Abstract
Malignant melanoma remains one of the fastest growing cancers worldwide. Although the primary cutaneous melanoma can be managed by surgery, the advanced metastatic melanoma cannot be managed by surgery alone and thus, requires better therapeutic approaches. In view of high mortality rates due to metastatic melanoma, better understanding of the molecular pathogenesis of malignant melanoma is urgently needed. Such information is expected to prove very valuable in early detection of potential metastatic lesions and developing newer therapeutic approaches in order to better manage this malignancy. This article reviews the available information on the molecular changes associated with malignant melanoma and discusses the potential of such information in facilitating the development of newer anti-melanoma therapeutics. Current state of knowledge and the future of traditional and newly approved anti-melanoma therapeutics are also discussed.
Keywords: Melanoma, BRAF, Vemurafenib, Dabrafenib, Trametinib, Cobimetinib
Introduction
Cutaneous melanoma is a malignant neoplasm of the melanocytes. Melanocytes, found in the basal layer of the epidermis, produce the pigment melanin that is responsible for skin color. It is estimated that in the US for the year 2014, about 76,100 new cases of melanoma of the skin will be diagnosed and about 9,710 people will die due to this disease this year alone (1). Among the Caucasian population, the incidence of malignant melanoma has increased 3–6% over the last few decades (2), making this one of the fastest growing cancers worldwide (3).
As one of the three major types of skin cancer, with basal cell carcinoma and squamous cell carcinoma being the other two, melanoma remains the deadliest, causing the vast majority of the skin cancer-related deaths even though it accounts for approximately 5% of the skin cancer cases (4, 5). Primary melanoma patients demonstrate approximately an 11% mortality rate (2, 6), whereas the mortality rate due to metastatic melanoma is significantly higher. Metastatic melanoma patients typically have a low survival rate due to the poor efficacies of current cancer therapies. The extreme difficulties encountered in therapeutic management of melanoma patients have prompted large scale efforts to elucidate the molecular pathogenesis of malignant melanoma in hopes to finding more effective treatment options.
Classification and Staging
There are four major forms of melanoma including (i) superficial spreading, (ii) nodular, (iii) lentigo maligna and (iv) acral lentiginous melanomas. Of these, the superficial spreading form remains the most common and accounts for about 70% of melanomas followed by nodular form that represents about 15–30% of melanoma cases. The lentigo maligna and acral lentiginous forms represent less than 10% of melanoma cases (7, 8). In terms of staging, four systems are followed including (i) the Clark scale, (ii) the Breslow scale, (iii) TNM staging and (iv) Number stages. The Clark scale evaluates the depth of lesion in terms of it affecting various skin layers. The Breslow scale evaluates as to how thick the melanoma is in the skin. The TNM (Tumor, Node, Metastases) staging is based on thickness of the lesion and evaluation of its spread to lymph nodes and different tissues in the body and is also used for clinical staging per the American Joint Committee on Cancer (AJCC). The number staging system (Stage 0 to Stage 4) couples information on depth of the lesion and the TNM staging. For example, Stage 0 implies that the lesion is confined to epidermis (in situ) without spread to deeper layers such as dermis whereas Stage 4, the other extreme, indicates spread to lymph nodes and metastases to distant parts of the body such as lung, liver or brain (7, 9).
Diagnosis
Diagnosis of melanoma can be accomplished through clinician assessment of the skin lesion with the unaided eye. Clinicians frequently assess lesions based on the “ABCDE rule” that is meant to indicate A: asymmetry, B: irregular border, C: color variations, D: diameter >6 mm, and E: elevated surface (5, 7, 10). However, diagnosis with the unaided eye is not always accurate as seen in the approximately 80% accurate diagnosis rate demonstrated amongst dermatologists, and approximately 30% rate for non-dermatological specialists (5). Detection processes have become more technical over time in order to improve efficacy and limit the number of false negative cases that would allow undetected melanoma to develop to dangerous stages. The use of a skin surface microscope or a dermoscope allows improved visualization of the lesion (11, 12). Development of advanced digital systems have allowed for the development of an automated melanoma diagnosis system termed MEDS, which integrates multiple classification algorithms and uses them to analyze different measurements and characteristics of the patient lesion to produce effective diagnoses (5).
Research in melanoma diagnostics has also focused on detecting melanoma-specific biological markers that may help predict the course of the disease. Assaying the blood of melanoma patients who had been deemed cancer free for melanoma cells and other mRNA markers helps predict the probability that the patient will experience remission of metastatic melanoma (13). Another molecular marker that can be used for diagnostic purposes is Microphthalmia transcription factor (Mitf) (14). Mitf has been found to be uniquely expressed in melanoma melanocytes. In addition, Mitf antibody has been reported to detect and stain melanocytic lesions that conventional markers HMB-45 or S-100 failed to stain (14).
Etiology and Pathogenesis
Ultraviolet Radiation
Epidemiological studies have done much to shed light on the possible causes of malignant melanoma. Studies have shown that a major risk factor for melanoma development is exposure to Ultraviolet (UV) radiation exposure. One or more blistering sunburn during childhood or adolescence more than doubles the risk for melanoma in later life (15). This suggests that UV exposure plays an important role in melanoma tumorigenesis. Additional experiments have shown that UV radiation frequently leads to DNA mutations, such as the formation of pyrimidine dimers or deamination of cytosine into thymidine (16, 17). Cutaneous melanoma samples demonstrate a high base mutation rate that exceeds that of almost every other form of solid cancer, which may be attributed to the potency of UV mutagenic effects (18–20). Individuals who have history of familial melanoma, which contributes to 8–12% of melanoma cases, demonstrate a high sensitivity to UV radiation (21–24). These individuals are more likely to develop melanoma earlier in age and to develop multiple melanoma lesions.
Melanocytic Nevi
Another risk factor for malignant melanoma is the melanocytic nevus. Nevi are benign lesions composed of a concentrated amount of melanocytes. This leads to a dark pigmented spot on the skin due to the large amount of melanin that is produced. Nevi are colloquially known as a mole or birthmark. Melanocytic nevi can be congenital or appear later in life. Atypical or excessive amounts of nevi acquired later in life are a potent risk factor or diagnostic feature indicating melanoma development (25). If nevi begin to change in shape, color, or texture then it becomes a strong indicator for malignant melanoma. In one study, about 81% of melanoma patients observed a changing nevus in the location of the malignant lesion (26). However, it is important to note that not all changing nevi lead to or are indicative of melanoma, and that the exact mechanism and cause for the changes are still not understood. The indication of changing nevi allows for early melanoma detection, and with the proper surgical excision, the cancer free survival rate is extremely high compared to metastatic melanoma.
Molecular Changes Associated with Malignant Melanoma
After the melanoma spreads or metastasizes from its origin into other cutaneous or subcutaneous tissues, the response rate to treatment plummets to approximately 5–20%, and the 10-year survival rate becomes only about 10% (27, 28). Metastatic melanoma has been especially difficult to treat, showing low cure and survival rates after surgical resection and radiation therapy (27). At the cellular level, cancer cells possess distinguishing molecular properties that allow for apoptosis evasion, limitless growth potential without the need for growth factors, angiogenesis, and metastasis (29). Identifying the specific molecular changes that allow melanoma cells to have a growth and survival advantage over others may allow for the development of more effective targeted therapies that can improve the prognosis of melanoma patients.
BRAF
Studies have shown that around 40–60% of all melanoma cases exhibit an activated BRAF mutation (30). BRAF is a proto-oncogene that encodes a serine/threonine protein kinase as part of the RAS-RAF-MEK-ERK kinase pathway, which promotes cell growth and proliferation (31, 32). Normally, BRAF participates in homo- or heterodimerization with another RAF kinase in response to growth signals. However, an activating mutation leads BRAF to act as a self-sufficient and continuously activated monomer (33). The result is uncontrolled cell proliferation and that may play a role in tumor development and growth. The V600E missense mutation, that changes valine into glutamic acid, is the most common BRAF alteration and constitutes about 90% of BRAF activating mutations or about 50% of all metastatic melanomas (30, 34, 35). The V600K is the second most common mutation that results in valine to lysine change. Interestingly, BRAF mutations, such as the V600E amino acid substitution are also exhibited in 68% of benign nevi (36). This evidence raises the possibility that BRAF mutations might not be involved in melanoma carcinogenesis. Because nevi are usually stable after their formation, some researchers instead suspect that BRAF mutations instead play a part in melanocytic neoplasm formation (36).
Further research has revealed that the early stage of melanoma formation, known as the radial growth phase, exhibits a low 10% BRAF mutation rate, which supports the hypothesis that BRAF does not play a part in melanoma initiation (37). At the same time, 60–70% of vertical growth lesions and metastasized melanomas possess BRAF mutations, which suggests that this oncogenic mutation may be involved in cancer progression (36–39). The existence of activating BRAF in metastasized melanoma is consistent with the proposed BRAF function of cell growth promotion. The high prevalence of the BRAFV600E mutation in melanoma patients makes this a prime target for anti-melanoma therapeutics.
NRAS
Another source of molecular changes that may allow for melanoma initiation or propagation is found in the NRAS GTPase or neuroblastoma RAS oncogene. NRAS GTPase mutations are found in 15–20% of melanoma patients and activating mutations are usually found in the Q60/61 and G12/13 codons (40–42). NRAS is also a constituent of the RAS-RAF-mitogen activating kinase pathway, and is specifically part of the class of Ras proteins that modulate Raf protein activity (32, 35). Therefore, both NRAS and BRAF are found on the same pathway. Interestingly, concurrent mutations in both BRAF and NRAS are rarely found in the same patient, although some melanoma patients who harbored BRAF mutations and failed BRAF inhibitor treatment did display NRAS mutations in melanomas that had progressed (43). These findings suggest that the underlying mechanisms of melanoma development may be redundant particularly in context to BRAF and NRAS.
The patients with NRAS mutations possess thick vertical growth tumors that researchers attribute to the heightened cell proliferations rates induced by the NRAS mutation (40). These characteristics that NRAS mutations confer to melanoma tumors are similar those of BRAF. Like BRAF mutations, NRAS mutations are also found in high rates in benign nevi, which may limit the possibility that a NRAS mutation acts in melanoma initiation (36, 41, 44). Mutation analysis of primary and metastatic tumor samples show that metastatic tumors exhibited a higher incidence of BRAF and NRAS mutations compared to primary tumors (32). In addition, BRAF and NRAS mutations in metastatic melanomas were correlated with lower survival rates (32). Collectively, these findings seem to suggest that both of these mutations in the MAPK pathway may be involved in promoting the transition of primary melanoma into the metastatic variant. Because NRAS mutations are also linked to uncontrolled proliferation, NRAS can also serve as a favorable target to develop anti-melanoma therapeutics.
PI3K-ATK/PTEN
The PI3K-AKT pathway is a separate mechanistic route that also plays a role in cell proliferation and survival (45). Similarly to the RAS-RAF-MEK-ERK pathway, constitutive activation of PI3K-AKT leads to a competitive growth advantage that makes way for melanoma proliferation and metastasis (46). PI3K-AKT pathway obtains advantageous gain of function changes through point mutations or changes in chromosome copy number for genes that encode key proteins in the pathway as seen in PIK3CA and AKT1(41). Analysis of melanoma tumor samples has identified about 3% PI3K missense mutation rate (41, 47, 48). Such a low rate makes this pathway appear as an unlikely contributor to melanoma development and progressions.
However, there are other ways the PI3K-AKT pathway can achieve hyper-activation. For example, high levels of PI3K-AKT activity can also result from activation via an NRAS mutation or a lack of inhibition due to PTEN inactivation (41). An NRAS-driven constitutive activation indicates that not only is the PI3K-AKT pathway upregulated, but the RAS-RAF-MEK-ERK pathway as well, which requires separate inhibitors to block both pathways because there remains a paucity of NRAS inhibitors. PTEN, or phosphatase and tensin homologues, is a tumor suppressor gene that produces a protein that dephosphorylates and inactivates the PI3K (45). One of PI3K-AKT pathway’s functions to maintain cell viability is to inhibit apoptosis. Thus, when this pathway is hyper-active or has lost its negative feedback regulation system, then chemotherapy and radiation therapy may lose their death inducing therapeutic potencies (49).
p53
Tumor suppressor gene p53 has been implicated in a variety of human malignancies, such as breast, colorectal, lung, and prostate cancer (50–60). The p53 protein is activated by cell stress or DNA damage and induces cell death (61). The role of p53 in melanoma is disputed. Immunohistochemistry analysis shows varying prevalence of p53 mutation, some reporting an 11% altered expression rate of p53, while other reporting as high as 85% (50, 62). Sequence analysis shows that the p53 gene locus does not appear to possess mutations and expression of wild type p53 is preserved (50). Despite the lack of p53 mutations, melanoma cells generally do not undergo apoptosis in response to gamma radiation or chemotoxic drugs, which suggests that perhaps p53 might not be functioning properly. Another hypothesis deduces that other protein(s) is/are inactivating p53, preventing it from carrying out its tumor suppression functions.
CDK4/CDKN2A
Oncogene Cyclin-dependent kinase 4 (CDK4) and tumor suppressor gene CDKN2A, which encodes p16INK4a have been implicated in familial melanoma development (63). In the case of familial melanoma, there is a genetic susceptibility to develop this disease (64). It has been observed that about 5–10% of melanoma patients have a relative who has also suffered from this cancer (64, 65). The proposed form of genetic inheritance of this predisposition is through autosomal dominant inheritance, with a 53% penetrance rate by the 8th decade (64, 66). Along with other D-type cylcins, CDK6, and pRb, CDK4 and p16INK4a regulate cell cycle progression form the G1 to S phase (67–72). Upon activation by Cyclin D1, CDK4 acts as a proto-oncogene that promotes the progression from G1 to S phase, allowing cell proliferation, whereas, p16INK4a inhibits CDK4 action, arresting cell division (24, 67). Studies have shown that about 25–60% of melanoma patients with a family history of melanoma have an inactivated p16INKA mutation found on 9p21 (63, 64, 73–80), while mutations found in the CDK4 gene occur at a much lower rate (81). In view of these molecular changes, the logical step would be to inhibit CDK4 to prevent further cell cycle progression and limit the uncontrolled growth in melanoma.
c-KIT
About 2–5% of all melanoma cases consist of the subtype acral lentiginous melanoma (8, 82, 83). Acral melanoma is characterized as cancerous lesions found on the palms and the soles of the feet and affects Blacks and Asian/Pacific Islanders at higher rates than Caucasian populations (82, 84). Mutations or chromosomal duplication in the c-KIT gene have been detected in about 23% of acral melanomas and in less than 2% of cutaneous melanomas (85). c-KIT encodes a tyrosine kinase transmembrane receptor, and in the case of melanocytes it is believed to contribute to melanogenesis and melanocyte cell division (86). Point mutations and gene duplications of c-KIT frequently lead to increased expression of the KIT protein, which may alter melanocyte proliferation, cell death, chemotaxis and adhesion, and contribute to tumorigenesis (86–88). Thus, c-KIT may serve as target to develop anti-melanoma therapeutics.
MC1R
Melanocortin 1 receptor (MC1R) acts as a G-protein coupled receptor in melanocytes and plays a critical role in determining skin pigmentation (84, 89). Exposure to UV radiation leads to generation of alpha-melanocyte stimulating hormone, which activates MC1R to generate melanin via the process of melanogenesis (89). Melanin is a light-absorbing polymer derived from oxidized tyrosine that determines the darkness and pigmentation of human skin in response to UV radiation (17, 90). Melanin is made up of two classes of dark brown-black eumelanin and red yellow pheomelanin that are both found in the skin (91). Studies have found that a high pheomelanin to eumelanin ratio along with an overall low level of melanin contributes to fair skin, red hair color (RHC), and the inability to tan phenotypes (92).
The full functions of melanin remain to be elucidated. Originally believed to shield from photo-radiation and protect the skin from DNA damage, melanin also possesses properties of a “thermoregulator, free radical sink, cation chelator, and an antioxidant” (90, 93). Fair skin individuals demonstrate lower amounts of melanin containing granules called melanosomes in their epidermis, which is believed to allow more penetration of UV radiation, leading to increased rates of “sun-induced” DNA damage in melanocytes and the formation of malignant neoplasms (92, 94). UV radiation, in addition to DNA mutagenesis, also leads to the production of reactive oxygen species (ROS), which can lead to oxidative stress within melanocytes and genetic mutations (93). In normal conditions, melanin along with the glutathione pathway acts as antioxidants that neutralize the destructive effects of ROS. However, in MC1R variant individuals, the decreased melanin production would lead to an increased state of oxidative stress and thus, DNA damage that may trigger mechanisms of melanoma initiation (93).
It has been reported that certain MC1R allele variants confer an increased susceptibility to melanoma development especially among some Caucasian populations (89). A case control study analyzing MC1R variants detected in melanoma patients and matched control subjects, found that 72% of melanoma patients possessed a MC1R polymorphism compared to only 56% of the controls (95). There are over 60 MC1R polymorphisms that have been grouped based upon their “strength of” correlation to the red hair phenotype (96). Each MC1R allele variants nearly doubles the risk for melanoma, but does not appear to be sufficient to initiate melanoma development (96). Instead, MC1R variants that cause reduced or complete loss of MC1R signaling capabilities increase the propensity to acquire additional genetic mutations that may promote melanoma development (89). Individuals with MC1R variant alleles that lead to low melanin production are more vulnerable to UV-induced DNA mutagenesis making them susceptible to acquiring mutations in genetic loci of BRAF, NRAS, or CDKN2A (89, 97, 98). Such variants have been found with constitutively activated BRAF melanoma patients who typically do not possess a history of chronic-sun damage of the skin (89). Rarely have wild-type MC1R alleles been found in BRAF mutant melanomas (89). BRAF mutations were seen to be 6.0 times more likely to occur in American individuals who possessed a MC1R polymorphism in non-chronic-sun-damaged melanomas typically found in young patients (96).
Analysis also found that 84% of melanoma tumors with MC1R allele variants also possessed CDKN2A mutations (99). CDKN2A mutations are frequently germ line mutations, present at birth and transmitted between parent and offspring, causing familial melanoma, thus it cannot be theorized that MC1R variants increase the chances of melanocytes to develop the CDKN2A mutation (95). Instead, it has been found that the MC1R variants may influence the penetrance, or the rate at which CDKN2A mutation patients develop melanoma, by increasing the penetrance as seen from the fact that 50% of MC1R wild type individuals with CDKN2A mutations developed cancer, whereas 83% of MC1R variant alleles with CDKN2A mutations developed melanoma (95). MC1R variants (Arg151Cys, Arg160Trp, and Asp284His) that were frequently implicated in these analyses were the same alleles that have been correlated with the RHC, pale skin, and inability to tan characteristics (89, 95, 96). Taking advantage of these trends may in the future allow for improved diagnostic procedures that utilize MC1R variant allele analyses or melanin measurements to provide more information on an individual’s likelihood of developing melanoma (100).
Cadherin
A distinguishing feature of malignant neoplasms is the ability to metastasize to other organs and tissues throughout the body. It can be envisioned that transitioning from vertical growth melanoma into metastatic melanoma may require new gain of function mutations. This is supported by the fact that analyses of primary and metastatic tumors reveal acquisition of larger number of mutations and chromosomal aberrations by metastatic tumors when compared to the primary tumors (101). In normal conditions, melanocytes are connected to undifferentiated basal keratinocytes via cell-cell adhesion molecules, such as transmembrane glycoprotein E-cadherin (102–104). This interaction allows for melanocyte homeostatic regulations as the keratinocyte acts to mediate cell growth, dendrite growth, and expression of cell surface receptors (102). When melanoma is transitioning from a primary tumor into a metastatic variant, it appears to undergo epithelial to mesenchymal transition (EMT) (105). EMT involves changes in cellular cytoskeleton and adhesion molecules that may facilitate cancer cell transition towards a metastatic variant.
One of the changes characterized in the EMT is the switch in the expression of E-cadherin to N-cadherin by the melanoma cells (105). The mutations accumulated seek to knock out the pathways that would induce apoptosis in melanoma cells, conferring a growth advantage and releasing growth restrictions on the tumor (102, 105). The loss of E-cadherin prevents melanocyte regulation by keratinocytes. The newly increased N-cadherin expression is believed to facilitate melanoma migration into the dermis and interactions with dermal fibroblasts and vascular endothelial cells that express the same cadherin. This sequence of events are believed to allow the melanoma to spread out of its epidermal origin (103). N-cadherin expression also leads to the activation of the PI3K-AKT pathway, which appears to inhibit the pro-apoptotic action of Bad and promoting the anti-apoptotic activity of beta-catenin. The N-cadherin-containing melanoma cells also adhere to vascular endothelial cells, which may provide the melanoma access to enter blood vessel and metastasize throughout the body (103).
Upreguated expression of VE-cadherin is another change that appears to promote the metastatic potential of melanoma cells (106). Compared to primary tumors that express VE-cadherin at very low levels, metastatic melanomas demonstrate high levels of expression, which is correlated to the phenomenon of vasculogenic mimicry. In three-dimensional culture, aggressive melanoma cells form tube-like extensions that are similar to embryonic vascularization networks (106). Vasculogenic mimicry has been correlated with VE-cadherin expression due to the fact that when VE-cadherin expression is limited, the formation of tubular networks decreases (106). Melanoma vasculogenic mimicry sheds light on the ability for melanoma tumors to gain access to blood supplies or their means of vascularization, which may lead to a major competitive growth advantage for the tumor (106, 107). Expression of VE-cadherin on melanoma cell membranes is believed to allow tumor cells to adhere to endothelial cells as well, which may facilitate for blood vessel penetration (106).
Melanoma Therapeutics
Traditional Therapies
Primary melanomas are typically treated with surgical excision, which yields a high survival rate. However, following metastasis, surgical excision of the tumor only yields about 10% five-year survival rate (27). Radiation therapy has also proven to be ineffective for treating metastatic melanoma (27). Thus, metastatic melanomas are medically managed by chemotherapeutics and dacarbazine has been used for this purpose. Dacarbazine, dimethyltriazeno-imidazol carboxamide (DTIC), is an alkylating agent and the only monochemotherapy approved by the FDA for treating melanomas (27, 108, 109). After being activated in the liver, DITC generates the metabolite MITC that possesses cytotoxic effects (110). MITC alkylates DNA bases, which prevents DNA replication and can induce mutations (111). Dacarbazine treatment demonstrates about 15–20% response rate that is sustained for an average of 3 months until resistance to dacarbazine develops (108, 111). Although this appears to be a relatively low success rate, dacarbazine is actually considered to be the best option for treating metastatic melanoma (112). In addition to dacarbazine, other single agent chemotherapies include antimicrotubule agents taxane and vindesine, platinum analog cisplatin, and nitrosoureaeas. Such alternatives all demonstrate a response rate of less than 20% when used alone (27, 108, 112). A combination of single agent chemotherapies, such as cisplatin, vindesine, and dacarbazine (CVD), fail to yield an increased response rate among metastatic patients (108, 113). Two major immunotherapies including interleukin 2 (IL-2) and interferon alpha (IFN-α) were developed in hopes to manipulate patients’ own immune activity to target melanoma cells (108, 114). Intravenous infusions of the cytokine IL-2, also called Proleukin and aldesleukin, promote natural killer lymphocytes to differentiate into lymphokine-activated killer cells (LAK) (108, 115, 116). LAK cells attack and lyse tumor cells by seeking out cells that do not display MHC I molecules (108). IL-2 approach yields a 16–17% response rate but is associated with increased morbidity and toxicities, the most frequent being capillary leak syndrome (117). IFN-α is an FDA approved adjuvant therapy, which is administered after surgery in order to help prevent any remaining melanoma cells from proliferating (118). IFN-α treatment leads to a long-lasting immune response by continuously activating CD4+ T-helper-1 lymphocytes, which activate macrophages and natural killer cells via the secretion of IFN-γ and IL-2 (108, 119). It has been noted that combination of immunotherapeutic agents, such as IL-2 and IFN-α, and cytotoxic chemotherapeutics, such as CVD, offers an increased response rate of about 31% (113).
Newer Therapeutic Approaches
Vemurafenib
Because BRAF is mutated and activated in a large number of melanoma cases, and linked to promoting cell proliferation, it was a logical target to develop therapeutics. Accordingly, Vemurafenib (PLX4032) was developed as BRAF kinase inhibitor. Vemurafenib (Figure 1) inhibits the mutant BRAF and thus, confers a degree of specificity towards melanoma harboring mutant form BRAF. In an international multicenter Phase III trial (BRIM-3), vemurafenib was compared with dacarbazine for unresectable stage IIIC and stage IV metastatic BRAFV600E positive melanomas. Previously untreated 675 patients were enrolled in the trial. Patients were given oral vemurafenib 960 mg BID and those on dacarbazine arm received dacarbazine 1000 mg/m2 IV every three weeks. The primary endpoints were progression-free survival (PFS) and overall survival (OS). The secondary endpoints were best overall response rate (BORR), time to response, safety and tolerability. It was concluded that vemurafenib treatment led to improvement in OS, PFS and BORR versus dacarbazine (34, 120).
Figure 1.

Structure of vemurafenib. From PubChem.
A multicenter phase II single arm trial was conducted on 132 previously treated patients. Patients had melanoma that harbored BRAFV600E or BRAFV600K mutation and had been treated with one or more prior therapies. Vemurafenib was given at 960 mg BID orally. The median duration of follow-up was12.9 months. The results achieved 53% response rate with 6% showing complete response and 47% showing partial response. The median progression-free survival was noted to be 6.8 months and the median overall survival 15.9 months (121). The results from the clinical trials indicated vemurafenib to be effective and well tolerated agent. Vermurafenib was duly approved by the FDA in August 2011 for unresectable stage III and IV or metastatic melanomas that harbor BRAFV600 mutations. It is prescribed at a dose of 960 mg BID orally and the duration of treatment is until disease progresses or unacceptable side effects develop. The side effects associated with vemurafenib include arthralgia, fatigue, photosensitivity, alopecia, nausea and diarrhea. Cutaneous squamous cell carcinoma (SCC) and keratoacanthoma or both can also occur. It can cause QT prolongation and thus, enhanced risk of ventricular arrhythmias and can also lead to new primary cutaneous melanoma. Vemurafenib is contraindicated for melanomas that harbor wild type BRAF. It is also contraindicated in electrolyte abnormalities, long QT syndrome with drugs that cause prolongation of QT interval.
Dabrafenib
Dabrafenib (Figure 2) is considered as a next generation agent and has a mechanism of action similar to that of vemurafenib. A phase III (BREAK-3) trial compared dabrafenib and dacarbazine in BRAFV600E-positive unresectable or metastatic (stage III or IV) previously untreated melanomas. Dabrafenib was given at 150 mg BID orally and dacarbazine at 1000 mg/m2 IV every three weeks. The trial enrolled 250 patients and the results indicted dabrafenib to have acceptable safety profile and showed improvement in PFS over dacarbazine. The FDA approved dabrafenib in 2013 for BRAFV600-positive unresectable or metastatic melanomas and the recommended dose is 150 mg BID orally. Dabrafenib is contraindicated in melanoma harboring wild type BRAF. The most common side effects include hyperkeratosis, headache, pyrexia, arthralgia, papilloma, alopecia, and palmar-plantar erhthrodysesthesia syndrome. The serious adverse events include the development of new primary squamous cell carcinoma, melanomas, and keratoacanthomas, febrile drug reactions, hyperglycemia, and uveitis, iritis (122, 123).
Figure 2.

Structure of dabrafenib. From PubChem.
Trametinib
Trametinib (Figure 3) was approved by the FDA in May 2013 for BRAFV600E or BRAFV600K positive unresectable or metastatic melanomas. Its mechanism of action is somewhat different from that of vemurafenib or dabrafenib. Trametinib inhibits MEK i.e. the extraceullular signal-regulated kinase that is downstream of BRAF. Its recommended dose is 2 mg orally once daily and the duration of treatment remains until disease progression or unacceptable side effects develop. The contraindications include melanoma harboring wild type BRAF and patients previously treated with BRAF inhibitors. The side effects associated with trametinib are cardiomyopathy, retinal pigment epithelial detachment, retinal vein occlusion, interstitial lung disease, serious skin toxicity involving rash, dermatitis, acneiform rash, palmar-plantar erythrodysesthesia syndrome, and erythema. Other common side effects are rash, diarrhea, lymphedema, dermatitis acneiform, stomatitis, hypertension, abdominal pain, hemorrhage, dry skin, pruritus and paronychia. It may also inhibit fertility in females.
Figure 3.

Structure of trametinib. From PubChem.
Although BRAF inhibitors have shown promise, acquired resistance does occur and believed to involve activation of MAPK pathway (124). BRAF inhibitors are also associated with other cutaneous malignancies that are thought to occur due to paradoxical activation of MAPK pathway in unaffected skin cells that do not harbor BRAF mutation (124). MEK resides downstream of BRAF and combination of BRAF inhibitor with MEK inhibitor seem to suggest a better approach to overcome the limitations of BRAF inhibitors as single agent therapeutics in this setting. In keeping with this idea, FDA granted (January 2014) accelerated approval to combination of dabrafenib and trametinib for unresectable or metastatic melanoma with BRAFV600 mutations. The accelerated approval was granted with the contingency that an ongoing phase III trial investigating the combination of dabrafenib and trametinib would be successfully completed. More recently, a study reported the results of a phase III trial comparing the effects of dabrafenib and trametinib combination with those of vemurafenib. The results indicated that dabrafenib and trametinib in combination improved OS significantly compared to vemurafenib alone in BRAFV600 mutant metastatic melanomas. Interestingly, only 1% of patients on combination therapy reported cutaneous squamous cell carcinoma and keratoacanthoma, while 18% of those on vemurafenib reported such abnormalities (124).
Another study reported the results also of a phase III trial that compared vemurafenib and cobimetinib combination with vemurafenib and placebo group. Just like trametinib, cobimetinib is a MEK inhibitor. It was noted that the combination of vemurafenib and cobimetinib improved significantly the PFS in metastatic melanoma harboring BRAFV600 mutation (125). Based on these studies, it is therefore, logical to think that the combination of BRAF and MEK inhibitors would appear to be superior to monotherapy going forward (Figure 4).
Figure 4. BRAF activation signaling events. Vemurafenib and dabrafenib inhibit BRAFV600 mutant form, whereas trametinib and cobimetinib inhibit MEK.
Adapted from Ascierto et al. (141).
Ipilimumab
Ipilimumab is a humanized monoclonal antibody that prevents T-cell activation via inhibition of cytotoxic T-lymphocyte antigen-4 (CTLA-4) (126–129). It was approved by the FDA in March 2011 for late stage metastatic melanoma. In normal immunomodulation, T-Cell Receptor ligation to its respective MHC presented antigen leads to CTLA-4 co-receptor expression on T cells (130, 131). CTLA-4 is very similar in structure to co-receptor CD28, which allows both these receptors to bind to CD80 and CD86 ligands found on antigen presenting cells (131). CD28-CD80/86 interaction activates the T-cell and upregulates T-cell proliferation, differentiation and cytokine production, whereas, high affinity CTLA-4-CD80/86 binding inhibits T-cell activation, limiting the immune response (130–134). Ipilimumab augments T-cell activation by inhibiting the CTLA-4 receptor interaction with CD80 or CD86, which prevents T-cells from becoming inactivated (128). Ultimately, anti-tumor immunity is boosted through Ipilimumab. Previously treated stage III unresectable melanoma and stage IV metastatic melanoma patients receiving a 10 mg/kg dosage of Ipilimumab demonstrated an 11.1% response rate with lower response rates at lower dosage levels, demonstrating a dose dependent relationship (135). Ipilimumab yielded a median 10-month survival rate (136). However, Ipilimumab has also shown to lead to autoimmune side effects in 15% of patients receiving the highest dosage, 10mg/kg, of the drug (135). These side effects include fatigue, diarrhea, skin rash, endocrine deficiencies and intestinal inflammation. It may be noted that this therapeutic modality does not work in all patients.
Pembrolizumab
Pemrolizumab is a humanized monoclonal IgG4 antibody. It targets human cell surface receptor PD-1 (programmed cell death-1) and interferes with PD-1 ligand binding. This interference results in activation of T cell-mediated response against tumors (137). The FDA approved pembrolizumab for management of advanced or unresectable melanomas that are refractory to other therapeutics (138). The common side effects include fatigue, cough, nausea, pruritus, rash, decreased appetite, constipation, arthralgia and diarrhea.
Nivolumab
Nivolumab is a fully human monoclonal antibody that targets PD-1. It functions in manner similar to pemrolizumab. FDA granted approval in December 2014 for treatment of metastatic melanomas that do not respond to other forms of treatment (139). The common side effects include rash, itching, cough, upper respiratory tract infections, and edema. The serious side effects engaging the immune system can involve major organs (140).
Future Trends and Conclusions
Historical trends have shown that treatments for stage III and stage IV metastatic melanoma are becoming more targeted and utilize the information gained from improved understanding of cellular and immunological pathway changes that are associated with this malignancy. Pinpointing the mutations in the various pathways that melanoma patients have acquired would allow for the continued development of more efficacious treatments that strive to prevent drug resistance and cancer recurrence. The convergence of immunology with other disciplines of biomedical research would lead to further improvement in the development of newer and more efficacious therapeutic approaches to manage this debilitating and deadly disease. From the preceding information, it is clear that not only have treatments become more molecularly sophisticated, but diagnosis and early detection techniques also have the opportunity to become increasingly specific. Discovery of specific markers of melanoma risk can allow for early detection and limit the amount of cytotoxic treatment that a patient must receive.
Acknowledgments
The work in M. Saeed Sheikh’s laboratory is supported in part by the NIH grant CA157168. The information on drugs provided in this article is for educational purpose and does not replace the advice of a healthcare provider.
Footnotes
Conflict of interests
YL has no conflicts of interest to declare. MSS owns securities of Bristol-Myers Squibb, GlaxoSmithKline and Merck.
References
- 1.SEER Stat Fact Sheets: Melanoma of the Skin [Internet] Bethesda, MD: National Cancer Institute; [updated 2014 Apr; cited 2014 Dec 29]. Available from: http://seer.cancer.gov/statfacts/html/melan.html. [Google Scholar]
- 2.Thompson JF, Scolyer RA, Kefford RF. Cutaneous melanoma. Lancet. 2005;365:687–701. doi: 10.1016/S0140-6736(05)17951-3. [DOI] [PubMed] [Google Scholar]
- 3.Lens MB, Dawes M. Global perspectives of contemporary epidemiological trends of cutaneous malignant melanoma. Br J Dermatol. 2004;150:179–85. doi: 10.1111/j.1365-2133.2004.05708.x. [DOI] [PubMed] [Google Scholar]
- 4.Miller AJ, Mihm MC., Jr Melanoma. N Engl J Med. 2006;355:51–65. doi: 10.1056/NEJMra052166. [DOI] [PubMed] [Google Scholar]
- 5.Sboner A, Eccher C, Blanzieri E, et al. A multiple classifier system for early melanoma diagnosis. Artif Intell Med. 2003;27:29–44. doi: 10.1016/s0933-3657(02)00087-8. [DOI] [PubMed] [Google Scholar]
- 6.Beddingfield FC., 3rd The melanoma epidemic: Res ipsa loquitur. Oncologist. 2003;8:459–65. doi: 10.1634/theoncologist.8-5-459. [DOI] [PubMed] [Google Scholar]
- 7.Heistein JB. Melanoma [Internet] Medscape. [updated 2014 Sept 5; cited 2014 Dec 29]. Available from: http://emedicine.medscape.com/article/1295718-overview#aw2aab6b2.
- 8.Cutaneous Melanoma, or Melanoma of the Skin. Washington, DC: Melanoma Research Foundation; [Internet] [updated 2014 Dec 29; cited 2014 Dec 30]. Available from: http://www.melanoma.org/understand-melanoma/what-is-melanoma/cutaneous-melanoma. [Google Scholar]
- 9.Stages of Melanoma. London, England: Cancer Research UK; [Internet] [updated 2014 Jan 17; cited 2014 Dec 30]. Available from: http://www.cancerresearchuk.org/about-cancer/type/melanoma/treatment/stages-of-melanoma. [Google Scholar]
- 10.Rigel DS, Friedman RJ, Kopf AW, Polsky D. ABCDE--an evolving concept in the early detection of melanoma. Arch Dermatol. 2005;141:1032–4. doi: 10.1001/archderm.141.8.1032. [DOI] [PubMed] [Google Scholar]
- 11.Kittler H, Pehamberger H, Wolff K, Binder M. Diagnostic accuracy of dermoscopy. Lancet Oncol. 2002;3:159–65. doi: 10.1016/s1470-2045(02)00679-4. [DOI] [PubMed] [Google Scholar]
- 12.Westerhoff K, McCarthy WH, Menzies SW. Increase in the sensitivity for melanoma diagnosis by primary care physicians using skin surface microscopy. Br J Dermatol. 2000;143:1016–20. doi: 10.1046/j.1365-2133.2000.03836.x. [DOI] [PubMed] [Google Scholar]
- 13.Hoon DS, Bostick P, Kuo C, et al. Molecular markers in blood as surrogate prognostic indicators of melanoma recurrence. Cancer Res. 2000;60:2253–7. [PubMed] [Google Scholar]
- 14.King R, Weilbaecher KN, McGill G, Cooley E, Mihm M, Fisher DE. Microphthalmia transcription factor: A sensitive and specific melanocyte marker for melanoma diagnosis. Am J Pathol. 1999;155:731–8. doi: 10.1016/S0002-9440(10)65172-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Weinstock MA, Colditz GA, Willett WC, et al. Nonfamilial cutaneous melanoma incidence in women associated with sun exposure before 20 years of age. Pediatrics. 1989;84:199–204. [PubMed] [Google Scholar]
- 16.Ikehata H, Ono T. The mechanisms of UV mutagenesis. J Radiat Res. 2011;52:115–25. doi: 10.1269/jrr.10175. [DOI] [PubMed] [Google Scholar]
- 17.Agar N, Young AR. Melanogenesis: A photoprotective response to DNA damage? Mutat Res. 2005;571:121–32. doi: 10.1016/j.mrfmmm.2004.11.016. [DOI] [PubMed] [Google Scholar]
- 18.Berger MF, Hodis E, Heffernan TP, et al. Melanoma genome sequencing reveals frequent PREX2 mutations. Nature. 2012;485:502–6. doi: 10.1038/nature11071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Pleasance ED, Cheetham RK, Stephens PJ, et al. A comprehensive catalogue of somatic mutations from a human cancer genome. Nature. 2010;463:191–6. doi: 10.1038/nature08658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hodis E, Watson IR, Kryukov GV, et al. A landscape of driver mutations in melanoma. Cell. 2012;150:251–63. doi: 10.1016/j.cell.2012.06.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Greene M, Fraumeni J. The hereditary variant of familial melanoma. In: Clark W, Goldman L, Mastrangelo M, editors. Human Malignant Melanoma. New York, NY: Grune and Stratton; 1979. pp. 139–166. [Google Scholar]
- 22.Grange F, Chompret A, Guilloud-Bataille M, et al. Comparison between familial and nonfamilial melanoma in france. Arch Dermatol. 1995;131:1154–9. [PubMed] [Google Scholar]
- 23.Aitken JF, Green AC, MacLennan R, Youl P, Martin NG. The queensland familial melanoma project: Study design and characteristics of participants. Melanoma Res. 1996;6:155–65. doi: 10.1097/00008390-199604000-00011. [DOI] [PubMed] [Google Scholar]
- 24.Soufir N, Avril MF, Chompret A, et al. Prevalence of p16 and CDK4 germline mutations in 48 melanoma-prone families in france. the french familial melanoma study group. Hum Mol Genet. 1998;7:209–16. doi: 10.1093/hmg/7.2.209. [DOI] [PubMed] [Google Scholar]
- 25.Batistatou A, Zioga A, Panelos J, Massi D, Agnantis NJ, Charalabopoulos K. A new concept of melanocytic neoplasia pathogenesis based on the phenotype of common acquired nevi. Med Hypotheses. 2007;69:1334–9. doi: 10.1016/j.mehy.2007.03.004. [DOI] [PubMed] [Google Scholar]
- 26.Rhodes AR, Weinstock MA, Fitzpatrick TB, Mihm MC, Jr, Sober AJ. Risk factors for cutaneous melanoma. A practical method of recognizing predisposed individuals. JAMA. 1987;258:3146–54. [PubMed] [Google Scholar]
- 27.Bhatia S, Tykodi SS, Thompson JA. Treatment of metastatic melanoma: An overview. Oncology (Williston Park) 2009;23:488–96. [PMC free article] [PubMed] [Google Scholar]
- 28.Balch CM, Buzaid AC, Soong SJ, et al. Final version of the american joint committee on cancer staging system for cutaneous melanoma. J Clin Oncol. 200;19:3635–48. doi: 10.1200/JCO.2001.19.16.3635. [DOI] [PubMed] [Google Scholar]
- 29.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
- 30.Flaherty KT, Puzanov I, Kim KB, et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med. 2010;363:809–19. doi: 10.1056/NEJMoa1002011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kumar R, Angelini S, Czene K, et al. BRAF mutations in metastatic melanoma: A possible association with clinical outcome. Clin Cancer Res. 2003;9:3362–8. [PubMed] [Google Scholar]
- 32.Houben R, Becker JC, Kappel A, et al. Constitutive activation of the ras-raf signaling pathway in metastatic melanoma is associated with poor prognosis. J Carcinog. 2004;3:6. doi: 10.1186/1477-3163-3-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Solit D, Sawyers CL. Drug discovery: How melanomas bypass new therapy. Nature. 2010;468:902–3. doi: 10.1038/468902a. [DOI] [PubMed] [Google Scholar]
- 34.Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–16. doi: 10.1056/NEJMoa1103782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.McCubrey JA, Steelman LS, Abrams SL, et al. Roles of the RAF/MEK/ERK and PI3K/PTEN/AKT pathways in malignant transformation and drug resistance. Adv Enzyme Regul. 2006;46:249–79. doi: 10.1016/j.advenzreg.2006.01.004. [DOI] [PubMed] [Google Scholar]
- 36.Pollock PM, Harper UL, Hansen KS, et al. High frequency of BRAF mutations in nevi. Nat Genet. 2003;33:19–20. doi: 10.1038/ng1054. [DOI] [PubMed] [Google Scholar]
- 37.Dong J, Phelps RG, Qiao R, Yao S, Benard O, Ronai Z, Aaronson SA. BRAF oncogenic mutations correlate with progression rather than initiation of human melanoma. Cancer Res. 2003;63:3883–5. [PubMed] [Google Scholar]
- 38.Brose MS, Volpe P, Feldman M, et al. BRAF and RAS mutations in human lung cancer and melanoma. Cancer Res. 2002;62:6997–7000. [PubMed] [Google Scholar]
- 39.Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–54. doi: 10.1038/nature00766. [DOI] [PubMed] [Google Scholar]
- 40.Kelleher FC, McArthur GA. Targeting NRAS in melanoma. Cancer J. 2012;18:132–6. doi: 10.1097/PPO.0b013e31824ba4df. [DOI] [PubMed] [Google Scholar]
- 41.Davies MA. The role of the PI3K-AKT pathway in melanoma. Cancer J. 2012;18:142–7. doi: 10.1097/PPO.0b013e31824d448c. [DOI] [PubMed] [Google Scholar]
- 42.Woodman SE, Lazar AJ, Aldape KD, Davies MA. New strategies in melanoma: Molecular testing in advanced disease. Clin Cancer Res. 2012;18:1195–200. doi: 10.1158/1078-0432.CCR-11-2317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Azvolinksy A. NRAS Mutation in Melanoma: A Challenging Target [Internet] Cancer Commons. [updated 2013 Jul 17; cited 2014 Dec 30]. Available from: http://www.cancercommons.org/2013/07/17/targeting-nras-mutations-in-melanoma/
- 44.Poynter JN, Elder JT, Fullen DR, et al. BRAF and NRAS mutations in melanoma and melanocytic nevi. Melanoma Res. 2006;16:267–73. doi: 10.1097/01.cmr.0000222600.73179.f3. [DOI] [PubMed] [Google Scholar]
- 45.Fresno Vara JA, Casado E, de Castro J, Cejas P, Belda-Iniesta C, Gonzalez-Baron M. PI3K/akt signalling pathway and cancer. Cancer Treat Rev. 2004;30:193–204. doi: 10.1016/j.ctrv.2003.07.007. [DOI] [PubMed] [Google Scholar]
- 46.Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov. 2005;4:988–1004. doi: 10.1038/nrd1902. [DOI] [PubMed] [Google Scholar]
- 47.Curtin JA, Stark MS, Pinkel D, Hayward NK, Bastian BC. PI3-kinase subunits are infrequent somatic targets in melanoma. J Invest Dermatol. 2006;126:1660–3. doi: 10.1038/sj.jid.5700311. [DOI] [PubMed] [Google Scholar]
- 48.Omholt K, Krockel D, Ringborg U, Hansson J. Mutations of PIK3CA are rare in cutaneous melanoma. Melanoma Res. 2006;16:197–200. doi: 10.1097/01.cmr.0000200488.77970.e3. [DOI] [PubMed] [Google Scholar]
- 49.Krasilnikov M, Adler V, Fuchs SY, et al. Contribution of phosphatidylinositol 3-kinase to radiation resistance in human melanoma cells. Mol Carcinog. 1999;24:64–9. doi: 10.1002/(sici)1098-2744(199901)24:1<64::aid-mc9>3.0.co;2-2. [DOI] [PubMed] [Google Scholar]
- 50.Stretch JR, Gatter KC, Ralfkiaer E, Lane DP, Harris AL. Expression of mutant p53 in melanoma. Cancer Res. 1991;5:5976–9. [PubMed] [Google Scholar]
- 51.Nigro JM, Baker SJ, Preisinger AC, et al. Mutations in the p53 gene occur in diverse human tumour types. Nature. 1989;342:705–8. doi: 10.1038/342705a0. [DOI] [PubMed] [Google Scholar]
- 52.Prosser J, Thompson AM, Cranston G, Evans HJ. Evidence that p53 behaves as a tumour suppressor gene in sporadic breast tumours. Oncogene. 1990;5:1573–9. [PubMed] [Google Scholar]
- 53.Iggo R, Gatter K, Bartek J, Lane D, Harris AL. Increased expression of mutant forms of p53 oncogene in primary lung cancer. Lancet. 1990;335:675–9. doi: 10.1016/0140-6736(90)90801-b. [DOI] [PubMed] [Google Scholar]
- 54.Baker SJ, Fearon ER, Nigro JM, et al. Chromosome 17 deletions and p53 gene mutations in colorectal carcinomas. Science. 1989;244:217–21. doi: 10.1126/science.2649981. [DOI] [PubMed] [Google Scholar]
- 55.Whibley C, Pharoah PD, Hollstein M. P53 polymorphisms: Cancer implications. Nat Rev Cancer. 2009;9:95–107. doi: 10.1038/nrc2584. [DOI] [PubMed] [Google Scholar]
- 56.Brown KM, Macgregor S, Montgomery GW, et al. Common sequence variants on 20q11. 22 confer melanoma susceptibility. Nat Genet. 2008;40:838–40. doi: 10.1038/ng.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Easton DF, Pooley KA, Dunning AM, et al. Genome-wide association study identifies novel breast cancer susceptibility loci. Nature. 2007;447:1087–93. doi: 10.1038/nature05887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Eeles RA, Kote-Jarai Z, Giles GG, et al. Multiple newly identified loci associated with prostate cancer susceptibility. Nat Genet. 2008;40:316–21. doi: 10.1038/ng.90. [DOI] [PubMed] [Google Scholar]
- 59.Hunter DJ, Kraft P, Jacobs KB, et al. A genome-wide association study identifies alleles in FGFR2 associated with risk of sporadic postmenopausal breast cancer. Nat Genet. 2007;39:870–4. doi: 10.1038/ng2075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Stacey SN, Manolescu A, Sulem P, et al. Common variants on chromosomes 2q35 and 16q12 confer susceptibility to estrogen receptor-positive breast cancer. Nat Genet. 2007;39:865–9. doi: 10.1038/ng2064. [DOI] [PubMed] [Google Scholar]
- 61.Smalley KS, Contractor R, Haass NK, et al. An organometallic protein kinase inhibitor pharmacologically activates p53 and induces apoptosis in human melanoma cells. Cancer Res. 2007;67:209–17. doi: 10.1158/0008-5472.CAN-06-1538. [DOI] [PubMed] [Google Scholar]
- 62.Albino AP, Vidal MJ, McNutt NS, et al. Mutation and expression of the p53 gene in human malignant melanoma. Melanoma Res. 1994;4:35–45. doi: 10.1097/00008390-199402000-00006. [DOI] [PubMed] [Google Scholar]
- 63.Liu L, Dilworth D, Gao L, et al. Mutation of the CDKN2A 5’ UTR creates an aberrant initiation codon and predisposes to melanoma. Nat Genet. 1999;21:128–32. doi: 10.1038/5082. [DOI] [PubMed] [Google Scholar]
- 64.FitzGerald MG, Harkin DP, Silva-Arrieta S, et al. Prevalence of germ-line mutations in p16, p19ARF, and CDK4 in familial melanoma: Analysis of a clinic-based population. Proc Natl Acad Sci U S A. 1996;93:8541–5. doi: 10.1073/pnas.93.16.8541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Tucker MA, Bale SJ. Clinical aspects of familial cutaneous malignant melanoma. Semin Oncol. 1988;15:524–8. [PubMed] [Google Scholar]
- 66.Cannon-Albright LA, Meyer LJ, Goldgar DE, et al. Penetrance and expressivity of the chromosome 9p melanoma susceptibility locus (MLM) Cancer Res. 1994;54:6041–4. [PubMed] [Google Scholar]
- 67.Bartkova J, Lukas J, Guldberg P, et al. The p16-cyclin D/Cdk4-pRb pathway as a functional unit frequently altered in melanoma pathogenesis. Cancer Res. 1996;56:5475–83. [PubMed] [Google Scholar]
- 68.Kamb A. Cell-cycle regulators and cancer. Trends Genet. 1995;11:136–40. doi: 10.1016/s0168-9525(00)89027-7. [DOI] [PubMed] [Google Scholar]
- 69.Sherr CJ, Roberts JM. Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev. 1995;9:1149–63. doi: 10.1101/gad.9.10.1149. [DOI] [PubMed] [Google Scholar]
- 70.Strauss M, Lukas J, Bartek J. Unrestricted cell cycling and cancer. Nat Med. 1995;1:1245–6. doi: 10.1038/nm1295-1245. [DOI] [PubMed] [Google Scholar]
- 71.Weinberg RA. The retinoblastoma protein and cell cycle control. Cell. 1995;81:323–30. doi: 10.1016/0092-8674(95)90385-2. [DOI] [PubMed] [Google Scholar]
- 72.Hall M, Peters G. Genetic alterations of cyclins, cyclin-dependent kinases, and cdk inhibitors in human cancer. Adv Cancer Res. 1996;68:67–108. doi: 10.1016/s0065-230x(08)60352-8. [DOI] [PubMed] [Google Scholar]
- 73.Hogg D, Brill H, Liu L, et al. Role of the cyclin-dependent kinase inhibitor CDKN2A in familial melanoma. J Cutan Med Surg. 1998;2:172–9. doi: 10.1177/120347549800200312. [DOI] [PubMed] [Google Scholar]
- 74.Hayward NK. The current situation with regard to human melanoma and genetic inferences. Curr Opin Oncol. 1996;8:136–42. doi: 10.1097/00001622-199603000-00011. [DOI] [PubMed] [Google Scholar]
- 75.Hussussian CJ, Struewing JP, Goldstein AM, et al. Germline p16 mutations in familial melanoma. Nat Genet. 1994;8:15–21. doi: 10.1038/ng0994-15. [DOI] [PubMed] [Google Scholar]
- 76.Kamb A, Shattuck-Eidens D, Eeles R, et al. Analysis of the p16 gene (CDKN2) as a candidate for the chromosome 9p melanoma susceptibility locus. Nat Genet. 1994;8:23–6. doi: 10.1038/ng0994-22. [DOI] [PubMed] [Google Scholar]
- 77.Gruis NA, Sandkuijl LA, van der Velden PA, Bergman W, Frants RR. CDKN2 explains part of the clinical phenotype in dutch familial atypical multiple-mole melanoma (FAMMM) syndrome families. Melanoma Res. 1995;5:169–77. doi: 10.1097/00008390-199506000-00005. [DOI] [PubMed] [Google Scholar]
- 78.Holland EA, Beaton SC, Becker TM, et al. Analysis of the p16 gene, CDKN2, in 17 australian melanoma kindreds. Oncogene. 1995;11:2289–94. [PubMed] [Google Scholar]
- 79.Walker GJ, Hussussian CJ, Flores JF, et al. Mutations of the CDKN2/p16INK4 gene in australian melanoma kindreds. Hum Mol Genet. 1995;4:1845–52. doi: 10.1093/hmg/4.10.1845. [DOI] [PubMed] [Google Scholar]
- 80.Liu L, Lassam NJ, Slingerland JM, et al. Germline p16INK4A mutation and protein dysfunction in a family with inherited melanoma. Oncogene. 1995;11:405–12. [PubMed] [Google Scholar]
- 81.Goldstein AM, Struewing JP, Chidambaram A, Fraser MC, Tucker MA. Genotype-phenotype relationships in U.S. melanoma-prone families with CDKN2A and CDK4 mutations. J Natl Cancer Inst. 2000;92:1006–10. doi: 10.1093/jnci/92.12.1006. [DOI] [PubMed] [Google Scholar]
- 82.Bradford PT, Goldstein AM, McMaster ML, Tucker MA. Acral lentiginous melanoma: Incidence and survival patterns in the united states, 1986–2005. Arch Dermatol. 2009;145:427–34. doi: 10.1001/archdermatol.2008.609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Markovic SN, Erickson LA, Rao RD, et al. Malignant melanoma in the 21st century, part 1: Epidemiology, risk factors, screening, prevention, and diagnosis. Mayo Clin Proc. 2007;82:364–80. doi: 10.4065/82.3.364. [DOI] [PubMed] [Google Scholar]
- 84.Reed R. New Concepts in Surgical Pathology of the Skin. New York, NY: Jon Wiley & Sons; 1976. pp. 89–90. [Google Scholar]
- 85.Beadling C, Jacobson-Dunlop E, Hodi FS, et al. KIT gene mutations and copy number in melanoma subtypes. Clin Cancer Res. 2008;14:6821–8. doi: 10.1158/1078-0432.CCR-08-0575. [DOI] [PubMed] [Google Scholar]
- 86.Rivera RS, Nagatsuka H, Gunduz M, et al. C-kit protein expression correlated with activating mutations in KIT gene in oral mucosal melanoma. Virchows Arch. 2008;452:27–32. doi: 10.1007/s00428-007-0524-2. [DOI] [PubMed] [Google Scholar]
- 87.Curtin JA, Busam K, Pinkel D, Bastian BC. Somatic activation of KIT in distinct subtypes of melanoma. J Clin Oncol. 2006;24:4340–6. doi: 10.1200/JCO.2006.06.2984. [DOI] [PubMed] [Google Scholar]
- 88.Lin SC, Liu CL, Wang TI, Chang WS, Tzen CY, Huang MJ. Clinical implications of C-kit gene mutation in patients with large gastrointestinal stromal tumors. J Gastroenterol Hepatol. 2006;21:1604–8. doi: 10.1111/j.1440-1746.2006.04322.x. [DOI] [PubMed] [Google Scholar]
- 89.Landi MT, Bauer J, Pfeiffer RM, et al. MC1R germline variants confer risk for BRAF-mutant melanoma. Science. 2006;313:521–2. doi: 10.1126/science.1127515. [DOI] [PubMed] [Google Scholar]
- 90.Riley PA. Melanin. Int J Biochem Cell Biol. 1997;29:1235–9. doi: 10.1016/s1357-2725(97)00013-7. [DOI] [PubMed] [Google Scholar]
- 91.Thody AJ, Higgins EM, Wakamatsu K, Ito S, Burchill SA, Marks JM. Pheomelanin as well as eumelanin is present in human epidermis. J Invest Dermatol. 1991;97:340–4. doi: 10.1111/1523-1747.ep12480680. [DOI] [PubMed] [Google Scholar]
- 92.Scott MC, Wakamatsu K, Ito S, et al. Human melanocortin 1 receptor variants, receptor function and melanocyte response to UV radiation. J Cell Sci. 2002;115:2349–55. doi: 10.1242/jcs.115.11.2349. [DOI] [PubMed] [Google Scholar]
- 93.Meyskens FL, Jr, Farmer P, Fruehauf JP. Redox regulation in human melanocytes and melanoma. Pigment Cell Res. 2001;14:148–54. doi: 10.1034/j.1600-0749.2001.140303.x. [DOI] [PubMed] [Google Scholar]
- 94.Menon IA, Persad S, Ranadive NS, Haberman HF. Effects of ultraviolet-visible irradiation in the presence of melanin isolated from human black or red hair upon ehrlich ascites carcinoma cells. Cancer Res. 1983;43:3165–9. [PubMed] [Google Scholar]
- 95.Palmer JS, Duffy DL, Box NF, et al. Melanocortin-1 receptor polymorphisms and risk of melanoma: Is the association explained solely by pigmentation phenotype? Am J Hum Genet. 2000;66:176–86. doi: 10.1086/302711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Hacker E, Hayward NK. Germline MC1R variants and BRAF mutant melanoma. J Invest Dermatol. 2008;128:2354–6. doi: 10.1038/jid.2008.236. [DOI] [PubMed] [Google Scholar]
- 97.Naysmith L, Waterston K, Ha T, et al. Quantitative measures of the effect of the melanocortin 1 receptor on human pigmentary status. J Invest Dermatol. 2004;122:423–8. doi: 10.1046/j.0022-202X.2004.22221.x. [DOI] [PubMed] [Google Scholar]
- 98.Duffy DL, Box NF, Chen W, et al. Interactive effects of MC1R and OCA2 on melanoma risk phenotypes. Hum Mol Genet. 2004;13:447–61. doi: 10.1093/hmg/ddh043. [DOI] [PubMed] [Google Scholar]
- 99.Box NF, Duffy DL, Chen W, et al. MC1R genotype modifies risk of melanoma in families segregating CDKN2A mutations. Am J Hum Genet. 200;69:765–73. doi: 10.1086/323412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Sturm RA. Skin colour and skin cancer - MC1R, the genetic link. Melanoma Res. 2002;12:405–16. doi: 10.1097/00008390-200209000-00001. [DOI] [PubMed] [Google Scholar]
- 101.Yokota J. Tumor progression and metastasis. Carcinogenesis. 2000;2:497–503. doi: 10.1093/carcin/21.3.497. [DOI] [PubMed] [Google Scholar]
- 102.Hsu MY, Meier FE, Nesbit M, et al. E-cadherin expression in melanoma cells restores keratinocyte-mediated growth control and down-regulates expression of invasion-related adhesion receptors. Am J Pathol. 2000;156:1515–25. doi: 10.1016/S0002-9440(10)65023-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Li G, Satyamoorthy K, Herlyn M. N-cadherin-mediated intercellular interactions promote survival and migration of melanoma cells. Cancer Res. 2001;61:3819–25. [PubMed] [Google Scholar]
- 104.Vleminckx K, Kemler R. Cadherins and tissue formation: Integrating adhesion and signaling. Bioessays. 1999;21:211–20. doi: 10.1002/(SICI)1521-1878(199903)21:3<211::AID-BIES5>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
- 105.Hao L, Ha JR, Kuzel P, Garcia E, Persad S. Cadherin switch from E- to N-cadherin in melanoma progression is regulated by the PI3K/PTEN pathway through twist and snail. Br J Dermatol. 2012;166:1184–97. doi: 10.1111/j.1365-2133.2012.10824.x. [DOI] [PubMed] [Google Scholar]
- 106.Hendrix MJ, Seftor EA, Meltzer PS, et al. Expression and functional significance of VE-cadherin in aggressive human melanoma cells: Role in vasculogenic mimicry. Proc Natl Acad Sci U S A. 2001;98:8018–23. doi: 10.1073/pnas.131209798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Hess AR, Seftor EA, Gruman LM, Kinch MS, Seftor RE, Hendrix MJ. VE-cadherin regulates EphA2 in aggressive melanoma cells through a novel signaling pathway: Implications for vasculogenic mimicry. Cancer Biol Ther. 2006;5:228–33. doi: 10.4161/cbt.5.2.2510. [DOI] [PubMed] [Google Scholar]
- 108.Bajetta E, Del Vecchio M, Bernard-Marty C, et al. Metastatic melanoma: Chemotherapy. Semin Oncol. 2002;29:427–45. doi: 10.1053/sonc.2002.35238. [DOI] [PubMed] [Google Scholar]
- 109.Dacarbazine. National Cancer Institute; [Internet] [updated 2013 Jul 10; cited 2014 Dec 30]. Available from: http://www.cancer.gov/cancertopics/druginfo/dacarbazine. [Google Scholar]
- 110.Tsang LL, Quarterman CP, Gescher A, Slack JA. Comparison of the cytotoxicity in vitro of temozolomide and dacarbazine, prodrugs of 3-methyl-(triazen-1-yl)imidazole-4-carboxamide. Cancer Chemother Pharmacol. 1991;27:342–6. doi: 10.1007/BF00688855. [DOI] [PubMed] [Google Scholar]
- 111.Sanada M, Takagi Y, Ito R, Sekiguchi M. Killing and mutagenic actions of dacarbazine, a chemotherapeutic alkylating agent, on human and mouse cells: Effects of mgmt and Mlh1 mutations. DNA Repair (Amst) 2004;3:413–20. doi: 10.1016/j.dnarep.2003.12.008. [DOI] [PubMed] [Google Scholar]
- 112.Gogas H, Bafaloukos D, Bedikian AY. The role of taxanes in the treatment of metastatic melanoma. Melanoma Res. 2004;14:415–20. doi: 10.1097/00008390-200410000-00013. [DOI] [PubMed] [Google Scholar]
- 113.Bajetta E, Del Vecchio M, Nova P, et al. Multicenter phase III randomized trial of polychemotherapy (CVD regimen) versus the same chemotherapy (CT) plus subcutaneous interleukin-2 and interferon-alpha2b in metastatic melanoma. Ann Oncol. 2006;17:571–7. doi: 10.1093/annonc/mdl007. [DOI] [PubMed] [Google Scholar]
- 114.Parmiani G, Castelli C, Rivoltini L, et al. Immunotherapy of melanoma. Semin Cancer Biol. 2003;13:391–400. doi: 10.1016/j.semcancer.2003.09.001. [DOI] [PubMed] [Google Scholar]
- 115.Drugs Approved for Melanoma. National Cancer Institute; [Internet] [updated 2013 Aug 16; cited 2014 Dec 30]. Available from: http://www.cancer.gov/cancertopics/druginfo/melanoma. [Google Scholar]
- 116.Mule JJ, Shu S, Schwarz SL, Rosenberg SA. Adoptive immunotherapy of established pulmonary metastases with LAK cells and recombinant interleukin-2. Science. 1984;225:1487–9. doi: 10.1126/science.6332379. [DOI] [PubMed] [Google Scholar]
- 117.Schwartz RN, Stover L, Dutcher J. Managing toxicities of high-dose interleukin-2. Oncology (Williston Park) 2002;16:11–20. [PubMed] [Google Scholar]
- 118.Immunotherapy for melanoma skin cancer [Internet] American Cancer Society. [updated 2013 Oct 29; cited 2014 Dec 30] [Google Scholar]
- 119.Tompkins WA. Immunomodulation and therapeutic effects of the oral use of interferon-alpha: Mechanism of action. J Interferon Cytokine Res. 1999;19:817–28. doi: 10.1089/107999099313325. [DOI] [PubMed] [Google Scholar]
- 120.Zelboraf (vemurafenib) Phase II trial Design. Zelboraf. [Internet] [updated 2014 Dec 19; cited 2014 Dec 30]. Available from: http://www.zelboraf.com/oncology/treatment/phase-ii-trial-design/
- 121.Sosman JA, Kim KB, Schuchter L, et al. Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib. N Engl J Med. 2012;366:707–14. doi: 10.1056/NEJMoa1112302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: A multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2012;380:358–65. doi: 10.1016/S0140-6736(12)60868-X. [DOI] [PubMed] [Google Scholar]
- 123.Pazdur R. National Cancer Institute; FDA Approval for Dabrafenib. [Internet] [updated 2014 Jan 10; cited 2014 Dec 30]. Available from: http://www.cancer.gov/cancertopics/druginfo/fda-dabrafenib. [Google Scholar]
- 124.Robert C, Karaszewska B, Schachter J, et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med. 2014 Nov 16; doi: 10.1056/NEJMoa1412690. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- 125.Larkin J, Ascierto PA, Dreno B, et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med. 2014;371:1867–76. doi: 10.1056/NEJMoa1408868. [DOI] [PubMed] [Google Scholar]
- 126.O’Day SJ, Maio M, Chiarion-Sileni V, et al. Efficacy and safety of ipilimumab monotherapy in patients with pretreated advanced melanoma: A multicenter single-arm phase II study. Ann Oncol. 2010;21:1712–7. doi: 10.1093/annonc/mdq013. [DOI] [PubMed] [Google Scholar]
- 127.Melero I, Hervas-Stubbs S, Glennie M, Pardoll DM, Chen L. Immunostimulatory monoclonal antibodies for cancer therapy. Nat Rev Cancer. 2007;7:95–106. doi: 10.1038/nrc2051. [DOI] [PubMed] [Google Scholar]
- 128.O’Day SJ, Hamid O, Urba WJ. Targeting cytotoxic T-lymphocyte antigen-4 (CTLA-4): A novel strategy for the treatment of melanoma and other malignancies. Cancer. 2007;110:2614–27. doi: 10.1002/cncr.23086. [DOI] [PubMed] [Google Scholar]
- 129.Peggs KS, Quezada SA, Korman AJ, Allison JP. Principles and use of anti-CTLA4 antibody in human cancer immunotherapy. Curr Opin Immunol. 2006;18:206–13. doi: 10.1016/j.coi.2006.01.011. [DOI] [PubMed] [Google Scholar]
- 130.Rudd CE, Taylor A, Schneider H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunol Rev. 2009;229:12–26. doi: 10.1111/j.1600-065X.2009.00770.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Alegre ML, Frauwirth KA, Thompson CB. T-cell regulation by CD28 and CTLA-4. Nat Rev Immunol. 2001;1:220–8. doi: 10.1038/35105024. [DOI] [PubMed] [Google Scholar]
- 132.Rudd CE, Schneider H. Unifying concepts in CD28, ICOS and CTLA4 co-receptor signalling. Nat Rev Immunol. 2003;3:544–56. doi: 10.1038/nri1131. [DOI] [PubMed] [Google Scholar]
- 133.Chikuma S, Bluestone JA. CTLA-4 and tolerance: The biochemical point of view. Immunol Res. 2003;28:241–53. doi: 10.1385/IR:28:3:241. [DOI] [PubMed] [Google Scholar]
- 134.Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol. 2005;23:515–48. doi: 10.1146/annurev.immunol.23.021704.115611. [DOI] [PubMed] [Google Scholar]
- 135.Wolchok JD, Neyns B, Linette G, et al. Ipilimumab monotherapy in patients with pretreated advanced melanoma: A randomised, double-blind, multicentre, phase 2, dose-ranging study. Lancet Oncol. 2010;11:155–64. doi: 10.1016/S1470-2045(09)70334-1. [DOI] [PubMed] [Google Scholar]
- 136.Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Pembrolizumab [Internet] National Cancer Institute; [cited 2014 Dec 30]. Available from: http://www.cancer.gov/drugdictionary?cdrid=695789. [Google Scholar]
- 138.FDA; FDA approves Keytruda for advanced melanoma. [Internet] [updated 2014 Sept 4; cited 2014 Dec 30]. Available from: http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm412802.htm. [Google Scholar]
- 139.FDA; FDA approves Opdivo for advanced melanoma. [Internet] [updated 2014 Dec 22; cited 2014 Dec 30]. Available from: http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm427716.htm. [Google Scholar]
- 140.Nivolumab. National Cancer Institute; [Internet] [cited 2014 Dec 30]. Available from: http://www.cancer.gov/drugdictionary?cdrid=539733. [Google Scholar]
- 141.vAscierto PA1, Kirkwood JM, Grob JJ, et al. The role of BRAF V600 mutation in melanoma. J Transl Med. 2012;10:85. doi: 10.1186/1479-5876-10-85. [DOI] [PMC free article] [PubMed] [Google Scholar]

