predisposes to familial and sporadic melanoma." Nature 480737599103
  207. 207. ZaidiM. RE. CDe FaboF. PNoonanand GMerlino2012Shedding light on melanocyte pathobiology in vivo." Cancer Res 72715911595
  208. 208. ZuoLJWegerQYangA. MGoldsteinM. ATuckeret al1996Germline mutations in the 16INK4abinding domain of CDK4 in familial melanoma." Nat Genet 12(1): 97-99.

Written By

John A. D’Orazio, Stuart Jarrett, Amanda Marsch, James Lagrew and Laura Cleary

Submitted: 25 April 2012 Published: 13 February 2013