Open Access

Molecular characteristics of uveal melanoma and intraocular tumors (Review)

  • Authors:
    • Periklis Katopodis
    • Mohammad S. Khalifa
    • Vladimir Anikin
  • View Affiliations

  • Published online on: November 3, 2020     https://doi.org/10.3892/ol.2020.12270
  • Article Number: 9
  • Copyright: © Katopodis et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Malignant melanomas within the eye present different types of metabolic and metastatic behavior. Uveal melanoma (UM) affects a quarter of a million individuals in the USA; however, the molecular pathogenesis in not well understood. Although UV radiation is a risk factor in cutaneous melanomas, it is not crucial for UM progression. Apart from chromosomal abnormalities, numerous major tumorigenic signaling pathways, including the PI3K/Akt, MAPK/ERK, Ras‑association domain family 1 isoform A and Yes‑associated protein/transcriptional co‑activator with PDZ‑binding motif signaling pathways, are associated with intraocular tumors. The present review describes the current insights regarding these signaling pathways that regulate the cell cycle and apoptosis, and could be used as potential targets for the treatment of UMs.

Introduction

Uveal melanoma (UM) is the most common primary malignant tumor of the eye in adults (1). Uveal, as well as cutaneous, melanomas have origins from the same precursor cell, the melanocyte, which migrates from the neural crest during embryonic development (2). UM represents 3–5% of all melanomas and it arises from proliferating atypical melanocytes situated in the choroid (85–90%), the ciliary body (5–8%) and the iris (5–8%) (1). Primary tumor location has an effect on UM progression: Melanoma originating from melanocytes in the iris is usually associated with good prognosis, while choroidal and ciliary ones have a poor prognosis, and in ≤50% of all cases lead to metastatic disease (3), which most commonly occurs in the liver (60.5%), lung (24.4%), skin (11%) and bone (8.4%) (1,4).

UM has a median diagnostic age of 62 years, with a peak between 70 and 79 years (5,6). It has been recorded to have 30% higher incidence in males; however, to the best of our knowledge, there is no known reason for this (7). Based on the Surveillance, Epidemiology, and End Results database (1973–2009), cases of UMs affect 5.1 in every million individuals (5). In Europe, the incidence varies between 2 and 8 individuals per million based on latitude according to the European Cancer Registry (1983–1994) (8). Additionally, UMs are less prevalent in Asian and black populations (9).

Similarly to other melanomas, the most common risk factors of UM are fair skin, light eyes, ocular melanocytosis, dysplastic nevus syndrome and multiple mutations (1012). Exposure to UV radiation is a major risk factor for the development of cutaneous melanoma; however, there is little evidence regarding its role in UM progression (13). Since UVA is mainly filtered by the cornea and lens, while UVB and UVC do not reach the choroid, ‘it is unlikely’ that UV radiation exposure is responsible for choroidal melanoma (14,15).

The primary tumor is often difficult to diagnose as a third of the cases are asymptomatic (6). In most cases, UM is manifested through blurred vision and a variety of other symptoms, such as elevated intraocular pressure, which causes glaucoma (16). UM is frequently misdiagnosed as glaucoma, since the latter is one of the potential side effects of the tumor (16). The median life expectancy after metastatic growth is 13.4 months with an 8% survival rate after 2 years (4).

Gene expression profiling is currently used to classify UMs into two distinct types depending on their ability to metastasize. Class 1 are tumors with a 1% chance of spreading, while class 2 are tumors that have a 25.9% chance of forming secondary tumors (17). Little is known regarding its molecular pathogenesis, and it has been considered that a variety of epigenetic alterations occur in the melanocyte to UM pathway (18). p53 and the retinoblastoma (Rb) signaling pathway are commonly inhibited, whereas the PI3K/Akt signaling pathway in mostly activated (18). The present review discusses these signaling pathways that regulate cell death and the cell cycle in UM. Increased understanding of these pathways may lead to the identification of the genetic profile of UM, enable the design of a personalized targeted therapy for the patient and, finally, an improved prognosis of patients with UM.

Chromosomal origin of UM

UM is often characterized by multiple chromosomal aberrations. Abnormalities on chromosomes 1, 3, 6 and 8 have been observed in 17–61% of UM cases (18). Additionally, these have been demonstrated to affect the prognosis and development of the tumor (19). The most common chromosomal aberrations result in loss of chromosome 3 and 6q, or in the gain of 6p and 8q (19). Monosomy 3 is observed in 50% of all tumors, 65% of UMs and in >70% of metastasizing UMs (20). Chromosome 3 loss results in a >50% reduction in the 5-year survival rate of patients (19). By contrast, patients with an intact pair of chromosome 3 have a 5-year survival rate of 90%, which is reduced to 37% by the loss of one sister chromatid (20). BRCA1 associated protein 1 (BAP1) is one of the genes that is mutated in 47% of all Ums, and, due to its location on chromosome 3, it is important for the understanding of the disease development (21). This also explains why monosomy 3 is associated with a poor prognosis. Furthermore, BAP1 is a single copy gene, which often results in inactivating mutations (21). In UM, this results in an earlier onset at the age of 30 to 59 years (22). Additionally, mutations in BAP1 have been associated with an 11% higher risk of secondary malignancies (22). Other genes located on chromosome 3 encode PI3K and Ras-association domain family 1 isoform A (RASSF1A), both of which are associated with essential molecular pathways that are mutated in cancer (23,24).

Other aberrations, including gain of chromosome 8, loss of chromosome 1 or polysomy 8q, have been associated with a reduced survival due to various factors, such as exposure to sun light, oculodermal melanocytosis and dysplastic nevi (25,26). Some of the genes located on chromosome 1 are associated with essential molecular pathways by encoding PI3K and Akt, or tumor suppressor genes (TSGs), such as centromere protein S (27). On the other hand, gain of chromosome 6p has been associated with good prognosis, despite the abundance of oncogenes (28).

One of the most common chromosomal abnormalities in UM is rearrangement of chromosome 8q. Copy number variations have been observed in 79% of UM cases (26). In patients with a normal 8q number the 5-year survival rate is 93%; however, with the increase of copy numbers, this rate is reduced to 67 and then 29% (20).

It has been noted that there are two distinct developmental pathways in UM. Class one exhibits disomy 3 with gain of chromosome 6p, while class 2 typically exhibits monosomy 3 and a high metastatic propensity (29,30). These chromosomal aberrations are present at early stages, while increased aneuploidy and changes in chromosome 8 are considered to be associated with later stages (31). While this two-class model is relatively simplistic, it provides a good basic understanding of the main chromosomal aberrations and their consequential effects.

Cell death regulation

One of the hallmarks of cancer is the ability of cells to evade death signals and proliferate indefinitely (32). Therefore, the regulation of the cell cycle and the induction of self-mediated cell death, also known as apoptosis, is vital.

The extrinsic apoptotic signaling pathway is activated when a death ligand binds to a death receptor on the cell membrane. These receptors have an intracellular death domain that recruits adapter proteins, which results in the formation of a binding site known as death-inducing signaling complex (DISC) (33). DISC assembles and activates pro-caspase-8, which initiates apoptosis by cleaving other caspases (34). Once activated, caspase 3 cleaves the caspase-activated deoxyribonuclease, which begins the process of DNA degradation (35). Finally, downstream caspases induce cleavage of protein kinases and proteins, and break down the cytoskeleton disturbing signaling pathways, which results in the typical morphological alterations of apoptosis (36).

The mitochondrial intrinsic pathway is initiated within the cell due to internal stimuli, such as genetic damage, hypoxia and oxidative stress (37). This results in the release of pro-apoptotic molecules, apoptosis inducing factor mitochondria associated 1, second mitochondria-derived activator of caspase, diablo IAP-binding mitochondrial protein, HtrA serine peptidase 2 and cytochrome c, which initiate apoptosis (38). Subsequently, cytochrome c and Apaf-1 assemble the apoptosome which activates caspase-9 (38). Alongside caspase-9 activation, caspase-3 is activated, leading to the same steps as the aforementioned extrinsic pathway (39). The Bcl-2 family proteins, which are directly controlled by p53, determine the cell fate through the balance of pro- and anti-apoptotic molecules (38).

The molecular and genetic makeup of UMs is considered to be more complicated than the aforementioned mutations. Therefore, the present review will explore the potential role of multiple molecular pathways and their role in UM development and pathogenesis.

G protein subunit αq and G protein subunit α11

Despite the chromosome abnormalities, most UMs are considered to be caused by point mutations in G protein α subunits, specifically in G protein subunit αq (GNAQ) and G protein subunit α11 (GNA11), regardless of tumor stage or chromosomal constellation (40). These mutations have similar effects as mutations in RAS, which are common in a number of other tumors (41). The G-α subunit is important due to its involvement in multiple essential cellular pathways, such as the MAPK (cell proliferation and apoptosis), PI3K-Akt (growth and homeostasis) and Hippo signaling pathways (42). GNAQ and GNA11 activate phospholipase C, which triggers a cascade of events resulting in the activation of protein kinase C (PKC). PKC then initiates a phosphorylation cascade, which activates Raf, MEK1/2 and ERK (43). This process results in the regulation of cell proliferation and survival (44). It has been hypothesized that these mutations are early events in the pathogenesis of UM and are necessary for tumor malignancy (45). On the other hand, mutant GNAQ and GNA11 are considered to be weak oncogenes and, therefore, cannot cause damage to melanocytes unless they are already deficient in the p53 and p16/CDK4/RB signaling pathways (46). Due to the importance of GNAQ and GNA11 in UM malignancy, the 5-oxo-ETE acid G-protein-coupled receptor 1 (GPCR) signaling pathway is a potential viable therapeutic target (42). The development of inhibitors for specific molecules, such as Gq/11 inhibitor YM-254890 and Arf6-inhibitor NAV-2729, is one of the main strategies that is currently being investigated (47,48). One such example is the inhibition of CysLT2R-L129Q, which is responsible for the constitutive activation of the Gq/11 signaling pathway in UM (47,49).

RAS interactions

The Ras superfamily consists of small GTPases that act as switches and modulate a vast array of cell functions by influencing signaling pathways. They are separated into six different groups of proteins, and are present in all cell types (50). One of the subfamilies, also referred to as RAS, consists of proteins that regulate cell proliferation. They have downstream effects on signaling pathways crucial to UM, such as the MAPK/ERK and PI3K/AKT/PTEN signaling pathways (51). In cancer, RAS is often mutated which affects a number of these pathways and makes them less sensitive to apoptosis triggers, thus increasing proliferation levels (Fig. 1) (52).

All three common RAS proteins in humans are highly conserved in the active regions and often undergo mutations in codons 12, 13 and 61 (53). The resulting point mutations lead to preferential binding to GTP over GDP, which in-turn leads to activation of proliferation pathways (54). Interestingly, RAS mutations are not usually associated with UM (55,56).

In general, RAS serve as activating proteins that remove GDP and allow GTP to bind its target (57). Ras-bound GTP goes on to activate Raf, which initiates the MAPK/ERK signaling pathway (58). In the case of PI3K, active Ras directly activates it without an intermediary protein (59).

PI3K/Akt/PTEN signaling pathway

The PI3K/Akt/PTEN is one of the main molecular pathways involved in cell proliferation. It is mutated in multiple types of cancer and is constitutively activated in most UMs (24). RAS directly activates PI3K, which then goes on to phosphorylate phosphatidylinositol 4,5-bisphosphate to produce phosphatidylinositol (3,4,5)-trisphosphate (PIP3) (60). PIP3 is dephosphorylated by PTEN to regulate PIP3 levels, which, when elevated, activates Akt. Subsequently, Akt goes on to phosphorylate a number of signaling pathways, such as the mTORC1, MDM2 proto-oncogene (MDM2), BAD and GTPase-activating protein signaling pathways (Fig. 1) (61).

Akt is an anti-apoptotic protein which serves an important role in cell survival and tumorigenesis (62). It is activated via phosphorylation, becoming phospho-Akt which inactivates several proteins, including members of the Bcl-2 family (BAD protein) and caspase-9 (63). Phospho-Akt is involved in the protection from apoptosis, but also in other cancer development processes, such as blockage of anti-proliferative signaling, facilitation of cell replication and angiogenesis (63). Using immunohistochemical testing, it has been demonstrated that phosphorylation of Akt is associated with a poor prognosis (62).

PTEN downregulation has also been associated with various types of cancer, including breast cancer, thyroid cancer, kidney cancer, endometrial cancer, colorectal cancer and melanoma (64,65). In UMs, loss of heterozygosity of PTEN has been observed in 76% of tumors, with 11% being within the PTEN coding region (66). Loss of cytoplasmic PTEN in primary UM tumors was associated with shortened disease-free survival (67). Its decreased expression can also result in increased aneuploidy and reduced survival (45).

Overall, these findings suggest that the PI3K/Akt/PTEN signaling pathway serves a vital role in UM progression; however, more research needs to be performed to fully understand its role.

MAPK/ERK signaling pathway

The MAPK/ERK pathway is crucial for mediating cell-cycle progression. In multiple cancer types, it is constitutively activated, resulting in proliferation of neoplastic cells (68,69). It has also been identified to serve an important role in melanocytic neoplasia (70).

MAPK signaling, similar to the PI3K/Akt signaling pathway, begins with the activation of RAS, which then recruits RAF (68). RAFs are a group of kinases that transduce signals along the MAPK signaling pathway (68). There are three isoforms that are expressed in humans: A-Raf proto-oncogene serine/threonine kinase, BRAF and Raf-1 proto-oncogene serine/threonine kinase (cRAF) (71). cRAF was the first to be discovered and BRAF has been the most extensively studied due to its high mutation rate in various types of cancer (72,73).

Activation of BRAF by RAS results in the phosphorylation of kinases, such as MEK1/2 and ERK1/2, which induces a multitude of proliferative and survival processes (74) via the consequent activation of transcription factors, such as ETS transcription factor ELK1. In cutaneous melanomas, RAS and BRAF often undergo activating mutations (75). These also appear in benign melanocytic naevi and, alongside activation of the MAPK signaling pathway, constitute early events of melanogenesis (51,76,77). In UM, the MAPK signaling pathway is upregulated, which advocates for the presence of upstream mutations (78). It is also known that mutations in RAS and BRAF are uncommon in UM (7981). Therefore, the constitutive activation is considered to be caused by mutations in the GNAQ family, which results in the upregulation of the pathways (82,83).

Yes-associated protein/transcriptional co-activator with PDZ-binding motif signaling pathway and its potential for treatment

Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) modulate regulation of cell proliferation, migration and survival (84). They are, in turn, negatively regulated by the Hippo signaling pathway (85), which acts as a tumor suppressor to limit cell proliferation and organ size regulation (86). As previously mentioned, mutations in GNAQ and GNA11 are present in >80% of UM cases. This is essential as GPCRs activate F-actin, which targets YAP/TAZ and, therefore, could result in the upregulation of the pathway (87). When this happens, YAP is activated independently of the Hippo signaling pathway, resulting in greater resistance to contact inhibition of growth (8890).

In contrast to MAPK-targeted therapy, which has no impact on the prognosis of patients with UM, YAP-targeted therapy is strongly associated with cancer metastasis (91) and shows promise as an ideal target (9193). For example, focal adhesion kinase-targeted therapy reduces YAP levels and counteracts the effects of the GNAQ/GNA11 mutation (43). This may be effective in treating UMs that exhibit such mutations (94).

p53

p53 is one of the key apoptotic regulators, which induces cell cycle arrest and consequently apoptosis. A study by Liu and Zhou (95) explored the role of p53 in the development of UM. They observed that p53 expression and prognosis were negatively associated. The mortality rate increased with p53 expression. Additionally, inhibition of p53 has been associated with inhibited invasion in UM (95). Other studies have mentioned that mutations in the p53 gene are rare and, in fact, most causes of disruption in the pathways are due to upstream or downstream mutations (2,96,97). One such cause could be MDM2 upregulation, which is common in UMs (98). MDM2 regulates p53 and reduces its expression (98).

Protection from apoptosis is a major factor in the metastatic cascade of most types of cancer, including UM. p53 does not appear to have a significant impact on UM (99). However, a previous study demonstrated that multiplication of chromosome 8 and c-myc expression are associated, suggesting that c-myc could be used as a prognostic factor (11). C-myc alone is involved in the regulation of cell proliferation and with p53-dependent mechanisms promotes apoptosis (100). On the other hand, Bcl-2, which is vital for the intrinsic apoptotic pathway, has been reported to be upregulated in most UMs (2,101). A strong inverse relationship has been observed between c-myc (nuclear and cytoplasmic) and Bcl-2, suggesting that the latter co-operates with c-myc to immortalize UM cells (79,101).

Brantley and Harbour (2) immunohistochemically analyzed the p53 and retinoblastoma protein (pRb) pathways and found that, in UM cases, most alterations are due to mutations in other proteins.

CDK and cyclin kinase inhibitors

A fine balance between cyclin kinase inhibitors (CDKs) and CKIs is required for a normal cell cycle (102). If toxic chemicals, oxidative stress (reactive oxygen species), ionizing radiation and other factors induce DNA damage, the cell must repair it and reenter the cell cycle (102). When the cell fails to repair the damage, it becomes senescent and the cell cycle is arrested in G1 phase (diploid DNA) or G2 phase (tetraploid DNA content). Deregulation of the cell cycle, especially at the G2/M phase, leads the cell to a more cancerous fate (103).

Cell cycle regulation is achieved through a family of serine/threonine kinase holoenzyme complexes consisting of regulatory cyclins that bind to and activate catalytic CDKs. The cyclins D1, D2, D3 and E are important for the G1-S cell cycle transition (104). Cyclin A is involved in DNA synthesis, S-phase completion and preparation for mitosis, while cyclins B1 and B2 control the onset, sequence of events and completion of mitosis (105). For example, the cyclin B1/CDK1 complex is a mitotic regulator that is responsible for the progression of the cell cycle (105).

On the other hand, cyclin-dependent kinase inhibitors (CDKIs) negatively regulate the kinase activity of the cyclin-CDK complexes (106). There are two known families of CDKIs: The cyclin dependent kinase inhibitor 2A (INK4) family, which includes p16/INK4A, p15/INK4B, p18/INK4C and p19 (p14)/INK4D, and the cardiac ISL1-interacting protein (CIP)/calcium and integrin binding 1 (KIP) family, which includes p21/CIP1, p27/KIP1 and p57/KIP2 (107109).

Different CKIs have been observed to affect different CDKs. For example, p15 and p16 inhibit CDK4 and CDK6 respectively, while p21 and p27 act on G1 CDK cyclins and S-phase CDK2 complexes (110,111). Both p21 and p27 inhibit DNA replication but through different mechanisms. p21 binds to and promotes CDK1 and CDK2 with cyclin D activity, while, under certain conditions, inhibiting CDK4 and CDK6 with cyclin E activity (112). p27 promotes CDK2 cyclin E complex and CDK4/6 cyclin D complex formation (113,114).

In uveal and choroidal melanoma cell lines, the expression levels of p21 and p27 are downregulated resulting in suppressed p16-CDK interaction (110,115). This results in more CDK activity, leading to cyclin D and CDKs phosphorylating pRb, which goes on to release E2F transcription factor 1 (E2F1) (116). E2F1 is a transcriptional factor that leads to the expression of numerous necessary factors for G1 to S phase progression (117). pRb is another one of the vital molecules in the cell cycle regulation of uveal and choroidal cells. It is encoded by the Rb gene and serves an important role as a tumor suppressor (118). Deregulation and inactivation of p16 and/or overexpression of cyclin D leads to inactivation of pRb by cyclin-dependent phosphorylation (119,120). In most UMs, the Rb protein is constitutively hyperphosphorylated and functionally inactivated (120). This has been attributed to cyclin D1 upregulation in 65% of cases and has also been associated with larger tumor sizes and poor prognosis (63,76,121).

E2F transcription factors are key regulators of cell division and, among them, E2F1, E2F2 and E2F3a are potent activators of E2F-responsive genes, but their transcriptional activity is inhibited by binding to pRb (122,123). pRb is functionally inactivated at the G1-S transition by cyclin D-CDK4/CDK6 and cyclin E-CDK2-mediated phosphorylation, thus enabling E2F transcription factors to activate their target genes (124).

RASSF1A

RASSF1A is a TSG that is required for death receptor-dependent apoptosis (80) and can be found at the 3p21.3 locus. RASSF1A inhibits the accumulation of cyclin D1 protein without affecting its mRNA levels (81). This results in suppressed proliferation via negative regulation of the cell cycle progression at the G1/S phase transition (125). It has been reported that the endogenous inactivation of RASSF1A leads to a decrease of p27 which is a negative cell cycle regulator at the protein level (111,125). The depletion of this gene in RASSF1A mouse mutants has been noted as an early event in the senescence of uveal melanocytes and is considered to contribute to the malignancy of UM (125). RASSF1A is frequently hypermethylated in UM, which results in its downregulation (126). Additionally, in 83% of the cases in a study by Calipel et al (125), the RASSF1A promoter was methylated which suppressed gene expression. Overall, the downregulation of RASSF1A is most likely explained by the loss of heterozygosity typical for UM.

Conclusion

UM is the most common intraocular tumor in adults and is caused by multiple molecular abnormalities. The most frequent mutations in GNAQ and GNA11 are considered to be the main driving events in UM. Due to the involvement of GPCRs in multiple molecular signaling pathways, the present review explored the various downstream effects that such mutations could trigger. Additionally, the potential effects of chromosomal abnormalities, and how the loss or gain of specific regions could improve or worsen prognosis, were described. All this information allowed the authors to pinpoint potential therapeutic targets which could be used to successfully treat patients with UM. Based on the understanding of the aforementioned pathways and DNA expression profiles of UM, prediction models can be produced, and this could lead to improved prognosis for patients with UM.

Acknowledgements

The authors wish to acknowledge the help provided by Ms. Heerni Halai (College of Health, Medicine and Life Sciences, Brunel University, UK).

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

PK produced and reviewed the manuscript and figure. MSK and VA reviewed the manuscript. VA sponsored the publication. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Krantz BA, Dave N, Komatsubara KM, Marr BP and Carvajal RD: Uveal melanoma: Epidemiology, etiology, and treatment of primary disease. Clin Ophthalmol. 11:279–289. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Brantley MA Jr and Harbour JW: Deregulation of the Rb and p53 pathways in uveal melanoma. Am J Pathol. 157:1795–1801. 2000. View Article : Google Scholar : PubMed/NCBI

3 

Weber A, Hengge UR, Urbanik D, Markwart A, Mirmohammadsaegh A, Reichel MB, Wittekind C, Wiedemann P and Tannapfel A: Absence of mutations of the BRAF gene and constitutive activation of extracellular-regulated kinase in malignant melanomas of the uvea. Lab Invest. 83:1771–1776. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Kuk D, Shoushtari AN, Barker CA, Panageas KS, Munhoz RR, Momtaz P, Ariyan CE, Brady MS, Coit DG, Bogatch K, et al: Prognosis of mucosal, uveal, acral, nonacral cutaneous, and unknown primary melanoma from the time of first metastasis. Oncologist. 21:848–854. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Andreoli MT, Mieler WF and Leiderman YI: Epidemiological trends in uveal melanoma. Br J Ophthalmol. 99:1550–1553. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Damato EM and Damato BE: Detection and time to treatment of uveal melanoma in the United Kingdom: An evaluation of 2,384 patients. Ophthalmology. 119:1582–1589. 2012. View Article : Google Scholar : PubMed/NCBI

7 

McLaughlin CC, Wu X, Jemal A, Martin HJ, Roche LM and Chen VW: Incidence of Noncutaneous Melanomas in the U.S. Cancer. 103:1000–1007. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Virgili G, Gatta G, Ciccolallo L, Capocaccia R, Biggeri A, Crocetti E, Lutz JM and Paci E; EUROCARE Working Group, : Incidence of uveal melanoma in Europe. Ophthalmology. 114:2309–2315. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Hu DN, Yu GP, Mccormick SA, Schneider S and Finger PT: Population-based incidence of uveal melanoma in various races and ethnic groups. Am J Ophthalmol. 140:612–617. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Nayman T, Bostan C, Logan P and Burnier MN Jr: Uveal melanoma risk factors: A systematic review of meta-analyses. Curr Eye Res. 42:1085–1093. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Kaliki S, Shields CL and Shields JA: Uveal melanoma: Estimating prognosis. Indian J Ophthalmol. 63:93–102. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Weis E, Shah CP, Lajous M, Shields JA and Shields CL: The association of cutaneous and iris nevi with uveal melanoma: A meta-analysis. Ophthalmology. 116:536–543.e2. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Gendron P, Desgarnier MD, Mallet JD and Rochette PJ: Implication of ultraviolet light in the etiology of uveal melanoma (Review). Photochem Photobiol. 90:15–21. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Mallet JD and Rochette PJ: Themed issue: Interaction of UV radiation with DNA. Photochem Photobiol Sci. 12:1245–1246. 2013.

15 

Mallet JD, Gendron SP, Drigeard Desgarnier MC and Rochettes PJ: Implication of ultraviolet light in the etiology of uveal melanoma: A review. Photochem Photobiol. 90:15–21. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Shields CL, Materin MA, Shields JA, Gershenbaum E, Singh sAD and Smith A: Factors associated with elevated intraocular pressure in eyes with iris melanoma. Br J Ophthalmol. 85:666–669. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Onken MD, Worley LA, Char DH, Augsburger JJ, Correa ZM, Nudleman E, Aaberg TM Jr, Altaweel MM, Bardenstein DS, Finger PT, et al: Collaborative Ocular Oncology Group report number 1: prospective validation of a multi-gene prognostic assay in uveal melanoma. Ophthalmology. 119:1596–1603. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Coupland SE, Lake SL, Zeschnigk M and Damato BE: Molecular pathology of uveal melanoma. Eye (Lond). 27:230–242. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Sisley K, Rennie IG, Parsons MA, Jacques R, Hammond DW, Bell SM, Potter AM and Rees RC: Abnormalities of chromosomes 3 and 8 in posterior uveal melanoma correlate with prognosis. Genes Chromosomes Cancer. 19:22–28. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Versluis M, de Lange MJ, van Pelt SI, Ruivenkamp CA, Kroes WG, Cao J, Jager MJ, Luyten GP and van der Velden PA: Digital PCR validates 8q dosage as prognostic tool in uveal melanoma. PLoS One. 10:e01163712015. View Article : Google Scholar : PubMed/NCBI

21 

Harbour JW, Onken MD, Roberson EDO, Duan S, Cao L, Worley LA, Council ML, Matatall KA, Helms C and Bowcock AM: Frequent mutation of BAP1 in metastasizing uveal melanomas. Science. 330:1410–1413. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Laíns I, Bartosch C, Mondim V, Healy B, Kim IK, Husain D and Miller JW: Second primary neoplasms in patients with uveal melanoma: A SEER Database Analysis. Am J Ophthalmol. 165:54–64. 2016. View Article : Google Scholar : PubMed/NCBI

23 

van der Weyden L and Adams DJ: The Ras-association domain family (RASSF) members and their role in human tumourigenesis. Biochim Biophys Acta. 1776:58–85. 2007.PubMed/NCBI

24 

Babchia N, Calipel A, Mouriaux F, Faussat AM and Mascarelli F: The PI3K/Akt and mTOR/P70S6K signaling pathways in human uveal melanoma cells: Interaction with B-Raf/ERK. Invest Ophthalmol Vis Sci. 51:421–429. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Rodríguez A, Dueñas-Gonzalez A and Delgado-Pelayo S: Clinical presentation and management of uveal melanoma. Mol Clin Oncol. 5:675–677. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Ewens KG, Kanetsky PA, Richards-yutz J, Al-Dahmash S, De Luca MC, Bianciotto CG, Shields CL and Ganguly A: Genomic profile of 320 uveal melanoma cases: Chromosome 8p-loss and metastatic outcome. Invest Ophthalmol Vis Sci. 54:5721–5729. 2013. View Article : Google Scholar : PubMed/NCBI

27 

van Gils W, Mensink HW, Kilic E, Vaarwater J, Verbiest MM, Paridaens D, Luyten GP, de Klein A and Brüggenwirth HT: Expression of APITD1 is not related to copy number changes of chromosomal region 1p36 or the prognosis of uveal melanoma. Invest Ophthalmol Vis Sci. 48:4919–4923. 2007. View Article : Google Scholar : PubMed/NCBI

28 

van Gils W, Kilic E, Brüggenwirth HT, Vaarwater J, Verbiest MM, Beverloo B, van Til-Berg ME, Paridaens D, Luyten GP and de Klein A: Regional deletion and amplification on chromosome 6 in a uveal melanoma case without abnormalities on chromosomes 1p, 3 and 8. Melanoma Res. 18:10–15. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Parrella P, Sidransky D and Merbs SL: Allelotype of posterior uveal melanoma: Implications for a bifurcated tumor progression pathway. Cancer Res. 59:3032–3037. 1999.PubMed/NCBI

30 

Tschentscher F, Hüsing J, Hölter T, Kruse E, Dresen IG, Jöckel KH, Anastassiou G, Schilling H, Bornfeld N, Horsthemke B, et al: Tumor classification based on gene expression profiling shows that uveal melanomas with and without monosomy 3 represent two distinct entities. Cancer Res. 63:2578–2584. 2003.PubMed/NCBI

31 

Harbour JW: The genetics of uveal melanoma: An emerging framework for targeted therapy. Pigment Cell Melanoma Res. 25:171–181. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Hanahan D and Weinberg RA: The hallmarks of cancer. Cell. 100:57–70. 2000. View Article : Google Scholar : PubMed/NCBI

33 

Kim JW, Choi EJ and Joe CO: Activation of death-inducing signaling complex (DISC) by pro-apoptotic C-terminal fragment of RIP. Oncogene. 19:4491–4499. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Tummers B and Green DR: Caspase-8: Regulating life and death. Immunol Rev. 277:76–89. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Wong RSY: Apoptosis in cancer: From pathogenesis to treatment. J Exp Clin Cancer Res. 30:872011. View Article : Google Scholar : PubMed/NCBI

36 

Wang RA, Li QL, Li ZS, Zheng PJ, Zhang HZ, Huang XF, Chi SM, Yang AG and Cui R: Apoptosis drives cancer cells proliferate and metastasize. J Cell Mol Med. 17:205–211. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Pistritto G, Trisciuoglio D, Ceci C, Garufi A and D'Orazi G: Apoptosis as anticancer mechanism: Function and dysfunction of its modulators and targeted therapeutic strategies. Aging (Albany NY). 8:603–619. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Wiman KG: Strategies for therapeutic targeting of the p53 pathway in cancer. Cell Death Differ. 13:921–926. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Parrish AB, Freel CD and Kornbluth S: Cellular mechanisms controlling caspase activation and function. Cold Spring Harb Perspect Biol. 5:52013. View Article : Google Scholar

40 

Cancer Genome Atlas Network, . Genomic Classification of cutaneous melanoma. Cell. 161:1681–1696. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Kalinec G, Nazarali AJ, Hermouet S, Xu N and Gutkind JS: Mutated alpha subunit of the Gq protein induces malignant transformation in NIH 3T3 cells. Mol Cell Biol. 12:4687–4693. 1992. View Article : Google Scholar : PubMed/NCBI

42 

Urtatiz O and Van Raamsdonk CD: Gnaq and Gna11 in the endothelin signaling pathway and melanoma. Front Genet. 7:592016. View Article : Google Scholar : PubMed/NCBI

43 

Croce M, Ferrini S, Pfeffer U and Gangemi R: Targeted therapy of uveal melanoma: Recent failures and new perspectives. Cancers (Basel). 11:8462019. View Article : Google Scholar

44 

Rozengurt E: Mitogenic signaling pathways induced by G protein-coupled receptors. J Cell Physiol. 213:589–602. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Landreville S, Agapova OA and Harbour JW: Emerging insights into the molecular pathogenesis of uveal melanoma. Future Oncol. 4:629–636. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Van Raamsdonk CD, Bezrookove V, Green G, Bauer J, Gaugler L, O'Brien JM, Simpson EM, Barsh GS and Bastian BC: Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature. 457:599–602. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Ceraudo E, Horioka M, Mattheisen J, Hitchman TD, Moore AR, Kazmi MA, Chi P, Chen Y, Sakmar TP and Huber T: Uveal melanoma oncogene CYSLTR2 encodes a constitutively active GPCR highly biased toward Gq signaling. bioRxiv. Jun 6–2019.(Epub ahead of print). doi: org/10.1101/663153.

48 

Chua V, Lapadula D, Randolph C, Benovic JL, Wedegaertner PB and Aplin AE: Dysregulated GPCR signaling and therapeutic options in uveal melanoma. Mol Cancer Res. 15:501–506. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Pandiani C, Béranger GE, Leclerc J, Ballotti R and Bertolotto C: Focus on cutaneous and uveal melanoma specificities. Genes Dev. 31:724–743. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Zenonos K and Kyprianou K: RAS signaling pathways, mutations and their role in colorectal cancer. World J Gastrointest Oncol. 5:97–101. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Zuidervaart W, van Nieuwpoort F, Stark M, Dijkman R, Packer L, Borgstein AM, Pavey S, van der Velden P, Out C, Jager MJ, et al: Activation of the MAPK pathway is a common event in uveal melanomas although it rarely occurs through mutation of BRAF or RAS. Br J Cancer. 92:2032–2038. 2005. View Article : Google Scholar : PubMed/NCBI

52 

Fernández-Medarde A and Santos E: Ras in cancer and developmental diseases. Genes Cancer. 2:344–358. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Prior IA, Lewis PD and Mattos C: A comprehensive survey of Ras mutations in cancer. Cancer Res. 72:2457–2467. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Muñoz-Maldonado C, Zimmer Y and Medová M: A comparative analysis of individual RAS mutations in cancer biology. Front Oncol. 9:10882019. View Article : Google Scholar : PubMed/NCBI

55 

Mooy CM, Van der Helm MJ, Van der Kwast TH, De Jong PT, Ruiter DJ and Zwarthoff EC: No N-ras mutations in human uveal melanoma: The role of ultraviolet light revisited. Br J Cancer. 64:411–413. 1991. View Article : Google Scholar : PubMed/NCBI

56 

Soparker CN, O'Brien JM and Albert DM: Investigation of the role of the ras protooncogene point mutation in human uveal melanomas. Invest Ophthalmol Vis Sci. 34:2203–2209. 1993.PubMed/NCBI

57 

Wennerberg K, Rossman KL and Der CJ: The Ras superfamily at a glance. J Cell Sci. 118:843–846. 2005. View Article : Google Scholar : PubMed/NCBI

58 

Kolch W: Meaningful relationships: The regulation of the Ras/Raf/MEK/ERK pathway by protein interactions. Biochem J. 351:289–305. 2000. View Article : Google Scholar : PubMed/NCBI

59 

Castellano E and Downward J: RAS interaction with PI3K: More than just another effector pathway. Genes Cancer. 2:261–274. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Liu P, Cheng H, Roberts TM and Zhao JJ: Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov. 8:627–644. 2009. View Article : Google Scholar : PubMed/NCBI

61 

O'Donnell JS, Massi D, Teng MW and Mandala M: PI3K-AKT-mTOR inhibition in cancer immunotherapy, redux. Semin Cancer Biol. 48:91–103. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Saraiva VS, Caissie AL, Segal L, Edelstein C and Burnier MN Jr: Immunohistochemical expression of phospho-Akt in uveal melanoma. Melanoma Res. 15:245–250. 2005. View Article : Google Scholar : PubMed/NCBI

63 

Cardone MH, Roy N, Stennicke HR, Salvesen GS, Franke TF, Stanbridge E, Frisch S and Reed JC: Regulation of cell death protease caspase-9 by phosphorylation. Science. 282:1318–1321. 1998. View Article : Google Scholar : PubMed/NCBI

64 

Milella M, Falcone I, Conciatori F, Cesta Incani U, Del Curatolo A, Inzerilli N, Nuzzo CM, Vaccaro V, Vari S, Cognetti F, et al: PTEN: Multiple functions in human malignant tumors. Front Oncol. 5:242015. View Article : Google Scholar : PubMed/NCBI

65 

Chang H, Cai Z and Roberts TM: The mechanisms underlying PTEN loss in human tumors suggest potential therapeutic opportunities. Biomolecules. 9:7132019. View Article : Google Scholar

66 

Woodman SE: Metastatic uveal melanoma: Biology and emerging treatments. Cancer J. 18:148–152. 2012. View Article : Google Scholar : PubMed/NCBI

67 

Abdel-Rahman MH, Yang Y, Zhou XP, Craig EL, Davidorf FH and Eng C: High frequency of submicroscopic hemizygous deletion is a major mechanism of loss of expression of PTEN in uveal melanoma. J Clin Oncol. 24:288–295. 2006. View Article : Google Scholar : PubMed/NCBI

68 

Dhillon AS, Hagan S, Rath O and Kolch W: MAP kinase signalling pathways in cancer. Oncogene. 26:3279–3290. 2007. View Article : Google Scholar : PubMed/NCBI

69 

Burotto M, Chiou VL, Lee JM and Kohn EC: The MAPK pathway across different malignancies: A new perspective. Cancer. 120:3446–3456. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Cohen C, Zavala-Pompa A, Sequeira JH, Shoji M, Sexton DG, Cotsonis G, Cerimele F, Govindarajan B, Macaron N and Arbiser JL: Mitogen-actived protein kinase activation is an early event in melanoma progression. Clin Cancer Res. 8:3728–3733. 2002.PubMed/NCBI

71 

Leicht DT, Balan V, Kaplun A, Singh-Gupta V, Kaplun L, Dobson M and Tzivion G: Raf kinases: Function, regulation and role in human cancer. Biochim Biophys Acta. 1773:1196–1212. 2007. View Article : Google Scholar : PubMed/NCBI

72 

Barras D: BRAF mutation in colorectal cancer: An update. Biomark Cancer. 7 (Suppl 1):9–12. 2015.PubMed/NCBI

73 

Zaman A, Wu W and Bivona TG: Targeting oncogenic BRAF: Past, present, and future. Cancers (Basel). 11:11972019. View Article : Google Scholar

74 

Gaudi S and Messina JL: Molecular bases of cutaneous and uveal melanomas. Pathol Res Int. 2011:1594212011. View Article : Google Scholar

75 

Glitza IC and Davies MA: Genotyping of cutaneous melanoma. Chin Clin Oncol. 3:272014.PubMed/NCBI

76 

Brose MS, Volpe P, Feldman M, Kumar M, Rishi I, Gerrero R, Einhorn E, Herlyn M, Minna J, Nicholson A, et al: Mutations in Human Lung Cancer and Melanoma. Cancer Res. 62:6997–7000. 2002.PubMed/NCBI

77 

Shinozaki M, Fujimoto A, Morton DL and Hoon DS: Incidence of BRAF oncogene mutation and clinical relevance for primary cutaneous melanomas. Clin Cancer Res. 10:1753–1757. 2004. View Article : Google Scholar : PubMed/NCBI

78 

Harbour JW: Genomic, prognostic, and cell-signaling advances in uveal melanoma. Am Soc Clin Oncol Educ book Am Soc Clin Oncol Annu Meet. 2013:388–391. 2013. View Article : Google Scholar

79 

Mooy CM, Luyten GP, de Jong PT, Luider TM, Stijnen T, van de Ham F, van Vroonhoven CC and Bosman FT: Immunohistochemical and prognostic analysis of apoptosis and proliferation in uveal melanoma. Am J Pathol. 147:1097–1104. 1995.PubMed/NCBI

80 

Merhavi E, Cohen Y, Avraham BC, Frenkel S, Chowers I, Pe'er J and Goldenberg-Cohen N: Promoter methylation status of multiple genes in uveal melanoma. Invest Ophthalmol Vis Sci. 48:4403–4406. 2007. View Article : Google Scholar : PubMed/NCBI

81 

Shivakumar L, Minna J, Sakamaki T, Pestell R and White MA: The RASSF1A tumor suppressor blocks cell cycle progression and inhibits cyclin D1 accumulation. Mol Cell Biol. 22:4309–4318. 2002. View Article : Google Scholar : PubMed/NCBI

82 

Kiliç E, Brüggenwirth HT, Verbiest MM, Zwarthoff EC, Mooy NM, Luyten GP and de Klein A: The RAS-BRAF kinase pathway is not involved in uveal melanoma. Melanoma Res. 14:203–205. 2004. View Article : Google Scholar : PubMed/NCBI

83 

Helgadottir H and Höiom V: The genetics of uveal melanoma: Current insights. Appl Clin Genet. 9:147–155. 2016. View Article : Google Scholar : PubMed/NCBI

84 

Liu H, Du S, Lei T, Wang H, He X, Tong R and Wang Y: Multifaceted regulation and functions of YAP/TAZ in tumors (Review). Oncol Rep. 40:16–28. 2018.PubMed/NCBI

85 

Meng Z, Moroishi T and Guan KL: Mechanisms of Hippo pathway regulation. Genes Dev. 30:1–17. 2016. View Article : Google Scholar : PubMed/NCBI

86 

Plouffe SW, Hong AW and Guan KL: Disease implications of the Hippo/YAP pathway. Trends Mol Med. 21:212–222. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Totaro A, Panciera T and Piccolo S: YAP/TAZ upstream signals and downstream responses. Nat Cell Biol. 20:888–899. 2018. View Article : Google Scholar : PubMed/NCBI

88 

Gumbiner BM and Kim NG: The Hippo-YAP signaling pathway and contact inhibition of growth. J Cell Sci. 127:709–717. 2014. View Article : Google Scholar : PubMed/NCBI

89 

Field MG and Harbour JW: GNAQ/11 mutations in uveal melanoma: Is YAP the key to targeted therapy? Cancer Cell. 25:714–715. 2014. View Article : Google Scholar : PubMed/NCBI

90 

Feng X, Degese MS, Iglesias-Bartolome R, Vaque JP, Molinolo AA, Rodrigues M, Zaidi MR, Ksander BR, Merlino G, Sodhi A, et al: Hippo-independent activation of YAP by the GNAQ uveal melanoma oncogene through a trio-regulated rho GTPase signaling circuitry. Cancer Cell. 25:831–845. 2014. View Article : Google Scholar : PubMed/NCBI

91 

Warren JS, Xiao Y and Lamar JM: YAP/TAZ activation as a target for treating metastatic Cancer. Cancers (Basel). 10:102018. View Article : Google Scholar

92 

Zanconato F, Battilana G, Cordenonsi M and Piccolo S: YAP/TAZ as therapeutic targets in cancer. Curr Opin Pharmacol. 29:26–33. 2016. View Article : Google Scholar : PubMed/NCBI

93 

Moroishi T, Hansen CG and Guan KL: The emerging roles of YAP and TAZ in cancer. Nat Rev Cancer. 15:73–79. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Feng X, Rigiracciolo D, Lee JS, Yeerna H, Arang N, Lubrano S, Schlaepfer DD, Tamayo P, Ruppin E and Gutkind JS: Abstract 968: Targeting FAK inhibits YAP-dependent tumor growth in uveal melanoma. Cancer Res. 78:9682018.

95 

Liu H and Zhou M: Evaluation of p53 gene expression and prognosis characteristics in uveal melanoma cases. OncoTargets Ther. 10:3429–3434. 2017. View Article : Google Scholar

96 

Hajkova N, Hojny J, Nemejcova K, Dundr P, Ulrych J, Jirsova K, Glezgova J and Ticha I: Germline mutation in the TP53 gene in uveal melanoma. Sci Rep. 8:76182018. View Article : Google Scholar : PubMed/NCBI

97 

Sun Y, Tran BN, Worley LA, Delston RB and Harbour JW: Functional analysis of the p53 pathway in response to ionizing radiation in uveal melanoma. Invest Ophthalmol Vis Sci. 46:1561–1564. 2005. View Article : Google Scholar : PubMed/NCBI

98 

Shi D and Gu W: Dual Roles of MDM2 in the regulation of p53: Ubiquitination dependent and ubiquitination independent mechanisms of MDM2 tepression of p53 sctivity. Genes Cancer. 3:240–248. 2012. View Article : Google Scholar : PubMed/NCBI

99 

Hussein MR: The relationships between p53 protein expression and the clinicopathological features in the uveal melanomas. Cancer Biol Ther. 4:57–59. 2005. View Article : Google Scholar : PubMed/NCBI

100 

Evan GI, Wyllie AH, Gilbert CS, Littlewood TD, Land H, Brooks M, Waters CM, Penn LZ and Hancock DC: Induction of apoptosis in fibroblasts by c-myc protein. Cell. 69:119–128. 1992. View Article : Google Scholar : PubMed/NCBI

101 

Schwartz LH, Ferrand R, Boelle PY, Maylin C and D'Hermies F, Virmont J and D'Hermies F: Lack of correlation between the location of choroidal melanoma and ultraviolet-radiation dose distribution. Radiat Res. 147:451–456. 1997. View Article : Google Scholar : PubMed/NCBI

102 

Lim S and Kaldis P: Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development. 140:3079–3093, 20132. View Article : Google Scholar : PubMed/NCBI

103 

Ding L, Cao J, Lin W, Chen H, Xiong X, Ao H, Yu M, Lin J and Cui Q: The roles of cyclin-dependent kinases in cell-cycle progression and therapeutic strategies in human breast cancer. Int J Mol Sci. 21:19602020. View Article : Google Scholar

104 

Ando K, Ajchenbaum-Cymbalista F and Griffin JD: Regulation of G1/S transition by cyclins D2 and D3 in hematopoietic cells. Proc Natl Acad Sci USA. 90:9571–9575. 1993. View Article : Google Scholar : PubMed/NCBI

105 

Bartkova J, Lukas J, Strauss M and Bartek J: Cyclin D3: Requirement for G1/S transition and high abundance in quiescent tissues suggest a dual role in proliferation and differentiation. Oncogene. 17:1027–1037. 1998. View Article : Google Scholar : PubMed/NCBI

106 

Bonelli P, Tuccillo FM, Borrelli A, Schiattarella A and Buonaguro FM: CDK/CCN and CDKI alterations for cancer prognosis and therapeutic predictivity. BioMed Res Int. 2014:3610202014. View Article : Google Scholar : PubMed/NCBI

107 

Li J, Poi MJ and Tsai MD: Regulatory mechanisms of tumor suppressor P16(INK4A) and their relevance to cancer. Biochemistry. 50:5566–5582. 2011. View Article : Google Scholar : PubMed/NCBI

108 

McConnell BB, Gregory FJ, Stott FJ, Hara E and Peters G: Induced expression of p16(INK4a) inhibits both CDK4- and CDK2-associated kinase activity by reassortment of cyclin-CDK-inhibitor complexes. Mol Cell Biol. 19:1981–1989. 1999. View Article : Google Scholar : PubMed/NCBI

109 

Satyanarayana A and Rudolph KL: p16 and ARF: Activation of teenage proteins in old age. J Clin Invest. 114:1237–1240. 2004. View Article : Google Scholar : PubMed/NCBI

110 

Macleod KF, Sherry N, Hannon G, Beach D, Tokino T, Kinzler K, Vogelstein B and Jacks T: p53-dependent and independent expression of p21 during cell growth, differentiation, and DNA damage. Genes Dev. 9:935–944. 1995. View Article : Google Scholar : PubMed/NCBI

111 

Mouriaux F, Maurage CA, Labalette P, Sablonnière B, Malecaze F and Darbon JM: Cyclin-dependent kinase inhibitory protein expression in human choroidal melanoma tumors. Invest Ophthalmol Vis Sci. 41:2837–2843. 2000.PubMed/NCBI

112 

Abbas T and Dutta A: p21 in cancer: Intricate networks and multiple activities. Nat Rev Cancer. 9:400–414. 2009. View Article : Google Scholar : PubMed/NCBI

113 

Abukhdeir AM and Park BH: P21 and p27: Roles in carcinogenesis and drug resistance. Expert Rev Mol Med. 10:e192008. View Article : Google Scholar : PubMed/NCBI

114 

Blain SW, Scher HI, Cordon-Cardo C and Koff A: p27 as a target for cancer therapeutics. Cancer Cell. 3:111–115. 2003. View Article : Google Scholar : PubMed/NCBI

115 

Mouriaux F, Casagrande F, Pillaire MJ, Manenti S, Malecaze F and Darbon JM: Differential expression of G1 cyclins and cyclin-dependent kinase inhibitors in normal and transformed melanocytes. Invest Ophthalmol Vis Sci. 39:876–884. 1998.PubMed/NCBI

116 

Narasimha AM, Kaulich M, Shapiro GS, Choi YJ, Sicinski P and Dowdy SF: Cyclin D activates the Rb tumor suppressor by mono-phosphorylation. eLife. 3:e028722014. View Article : Google Scholar

117 

Rayess H, Wang MB and Srivatsan ES: Cellular senescence and tumor suppressor gene p16. Int J Cancer. 130:1715–1725. 2012. View Article : Google Scholar : PubMed/NCBI

118 

Nagarkatti-Gude N, Wang Y, Ali MJ, Honavar SG, Jager MJ and Chan CC: Genetics of primary intraocular tumors. Ocul Immunol Inflamm. 20:244–254. 2012. View Article : Google Scholar : PubMed/NCBI

119 

Bartek J, Bartkova J and Lukas J: The retinoblastoma protein pathway in cell cycle control and cancer. Exp Cell Res. 237:1–6. 1997. View Article : Google Scholar : PubMed/NCBI

120 

Brantley MA Jr and Harbour JW: Inactivation of retinoblastoma protein in uveal melanoma by phosphorylation of sites in the COOH-terminal region. Cancer Res. 60:4320–4323. 2000.PubMed/NCBI

121 

Pollock PM, Harper UL, Hansen KS, Yudt LM, Stark M, Robbins CM, Moses TY, Hostetter G, Wagner U, Kakareka J, et al: High frequency of BRAF mutations in nevi. Nat Genet. 33:19–20. 2003. View Article : Google Scholar : PubMed/NCBI

122 

Hollern DP, Honeysett J, Cardiff RD and Andrechek ER: The E2F transcription factors regulate tumor development and metastasis in a mouse model of metastatic breast cancer. Mol Cell Biol. 34:3229–3243. 2014. View Article : Google Scholar : PubMed/NCBI

123 

Timmers C, Sharma N, Opavsky R, Maiti B, Wu L, Wu J, Orringer D, Trikha P, Saavedra HI and Leone G: E2f1, E2f2, and E2f3 control E2F target expression and cellular proliferation via a p53-dependent negative feedback loop. Mol Cell Biol. 27:65–78. 2007. View Article : Google Scholar : PubMed/NCBI

124 

Lundberg AS and Weinberg RA: Functional inactivation of the retinoblastoma protein requires sequential modification by at least two distinct cyclin-cdk complexes. Mol Cell Biol. 18:753–761. 1998. View Article : Google Scholar : PubMed/NCBI

125 

Calipel A, Abonnet V, Nicole O, Mascarelli F, Coupland SE, Damato B and Mouriaux F: Status of RASSF1A in uveal melanocytes and melanoma cells. Mol Cancer Res. 9:1187–1198. 2011. View Article : Google Scholar : PubMed/NCBI

126 

Yang ZK, Yang JY, Xu ZZ and Yu WH: DNA Methylation and Uveal Melanoma. Chin Med J (Engl). 131:845–851. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2021
Volume 21 Issue 1

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Katopodis P, Khalifa MS and Anikin V: Molecular characteristics of uveal melanoma and intraocular tumors (Review). Oncol Lett 21: 9, 2021
APA
Katopodis, P., Khalifa, M.S., & Anikin, V. (2021). Molecular characteristics of uveal melanoma and intraocular tumors (Review). Oncology Letters, 21, 9. https://doi.org/10.3892/ol.2020.12270
MLA
Katopodis, P., Khalifa, M. S., Anikin, V."Molecular characteristics of uveal melanoma and intraocular tumors (Review)". Oncology Letters 21.1 (2021): 9.
Chicago
Katopodis, P., Khalifa, M. S., Anikin, V."Molecular characteristics of uveal melanoma and intraocular tumors (Review)". Oncology Letters 21, no. 1 (2021): 9. https://doi.org/10.3892/ol.2020.12270