Next Article in Journal
Receptor-Mediated Mitophagy Rescues Cancer Cells under Hypoxic Conditions
Next Article in Special Issue
Soluble Biomarkers with Prognostic and Predictive Value in Advanced Non-Small Cell Lung Cancer Treated with Immunotherapy
Previous Article in Journal
Leveraging Genomics, Transcriptomics, and Epigenomics to Understand the Biology and Chemoresistance of Ovarian Cancer
Previous Article in Special Issue
Predictors of Invasive Adenocarcinomas among Pure Ground-Glass Nodules Less Than 2 cm in Diameter
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Prognostic Immunohistochemical Markers in Uveal Melanoma-Literature Review

by
Malgorzata Gajdzis
1,
Radoslaw Kaczmarek
1,* and
Pawel Gajdzis
2
1
Department of Ophthalmology, Wroclaw Medical University, 50-556 Wroclaw, Poland
2
Department of Clinical and Experimental Pathology, Wroclaw Medical University, 50-556 Wroclaw, Poland
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(16), 4031; https://doi.org/10.3390/cancers13164031
Submission received: 13 July 2021 / Revised: 8 August 2021 / Accepted: 9 August 2021 / Published: 10 August 2021
(This article belongs to the Special Issue Bio-Pathological Markers in the Diagnosis and Therapy of Cancer)

Abstract

:

Simple Summary

The following study provides an overview of the English-language literature on the search for new prognostic factors for uveal melanoma. Uveal melanoma is the most common primary intraocular tumor in adults, and although it is a relatively rare disease, it poses a significant health problem. About half of the patients develop distant metastases, and unfortunately there are currently no effective treatments for the disease at such an advanced stage. The search for new prognostic factors is important to understand the biology of the disease and to be able to monitor patients more effectively. At the same time, it creates an opportunity to find new therapeutic targets. We focused our attention on immunohistochemical research because it is a widely used method, relatively cheap, technically simple, and gives reproducible results. The analysis of this study will enable other researchers to verify their research plans and may also be a source of inspiration for creating new ones.

Abstract

Uveal melanoma is the most common primary intraocular neoplasm in adults. As there are currently no effective methods of treating the disease in the metastatic stage, there is a need to search for new prognostic factors that would enable a reliable assessment of the patient’s condition and constitute a possible therapeutic target. In this review, we have prepared the results of English-language studies on new prognostic factors determined with immunohistochemical methods. We found 64 articles in which the expression of various proteins was associated in a statistically significant manner with the histopathological and clinical prognostic factors identified by AJCC. The results of our work clearly show that the biology of uveal melanoma is extraordinarily complex. Numerous studies have shed new light on the complexity of the processes involved in the development of this cancer. Moreover, a careful analysis of the expression of individual proteins may allow the identification of homogeneous groups of patients requiring different treatment regimens.

1. Introduction

Uveal melanoma (UM) is the most common primary intraocular malignancy in adults and constitutes a significant problem in ophthalmic oncology. The incidence remains stable over many years of follow-up, at approximately 5.1 cases per million per year [1]. The most important risk factors are ethnicity, age, fair skin, light eye color, tendency to sunburn, ocular melanocytosis, iris or choroidal nevus, and BRCA1-associated protein 1 mutation [1].
The uvea forms the middle layer of the eye, bordering on the inside with the retina and on the outside with the sclera. It consists of the iris, the ciliary body, and the choroid [2]. The uvea is a richly perfused structure consisting of blood vessels and the stroma surrounding them. Within the stroma there are melanocytes from the neural crest, from which melanomas arise [3]. Choroidal melanomas account for 85–90% of all UM. Much less often the disease develops in the ciliary body (5–8%) and the iris (3–5%) [4].
There is no basement membrane within the uvea, so UM is in direct contact with the blood vessels, making it much easier for the disease to spread [2]. Metastases to the regional lymph nodes (parotid, submandibular, or cervical) appear late and only when the outer parts of the sclera and conjunctiva are affected. The vast majority of distant metastases occur in the liver (60–90%), followed by the lungs, subcutaneous tissue, bones, and brain [5]. The COMS study showed that 93% of patients have liver metastases at death, and in cases with only one site of metastasis, the liver is involved in 95% [6].
Five-year survival rates in UM range from 25% to 97% (mean 69%), depending on the clinical stage of the disease (staging) [7]. The prognosis worsens significantly in the presence of distant metastases—then the survival rates are as low as 8% after two years [5]. The average survival time from detection of the presence of metastases is 4–15 months [8]. Over the last thirty years of observation, the survival times have not changed significantly. Many treatments modalities have been evaluated for effectiveness in metastatic disease; unfortunately, none of them have resulted in an increase in survival [9]. A meta-analysis of 29 studies conducted between 1988 and 2015 showed that the average progression-free survival and overall survival have not changed over the years, regardless of the therapies used [3].
Currently, the treatment of UM is focused on the primary tumor. Radiotherapy is the most commonly used; less often, tumor resection, or in the case of advanced disease, removal of the eyeball (enucleation) [3]. In small lesions, transpupillary thermotherapy or photocoagulation can be used, but their usefulness is limited due to numerous side effects [10]. There are also attempts to administer drugs by injection into the vitreous chamber [3]. Topical treatment is highly effective. Unfortunately, half of the patients develop metastases, often many years after the primary tumor has been treated [11]. This inevitably leads to death as there is no effective treatment for the systemic disease.
The search for new reliable prognostic factors is important as it allows the identification of groups of patients more prone to metastasis. It acquires additional significance in the context of the lack of clear guidelines regarding the intervals between control tests and the type of tests that should be performed during the control. Assigning patients to appropriate groups allows for more frequent monitoring of patients from higher risk groups. Detecting distant metastases at an early stage makes it possible to remove them surgically, which is currently the only proven way to extend patients’ lives [12]. Therefore, it seems justified to search for new markers, especially in the context of a holistic approach to the patient and adjusting the therapy to individual needs. Moreover, potential prognostic factors may also become a target of therapy.
In recent years, significant progress has been made in the development of molecular methods that make it possible to predict the course of the disease based on the analysis of genetic material. Initially, based on the gene expression profile, UM were divided into two groups—class 1 associated with low risk of metastasis, class 2 with high. However, it quickly turned out that a large proportion of patients classified as class 1 died because of the development of distant metastases. For this reason, class 1 was divided into two subclasses—1A and 1B, based on the expression of the PRAME marker [13]. In class 1A, the risk of developing metastases within 5 years is 2%, while in class 1B it is 21%. A recently published report, part of the Cancer Genome Atlas project, categorized UM into four groups [14]. The results of the report indicate a high prognostic value of genetic abnormalities in UM, but no prospective studies to support the results are yet available. Proponents of molecular methods are of the opinion that genetic testing is sufficient to determine the prognosis and identify groups of patients at increased risk. However, it has been shown that the most accurate prediction can be obtained by combining the TNM classification with genetic tests, in particular the loss of chromosome 3 [15].
The AJCC tumor staging is a recognized, long-established system, thoroughly tested and regularly updated. The latest, 8th edition, describes the basic, recognized prognostic factors on which the TNM classification is based, as well as other, relatively new, independent prognostic factors, including the gene expression profile [7]. AJCC members emphasize that genetic testing, while highly promising, cannot be the primary and only prognostic factor. The main weaknesses are the short follow-up time of patients (on average less than 5 years) and the small number of patients in many T-categories. It should also be remembered that the AJCC anatomical classification will remain the gold standard for patients where genetic testing will not be available.
The AJCC classification takes into account the prognostic factors necessary for the stage grouping (TNM) and additional factors recommended for clinical care—size, location, extraocular extension, cell type, chromosomal analysis, gene expression profiling, mitotic count, extravascular matrix loops and networks, microvascular density, and tumor-infiltrating macrophages and lymphocytes. For the purposes of this study, we have researched the English-language literature available in the PubMed medical database. We searched for articles on protein expression determined by immunohistochemistry (IHC), with statistically significant relationships with the AJCC-listed prognostic factors for uveal melanoma. We decided to focus on IHC, because it is a technique that is widespread all over the world, gives highly reproducible results, and is relatively cost-effective [16]. This makes it one of the most universal tools in cancer diagnosis and prognosis. We focused on new, promising proteins that could also be an important therapeutic target in the future. A summary table in alphabetical order can be found at the end of the article (Table 1).

2. Tumor Size

Largest basal tumor diameter is the predominant clinical predictor of prognosis, strongly associated with risk of metastasis (AJCC Level of Evidence: I) [7]. Studies also show that tumor thickness may be an independent prognostic factor [72]. The most accurate measurements are obtained by analyzing fundus photography and ultrasonography. In the event of enucleation, the measurement may be understated due to tissue shrinkage during material processing.
Ataxia-telangiectasia mutated protein (ATM) is an activator of DNA damage response. It is predominantly nuclear protein. Loss of expression correlates with both large tumor diameter and tumor thickness [21]. With tumor thickness correlate high expression of c-REL and CCR7 [26,35]. c-REL is a member of the nuclear factor κB (NF-κB) transcription factor family and an emerging regulator of tumorigenesis. CCR7 (C-C Motif Chemokine Receptor 7) is a receptor mainly expressed in lymphoid cells. It mediates cell migration of naïve lymphocytes and mature dendritic cells to secondary lymphoid organs and regulates the transport of cancer cells through the extracellular matrix.
In the literature, we found several proteins for which the relationship of expression with the largest tumor diameter has been demonstrated. nm23, a product of the metastasis suppressor gene (NM23), is expressed in cytoplasm. Low expression correlates with the largest tumor diameter [57]. In other cases, tumor diameter is related to high protein expression. They are Hsp90, PARP, and PD-1 [46,60,63]. Heat shock protein 90 (Hsp90) plays a role in folding, intracellular transport, maintenance, and degradation of proteins, and facilitates cell signaling. Poly (ADP-ribose) polymerase (PARP), a nuclear protein, participates in the DNA repair processes. Programmed cell death receptor-1 (PD-1) is a membrane-bound receptor and plays a role in regulation immune system’s response to the cells of the human body.
High expression of cluster of differentiation 147 (CD147), a member of the immunoglobulin superfamily, which plays a role in intercellular recognition, various immunologic phenomena, differentiation, and development, also correlates with larger tumor diameter, but only in the nonmetastatic sub-group [28].
Another relationship occurs in the case of EphA1, the erythropoietin-producing human hepatocellular receptor A1, where high expression is associated with a smaller tumor diameter [43]. Thus, high expression is a favorable prognostic factor in this case. EphA1 is an RTK receptor and plays a role in the regulation of a processes critical to embryonic development, including axon guidance, formation of tissue boundaries, cell migration, segmentation, proliferation, and angiogenesis.
Proteins whose expression is related to tumor size are listed in Table 2.

3. Tumor Location

Involvement of the ciliary body in the disease process is associated with the least favorable prognosis. It is independently associated with the risk of metastasis (AJCC Level of Evidence: I) [72]. Due to its location, the ciliary body is poorly accessible in the examination. For this reason, possible neoplastic changes may develop long before they are detected. It is nearly impossible to visualize the ciliary body directly. Its condition can be most accurately assessed in anterior segment ultrasound imaging. Other options available are gonioscopy, transillumination, and optical coherence tomography of the anterior segment of the eye [7].
In the literature, we found four proteins whose high expression is associated with the involvement of the ciliary body. These include the previously mentioned PD-1 and CD147, but in the case of CD147, the relationship relates only to the metastatic sub-group [28,62]. The other two are HERC2 (HECT and RLD Domain-Containing E3 Ubiquitin Protein Ligase 2), a predominantly nuclear and cytoplasm protein, which plays a role in DNA repair regulation, pigmentation, and neurological disorders, and P-protein (pink-eyed dilution protein), which plays a role in melanin synthesis in melanocytes and retinal pigment epithelium [45].
Proteins whose expression is related to ciliary body involvement are listed in Table 3.

4. Extraocular Extension

The prevalence of extrascleral infiltration is an unfavorable prognostic factor (AJCC Level of Evidence: II). If it occurs at the front part of the eyeball, it can be assessed by a basic ophthalmological examination. Posterior infiltration can be assessed by imaging studies—ultrasound, computed tomography, or magnetic resonance imaging [7]. In the case of removal of the eyeball, histopathological examination can assess scleral invasion, which is an event immediately preceding the formation of extrascleral infiltration [72].
Low expression of the above-mentioned nm23 is associated with the presence of a deeper scleral invasion [57]. On the other hand, in the case of BNIP3 (BCL2 19 kD protein-interacting protein 3), protein, which regulates cell death, autophagy, and cytoprotection, high expression in the cytoplasm is associated with a deeper scleral invasion [24].
High EphA1 expression is a favorable prognostic factor as it is associated with a lower incidence of exstrascleral infiltration [43]. In contrast, in the case of Cripto-1 (teratocarcinoma-derived growth factor-1) and Cyclin1, high expression increases the risk of the presence of extrascleral infiltration. Cripto-1 is an oncogenic growth factor involving tumorigenesis and cancer cell proliferation and survival [36]. Cyclin1 is a predominantly nuclear protein and regulator of the cell cycle. Its relationship with the formation of extrascleral infiltrates has been demonstrated in two studies [38,39]. Finally, low expression of adiponectin correlates with extrascleral infiltration. Adiponectin is a protein hormone involved in regulating glucose levels, fatty acid breakdown, and plays a role in limiting cell proliferation and reducing inflammation [19].
Proteins whose expression is related to scleral and extrascleral infiltration are listed in Table 4.

5. Cell Type

The type of cells present in the tumor tissue is an independent prognostic factor associated with the risk of metastasis (AJCC Level of Evidence: I) [7]. There are two types of cells in UM—spindle and epithelioid cells. Both types of cells can appear within the tumor, and the presence of epithelioid cells is associated with a worse prognosis. There is no consensus on the proportions of individual cells required to qualify a given tumor as mixed or epithelioid type. Many pathologists do not try to classify individual types, and instead only determine the presence of epithelioid cells [7].
The presence of epithelioid cells is associated with high expression of the aforementioned c-REL, CyclinD1, HERC2, PD-1, and P-protein [35,39,45,62]. In the case of PARP, high expression correlates with a higher histopathological grade [60]. In turn, low expression of ATM correlate with the presence of epithelioid cells [21]. Low expression of p16 (cyclin-dependent kinase inhibitor 2A, CDKN2A, multiple tumor suppressor 1), an inhibitor of cyclin-dependent kinases responsible for slowing the progression of the cell cycle from G1 phase to the S phase, is associated with mixed cell type [59].
With the presence of epithelioid cells, high expression of LOX (lysyl oxidase), an extracellular enzyme, also correlates, playing a role in embryonic development, wound healing, and adult tissue remodeling [50]. Nestin (neural stem cell protein), a cytoplasmic and membrane-bound protein, a member of the intermediate filament (IF) class VI protein family, correlates with the presence of epithelioid cells too [56]. Moreover, high CEACAM expression (carcinoembryonic antigen cell adhesion molecule-1), a transmembrane glycoprotein, playing a role in the intercellular interactions, regulation of cell growth, angiogenesis, immune modulation, and hepatic insulin clearance, is associated with the presence of epithelioid cells, as is the case with CXCR4 (C-X-C motif chemokine receptor 4), an alpha-chemokine receptor specific for stromal-derived-factor-1, a molecule endowed with potent chemotactic activity for lymphocytes [29,37].
In contrast, in the case of NEMO/IKKγ (nuclear factor κB essential modulator, inhibitor of nuclear factor kappa B kinase subunit gamma), a protein essential for the activation of transcription factor NFκB, which regulates the cellular responses to inflammation, immunity and cell survival, low expression correlates with presence of epithelioid cells [55].
Proteins whose expression is related to presence of various cell type are listed in Table 5.

6. Chromosomal Analysis

Another independent prognostic factor associated with the risk of metastasis is the loss of chromosome 3, especially with the frequently coexisting gain in chromosome 8q (AJCC Level of Evidence: II). The combination of both, monosomy 3 and chromosome 8q gain, are associated with the worst prognosis. Chromosome status is typically determined by karyotyping or fluorescent in situ hybridization [7].
We found only five studies in the literature in which the expression of specific proteins was related to the status of chromosomes. High EphA5 expression seems to be a favorable prognostic factor as it correlates with less frequent loss of chromosome 3 [43]. In turn, high PARP expression is associated with more frequent loss of chromosome 3 [60]. The worst prognosis induces high nestin expression, as it is associated with more frequent loss of chromosome 3 and chromosome 8q gain [56].
The next study we found is about PERP protein (p53 apoptosis effector related to PMP-22), playing a role in inducing cell death. In his case, low expression is associated with more frequent loss of chromosome 3 [64]. An analogous situation occurs in the case of adiponectin, where low expression also correlates with more frequent loss of chromosome 3 [19].
Proteins whose expression is related to chromosome status are listed in Table 6.

7. Mitotic Count

Mitotic count is independently associated with metastatic risk (AJCC Level of Evidence: II) [7]. Higher counts are associated with shorter survival. Mitotic count is assessed with a light microscope, counting at least 40 high-power fields [73]. Alternatively, mitotic cells can be assessed using immunohistochemical methods.
High expression of nestin correlates with higher mitotic count [56]. The same relationship occurs in the case of EGFR (epidermal growth factor receptor), a transmembrane protein that plays a role in epithelial tissue development and homeostasis [40]. High expression of EphA1 correlates with a lower mitotic count, while low expression of NEMO/IKKγ correlates with a higher mitotic count [43,55].
Proteins whose expression is related to mitotic count are listed in Table 7.

8. Extravascular Matrix Loops and Networks

Two types of extravascular matrix pattern have been documented that are associated with a higher risk of metastasis (AJCC Level from Evidence: II) [13]. These are networks and loops formed by the tumor vessels. The presence of patterns is determined under a light microscope [7].
High CEACAM expression is associated with the presence of the networks [29]. In turn, high expression of MMP-9 and VEGF-A is associated with the presence of both loops and networks [52]. MMP-9 (matrix metalloproteinase-9) is secreted along with membrane-associated neutral endopeptidase, and plays a role in degrading extracellular matrix proteins, cell proliferation, migration, differentiation, angiogenesis, apoptosis, and host defense. VEGF-A (vascular endothelial growth factor-A) acts specifically on endothelial cells, mediates increased vascular permeability, induces angiogenesis, vasculogenesis, and endothelial cell growth, promotes cell migration, and inhibits apoptosis.
Low expression of NEMO/IKKγ and high expression of nestin correlates with the presence of vascular loops [55,56].
Proteins whose expression is related to extravascular matrix loops and networks are listed in Table 8.

9. Tumor-Infiltrating Macrophages and Lymphocytes

Tumor-infiltrating macrophages (TIMs) and tumor-infiltrating lymphocytes (TILs) are other important prognostic factors in uveal melanoma. Generally, an inflammatory infiltrate is associated with a worse prognosis [13]. The AJCC lists the presence of TIMs as being independently associated with the risk of metastasis (AJCC level of Evidence: II) [7].
High expression of the three proteins is associated with a more intense infiltration of TIMs. They are PD-1, PD-L1, and EMAP-II [42,62]. EMAP-II (endothelial monocyte-activating polypeptide II) is a proinflammatory cytokine and chemoattractant of macrophages, expressed on the cell surface. In the case of TILs, a relationship was also demonstrated between the intensity of infiltration and high expression of the three proteins. They are CCR7, MMP-9, and VEGF-A [26,52].
Proteins whose expression is related to TIMs, and TILs infiltration are listed in Table 9.

10. Other Prognostic Factors

In most studies on prognostic factors, the authors try to demonstrate the relationship of the studied proteins with as many parameters as possible. Some of them do not currently have a confirmed prognostic value, but they are certainly worth paying attention to, as they constitute a significant contribution to expanding knowledge about the biology of UM. In this section, we list proteins whose expression is associated with these parameters. We also included some research related to the AJCC staging group.
High expression of BNIP3, HERC2, and P-protein is associated with increased pigmentation within the tumor [24,45]. The amount of the pigment probably does not influence the prognosis in melanoma in any way, but it has some diagnostic significance as it hinders the immunohistochemical assessment of the examined tissues. High P-protein expression is also associated with more advanced clinical tumor staging [45]. A similar relationship occurs in the case of PD-L1 and CD147, with the latter only in the nonmetastatic subgroup [28,62]. The inverse relationship is observed in the case of PLK-1 (polo-like kinase-1), a regulator of mitotic entry and cytokines. Its low expression correlates with the higher AJCC prognostic stage group [66].
Another parameter is the severity of necrosis in the tumor tissues. Necrosis usually occurs in advanced tumors and is therefore associated with poor prognosis [74]. In UM, the severity of necrosis was associated with high CCR7, MMP-9, and VEGF-A expression [26,52].
High expression of EphA1 and EphA5 is associated with more frequent hemorrhages in the vitreous chamber in patients with UM [43]. This dependence probably results from the role played by Eph receptors in the angiogenesis process. In turn, low NEMO/IKKγ expression is associated with the presence of neovascularization in the tumor [55]. The latest relationships we found are high expression CyclinD1 related to the tumor cell MIB-1 positivity and high PD-1 expression related to the absence of BAP-1 staining [39,62].
Table 10 summarizes the relationships discussed above.

11. Metastasis

As mentioned earlier, the development of metastases significantly worsens the prognosis for patients with UM. Because metastases can appear up to 30 years after primary tumor treatment, patients must be closely monitored for the rest of their lives [7]. There are no strict guidelines for the management of patients after UM is detected. As agreed by COMS, a physical examination and liver function test are not sufficient to detect possible metastases. Usually, imaging tests (ultrasound, MR imaging, or CT) are performed annually, but the procedures may differ in individual centers [75]. Since only a quick detection of metastases in the liver followed by their surgical removal may slightly extend the survival time of patients, it is important to search for new prognostic factors that would allow to select groups of patients at higher risk. Therefore, most of the studied proteins are tested for dependence on the occurrence of metastasis.
In most of the studied cases, high expression of a given protein is associated with more frequent metastases. Of the proteins mentioned earlier, this is the case with CCR7, c-REL, Cripto-1, CyclinD1, MMP-2, MMP-9, PD-1, PD-L1, P-protein, and VEGF-A [27,28,35,36,39,45,52,62]. High expression of many consecutive proteins is also associated with more frequent occurrence of metastases. ABCB5 (ATP-binding cassette sub-family B member 5), human transmembrane P-glycoprotein, plays a role in transmembrane transport (including chemotherapeutic drugs), which makes it particularly important in the context of drug resistance [17]. ADAM10 (a disintegrin and metalloproteinase domain-containing protein 10), a transmembrane protein, controls membrane fusion and cell-cell and cell-matrix interactions [18]. c-Met (Tyrosine-protein kinase Met or hepatocyte growth factor receptor (HGFR)), a transmembrane RTK receptor, plays a role in embryonic development, organogenesis and wound healing, angiogenesis, and metastasis formation [30]. C-NFκB proteins (canonical nuclear factor-κB proteins (p65 and p50)), coordinate innate immunity and inflammation processes, particularly important in neoplastic transformation [32]. Other proteins associated with the presence of metastasis are COX-2 (Cyclooxygenase-2), an enzyme which catalyzes the prostanoid synthesis reaction [34], and p53, a cell-cycle regulatory protein [39]. PCNA (proliferating cell nuclear antigen, ATLD2), the DNA polymerase auxiliary protein involved in the control of DNA replication, is also associated with the presence of metastases [61]. Similar relationships are observed in the case of phospho-Akt, a cytoplasmic protein that plays a role in phosphorylation and inactivation of several proteins involved in apoptosis [65], and PRDX3 (thioredoxin-dependent peroxidase reductase), a cytoplasmic protein involved in redox regulation of the cell and protects radical-sensitive enzymes from oxidative damage [67]. Finally, SPANX-C (SPANX family member C; Sperm protein associated with the nucleus on the X chromosome C), a cytoplasmic protein, is expressed in highly metastatic cell lines [69].
For a few proteins, high expression is associated with a higher likelihood of death from metastasis. These are the previously described EGFR and COX-2, and IGF-1R and MCAM [33,41]. IGF-1R (insulin-like growth factor 1 receptor), a transmembrane receptor implicated in insulin signaling, plays a role in several cancer development [48]. MCAM (melanoma cell adhesion molecule, MUC18, Mel-Cam, CD146), an adhesion molecule, plays a role in intracellular signaling cascades [51]. In turn, high expression of LOX and NC-NFκB proteins is associated with reduced metastasis-free survival time [50,54]. NC-NFκB proteins (p52, RelB, and co-expression of p52/RelB) are nuclear proteins; they play a role in promoting cancer proliferation and progression. In the case of syntenin (syndecan binding protein syntenin-1, melanoma differentiation-associated gene 9, mda-9), a predominantly cytoplasm protein playing a role in clustering of membrane receptors, intracellular trafficking, Sox4 activation, and signal transduction, high expression correlates with the risk of recurrence of metastasis [71].
Low adiponectin expression is associated with the presence of metastases [19]. A similar relationship occurs in the case of RKIP (Raf kinase inhibitor protein), a regulator of proliferative pathways within the cell [68]. The presence of metastases is also associated with the loss of ICAM expression (intercellular cell adhesion molecule-1), an adhesion molecule and ligand for leukocyte function-associated antigen-1, involved in the process of inflammation, the circulation of blood cells, and in the immune surveillance of the host [47].
High expression of the aforementioned EphA5 is associated with less frequent occurrence of metastases [43]. A similar situation occurs in the case of beclin, an autophagy related protein, performing a central role in the autophagic process as a major member of the macro-autophagic phase [23].
Table 11 summarizes the relationships discussed above in alphabetical order.

12. Survival Times

From the point of view of oncology, establishing survival times is of fundamental importance. It is an extremely clear picture of the prognosis for the patient. For this reason, many studies have focused on linking the expression of various proteins with survival times, even though such studies require a long time of observation.
In UM, most of the proteins tested show a reduction in survival times with high expression. Such relationships have been demonstrated for BNIP3, CCR7, c-Met, C-NFκB proteins, c-REL, MMP-2, and MMP-9, Nestin, PARP, PD-1, and PRDX3 [24,26,28,30,32,35,56,60,63,67]. Proteins whose high expression also is associated with a reduction in survival times are also AIF, JARID1B, legumain, Nbs1, and NC-NFκB proteins.
AIF (apoptosis inducing factor) is ubiquitous protein and plays a proapoptotic function in the nucleus and redox activity in mitochondria [20]. JARID1B (Jumonji AT-rich interactive domain 1B), a demethylase enzyme, induces demethylation of tri- and di-methylated lysines in the 4 position of histone 3 [49]. Legumain (asparagine endopeptidase (AEP)), a proteolytic enzyme, plays a role in the functioning of the immune system [76]. Nbs1 (Nibrin, NBN), an intracellular protein, plays a role in the repair of double strand breaks and telomere maintenance [53]. NC-NFκB proteins (p52, RelB, and co-expression of p52/RelB), which are nuclear proteins, plays a role in promoting cancer proliferation and progression [54].
At the opposite site, there are relationships where high expression in immunohistochemical studies is associated with longer survival. Among the previously mentioned proteins, this is the case of Beclin and EphA5 [23,43]. Other factors are BTNL9, HER3, nm23-H1, SSR, TIMP-1, and TIMP-2.
BTNL9 (butyrophilin-like protein 9) is a modulator of the T cell response [25]. HER3 (human epidermal growth factor receptor 3 or receptor tyrosine-protein kinase erbB-3), a transmembrane RTK receptor, is implicated in growth, proliferation, chemotherapeutic resistance, and the promotion of invasion and metastasis [44]. nm23-H1 is a cytoplasm protein, the product of a metastasis suppressor gene (NM23) [58]. SSR (somatostatin receptor) through the binding of somatostatin starts a signaling pathway that leads to arrest of cell growth or apoptosis [70]. TIMP-1 and TIMP-2 (tissue inhibitor of metalloproteinase-1 and -2) are metalloproteinase inhibitors [28].
Moreover, low or loss of expression may be associated with a reduction in survival time. Such relationships have been demonstrated for ATM, NEMO/IKKγ, and PLK-1 [18,39,45]. In the literature, we also found articles on the relationship of high c-Met and IGF-1R expression to melanoma-specific mortality [30,40].
Table 12 summarizes the relationships discussed above in alphabetical order.

13. Therapeutic Perspectives

The described protein markers of UM are not only a diagnostic tool but may also be a potential target in anti-cancer therapy, especially in the case of advanced UM. As there is currently no effective pharmacological treatment for such patients, they may be crucial for favorable long-term survival. Some of them have already been tried to be used for therapy, and others are still waiting for their chance. One of the better described in this regard are immune checkpoint inhibitors (ICIs). Their introduction started a new era in the treatment of cancer. ICIs restore the immune system’s ability to perform a standard, cytotoxic response against cancer cells. Antibodies targeting CTLA-4 and PD-1 in the treatment of cutaneous melanoma (CM) significantly improved treatment outcomes in metastatic CM. Unfortunately, these results were not confirmed in clinical trials of UM patients. Studies with ipilimumab, tremelimumab, nivolumab, and pembrolizumab were somewhat disappointing [77,78,79,80,81,82,83]. Slightly better results were achieved in combination therapy in which drugs against both types of ICIs receptors were used: CTLA-4 and PD-1, but still without a satisfactory breakthrough [84,85].
Another group of biomarkers targeted for cancer therapy is RTKs: IGFR, EGFR VEGFR, c-Met, and its ligands. In recent years, numerous studies have been published on the effectiveness of their use in treating metastatic UM, with both monoclonal antibodies or small molecule drugs targeting the extracellular domains of RTKs or RTK ligands. These studies, however, often concerned unselected, small groups of patients. For example, in the case of IGFR, a phase I trial with a monoclonal antibody against IGF-1R in patients with relapsed multiple myelomas did not provide satisfactory results [86]. However, more recent in vitro reports of ICF-1R blocking with pristimerin are more promising [87].
In the EGFR inhibitor, gefitinib, a decrease in UM cells survival was found in in vivo studies [88]. IGFR inhibition with gefitinib in the phase II study showed only low clinical efficacy in the group of unselected patients [89]. Intraocular VEGF inhibitors have revolutionized the treatment of the wet form of AMD, but their pedigree comes from oncological therapy, where they have so far been found in the treatment of numerous solid tumors. They were also tested for use in UM. However, a retrospective analysis of metastatic UMs treated with one of the anti-VEGF antibodies, bevacizumab, compared to the group not receiving this antibody, showed no statistical difference [90]. The other axitinib and pazopanib have also failed to prove their effectiveness in the treatment of melanoma [91,92].
For c-Met or its HGF ligand, the preclinical study has shown promising results, but so far only partially confirmed in clinical trials [93,94,95]. One such inhibitor, crizotinib, was shown to be potent in preventing metastatic UM development [94]. Another kinase inhibitor, cabozantinib, showed potential clinical benefit in randomized discontinuation studies including 23 patients with metastatic UM [96]. However, these have not been confirmed in phase II randomized trials comparing the benefits of cabozantinib versus chemotherapy [97]. Another selective c-Met inhibitor, tivantinib, also showed promising results in a phase I study in patients with advanced solid tumors, including 19 with melanoma patients; Another phase I study was carried out with tepotinib in 149 patients with advanced solid tumors [98,99].
Despite many promising research results, there is still no standard of care therapy for metastatic UM with satisfactory effectiveness. More research is needed to develop new substances, better select patients for whom known treatments may be particularly effective and develop combination therapies that combine the advantages of various anti-cancer effects. It is worth mentioning tebentafusp, a bispecific fusion protein designed to target gp100 (a melanoma-associated antigen), which redirects T cells to kill tumor cells [100]. gp100 is expressed strongly in melanomas and weakly in normal melanocytes [100]. Initial clinical trial results are highly promising, even though it is restricted for patients with a particular HLA allele so far. Moreover, the technology used in the production of tebentafusp could allow any other protein to be used as a target, potentially opening the way for the production of further drugs [101].

14. Conclusions

UM is a relatively rare but deadly cancer. Researchers around the world continue their efforts to better understand its biology, which should translate into more effective treatment, especially of metastatic disease. The IHC on which we focused this article remains the gold standard in tissue testing. This is due to its wide availability and proven methodology. However, there are other new research methods that are also contributing to advances in the treatment of UM.
It is worth looking at the research on long non-coding RNA, which can also serve as a potential prognostic factor [102]. The use of Multi-Platform OMICS Analysis also seems prospective [103]. Single-cell analysis, single-cell RNA sequencing, and molecular profiling are also highly promising and interesting methods [104,105,106]. The development of science and wider access to new technologies enable a better understanding of the mechanisms leading to the development of all diseases, especially cancer. However, do not forget about older, proven methods such as IHC, as usually all types of tests complement each other perfectly, thus guaranteeing the best results.

Author Contributions

Conceptualization, M.G., R.K. and P.G.; methodology, M.G., R.K. and P.G.; writing—original draft preparation, M.G. and R.K.; writing—review and editing, R.K. and P.G.; visualization, M.G.; supervision, P.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kaliki, S.; Shields, C.L. Uveal melanoma: Relatively rare but deadly cancer. Eye 2017, 31, 241–257. [Google Scholar] [CrossRef] [Green Version]
  2. Nickla, D.L.; Wallman, J. The multifunctional choroid. Prog. Retin. Eye Res. 2010, 29, 144–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Yang, J.; Manson, D.K.; Marr, B.P.; Carvajal, R.D. Treatment of uveal melanoma: Where are we now? Ther. Adv. Med. Oncol. 2018. [Google Scholar] [CrossRef]
  4. Wu, M.Y.; Lai, T.T.; Liao, W.T.; Li, C.J. Clinicopathological and prognostic significance and molecular mechanisms governing uveal melanoma. Ther. Adv. Med. Oncol. 2020, 12. [Google Scholar] [CrossRef]
  5. Krantz, B.A.; Dave, N.; Komatsubara, K.M.; Marr, B.P.; Carvajal, R.D. Uveal melanoma: Epidemiology, etiology, and treatment of primary disease. Clin. Ophthalmol. 2017, 11, 279–289. [Google Scholar] [CrossRef] [Green Version]
  6. Collaborative Ocular Melanoma Study Group. Assessment of metastatic disease status at death in 435 patients with large choroidal melanoma in the Collaborative Ocular Melanoma Study (COMS): COMS report no. 15. Arch. Ophthalmol. 2001, 119, 670–676. [Google Scholar] [CrossRef] [PubMed]
  7. Kivela, T.; Simpson, E.R.; Grossniklaus, H.E.; Jager, M.J.; Singh, A.D.; Caminal, J.M.; Pavlick, A.C.; Kujala, E.; Coupland, S.E.; Finger, P.T. Uveal Melanoma. In AJCC Cancer Staging Manual, 8th ed.; Amin, M.B., Edge, S., Greene, F., Byrd, D.R., Brookland, R.K., Washington, M.K., Gershenwald, J.E., Compton, C.C., Hess, K.R., Sullivan, D.C., et al., Eds.; Springer Publishing Company: New York, NY, USA, 2017; pp. 805–817. [Google Scholar]
  8. Carvajal, R.D.; Schwartz, G.K.; Tezel, T.; Marr, B.; Francis, J.H.; Nathan, P.D. Metastatic disease from uveal melanoma: Treatment options and future prospects. Br. J. Ophthalmol. 2017, 101, 38–44. [Google Scholar] [CrossRef] [Green Version]
  9. Blum, E.S.; Yang, J.; Komatsubara, K.M.; Carvajal, R.D. Clinical Management of Uveal and Conjunctival Melanoma. Oncology 2016, 30, 29–32, 34–43. [Google Scholar] [PubMed]
  10. Mashayekhi, A.; Shields, C.L.; Rishi, P.; Atalay, H.; Pellegrini, M.; McLaughlin, J.P.; Patrick, K.A.; Morton, S.J.; Remmer, M.H.; Parendo, A.; et al. Primary transpupillary thermotherapy for choroidal melanoma in 391 cases: Importance of risk factors in tumor control. Ophthalmology 2015, 122, 600–609. [Google Scholar] [CrossRef]
  11. Kujala, E.; Mäkitie, T.; Kivelä, T. Very long-term prognosis of patients with malignant uveal melanoma. Investig. Ophthalmol. Vis. Sci. 2003, 44, 4651–4659. [Google Scholar] [CrossRef] [Green Version]
  12. Agarwala, S.S.; Eggermont, A.M.M.; O’Day, S.; Zager, J.S. Metastatic melanoma to the liver: A contemporary and comprehensive review of surgical, systemic, and regional therapeutic options. Cancer 2014, 120, 781–789. [Google Scholar] [CrossRef]
  13. Berus, T.; Halon, A.; Markiewicz, A.; Orlowska-Heitzman, J.; Romanowska-Dixon, B.; Donizy, P. Clinical, Histopathological and Cytogenetic Prognosticators in Uveal Melanoma—A Comprehensive Review. Anticancer Res. 2017, 37, 6541–6549. [Google Scholar] [CrossRef]
  14. Robertson, A.G.; Shih, J.; Yau, C.; Gibb, E.A.; Oba, J.; Mungall, K.L.; Hess, J.M.; Uzunangelov, V.; Walter, V.; Danilova, L.; et al. Integrative Analysis Identifies Four Molecular and Clinical Subsets in Uveal Melanoma. Cancer Cell 2017, 32, 204–220.e15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Dogrusöz, M.; Bagger, M.; van Duinen, S.G. The Prognostic Value of AJCC Staging in Uveal Melanoma Is Enhanced by Adding Chromosome 3 and 8q Status. Investig. Opthalmol. Vis. Sci. 2017, 58, 833–842. [Google Scholar] [CrossRef] [Green Version]
  16. Sukswai, N.; Khoury, J.D. Immunohistochemistry Innovations for Diagnosis and Tissue-Based Biomarker Detection. Curr. Hematol. Malig. Rep. 2019, 14, 368–375. [Google Scholar] [CrossRef] [PubMed]
  17. Broggi, G.; Musumeci, G.; Puzzo, L.; Russo, A.; Reibaldi, M.; Ragusa, M.; Longo, A.; Caltabiano, R. Immunohistochemical Expression of ABCB5 as a Potential Prognostic Factor in Uveal Melanoma. Appl. Sci. 2019, 9, 1316. [Google Scholar] [CrossRef] [Green Version]
  18. Caltabiano, R.; Puzzo, L.; Barresi, V.; Ieni, A.; Loreto, C.; Musumeci, G.; Castrogiovanni, P.; Ragusa, M.; Foti, P.; Russo, A.; et al. ADAM 10 expression in primary uveal melanoma as prognostic factor for risk of metastasis. Pathol. Res. Pract. 2016, 212, 980–987. [Google Scholar] [CrossRef]
  19. Tura, A.; Thieme, C.; Brosig, A.; Merz, H.; Ranjbar, M.; Vardanyan, S.; Zuo, H.; Maassen, T.; Kakkassery, V.; Grisanti, S. Lower levels of adiponectin and its receptor adipor1 in the uveal melanomas with monosomy-3. Investig. Ophthalmol. Vis. Sci. 2020, 61. [Google Scholar] [CrossRef]
  20. Krasnik, V.; Furdova, A.; Svetlosakova, Z.; Kobzova, D.; Gergisakova, H.; Feketeova, L.; Svetlosak, M.; Barta, A.; Babal, P. Prognostic value of apoptosis inducing factor in uveal melanoma. Neoplasma 2017, 64, 262–268. [Google Scholar] [CrossRef]
  21. Jha, J.; Singh, M.K.; Singh, L.; Pushker, N.; Bajaj, N.; Sen, S.; Kashyap, S. Prognostic relevance of ATM protein in uveal melanoma and its association with clinicopathological factors. Int. J. Clin. Oncol. 2019, 24, 1526–1535. [Google Scholar] [CrossRef]
  22. Jha, J.; Singh, M.K.; Singh, L.; Pushker, N.; Bajaj, M.S.; Seema Sen, S.; Kashyap, S. Expression of BAP1 and ATM proteins: Association with AJCC tumor category in uveal melanoma. Ann. Diagn. Pathol. 2020, 44, 151432. [Google Scholar] [CrossRef]
  23. Broggi, G.; Ieni, A.; Russo, D.; Varricchio, S.; Puzzo, L.; Russo, A.; Reibaldi, M.; Longo, A.; Tuccari, G.; Staibano, S.; et al. The Macro-Autophagy-Related Protein Beclin-1 Immunohistochemical Expression Correlates with Tumor Cell Type and Clinical Behavior of Uveal Melanoma. Front. Oncol. 2020, 10, 2515. [Google Scholar] [CrossRef] [PubMed]
  24. Jiang, Z.; Yu, F.; Li, M. Upregulation of BCL2 19 kD protein-interacting protein 3 (BNIP3) is predictive of unfavorable prognosis in uveal melanoma. Med. Sci. Monit. 2018, 24, 4711–4717. [Google Scholar] [CrossRef]
  25. Jiang, Z.; Liu, F. Butyrophilin-like 9 (BTNL9) suppresses invasion and correlates with favorable prognosis of uveal melanoma. Med. Sci. Monit. 2019, 25, 3190–3198. [Google Scholar] [CrossRef]
  26. van den Bosch, T.; Koopmans, A.E.; Vaarwater, J.; van den Berg, M.; de Klein, A.; Verdijk, R.M. Chemokine receptor CCR7 expression predicts poor outcome in uveal melanoma and relates to liver metastasis whereas expression of CXCR4 is not of clinical relevance. Investig. Ophthalmol. Vis. Sci. 2013, 54, 7354–7361. [Google Scholar] [CrossRef] [Green Version]
  27. Dobner, B.C.; Riechardt, A.I.; Joussen, A.M.; Englert, S.; Bechrakis, N.E. Expression of haematogenous and lymphogenous chemokine receptors and their ligands on uveal melanoma in association with liver metastasis. Acta Ophthalmol. 2012, 90, e638–e644. [Google Scholar] [CrossRef] [PubMed]
  28. Lüke, J.; Vukoja, V.; Brandenbusch, T.; Nassar, K.; Rohrbach, J.M.; Grisanti, S.; Lüke, M.; Tura, A. CD147 and matrix-metalloproteinase-2 expression in metastatic and non-metastatic uveal melanomas. BMC Ophthalmol. 2016, 16, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Khatib, N.; Pe’er, J.; Ortenberg, R.; Schachter, J.; Frenkel, S.; Markel, G.; Amer, R. Carcinoembryonic antigen cell adhesion molecule-1 (CEACAM1) in posterior uveal melanoma: Correlation with clinical and histological survival markers. Investig. Ophthalmol. Vis. Sci. 2011, 52, 9368–9372. [Google Scholar] [CrossRef] [PubMed]
  30. Mallikarjuna, K.; Pushparaj, V.; Biswas, J.; Krishnakumar, S. Expression of epidermal growth factor receptor, ezrin, hepatocyte growth factor, and c-Met in uveal melanoma: An immunohistochemical study. Curr. Eye Res. 2007, 32, 281–290. [Google Scholar] [CrossRef]
  31. Economou, M.A.; All-Ericsson, C.; Bykov, V.; Girnita, L.; Bartolazzi, A.; Larsson, O.; Seregard, S. Receptors for the liver synthesized growth factors IGF-1 and HGF/SF in uveal melanoma: Intercorrelation and prognostic implications. Acta Ophthalmol. 2008, 86, 20–25. [Google Scholar] [CrossRef]
  32. Singh, M.K.; Singh, L.; Pushker, N.; Saini, N.; Meel, R.; Chosdol, K.; Bakhshi, S.; Sen, S.; Venkatesh, P.; Chawla, B.; et al. Identification of canonical NFκB (C-NFκB) pathway in uveal melanoma and their relation with patient outcome. Clin. Exp. Metastasis 2019, 36, 271–290. [Google Scholar] [CrossRef]
  33. Cryan, L.M.; Paraoan, L.; Hiscott, P.; Damato, B.E.; Grierson, I.; Gray, D.; Farrell, M.; Doherty, G.A.; Fitzgerald, D.J.; O’Brien, C. Expression of COX-2 and prognostic outcome in uveal melanoma. Curr. Eye Res. 2008, 33, 177–184. [Google Scholar] [CrossRef]
  34. Suesskind, D.; Paulsen, F.; Buchgeister, M.; Spitzer, B.; Rohrbach, J.M.; Bartz-Schmidt, K.U.; Spitzer, M.S. Cyclo-oxygenase-2 expression in photon-radiated and non-radiated uveal melanomas. Acta Ophthalmol. 2010, 88, 582–587. [Google Scholar] [CrossRef]
  35. Singh, M.K.; Singh, L.; Pushker, N.; Chosdol, K.; Bakhshi, S.; Meel, R.; Sen, S.; Kashyap, S. Constitutive expression of c-REL in uveal melanoma patients: Correlation with clinicopathological parameters and patient outcome. Clin. Transl. Oncol. 2020, 22, 1193–1204. [Google Scholar] [CrossRef] [PubMed]
  36. Mallikarjuna, K.; Vaijayanthi, P.; Krishnakumar, S. Cripto-1 expression in uveal melanoma: An immunohistochemical study. Exp. Eye Res. 2007, 84, 1060–1066. [Google Scholar] [CrossRef]
  37. Scala, S.; Ieranò, C.; Ottaiano, A.; Franco, R.; La Mura, A.; Liguori, G.; Mascolo, M.; Staibano, S.; Ascierto, P.A.; Botti, G.; et al. CXC chemokine receptor 4 is expressed in uveal malignant melanoma and correlates with the epithelioid-mixed cell type. Cancer Immunol. Immunother. 2007, 56, 1589–1595. [Google Scholar] [CrossRef] [PubMed]
  38. Coupland, S.E.; Bechrakis, N.; Schüler, A.; Anagnostopoulos, I.; Hummel, M.; Bornfeld, N.; Stein, H. Expression patterns of cyclin D1 and related proteins regulating G1-S phase transition in uveal melanoma and retinoblastoma. Br. J. Ophthalmol. 1998, 82, 961–970. [Google Scholar] [CrossRef]
  39. Coupland, S.E.; Anastassiou, G.; Stang, A.; Anagnostopoulos, I.; Schilling, H.; Bornfeld, N.; Stein, H. The prognostic value of cyclin D1, p53, and MDM2 protein expression in uveal melanoma. J. Pathol. 2000, 191, 120–126. [Google Scholar] [CrossRef]
  40. Topcu-Yilmaz, P.; Kiratli, H.; Saglam, A.; Söylemezoglu, F.; Hascelik, G. Correlation of clinicopathological parameters with HGF, c-Met, EGFR, and IGF-1R expression in uveal melanoma. Melanoma Res. 2010, 20, 126–132. [Google Scholar] [CrossRef]
  41. Hurks, H.M.H.; Metzelaar-Blok, J.A.W.; Barthen, E.R.; Zwinderman, A.H.; De Wolff-Rouendaal, D.; Keunen, J.E.; Jager, M.J. Expression of epidermal growth factor receptor: Risk factor in uveal melanoma. Investig. Ophthalmol. Vis. Sci. 2000, 41, 2023–2027. [Google Scholar]
  42. Clarijs, R.; Schalkwijk, L.; Ruiter, D.J.; De Waal, R.M.W. EMAP-II expression is associated with macrophage accumulation in primary uveal melanoma. Investig. Ophthalmol. Vis. Sci. 2003, 44, 1801–1806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Gajdzis, M.; Theocharis, S.; Gajdzis, P.; Cassoux, N.; Gardrat, S.; Donizy, P.; Klijanienko, J.; Kaczmarek, R. Ephrin receptors (EPH): EphA1, EphA5, and EphA7 expression in uveal melanoma—associations with clinical parameters and patient survival. Life 2020, 10, 225. [Google Scholar] [CrossRef] [PubMed]
  44. Trocmé, E.; Mougiakakos, D.; Johansson, C.C.; All-Eriksson, C.; Economou, M.A.; Larsson, O.; Seregard, S.; Kiessling, R.; Lin, Y. Nuclear HER3 is associated with favorable overall survival in uveal melanoma. Int. J. Cancer 2012, 130, 1120–1127. [Google Scholar] [CrossRef]
  45. Kashyap, S.; Singh, M.K.; Jha, J.; Singh, J.; Pushker, N.; Sen, S.; Venkatesh, P.; Meel, R.; Lomi, N. Prognostic impact of HERC2 protein and pink-eyed dilution protein in uveal melanoma. Hum. Cell 2020, 33, 1264–1272. [Google Scholar] [CrossRef]
  46. Faingold, D.; Marshall, J.C.; Antecka, E. Immune expression and inhibition of heat shock protein 90 in uveal melanoma. Clin. Cancer Res. 2008, 14, 847–855. [Google Scholar] [CrossRef] [Green Version]
  47. Anastassiou, G.; Schilling, H.; Stang, A.; Djakovic, S.; Heiligenhaus, A.; Bornfeld, N. Expression of the cell adhesion molecules ICAM-1, VCAM-1 and NCAM in uveal melanoma: A clinicopathological study. Oncology 2000, 58, 83–88. [Google Scholar] [CrossRef]
  48. All-Ericsson, C.; Girnita, L.; Seregard, S.; Bartolazzi, A.; Jager, M.J.; Larsson, O. Insulin-like growth factor-1 receptor in uveal melanoma: A predictor for metastatic disease and a potential therapeutic target. Investig. Ophthalmol. Vis. Sci. 2002, 43, 1–8. [Google Scholar]
  49. Radberger, P.; Radberger, A.; Vladimir, J.N.B.; Seregard, S.; Economou, M.A. JARID1B Protein Expression and Prognostic Implications in Uveal Melanoma. Investig. Ophthalmol. Vis. Sci. 2012, 53, 4442–4449. [Google Scholar] [CrossRef] [PubMed]
  50. Abourbih, D.A.; Di Cesare, S.; Orellana, M.E.; Antecka, E.; Martins, C.; Petruccelli, L.A.; Burnier Jr., B.M. Lysyl oxidase expression and inhibition in uveal melanoma. Melanoma Res. 2010, 20, 97–106. [Google Scholar] [CrossRef]
  51. Beutel, J.; Wegner, J.; Wegner, R.; Ziemssen, F.; Nassar, K.; Rohrbach, J.M.; Hilgers, R.; Lüke, M.; Grisanti, S. Possible implications of MCAM expression in metastasis and non-metastatic of primary uveal melanoma patients. Curr. Eye Res. 2009, 34, 1004–1009. [Google Scholar] [CrossRef]
  52. Sahin, A.; Kiratli, H.; Soylemezoglu, F.; Tezel, G.G.; Bilgic, S.; Saracbasi, O. Expression of vascular endothelial growth factor-A, matrix metalloproteinase-9, and extravascular matrix patterns and their correlations with clinicopathologic parameters in posterior uveal melanomas. Jpn. J. Ophthalmol. 2007, 51, 325–331. [Google Scholar] [CrossRef] [PubMed]
  53. Ehlers, J.P.; Harbour, J.W. NBS1 expression as a prognostic marker in uveal melanoma. Clin. Cancer Res. 2005, 11, 1849–1853. [Google Scholar] [CrossRef] [Green Version]
  54. Singh, M.K.; Singh, L.; Chosdol, K.; Pushker, N.; Saini, N.; Meel, R.; Bakhshi, S.; Sen, S.; Kashyap, S. Differential expression of p52 and RelB proteins in the metastatic and non-metastatic groups of uveal melanoma with patient outcome. J. Cancer Res. Clin. Oncol. 2019, 145, 2969–2982. [Google Scholar] [CrossRef]
  55. Singh, M.K.; Pushker, N.; Meel, R.; Chodsol, K.; Sen, S.; Bakhshi, S.; Singh, L.; Kashyap, S. Does NEMO/IKKγ protein have a role in determining prognostic significance in uveal melanoma? Clin. Transl. Oncol. 2018, 20, 1592–1603. [Google Scholar] [CrossRef] [PubMed]
  56. Djirackor, L.; Shakir, D.; Kalirai, H.; Petrovski, G.; Coupland, S.E. Nestin expression in primary and metastatic uveal melanoma—Possible biomarker for high-risk uveal melanoma. Acta Ophthalmol. 2018, 96, 503–509. [Google Scholar] [CrossRef] [Green Version]
  57. Greco, I.M.; Calvisi, G.; Ventura, L.; Cerrito, F. An immunohistochemical analysis of nm23 gene product expression in uveal melanoma. Melanoma Res. 1997, 7, 231–236. [Google Scholar] [CrossRef] [PubMed]
  58. Bakalian, S.; Marshall, J.C.; Faingold, D.; Logan, P.; Antecka, E.; Burnier, M.N. Expression of nm23-H1 in uveal melanoma. Melanoma Res. 2007, 17, 284–290. [Google Scholar] [CrossRef]
  59. Lamperska, K.; Mackiewicz, K.; Kaczmarek, A.; Kwiatkowska, E.; Starzycka, M.; Romanowska, B.; Heizman, J.; Stachura, J.; Mackiewicz, A. Expression of p16 in sporadic primary uveal melanoma. Acta Biochim. Pol. 2002, 49, 377–385. [Google Scholar] [CrossRef] [Green Version]
  60. Gajdzis, M.; Theocharis, S.; Klijanienko, J.; Cassoux, N.; Gardrat, S.; Donizy, P.; Kaczmarek, R.; Gajdzis, P. The Prognostic Values of PARP-1 Expression in Uveal Melanoma. Cells 2021, 10, 285. [Google Scholar] [CrossRef] [PubMed]
  61. Seregard, S.; Oskarsson, M.; Spångberg, B. PC-10 as a predictor of prognosis after antigen retrieval in posterior uveal melanoma. Investig. Ophthalmol. Vis. Sci. 1996, 37, 1451–1458. [Google Scholar]
  62. Singh, L.; Singh, M.K.; Kenney, M.C.; Jager, M.J.; Rizvi, M.A.; Meel, R.; Lomi, N.; Bakhshi, S.; Sen, S.; Kashyap, S. Prognostic significance of PD-1/PD-L1 expression in uveal melanoma: Correlation with tumor-infiltrating lymphocytes and clinicopathological parameters. Cancer Immunol. Immunother. 2021, 70, 1291–1303. [Google Scholar] [CrossRef]
  63. Jiang, Z.; Yan, Y.; Dong, J.; Duan, L. PD-1 expression on uveal melanoma induces tumor proliferation and predicts poor patient survival. Int. J. Biol. Markers 2020, 35, 50–58. [Google Scholar] [CrossRef]
  64. Paraoan, L.; Gray, D.; Hiscott, P.; Ebrahimi, B.; Damato, B.; Grierson, I. Expression of p53-induced apoptosis effector PERP in primary uveal melanomas: Downregulation is associated with aggressive type. Exp. Eye Res. 2006, 83, 911–919. [Google Scholar] [CrossRef]
  65. Saraiva, V.S.; Caissie, A.L.; Segal, L.; Edelstein, C.; Burnier, M.N. Immunohistochemical expression of phospho-Akt in uveal melanoma. Melanoma Res. 2005, 15, 245–250. [Google Scholar] [CrossRef] [PubMed]
  66. Berus, T.; Markiewicz, A.; Zwierzchowska, K.; Biecek, P.; Orlowska-Heitzman, J.; Romanowska-Dixon, B.; Donizy, P. Downregulation of polo-like kinase-1 (Plk-1) expression is associated with poor clinical outcome in uveal melanoma patients. Folia Histochem. Cytobiol. 2020, 58, 108–116. [Google Scholar] [CrossRef]
  67. Ramasamy, P.; Larkin, A.; Linge, A.; Tiernan, D.; McAree, F.; Horgan, N.; Moriarty, P.; Beatty, S.; Murphy, C.C.; Clynes, M.; et al. PRDX3 is associated with metastasis and poor survival in uveal melanoma. J. Clin. Pathol. 2020, 73, 408–412. [Google Scholar] [CrossRef]
  68. Caltabiano, R.; Puzzo, L.; Barresi, V.; Cardile, V.; Loreto, C.; Ragusa, M.; Russo, A.; Reibaldi, M.; Longo, A. Expression of Raf Kinase Inhibitor Protein (RKIP) is a predictor of uveal melanoma metastasis. Histol. Histopathol. 2014, 29, 1325–1334. [Google Scholar] [CrossRef]
  69. Salvatorelli, L.; Puzzo, L.; Russo, A.; Reibaldi, M.; Longo, A.; Ragusa, M.; Aldo, C.; Rappazzo, G.; Caltabiano, R.; Salemi, M. Immunoexpression of SPANX-C in metastatic uveal melanoma. Pathol. Res. Pract. 2019, 215. [Google Scholar] [CrossRef] [PubMed]
  70. Ardjomand, N.; Ardjomand, N.; Schaffler, G.; Radner, H.; El-Shabrawi, Y. Expression of somatostatin receptors in uveal melanomas. Investig. Ophthalmol. Vis. Sci. 2003, 44, 980–987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Gangemi, R.; Mirisola, V.; Barisione, G.; Fabbi, M.; Brizzolara, A.; Lanza, F.; Mosci, C.; Salvi, S.; Gualco, M.; Truini, M.; et al. Mda-9/syntenin is expressed in uveal melanoma and correlates with metastatic progression. PLoS ONE 2012, 7, e29989. [Google Scholar] [CrossRef] [PubMed]
  72. Kujala, E.; Damato, B.; Coupland, S.E.; Desjardins, L.; Bechrakis, N.E.; Grange, J.D.; Kivelä, T. Staging of ciliary body and choroidal melanomas based on anatomic extent. J. Clin. Oncol. 2013, 31, 2825–2831. [Google Scholar] [CrossRef]
  73. Angi, M.; Damato, B.; Kalirai, H.; Dodson, A.; Taktak, A.; Coupland, S.E. Immunohistochemical assessment of mitotic count in uveal melanoma. Acta Ophthalmol. 2011, 89, e155–e160. [Google Scholar] [CrossRef] [PubMed]
  74. Liu, Z.G.; Jiao, D. Necroptosis, tumor necrosis and tumorigenesis. Cell Stress 2020, 4, 1–8. [Google Scholar] [CrossRef] [PubMed]
  75. Diener-West, M.; Reynolds, S.M.; Agugliaro, D.J.; Caldwell, R.; Cumming, K.; Earle, J.D.; Green, D.L.; Hawkins, B.S.; Hayman, J.; Jaiyesimi, I.; et al. Screening for metastasis from choroidal melanoma: The Collaborative Ocular Melanoma Study Group Report 23. J. Clin. Oncol. 2004, 22, 2438–2444. [Google Scholar] [CrossRef]
  76. Wu, T.; Sun, L.; Wu, Y.; Xiang, R.; Li, Y.; Rong, W.; Sun, F.; Wang, N. Prognostic value of legumain in uveal melanoma. Mol. Med. Rep. 2016, 13, 2377–2384. [Google Scholar] [CrossRef] [Green Version]
  77. Rozeman, E.A.; Prevoo, W.; Meier, M.A.J.; Sikorska, K.; Van, T.M.; van de Wiel, B.A.; van der Wal, J.E.; Mallo, H.A.; Grijpink-Ongering, L.G.; Broeks, A.; et al. Phase Ib/II trial testing combined radiofrequency ablation and ipilimumab in uveal melanoma (SECIRA-UM). Melanoma Res. 2020, 30, 252–260. [Google Scholar] [CrossRef] [PubMed]
  78. Jung, M.; Lee, J.; Kim, T.M.; Lee, D.H.; Kang, J.H.; Oh, S.Y.; Lee, S.J.; Shin, S.J. Ipilimumab real-world efficacy and safety in Korean melanoma patients from the Korean named-patient program cohort. Cancer Res. Treat. 2017, 49, 44–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Fountain, E.; Bassett, R.L.; Cain, S.; Posada, L.; Gombos, D.S.; Hwu, P.; Bedikian, A.; Patel, S.P. Adjuvant ipilimumab in high-risk Uveal melanoma. Cancers 2019, 11, 152. [Google Scholar] [CrossRef] [Green Version]
  80. Joshua, A.M.; Monzon, J.G.; Mihalcioiu, C.; Hogg, D.; Smylie, M.; Cheng, T. A phase 2 study of tremelimumab in patients with advanced uveal melanoma. Melanoma Res. 2015, 25, 342–347. [Google Scholar] [CrossRef] [PubMed]
  81. Schadendorf, D.; Ascierto, P.A.; Haanen, J.B.A.G.; Espinosa, E.; Demidov, L.V.; Garbe, C.; Lorigan, P.; Gogas, H.; Hoeller, C.; Guren, T.K.; et al. Efficacy and safety of nivolumab (NIVO) in patients with advanced melanoma (MEL) and poor prognostic factors who progressed on or after ipilimumab (IPI): Results from a phase II study (CheckMate 172). J. Clin. Oncol. 2017, 35, 9524. [Google Scholar] [CrossRef]
  82. Namikawa, K.; Takahashi, A.; Mori, T.; Tsutsumida, A.; Suzuki, S.; Motoi, N.; Jinnai, S.; Kage, Y.; Mizuta, H.; Muto, Y.; et al. Nivolumab for patients with metastatic uveal melanoma previously untreated with ipilimumab: A single-institution retrospective study. Melanoma Res. 2020, 30, 76–84. [Google Scholar] [CrossRef]
  83. Rossi, E.; Pagliara, M.M.; Orteschi, D.; Dosa, T.; Sammarco, M.G.; Caputo, C.G.; Petrone, G.; Rindi, G.; Zollino, M.; Blasi, M.A.; et al. Pembrolizumab as first-line treatment for metastatic uveal melanoma. Cancer Immunol. Immunother. 2019, 68, 1179–1185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Shoushtari, A.N.; Navid-Azarbaijani, P.; Friedman, C.F.; Panageas, K.; Postow, M.A.; Callahan, M.K.; Parisa Momtaz, P.; Campbell, S.C.; Shames, Y.; Prempeh-Keteku, N.A.; et al. Efficacy of nivolumab and ipilimumab (Nivo + Ipi) combination in melanoma patients (pts) treated at a single institution on an expanded-access program (EAP). J. Clin. Oncol. 2016, 34, 9554. [Google Scholar] [CrossRef]
  85. Itchins, M.; Ascierto, P.A.; Menzies, A.M.; Oatley, M.; Lo, S.; Douraghi-Zadeh, D.; Harrington, T.; Maher, R.; Grimaldi, A.M.; Guminski, A. A multireferral centre retrospective cohort analysis on the experience in treatment of metastatic uveal melanoma and utilization of sequential liver-directed treatment and immunotherapy. Melanoma Res. 2017, 27, 243–250. [Google Scholar] [CrossRef] [PubMed]
  86. Moreau, P.; Cavallo, F.; Leleu, X.; Hulin, C.; Amiot, M.; Descamps, G.; Facon, T.; Boccadoro, M.; Mignard, D.; Harousseau, J.L. Phase i study of the anti insulin-like growth factor 1 receptor (IGF-1R) monoclonal antibody, AVE1642, as single agent and in combination with bortezomib in patients with relapsed multiple myeloma. Leukemia 2011, 25, 872–874. [Google Scholar] [CrossRef] [Green Version]
  87. Xie, X.; Xie, S.; Xie, C.; Fang, Y.; Li, Z.; Wang, R.; Jiang, W. Pristimerin attenuates cell proliferation of uveal melanoma cells by inhibiting insulin-like growth factor-1 receptor and its downstream pathways. J. Cell Mol. Med. 2019, 23, 7545–7553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Amaro, A.; Mirisola, V.; Angelini, G.; Musso, A.; Tosetti, F.; Esposito, A.I.; Perri, P.; Lanza, F.; Nasciuti, F.; Mosci, C.; et al. Evidence of epidermal growth factor receptor expression in uveal melanoma: Inhibition of epidermal growth factor-mediated signalling by Gefitinib and Cetuximab triggered antibody-dependent cellular cytotoxicity. Eur. J. Cancer 2013, 49, 3353–3365. [Google Scholar] [CrossRef]
  89. Patel, S.P.; Kim, K.B.; Papadopoulos, N.E.; Hwu, W.; Hwu, P.; Prieto, V.G.; Bar-Eli, M.; Zigler, M.; Dobroff, A.; Bronstein, Y.; et al. A phase II study of gefitinib in patients with metastatic melanoma. Melanoma Res. 2011, 21, 357–363. [Google Scholar] [CrossRef] [Green Version]
  90. Moser, J.C.; Pulido, J.S.; Dronca, R.S.; McWilliams, R.R.; Markovic, S.N.; Mansfield, A.S. The Mayo Clinic experience with the use of kinase inhibitors, ipilimumab, bevacizumab, and local therapies in the treatment of metastatic uveal melanoma. Melanoma Res. 2015, 25, 59–63. [Google Scholar] [CrossRef] [PubMed]
  91. Sheng, X.; Yan, X.; Chi, Z.; Si, L.; Cui, C.; Tang, B.; Li, S.; Mao, L.; Lian, B.; Wang, X.; et al. Axitinib in combination with toripalimab, a humanized immunoglobulin G 4 monoclonal antibody against programmed cell death-1, in patients with metastatic mucosal melanoma: An open-label phase Ib trial. J. Clin. Oncol. 2019, 37, 2987–2999. [Google Scholar] [CrossRef]
  92. Dayer, L.E.; Hutchins, L.F.; Johnson, J.T. Treatment of metastatic melanoma with pazopanib: A report of five patient cases. J. Oncol. Pharm. Pract. 2014, 21, 224–231. [Google Scholar] [CrossRef]
  93. De Silva, D.M.; Roy, A.; Kato, T.; Cecchi, F.; Lee, Y.H.; Matsumoto, K.; Bottaro, D.P. Targeting the hepatocyte growth factor/Met pathway in cancer. Biochem. Soc. Trans. 2017, 45, 855–870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Schöffski, P.; Wozniak, A.; Escudier, B.; Rutkowski, P.; Anthoney, A.; Bauer, S.; Sufliarsky, J.; van Herpen, C.; Lindner, L.H.; Grünwald, V.; et al. Crizotinib achieves long-lasting disease control in advanced papillary renal-cell carcinoma type 1 patients with MET mutations or amplification. EORTC 90101 CREATE trial. Eur. J. Cancer 2017, 87, 147–163. [Google Scholar] [CrossRef] [PubMed]
  95. Cheng, H.; Chua, V.; Liao, C.; Purwin, T.J.; Terai, M.; Kageyama, K.; Davies, M.A.; Sato, T.; Aplin, A.E. Co-Targeting HGF/cMET Signaling with MEK Inhibitors in metastatic uveal melanoma. Mol. Cancer Ther. 2017, 16, 516–528. [Google Scholar] [CrossRef] [Green Version]
  96. Daud, A.; Kluger, H.M.; Kurzrock, R.; Schimmoller, F.; Weitzman, A.L.; Samuel, T.A.; Moussa, A.H.; Gordon, M.S.; Shapiro, G.I. Phase II randomised discontinuation trial of the MET/VEGF receptor inhibitor cabozantinib in metastatic melanoma. Br. J. Cancer 2017, 116, 432–440. [Google Scholar] [CrossRef] [Green Version]
  97. Luke, J.J.; Olson, D.J.; Allred, J.B.; Strand, C.A.; Bao, R.; Zha, Y.; Carll, T.; Labadie, B.W.; Bastos, B.R.; Butler, M.O.; et al. Randomized Phase II trial and tumor mutational spectrum analysis from cabozantinib versus chemotherapy in metastatic uveal melanoma (Alliance A091201). Clin. Cancer Res. 2020, 26, 804–811. [Google Scholar] [CrossRef] [Green Version]
  98. Puzanov, I.; Sosman, J.; Santoro, A.; Saif, M.W.; Goff, L.; Dy, G.K.; Zucali, P.; Means-Powell, J.A.; Ma, W.W.; Simonelli, M.; et al. Phase 1 trial of tivantinib in combination with sorafenib in adult patients with advanced solid tumors. Investig. N. Drugs 2015, 33, 159–168. [Google Scholar] [CrossRef] [Green Version]
  99. Falchook, G.S.; Kurzrock, R.; Amin, H.M.; Xiong, W.; Fu, S.; Piha-Paul, S.A.; Janku, F.; Eskandari, G.; Catenacci, D.V.; Klevesath, M.; et al. First-in-man phase I trial of the selective MET inhibitor tepotinib in patients with advanced solid tumors. Clin. Cancer Res. 2020, 26, 1237–1246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Middleton, M.R.; McAlpine, C.; Woodcock, V.K.; Corrie, P.; Infante, J.R.; Steven, N.M.; Evans, T.R.J.; Anthoney, A.; Shoushtari, A.N.; Hamid, O.; et al. Tebentafusp, A TCR/Anti-CD3 Bispecific Fusion Protein Targeting gp100, Potently Activated Antitumor Immune Responses in Patients with Metastatic Melanoma. Clin. Cancer Res. 2020, 26, 5869–5878. [Google Scholar] [CrossRef] [PubMed]
  101. Damato, B.E.; Dukes, J.; Goodall, H.; Carvajal, R.D. Tebentafusp: T Cell Redirection for the Treatment of Metastatic Uveal Melanoma. Cancers 2019, 11, 971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Barbagallo, C.; Caltabiano, R.; Broggi, G.; Russo, A.; Puzzo, L.; Avitabile, T.; Longo, A.; Reibaldi, M.; Barbagallo, D.; Di Pietro, C.; et al. LncRNA LINC00518 Acts as an Oncogene in Uveal Melanoma by Regulating an RNA-Based Network. Cancers 2020, 12, 3867. [Google Scholar] [CrossRef] [PubMed]
  103. Kim, Y.J.; Park, S.J.; Maeng, K.J.; Lee, S.C.; Lee, C.S. Multi-Platform Omics Analysis for Identification of Molecular Characteristics and Therapeutic Targets of Uveal Melanoma. Sci. Rep. 2019, 9, 19235. [Google Scholar] [CrossRef] [PubMed]
  104. Durante, M.A.; Rodriguez, D.A.; Kurtenbach, S.; Kuznetsov, J.N.; Sanchez, M.I.; Decatur, C.L.; Snyder, H.; Feun, L.G.; Livingstone, A.S.; Harbour, J.W. Single-cell analysis reveals new evolutionary complexity in uveal melanoma. Nat. Commun. 2020, 11, 496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Pandiani, C.; Strub, T.; Nottet, N.; Cheli, Y.; Gambi, G.; Bille, K.; Husser, C.; Dalmasso, M.; Béranger, G.; Lassalle, S.; et al. Single-cell RNA sequencing reveals intratumoral heterogeneity in primary uveal melanomas and identifies HES6 as a driver of the metastatic disease. Cell Death Diff. 2021, 28, 1990–2000. [Google Scholar] [CrossRef]
  106. Karlsson, J.; Nilsson, L.M.; Mitra, S.; Alsén, S.; Shelke, G.V.; Sah, V.R.; Forsberg, E.M.V.; Stierner, U.; All-Eriksson, C.; Einarsdottir, B.; et al. Molecular profiling of driver events in metastatic uveal melanoma. Nat. Commun. 2020, 11, 1894. [Google Scholar] [CrossRef] [Green Version]
Table 1. Summary of all markers whose expression is related to different prognostic factors in uveal melanoma.
Table 1. Summary of all markers whose expression is related to different prognostic factors in uveal melanoma.
MarkerProtein FunctionStudy Group SizeConclusion
ABCB5 (ATP-binding cassette sub-family B member 5) [17]Human transmembrane P-glycoprotein, plays a role in transmembrane transport (including chemotherapeutic drugs).55High expression correlates with presence of metastasis.
ADAM10 (A disintegrin and metalloproteinase domain-containing protein 10) [18]Transmembrane protein, controls membrane fusion and cell-cell and cell-matrix interactions.52High expression correlates with presence of metastasis.
Adiponectin (GBP-28, apM1, AdipoQ and Acrp30) [19]Protein hormone involved in regulating glucose levels, fatty acid breakdown, plays a role in limiting cell proliferation and reducing inflammation.34Low expression correlates with extrascleral extension, more frequent chromosome 3 loss and presence of metastasis.
AIF (Apoptosis inducing factor) [20]Ubiquitous protein, plays a proapoptotic function in the nucleus and redox activity in mitochondria.54High expression correlates with reduced survival time.
ATM (Ataxia-telangiectasia mutated protein) [21]Predominantly nuclear protein, an activator of DNA damage response.69Loss of expression correlates with larger tumor diameter, tumor thickness, presence of epithelioid cells, reduced disease-free survival time.
ATM (Ataxia-telangiectasia mutated protein) [22]Predominantly nuclear protein, an activator of DNA damage response.69Loss of expression correlates with reduced disease-free survival time.
Beclin [23]Autophagy related protein, plays a central role in the autophagic process as a major member of the macro-autophagic phase.85High expression correlates with less frequent presence of metastasis and longer disease-free survival time.
BNIP3 (BCL2 19 kD protein-interacting protein 3) [24]Cytoplasm protein, regulates cell death, autophagy, and cytoprotection.47High expression correlates with deeper scleral invasion, increased pigmentation and reduced overall survival time.
BTNL9 (Butyrophilin-like protein 9) [25]Modulator of the T cell response.62High expression correlates with longer overall survival time.
CCR7 (C-C Motif Chemokine Receptor 7) [26]Receptor mainly expressed in lymphoid cells, mediate cell migration of naıve lymphocytes and mature dendritic cells to secondary lymphoid organs and regulate the transport of cancer cells through the extracellular matrix.49High expression correlates with higher tumor thickness, presence of epithelioid cells, lymphocytic infiltration, presence of necrosis and reduced overall survival time.
CCR7 (C-C Motif Chemokine Receptor 7) [27]Receptor mainly expressed in lymphoid cells, mediate cell migration of naıve lymphocytes and mature dendritic cells to secondary lymphoid organs and regulate the transport of cancer cells through the extracellular matrix.70High expression correlates with presence of metastasis.
CD147 (Cluster of differentiation 147, Basigin (BSG), extracellular matrix metalloproteinase inducer (EMMPRIN)) [28]Member of the immunoglobulin superfamily, plays a role in intercellular recognition, various immunologic phenomena, differentiation, and development.49High expression in the nonmetastatic sub-group correlates with larger tumor diameter and TNM stage. In the metastatic sub-group, the presence of nested CD147 positive cells correlates with ciliary body involvement.
CEACAM (Carcinoembryonic antigen cell adhesion molecule-1) [29]Transmembrane glycoprotein, plays a role in the intercellular interactions, regulation of cell growth, angiogenesis, immune modulation, and hepatic insulin clearance.79High expression correlates with presence of epithelioid cells and network extracellular matrix pattern.
c-Met (Tyrosine-protein kinase Met or hepatocyte growth factor receptor (HGFR)) [30]Transmembrane RTK receptor, plays a role in embryonic development, organogenesis and wound healing, angiogenesis, and metastasis formation.60High expression correlates with presence of metastasis and reduced overall survival time.
c-Met (Tyrosine-protein kinase Met or hepatocyte growth factor receptor (HGFR)) [31]Transmembrane RTK receptor, plays a role in embryonic development, organogenesis and wound healing, angiogenesis, and metastasis formation.132High expression correlates with melanoma-specific mortality.
C-NFκB proteins (Canonical nuclear factor-κB proteins (p65 and p50)) [32]Nuclear protein, coordinator of innate immunity and inflammation.75High expression of p65 and p50 correlates with presence of metastasis and reduced survival time.
COX-2 (Cyclooxygenase-2) [33]Enzyme, which catalyze the prostanoid synthesis reaction.32High expression correlates with metastatic death.
COX-2 (Cyclooxygenase-2) [34]Enzyme, which catalyze the prostanoid synthesis reaction.43High expression correlates with presence of metastasis.
c-REL [35]Member of the nuclear factor κB (NF-κB) transcription factor family and an emerging regulator of tumorigenesis.75High expression correlates with tumor thickness, presence of epithelioid cells, presence of metastasis and reduced overall survival time.
Cripto-1 (Teratocarcinoma-derived growth factor-1) [36]An oncogenic growth factor involving tumorigenesis and cancer cell proliferation and survival.36High expression correlates with extrascleral extension and presence of metastasis.
CXCR4 (C-X-C motif chemokine receptor 4) [37]Alpha-chemokine receptor specific for stromal-derived-factor-1, a molecule endowed with potent chemotactic activity for lymphocytes.44High expression correlates with presence of epithelioid cells.
CyclinD1 [38]Predominantly nuclear protein, regulator of cell cycle.66High expression correlates with extrascleral extension and presence of epithelioid cells.
CyclinD1 [39]Predominantly nuclear protein, regulator of cell cycle.96High expression correlates with extrascleral extension, presence of the mixed or epithelioid cells the tumor cell MIB-1 positivity and presence of metastasis.
EGFR (Epidermal growth factor receptor) [40]Transmembrane protein, plays a role in epithelial tissue development and homeostasis.40High expression correlates with higher mitotic activity.
EGFR (Epidermal growth factor receptor) [41]Transmembrane protein, plays a role in epithelial tissue development and homeostasis.22High expression correlates with metastatic death.
EMAP-II (Endothelial monocyte-activating polypeptide II) [42]Proinflammatory cytokine and chemoattractant of macrophages, expressed on the cell surface.25High expression correlates with macrophage infiltration.
EphA1 (Eph-A1 receptor, erythropoietin-producing human hepatocellular receptor A1) [43]RTK receptor, plays a role in the regulation of a processes critical to embryonic development including axon guidance, formation of tissue boundaries, cell migration, segmentation, proliferation, and angiogenesis.94High expression correlates with smaller tumor diameter, less frequently occurring extrascleral extension, lower mitotic activity, and presence of vitreous hemorrhage.
EphA5 (Eph-A5 receptor, erythropoietin-producing human hepatocellular receptor A5) [43]RTK receptor, plays a role in the regulation of a processes critical to embryonic development including axon guidance, formation of tissue boundaries, cell migration, segmentation, proliferation, and angiogenesis.94High expression correlates with less frequent chromosome 3 loss, more frequent occurrence of vitreous hemorrhage, absence of distant metastases and longer overall survival time.
HER3 (Human epidermal growth factor receptor 3 or receptor tyrosine-protein kinase erbB-3) [44]Transmembrane RTK receptor, implicated in growth, proliferation, chemotherapeutic resistance, and the promotion of invasion and metastasis.128High nuclear expression correlates with longer overall survival time.
HERC2 (HECT and RLD Domain Containing E3 Ubiquitin Protein Ligase 2) [45]Predominantly nuclear and cytoplasm protein, plays a role in DNA repair regulation, pigmentation, and neurological disorders.52High expression correlates with ciliary body involvement, presence of epithelioid cells and increased pigmentation.
Hsp90 (Heat shock protein 90) [46]Cytoplasmic protein, plays a role in folding, intracellular transport, maintenance, and degradation of proteins, and facilitating cell signaling.44High expression correlates with larger tumor diameter.
ICAM-1 (Intercellular cell adhesion molecule-1) [47]Adhesion molecule, ligand for leukocyte function-associated antigen-1, involved in the process of inflammation, the circulation of blood cells, and in the immune surveillance of the host.90Loss of expression correlates with presence of metastasis.
IGF-1R (Insulin-like growth factor 1 receptor) [31]Transmembrane receptor, implicated in insulin signaling, plays a role in several cancer development.132High expression correlates with melanoma-specific mortality.
IGF-1R (Insulin-like growth factor 1 receptor) [48]Transmembrane receptor, implicated in insulin signaling, plays a role in several cancer development.36High expression correlates with death of disease.
JARID1B (Jumonji AT-rich interactive domain 1B) [49]Demethylase enzyme, induce demethylation of tri- and di-methylated lysines in the 4 position of histone 3.121High expression correlates with reduced survival time.
LOX (Lysyl oxidase) [50]Extracellular enzyme, plays a role in embryonic development, wound healing and adult tissue remodeling.33High expression correlates with presence of epithelioid cells and reduced metastasis-free survival time.
MCAM (Melanoma cell adhesion molecule, MUC18, Mel-Cam, CD146) [51]Adhesion molecule, plays a role in intracellular signaling cascades.35High expression correlates with death of disease.
MMP-2 and MMP-9 (Matrix metalloproteinase-2 and -9) [28]Secreted and membrane-associated neutral endopeptidase, plays a role in degrading extracellular matrix proteins, cell proliferation, migration, differentiation, angiogenesis, apoptosis, and host defense.26High expression correlates with presence of metastasis and reduced survival time.
MMP-9 (Matrix metalloproteinase 9) [52]Secreted and membrane-associated neutral endopeptidase, plays a role in degrading extracellular matrix proteins, cell proliferation, migration, differentiation, angiogenesis, apoptosis, and host defense.100High expression correlates with presence of loop and/or network patterns, lymphocytic infiltration, presence of necrosis and presence of metastasis.
Nbs1 (Nibrin, NBN) [53]Intracellular protein, plays a role in the repair of double strand breaks and telomere maintenance.49High expression correlates with reduced survival time.
NC-NFκB proteins (p52, RelB, and co-expression of p52/RelB) [54]Nuclear protein, plays a role in promoting cancer proliferation and progression.75High expression correlates with reduced metastasis-free survival time and reduced overall survival time.
NEMO/IKKγ (Factor κB essential modulator, inhibitor of nuclear factor kappa B kinase subunit gamma) [55]Protein essential for the activation of transcription factor NFκB, which regulates the cellular responses to inflammation, immunity, and cell survival.75Low expression correlates with presence of epithelioid cells, higher mitotic activity, presence of vascular loop, neovascularization and reduced overall survival time.
Nestin (Neural stem cell protein) [56]Cytoplasm and membrane-bound protein, member of the intermediate filament (IF) class VI protein family167High expression correlates with presence of epithelioid cells, more frequent chromosome 3 loss and chromosome 8q gain, higher mitotic activity, presence of vascular loop and reduced survival time.
nm23 (Nucleoside diphosphate kinase A) [57]Cytoplasm protein, product of metastasis suppressor gene (NM23).33Low expression correlates with larger tumor diameter and deeper scleral invasion.
nm23-H1 (Nucleoside diphosphate kinase A) [58]Cytoplasm protein, product of metastasis suppressor gene (NM23).32The increased immunostaining intensity correlates with longer survival time.
p16 (Cyclin-dependent kinase inhibitor 2A, CDKN2A, multiple tumor suppressor 1) [59]Predominantly nuclear protein, slowing the progression of the cell cycle from G1 phase to the S phase.41Low expression correlates with mixed cell type.
p53 [39]Cell-cycle regulatory protein.96High expression correlates with presence of metastasis.
PARP (Poly (ADP-ribose) polymerase) [60]Nuclear protein, which participate in the DNA repair processes.91High expression correlates with larger tumor diameter, higher histopathological grade, more frequent chromosome 3 loss, reduced overall survival time and disease-free survival time.
PCNA (Proliferating cell nuclear antigen, ATLD2) [61]The DNA polymerase auxiliary protein involved in the control of DNA replication.212High expression correlates with presence of metastasis.
PD-1 (Programmed cell death receptor-1) [62]Membrane-bound receptor, plays a role in regulation immune system’s response to the cells of the human body.71High expression correlates with ciliary body involvement, presence of epithelioid cells, macrophage infiltration, absence of BAP-1 staining and presence of metastasis.
PD-1 (Programmed cell death receptor-1) [63]Membrane-bound receptor, plays a role in regulation immune system’s response to the cells of the human body.82High expression correlates with larger tumor diameter and reduced survival time.
PD-L1 (Programmed death-ligand 1) [62]Ligand for PD-1 (programmed cell death receptor-1), plays a role in regulation immune system’s response to the cells of the human body.71High expression correlates with macrophage infiltration, higher AJCC prognostic stage group and presence of metastasis.
PERP (p53 apoptosis effector related to PMP-22) [64]Cytoplasm protein, plays a role in inducing cell death.16Low expression correlates with more frequent chromosome 3 loss.
Phospho-Akt [65]Cytoplasmic protein, plays a role in phosphorylation and inactivation of several proteins involved in apoptosis.34High expression correlates with presence of metastasis.
PLK-1 (Polo-like kinase-1) [66]Regulator of mitotic entry and cytokinesis.158Low expression correlates with higher clinical tumor stage, higher AJCC prognostic stage group and reduced overall survival time.
P-protein (Pink-eyeddilution protein) [45]Membrane protein, plays a role in melanin synthesis in melanocytes and retinal pigment epithelium.52High expression correlates with ciliary body involvement, presence of epithelioid cells, increased pigmentation, and advanced clinical tumor staging. High cytoplasmic expression correlates with presence of metastasis.
PRDX3 (Thioredoxin-dependent peroxidase reductase) [67]Cytoplasmic protein, involved in redox regulation of the cell and protects radical-sensitive enzymes from oxidative damage.92High expression correlates with presence of metastasis and reduced survival time.
RKIP (Raf Kinase Inhibitor Protein) [68]Regulator of proliferative pathways within the cell.44Low expression correlates with presence of metastasis.
SPANX-C (SPANX family member C; Sperm protein associated with the nucleus on the X chromosome C) [69]Cytoplasmic protein, expressed in highly metastatic cell lines.55High expression correlates with presence of metastasis.
SSR (Somatostatin receptor, SSTR) [70]The binding of somatostatin to its membrane receptor starts a signaling pathway that leads to arrest of cell growth or apoptosis.25High expression correlates with longer survival time.
Syntenin (Syndecan binding protein syntenin-1, melanoma differentiation-associated gene 9, mda-9) [71]Predominantly cytoplasm protein, plays a role in clustering of membrane receptors, intracellular trafficking, Sox4 activation, and signal transduction.29High expression correlates with risk of metastasis recurrence.
TIMP-1 and TIMP-2 (Tissue inhibitor of metalloproteinase-1 and -2) [28]Metalloproteinase inhibitors.26High expression correlates with longer survival time.
VEGF-A (Vascular endothelial growth factor-A) [52]Acts specifically on endothelial cells, mediates increased vascular permeability, induces angiogenesis, vasculogenesis and endothelial cell growth, promotes cell migration, and inhibits apoptosis.100High expression correlates with presence of vascular loos and/or network patterns, lymphocytic infiltration, necrosis, and presence of metastasis.
Table 2. Summary of markers whose expression is related to tumor size.
Table 2. Summary of markers whose expression is related to tumor size.
MarkerStudy Group SizeConclusion
EphA1 (Eph-A1 receptor, erythropoietin-producing human hepatocellular receptor A1) [43]94High expression correlates with smaller tumor diameter.
nm23 (Nucleoside diphosphate kinase A) [57]33Low expression correlates with larger tumor diameter.
CD147 (Cluster of differentiation 147, Basigin (BSG), extracellular matrix metalloproteinase inducer (EMMPRIN)) [28]49High expression in the nonmetastatic sub-group correlates with larger tumor diameter.
Hsp90 (Heat shock protein 90) [46]44High expression correlates with larger tumor diameter.
PARP (Poly (ADP-ribose) polymerase) [60]91High expression correlates with larger tumor diameter.
PD-1 (Programmed cell death receptor-1) [63]82High expression correlates with larger tumor diameter.
ATM (Ataxia-telangiectasia mutated protein) [21]69Loss correlates with larger tumor diameter and tumor thickness.
CCR7 (C-C Motif Chemokine Receptor 7) [26]49High expression correlates with tumor thickness.
c-REL [35]75High expression correlates with tumor thickness.
Table 3. Summary of markers whose expression is related to ciliary body involvement.
Table 3. Summary of markers whose expression is related to ciliary body involvement.
MarkerStudy Group SizeConclusion
CD147 (Cluster of differentiation 147, Basigin (BSG), extracellular matrix metalloproteinase inducer (EMMPRIN)) [28]49In the metastatic sub-group, the presence of nested CD147 positive cells correlates with ciliary body involvement.
HERC2 (HECT and RLD Domain Containing E3 Ubiquitin Protein Ligase 2) [45]52High expression correlates with ciliary body involvement.
PD-1 (Programmed cell death receptor-1) [62]71High expression correlates with ciliary body involvement.
P-protein (Pink-eyed dilution protein) [45]52High expression correlates with ciliary body involvement.
Table 4. Summary of markers whose expression is related to scleral and extrascleral infiltration.
Table 4. Summary of markers whose expression is related to scleral and extrascleral infiltration.
MarkerStudy Group SizeConclusion
BNIP3 (BCL2 19 kD protein-interacting protein 3) [24]47High expression correlates with deeper scleral invasion.
nm23 (Nucleoside diphosphate kinase A) [57]33Low expression correlates with deeper scleral invasion.
Adiponectin (GBP-28, apM1, AdipoQ and Acrp30) [19]34Low expression correlates with extrascleral extension.
Cripto-1 (Teratocarcinoma-derived growth factor-1) [36]36High expression correlates with extrascleral extension.
CyclinD1 [38]66High expression correlates with extrascleral extension.
CyclinD1 [39]96High expression correlates with extrascleral extension.
EphA1 (Eph-A1 receptor, erythropoietin-producing human hepatocellular receptor A1) [43]94High expression correlates with less frequently occurring extrascleral extension.
Table 5. Summary of markers whose expression is related to presence of various cell types.
Table 5. Summary of markers whose expression is related to presence of various cell types.
MarkerStudy Group SizeConclusion
ATM (Ataxia-telangiectasia mutated protein) [21]69Low expression correlates with presence of epithelioid cells.
CCR7 (C-C Motif Chemokine Receptor 7) [26]49High expression correlates with presence of epithelioid cells.
CEACAM (Carcinoembryonic antigen cell adhesion molecule-1) [29]79High expression correlates with presence of epithelioid cells.
c-REL [35]75High expression correlates with presence of epithelioid cells.
CXCR4 (C-X-C motif chemokine receptor 4) [37]44High expression correlates with presence of epithelioid cells.
CyclinD1 [38]66High expression correlates with presence of epithelioid cells.
HERC2 (HECT and RLD Domain Containing E3 Ubiquitin Protein Ligase 2) [45]52High expression correlates with presence of epithelioid cells.
LOX (Lysyl oxidase) [50]33High expression correlates with presence of epithelioid cells.
NEMO/IKKγ (Nuclear factor κB essential modulator, inhibitor of nuclear factor kappa B kinase subunit gamma) [55]75Low expression correlates with presence of epithelioid cells.
Nestin (Neural stem cell protein) [56]167High expression correlates with presence of epithelioid cells.
PD-1 (Programmed cell death receptor-1) [62]71High expression correlates with presence of epithelioid cells.
P-protein (Pink-eyed dilution protein) [45]52High expression correlates with presence of epithelioid cells.
CyclinD1 [39]96High expression correlates with presence of mixed or epithelioid cells.
p16 (Cyclin-dependent kinase inhibitor 2A, CDKN2A, multiple tumor suppressor 1) [59]41Low expression correlates with mixed cell type.
PARP (Poly (ADP-ribose) polymerase) [60]91High expression correlates with higher histopathological grade.
Table 6. Summary of markers whose expression is related to chromosome status.
Table 6. Summary of markers whose expression is related to chromosome status.
MarkerStudy Group SizeConclusion
EphA5 (Eph-A5 receptor, erythropoietin-producing human hepatocellular receptor A5) [43]94High expression correlates with less frequent chromosome 3 loss.
Nestin (Neural stem cell protein) [56]167High expression correlates with more frequent chromosome 3 loss and chromosome 8q gain.
PARP (Poly (ADP-ribose) polymerase) [60]91High expression correlates with more frequent chromosome 3 loss.
Adiponectin (GBP-28, apM1, AdipoQ and Acrp30) [19]34Low expression correlates with more frequent chromosome 3 loss.
PERP (p53 apoptosis effector related to PMP-22) [64]16Low expression correlates with more frequent chromosome 3 loss.
Table 7. Summary of markers whose expression is related to mitotic count.
Table 7. Summary of markers whose expression is related to mitotic count.
MarkerStudy Group SizeConclusion
EGFR (Epidermal growth factor receptor) [40]40High expression correlates with higher mitotic count.
EphA1 (Eph-A1 receptor, erythropoietin-producing human hepatocellular receptor A5) [43]94High expression correlates with lower mitotic count.
NEMO/IKKγ (Nuclear factor κB essential modulator, inhibitor of nuclear factor kappa B kinase subunit gamma) [55]75Low expression correlates with higher mitotic count.
Nestin (Neural stem cell protein) [56]167High expression correlates with higher mitotic count.
Table 8. Summary of markers whose expression is related to extravascular matrix loops and networks.
Table 8. Summary of markers whose expression is related to extravascular matrix loops and networks.
MarkerStudy Group SizeConclusion
CEACAM (Carcinoembryonic antigen cell adhesion molecule-1) [29]79High expression correlates with network extracellular matrix pattern.
MMP-9 (Matrix metalloproteinase-9) [52]100High expression correlates with presence of vascular loops and network patterns.
VEGF-A (Vascular endothelial growth factor-A) [52]100High expression correlates with presence of vascular loops and network patterns.
NEMO/IKKγ (Nuclear factor κB essential modulator, inhibitor of nuclear factor kappa B kinase subunit gamma) [55]75Low expression correlates with presence of vascular loops.
Nestin (Neural stem cell protein) [56]167High expression correlates with presence of vascular loops.
Table 9. Summary of markers whose expression is related to TIMs and TILs infiltration.
Table 9. Summary of markers whose expression is related to TIMs and TILs infiltration.
MarkerStudy Group SizeConclusion
EMAP-II (Endothelial monocyte-activating polypeptide II) [42]25High expression correlates with macrophage infiltration.
PD-1 (Programmed cell death receptor-1) [62]71High expression correlates with macrophage infiltration.
PD-L1 (Programmed death-ligand 1) [62]71High expression correlates with macrophage infiltration.
CCR7 (C-C Motif Chemokine Receptor 7) [26]49High expression correlates with lymphocytic infiltration.
MMP-9 (Matrix metalloproteinase-9) [52]100High expression correlates with lymphocytic infiltration.
VEGF-A (Vascular endothelial growth factor-A) [52]100High expression correlates with lymphocytic infiltration.
Table 10. Summary of markers whose expression is related to other prognostic factors.
Table 10. Summary of markers whose expression is related to other prognostic factors.
MarkerStudy Group SizeConclusion
BNIP3 (BCL2 19 kD protein-interacting protein 3) [24]47High expression correlates with increased pigmentation.
HERC2 (HECT and RLD Domain Containing E3 Ubiquitin Protein Ligase 2) [45]52High expression correlates with increased pigmentation.
P-protein (Pink-eyed dilution protein) [45]52High expression correlates with increased pigmentation and advanced clinical tumor staging.
PD-L1 (Programmed death-ligand 1) [63]71High expression correlates with higher AJCC prognostic stage group.
PLK-1 (Polo-like kinase-1) [66]158Low expression correlates with higher AJCC prognostic stage group.
CD147 (Cluster of differentiation 147, Basigin (BSG), extracellular matrix metalloproteinase inducer (EMMPRIN)) [28]49High expression in the nonmetastatic sub-group correlates with TNM stage.
CCR7 (C-C Motif Chemokine Receptor 7) [26]49High expression correlates with necrosis.
MMP-9 (Matrix metalloproteinase-9) [52]100High expression correlates with degree of necrosis.
VEGF-A (Vascular endothelial growth factor-A) [52]100High expression correlates with degree of necrosis.
EphA1 (Eph-A1 receptor, erythropoietin-producing human hepatocellular receptor A1) [43]94High expression correlates with more frequent presence of hemorrhage in the vitreous chamber.
EphA5 (Eph-A5 receptor, erythropoietin-producing human hepatocellular receptor A5) [43]94High expression correlates with more frequent presence of hemorrhage in the vitreous chamber.
NEMO/IKKγ (Nuclear factor κB essential modulator, inhibitor of nuclear factor kappa B kinase subunit gamma) [55]75Low expression correlates with neovascularization.
CyclinD1 [39]96High expression correlates with the tumor cell MIB-1 positivity.
PD-1(Programmed cell death receptor-1) [62]71High expression correlates with absence of BAP-1 staining.
Table 11. Summary of markers whose expression is related to metastasis presence.
Table 11. Summary of markers whose expression is related to metastasis presence.
MarkerStudy Group SizeConclusion
ABCB5 (ATP-binding cassette sub-family B member 5) [17]55High expression correlates with presence of metastasis.
ADAM10 (A disintegrin and metalloproteinase domain-containing protein 10) [18]52High expression correlates with presence of metastasis.
Adiponectin (GBP-28, apM1, AdipoQ and Acrp30) [19]34Low expression correlates with presence of metastasis.
Beclin [23]85High expression correlates with less frequent metastasis.
CCR7 (C-C Motif Chemokine Receptor 7) [27]70High expression correlates with presence of metastasis.
c-Met (Tyrosine-protein kinase Met or hepatocyte growth factor receptor (HGFR)) [30]60High expression correlates with presence of metastasis.
C-NFκB proteins (Canonical nuclear factor-κB proteins (p65 and p50)) [32]75High expression correlates with presence of metastasis.
COX-2 (Cyclooxygenase-2) [33]32High expression correlates with metastatic death.
COX-2 (Cyclooxygenase-2) [34]43High expression correlates with presence of metastasis.
c-REL [35]75High expression correlates with presence of metastasis.
Cripto-1 (Teratocarcinoma-derived growth factor-1) [36]36High expression correlates with presence of metastasis.
CyclinD1 [39]96High expression correlates with presence of metastasis.
EGFR (Epidermal growth factor receptor) [41]22High expression correlates with metastatic death.
EphA5 (Eph-A5 receptor, erythropoietin-producing human hepatocellular receptor A5) [43]94High expression correlates with less frequent metastasis.
ICAM-1 (Intercellular cell adhesionmolecule-1) [47]90Loss of expression correlates with presence of metastasis.
IGF-1R (Insulin-like growth factor 1 receptor) [48]36High expression correlates with metastatic death.
LOX (Lysyl oxidase) [50]33High expression correlates with reduced metastasis-free survival time.
MCAM (Melanoma cell adhesion molecule, MUC18, Mel-Cam, CD146) [51]35High expression correlates with metastatic death.
MMP-2 and MMP-9 (Matrix metalloproteinase-2 and -9) [28]26High expression correlates with presence of metastasis.
MMP-9 (Matrix metalloproteinase-9) [52]100High expression correlates with presence of metastasis.
NC-NFκB proteins (p52, RelB, and co-expression of p52/RelB) [54]75High expression correlates with reduced metastasis-free survival time.
p53 [39]96High expression correlates with presence of metastasis.
PCNA (Proliferating cell nuclear antigen, ATLD2) [61]212High expression correlates with presence of metastasis.
PD-1 (Programmed cell death receptor-1) [62]71High expression correlates with presence of metastasis.
PD-L1 (Programmed death-ligand 1) [62]71High expression correlates with presence of metastasis.
phospho-Akt [65]34High expression correlates with presence of metastasis.
P-protein (Pink-eyed dilution protein) [45]52High cytoplasmic expression correlates with presence of metastasis.
PRDX3 (Thioredoxin-dependent peroxidase reductase) [67]92High expression correlates with presence of metastasis.
RKIP (Raf Kinase Inhibitor Protein) [68]44Low expression correlates with presence of metastasis.
SPANX-C (SPANX family member C; Sperm protein associated with the nucleus on the X chromosome C) [69]55High expression correlates with presence of metastasis.
Syntenin (Syndecan binding protein syntenin-1, melanoma differentiation-associated gene 9, mda-9) [71]29High expression correlates with risk of metastasis recurrence.
VEGF-A (Vascular endothelial growth factor-A) [52]100High expression correlates with presence of metastasis.
Table 12. Summary of markers whose expression is related to survival times.
Table 12. Summary of markers whose expression is related to survival times.
MarkerStudy Group SizeConclusion
AIF (Apoptosis inducing factor) [20]54High expression correlates with reduced survival time.
ATM (Ataxia-telangiectasia mutated protein) [21]69Loss of expression correlates with reduced disease-free survival time.
ATM (Ataxia-telangiectasia mutated protein) [22]69Loss of expression correlates with reduced disease-free survival time.
Beclin [23]85High expression correlates with longer disease-free survival time.
BNIP3 (BCL2 19 kD protein-interacting protein 3) [24]47High expression correlates with reduced overall survival time.
BTNL9 (Butyrophilin-like protein 9) [25]62High expression correlates with longer overall survival time.
CCR7 (C-C Motif Chemokine Receptor 7) [26]49High expression correlates with reduced overall survival time.
c-Met (Tyrosine-protein kinase Met or hepatocyte growth factor receptor (HGFR)) [30]60High expression correlates with reduced overall survival time.
c-Met (Tyrosine-protein kinase Met or hepatocyte growth factor receptor (HGFR)) [31]132High expression correlates with melanoma-specific mortality.
C-NFκB proteins (Canonical nuclear factor-κB proteins (p65 and p50)) [32]75High expression correlates with reduced survival time.
c REL [35]75High expression correlates with reduced overall survival time.
EphA5 (Eph-A5 receptor, erythropoietin-producing human hepatocellular receptor A5) [43]94High expression correlates with longer overall survival time.
HER3 (Human epidermal growth factor receptor 3 or receptor tyrosine-protein kinase erbB-3) [44]128High nuclear expression correlates with longer overall survival time.
IGF-1R (Insulin-like growth factor 1 receptor) [31]132High expression correlates with melanoma-specific mortality.
JARID1B (Jumonji AT-rich interactive domain 1B) [49]121High expression correlates with reduced survival time.
Legumain (Asparagine endopeptidase (AEP)) [76]82High expression correlates with reduced survival time.
MMP-2 and MMP-9 (Matrix metalloproteinase-2 and -9) [28]26High expression correlates with reduced survival time.
Nbs1 (Nibrin, NBN) [53]49High expression correlates with reduced survival time.
NC-NFκB proteins (p52, RelB, and co-expression of p52/RelB) [54]75High expression correlates with reduced overall survival time.
NEMO/IKKγ (Nuclear factor κB essential modulator, inhibitor of nuclear factor kappa B kinase subunit gamma) [55]75Low expression correlates with reduced overall survival time.
Nestin (Neural stem cell protein) [56]167High expression correlates with reduced survival time.
nm23-H1 (Nucleoside diphosphate kinase A) [58]32The increased immunostaining intensity correlates with longer survival time.
PARP (Poly (ADP-ribose) polymerase) [60]91High expression correlates with reduced overall survival time and disease-free survival time.
PD-1 (Programmed cell death receptor-1) [63]82High expression correlates with reduced survival time.
PLK-1 (Polo-like kinase-1) [66]158Low expression correlates with reduced overall survival time.
PRDX3 (Thioredoxin-dependent peroxidase reductase) [67]92High expression correlates with reduced survival time.
SSR (Somatostatin receptor, SSTR) [70]25High expression correlates with longer survival time.
TIMP-1 and TIMP-2 (Tissue inhibitor of metalloproteinase-1 and -2) [28]26High expression correlates with longer survival time.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gajdzis, M.; Kaczmarek, R.; Gajdzis, P. Novel Prognostic Immunohistochemical Markers in Uveal Melanoma-Literature Review. Cancers 2021, 13, 4031. https://doi.org/10.3390/cancers13164031

AMA Style

Gajdzis M, Kaczmarek R, Gajdzis P. Novel Prognostic Immunohistochemical Markers in Uveal Melanoma-Literature Review. Cancers. 2021; 13(16):4031. https://doi.org/10.3390/cancers13164031

Chicago/Turabian Style

Gajdzis, Malgorzata, Radoslaw Kaczmarek, and Pawel Gajdzis. 2021. "Novel Prognostic Immunohistochemical Markers in Uveal Melanoma-Literature Review" Cancers 13, no. 16: 4031. https://doi.org/10.3390/cancers13164031

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop