Indian Journal of Ophthalmology

SYMPOSIUM
Year
: 2015  |  Volume : 63  |  Issue : 2  |  Page : 93--102

Uveal melanoma: Estimating prognosis


Swathi Kaliki1, Carol L Shields2, Jerry A Shields2,  
1 Institute for Eye Cancer, L V Prasad Eye Institute, Banjara Hills, Support provided by Operation Eyesight Institute for Eye Cancer (SK) and Hyderabad Eye Research Foundation (SK), Hyderabad, India
2 Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, USA

Correspondence Address:
Dr. Swathi Kaliki
Institute for Eye Cancer, L V Prasad Eye Institute, Hyderabad - 500 034, Telangana
India

Abstract

Uveal melanoma is the most common primary malignant tumor of the eye in adults, predominantly found in Caucasians. Local tumor control of uveal melanoma is excellent, yet this malignancy is associated with relatively high mortality secondary to metastasis. Various clinical, histopathological, cytogenetic features and gene expression features help in estimating the prognosis of uveal melanoma. The clinical features associated with poor prognosis in patients with uveal melanoma include older age at presentation, male gender, larger tumor basal diameter and thickness, ciliary body location, diffuse tumor configuration, association with ocular/oculodermal melanocytosis, extraocular tumor extension, and advanced tumor staging by American Joint Committee on Cancer classification. Histopathological features suggestive of poor prognosis include epithelioid cell type, high mitotic activity, higher values of mean diameter of ten largest nucleoli, higher microvascular density, extravascular matrix patterns, tumor-infiltrating lymphocytes, tumor-infiltrating macrophages, higher expression of insulin-like growth factor-1 receptor, and higher expression of human leukocyte antigen Class I and II. Monosomy 3, 1p loss, 6q loss, and 8q and those classified as Class II by gene expression are predictive of poor prognosis of uveal melanoma. In this review, we discuss the prognostic factors of uveal melanoma. A database search was performed on PubMed, using the terms �DQ�uvea,�DQ� �DQ�iris,�DQ� �DQ�ciliary body,�DQ� �DQ�choroid,�DQ� �DQ�melanoma,�DQ� �DQ�uveal melanoma�DQ� and �DQ�prognosis,�DQ� �DQ�metastasis,�DQ� �DQ�genetic testing,�DQ� �DQ�gene expression profiling.�DQ� Relevant English language articles were extracted, reviewed, and referenced appropriately.



How to cite this article:
Kaliki S, Shields CL, Shields JA. Uveal melanoma: Estimating prognosis.Indian J Ophthalmol 2015;63:93-102


How to cite this URL:
Kaliki S, Shields CL, Shields JA. Uveal melanoma: Estimating prognosis. Indian J Ophthalmol [serial online] 2015 [cited 2024 Mar 29 ];63:93-102
Available from: https://journals.lww.com/ijo/pages/default.aspx/text.asp?2015/63/2/93/154367


Full Text

Uveal melanoma represents 79-81% of ocular melanomas and 3-5% of all melanomas. [1],[2],[3] In the United States, the incidence of uveal melanoma is 5/million population. [1] In Europe, the incidence of uveal melanoma follows a north-to-south decreasing gradient ranging from 2 to 8/million population. [2] Over the years, with advances in the treatment strategies, there is an improvement in the rate of local tumor control and globe salvage, but survival rate remains relatively unchanged. [1],[4],[5]

Uveal melanoma has a high tendency to metastasize resulting in high mortality. [1],[5],[6],[7],[8],[9],[10] The common sites of metastasis include liver (89%), lung (29%), and bone (17%). [8] Approximately, 50% of patients with uveal melanoma succumb to metastasis within 10 years of diagnosis, irrespective of the type of treatment. [1],[5],[6],[7],[8],[9],[10] Median survival after metastasis is 6 to 12 months, though long-term survival has been reported. [8],[11] Though, the overall survival rate of patients with metastatic uveal melanoma is poor, median survival of patients receiving treatment for metastasis is better than those receiving no treatment. [8],[12],[13],[14],[15],[16]

Various clinical, histopathological, and cytogenetic features of uveal melanoma can identify those patients who are at high risk of developing metastasis and probably benefit from appropriate prophylactic/therapeutic adjuvant and adjunctive treatments [Table 1]. In this review, we describe the features predictive of poor prognosis in patients with uveal melanoma. A database search was performed on PubMed, using the terms "uvea," "iris," "ciliary body," "choroid," "melanoma," "uveal melanoma" and "prognosis," "metastasis," "genetic testing," "gene expression profiling (GEP)". Relevant English language articles were extracted, reviewed, and referenced appropriately.{Table 1}

 Clinical Features



The clinical features predicting prognosis in patients with uveal melanoma include age at presentation, gender, tumor size, tumor location, tumor configuration, presence or absence of ocular/oculodermal melanocytosis (OM), extraocular tumor extension (EOE), and American Joint Committee on Cancer (AJCC) classification. [17],[18],[19],[20],[21],[22],[23],[24],[25],[26],[27],[28],[29],[30],[31],[32],[33],[34],[35],[36],[37],[38],[39],[40],[41],[42],[43],[44],[45],[46],[47],[48],[49],[50],[51],[52],[53],[54],[55],[56],[57],[58],[59],[60],[61],[62],[63],[64],[65]

Age at presentation

A few studies have concluded that the age of presentation has no influence on the prognosis of uveal melanoma. [17],[18],[19] In contrast, other studies have indicated that the life prognosis is more favorable in children with uveal melanoma compared with adults. [21],[22],[23],[24],[25],[26] The favorable prognosis of uveal melanoma in children is attributed to a bias secondary to confounding factors such as higher percentage of iris melanoma, smaller tumor size at diagnosis, lower incidence of EOE, and shorter follow-up duration in children compared to adults. [20],[21],[22] In an attempt to overcome these confounding factors, Kaliki et al. matched clinical predictive factors for metastasis such as gender, tumor location, tumor basal diameter, tumor thickness, extraocular extension, and follow-up duration in 122 patients in each age category (young [≤20 years], mid-adults [21-60 years], older adults [>60 years]) and found that younger patient age at the time of diagnosis of uveal melanoma is associated with lower rate of metastasis compared with mid-adults and older adults. [27] Evaluation by decade of presentation in 8,033 patients with uveal melanoma by Shields et al. showed similar gradual increase in risk for metastasis with increasing age. [26] At 10 years, metastasis was 10% in patients aged 11-20 years, 21% for 41-50 years, and 30% for 71-80 years. [26]

Gender

The Collaborative Ocular Melanoma Study (COMS) study group found no difference in uveal melanoma-related metastasis and death between men and women. [26] Other study groups made similar observations. [9],[28] However, in a study of 723 uveal melanoma patients, Zloto et al. found significant gender differences in prognosis. [29] Male patients had worse prognosis with higher melanoma-related metastasis and death than female patients. Melanoma-related mortality in the first 10 years was two-fold higher in males compared with females. [29] By multivariable regression analysis, Rietschel et al. found that male gender was associated with significantly higher risk of melanoma-related mortality than female gender. [30] The lower metastatic rate in females could be related to hormonal factors. [29],[31] Zloto et al. suggested that estrogen may indirectly influence the tissue through the regulation of other factors that directly affect the melanoma or could be related to inhibitory action of estrogen on the growth of micro-metastases within the liver. [29]

Tumor size

Tumor size (largest basal diameter and thickness) is one of the most important clinical prognostic feature of uveal melanoma. [6],[7],[9],[30],[32],[33],[34],[35],[36],[37] In a meta-analysis of 8 articles by Diener-West et al., the combined weighted estimates of 5-year mortality rates associated with uveal melanoma were 16% for small tumors (<2 or 3 mm tumor thickness and <10 or 11 mm basal diameter), 32% for medium tumors (3-8 mm tumor thickness and <15 or 16 mm basal diameter), and 53% for large tumors (>8 mm tumor thickness and >15 mm basal diameter). [7] The medium sized tumor trial (2.5-10 mm tumor thickness and <16 mm basal diameter) by COMS group revealed 5, 10, and 12 years melanoma-related mortality at 10%, 18%, and 21%, respectively, for patients in the iodine - 125 brachytherapy treatment arm and 11%, 17%, and 17%, respectively, for those in the enucleation treatment arm. [6] In the large tumor trial (>10 mm tumor thickness or >2 mm tumor thickness and >16 mm basal diameter) by COMS group, melanoma-related mortality at 5 and 10 years was 28% and 40%, respectively, for patients in the enucleation treatment arm and 26% and 45% in the external beam radiotherapy preceding enucleation treatment arm. [32],[33]

In a long-term study of 289 patients with uveal melanoma, Kujala et al. found significant association between largest basal diameter of the tumor and melanoma-related mortality. [9] By competing risks regression analysis, the Hazard ratio was 1.08 for each millimeter increase in tumor diameter. The cumulative incidence estimates of melanoma-related mortality increased with increasing tumor basal diameter at 18% for small tumors (<10 mm basal diameter), 52% for medium tumors (10-15 mm basal diameter), and 59% for large tumors (≥16 mm basal diameter) at 25 years. [7]

In a study of 8,033 uveal melanoma patients by Shields et al., increasing tumor thickness of uveal melanoma was found to be associated with increasing risk for metastasis. [34] Kaplan-Meier estimates of metastasis at 5, 10, and 20 years was 6%, 12%, and 20% for small melanoma (<3 mm tumor thickness), 14%, 26%, and 37% for medium melanoma (3.1-8 mm), and 35%, 49%, and 67% for large melanoma (>8 mm) respectively. Each millimeter increase in tumor thickness was associated with approximately 5% increased risk for metastasis at 10 years and a hazard ratio of 1.08. [34]

Tumor location

Uveal melanoma can arise in the iris, ciliary body, or choroid. Iris melanoma has a better prognosis and ciliary body melanoma has the worst prognosis. [34],[35],[36],[37],[38],[39],[40],[41],[42],[43],[44],[45] In a study of 8,033 patients with uveal melanoma, metastasis at 5 and 10 years was 4% and 7% for iris melanoma, 19% and 33% for ciliary body melanoma, and 15% and 25% for choroidal melanoma, respectively. [34]

The lower metastasis rate of iris melanoma is related to lower biologic activity or smaller tumor size of iris melanoma. [34],[37],[42] In a study of 3432 cases of uveal melanoma, iris melanoma was reported to have 10 times lower mortality compared with ciliary body and choroidal melanoma. [37] In another study of 8033 patients with uveal melanoma, the 10-year metastatic rate from iris melanoma was approximately 5 times less than ciliary body melanoma and 4 times less than choroidal melanoma. [34],[38]

Patients with ciliary body melanoma greater than 7 mm in thickness are at 2.5 times greater risk than patients with thinner tumors for metastatic disease and melanoma-related death. [43] According to Li et al., there is a significant relation between degree of ciliary body involvement (% of tumor base within the ciliary body) and melanoma-related metastasis. [44] According to this study, a melanoma of presumed ciliary body origin (>50% of the tumor base within the ciliary body) had 1.6-2.3 times higher chance of metastasis than a choroidal (<50% of the tumor base within the ciliary body) tumor. Tumors with 100% ciliary body involvement had 3.6 times higher chance of metastasis than choroidal melanoma. [44] The poor prognosis of ciliary body melanoma has been related to larger tumor size, predilection for monosomy 3 and 8q gain, and tumor microvascular patterns. [35],[37],[39],[41],[45] However, ciliary body involvement has been an independent predictor of survival in several multivariate models. [36],[40],[45]

Tumor configuration

Diffuse configuration of uveal melanoma is associated with poor prognosis. [46],[47],[48],[49],[50] Diffuse uveal melanoma represents horizontal, flat growth pattern of uveal melanoma, including diffuse iris melanoma, ring melanoma of ciliary body, and diffuse choroidal melanoma. [51]

Diffuse iris melanoma is a rare variant of iris melanoma representing 11% cases. [47],[48] Diffuse iris melanoma is associated with greater metastatic potential compared with non-diffuse iris melanoma. In an analysis of 25 cases of diffuse iris melanoma, metastasis occurred in 13% cases at 6 years follow-up, [48] compared to 2-4% metastasis in cases with nondiffuse iris melanoma. [34],[38],[52] The higher metastatic rate in diffuse iris melanoma is associated with high incidence of epithelioid cells, elevated intraocular pressure, posterior tumor margin at iris root or angle, and extraocular extension. [48],[53]

Ring melanoma of the ciliary body is a rare variant of uveal melanoma occurring in <1% cases. [49] Metastasis in these cases is as high as 52% at 5-year follow-up, [49] compared to 19% in cases with nondiffuse variant. [34] Poor prognosis in cases with ring melanoma of ciliary body is attributed to difficult and delayed diagnosis and treatment.

Diffuse choroidal melanoma represents 3-17% of all choroidal melanomas. [50] Diffuse choroidal melanoma carries a substantial risk for metastasis despite its flat appearance. [54] In a comparative study of diffuse versus nondiffuse choroidal melanoma in 2121 patients, Kaplan-Meier estimates of melanoma-related metastasis (diffuse vs. nondiffuse) was 8% versus 4% at 5 years and 17% versus 10% at 10 years. [50] The poor prognosis in diffuse choroidal melanoma may be related to delayed diagnosis, a greater proportion of epithelioid cells, and its tendency for extraocular extension. [46],[50]

Ocular/oculodermal melanocytosis

Ocular/OM is associated with increased risk of development of uveal melanoma, estimated at 1 in 400 affected patients. [54] The influence of ocular/OM on the prognosis of uveal melanoma has been recently explored. [55],[56]

In a study of 7872 patients with uveal melanoma, patients with associated OM had double the risk for metastasis compared with those with no OM. [55] By Kaplan-Meier estimates, metastasis in patients with OM versus no OM was 27% versus 15% at 5 years, and 48% versus 24% at 10 years. [55] Similar findings were recorded in a matched study where each patient with uveal melanoma associated with OM was matched for factors age, gender, tumor thickness, tumor basal diameter, location of tumor epicenter, and location of anterior tumor margin. [56] In that analysis, Kaplan-Meier estimates for systemic metastasis in the melanocytosis group at 5 and 15 years were 27% and 59% (respectively) compared with 15% and 33% in the no melanocytosis group. [56]

Extraocular tumor extension

Extraocular tumor extension is a poor prognostic factor for uveal melanoma, occurring in 8-15% cases. [57],[58],[59],[60],[61] EOE is more commonly associated with larger tumors, anterior tumor extension, large basal tumor diameter, diffuse uveal melanoma, epithelioid cellularity, closed vascular loops, high mitotic rate, and monosomy 3, resulting in poor prognosis. [50],[54],[57],[58],[59],[60],[61]

The overall survival could be related to the characteristics of the intraocular portion of the tumor rather than the EOE, except when the size of the EOE is large (>5 mm). [58] In a study of 610 patients with uveal melanoma, the 5-year mortality rate for patients with a microscopic extension and small EOE (1-4 mm) were 37% and 24%, respectively. [58] These numbers were markedly higher for patients with large EOE with 5-year mortality rate of 78%. [58]

American Joint Committee on Cancer classification

The AJCC (7 th edition) is an attempt to unify the clinical prognostic factors into a single classification system. [62] In this classification, iris melanoma is graded according to tumor extent, associated secondary glaucoma, and EOE. Posterior uveal (ciliary body and choroid) melanoma is graded according to tumor basal diameter and thickness, ciliary body involvement, and extraocular extension. [62] The patients with advanced AJCC tumor staging exhibit poor prognosis. [63]

In a study of 452 patients with iris melanoma based on AJCC classification, the 10-year metastatic rate was 2% for stage I tumors, 6% for stage II, and 41% for stage III. [63] In a study of 7731 patients with posterior uveal melanoma based on T category of AJCC classification, the 10-year metastatic rate was 15% for T1 tumors, 25% for T2, and 49% for T3, and 63% for T4. [63] The risk for metastasis and death increased two-fold with each increasing tumor category. [64] Based on AJCC staging for posterior uveal melanoma, 10-year metastatic rate was 12% for stage I tumors, 29% for stage II, and 61% for stage III. The risk for metastasis and death increased three-fold with each increasing melanoma staging. [64]

 Histopathological Features



The histopathologic features predicting prognosis of uveal melanoma include tumor cell type, mitotic activity, mean diameter of ten largest nucleoli, microvascular density (MVD), extravascular matrix patterns, tumor-infiltrating lymphocytes, tumor-infiltrating macrophages, insulin-like growth factor-1 receptor (IGF-1R), and human leukocyte antigen (HLA) Class I expression. [65],[66],[67],[68],[69],[70],[71],[72],[73],[74],[75],[76],[77],[78],[79],[80],[81],[82],[83],[84],[85],[86],[87],[88],[89],[90],[91],[92],[93],[94],[95],[96],[97],[98],[99],[100],[101],[102],[103],[104],[105],[106],[107],[108],[109],[110],[111],[112],[113]

Tumor cell type

Tumor cell type is an important prognostic factor. Callender initially proposed a classification system for uveal melanoma including spindle A, spindle B, epithelioid, mixed, fascicular, and necrotic types. [65] This was later modified to include spindle A, spindle B, epithelioid, and mixed tumors. [66] The modified Callender classification showed an improved correlation between the cell type and the mortality. [66]

In a study of 2652 enucleated eyes with uveal melanoma by Paul et al., the 15-year mortality for spindle A tumor was 19%, spindle B was 26%, mixed spindle B and epithelioid was 59%, and epithelioid tumor was 72%. [67] The 15-year mortality of patients with melanomas of mixed cell type is three times that of patients with tumors of pure spindle cell type. [37] Various studies have established that the spindle cell uveal melanoma has the best prognosis, mixed cell melanoma an intermediate, and epithelioid cell melanoma has the worst prognosis. [37],[40],[61],[67],[68],[69],[70] The prognosis worsens with increasing number of epithelioid cells per high power field (HPF). [69] In a study of 232 enucleated eyes from patients with uveal melanoma, the 10-year survival was 82% in patients with <0.5 epithelioid cells/HPF, 55% for 0.5 to 4.9 epithelioid cells/HPF, and 33% in patients with >5 epithelioid cells/HPF. [69]

Mitotic activity

Tumors with high cellular proliferation have a poor prognosis. Cellular proliferation in uveal melanoma can be determined by counting number of mitoses per 40 HPFs. Tumors with a low mitotic activity have a relatively good prognosis compared to those showing high mitotic activity. [71] In a study of 217 small malignant melanomas by McLean et al., increase in the number of mitoses/40 HPFs was significantly associated with increasing mortality rate. Six-year mortality was 16% for 0 mitosis/40 HPSs, 23% for 1mitosis/40 HPFs, 40% for 2-4, 47% for 5-8, and 56% for 9-48 mitoses/40 HPFs. [71]

In recent years immunohistochemical markers, PC-10 for proliferating cell nuclear antigen and MIB-1 for Ki-67 are used to examine cellular proliferation in conventionally processed histological preparations. [72],[72],[74],[75],[76],[77] These markers seem to be reliable and easy tools for evaluating cellular activity. A high fraction of PC-10 and Ki-67 in uveal melanoma cells is associated with decreased melanoma-specific survival. [77],[78]

Mean diameter of ten largest nucleoli

A large mean diameter of ten largest nucleoli (MLN) is associated with poor prognosis. [37],[79],[80],[81],[82],[83],[84],[85] MLN can be measured on silver or hematoxylin-eosin stained sections. Larger MLN is found in tumors with epithelioid cells and in those with increasing MVD, which could influence poor prognosis. [79],[81],[85] However, studies have shown that larger MLN is an independent predictor of melanoma-related mortality. [79],[83],[84] There is a 0.58-1.27 times increase in melanoma-related mortality for each 1-um increase in MLN. [79],[80],[81],[82],[83],[84]

In a study of 167 specimens for MLN, the median MLN was 4.05um (range, 2.60-6.18sum). [79] The 10-year Kaplan-Meier estimate for melanoma-specific survival was 74% for small, 60% for medium, and 42% for large MLN. [79] MLN remained an independent predictor of prognosis, when adjusted in turn for the effect of ciliary body involvement, largest tumor basal diameter, presence of epithelioid cells, and microvascular loops and networks. When adjusted for MVD, it was of borderline significance. However, combined MLN with cell type and MVD best predicted melanoma-specific survival. [79]

Microvascular density

MVD is a quantitative measurement of tumor vascularity. Microvessels are more distinct and easier to count from sections immunolabeled for CD34 epitope or FVIII-Rag. [86],[87] Noninvasive methods of detection of MVD by ultrasound parameter imaging and confocal microscopy are also described. [88],[89],[90] High MVD is associated with a shortened survival of patients with uveal melanoma. [79],[86],[87],[91] There is a significant association between high MVD and presence of microvascular loops and networks, epithelioid cells, and largest basal tumor diameter, thus influencing poor prognosis. [86],[87] High MVD alone can also serve as an independent risk factor for melanoma-related metastasis and death. [79],[87]

In a study of 162 consecutive enucleation specimens by Mäkitie et al., the median MVD was 40 vessels/0.313 mm 2 (range, 5-121). [87] The 10-year melanoma-specific mortality increased from quartile to quartile with increasing MVD, at 9%, 29%, 59%, and 64% according to quartiles. [87] Similar results were shown by Foss et al., with estimated 9-year cumulative probabilities of survival for the four quartiles at 85%, 55%, 44%, and 27%, respectively. [86]

Extravascular matrix patterns

The concept of microvascular patterns was introduced by Folberg et al., who suggested that microvessel architecture has a strong association with prognosis of uveal melanoma. [92] In addition to normal vessels incorporated into the tumor stroma and focal avascular zones, the tumors contain straight vessels, parallel straight vessels, parallel vessels that cross-link, vascular arcs (incomplete loops), arcs with branching, closed vascular loops that encircle small clusters of tumor cells, and microvascular networks composed of back-to-back loops. [92] The patterns are assessed with light microscopy on periodic-acid-schiff stained tissues.

The presence of microvascular loops and networks surrounding nests of tumor cells can independently predict melanoma-related tumor death. [92],[93],[94] In a matched case control study by Folberg et al., it was shown that the presence of at least one closed vascular loop in a uveal melanoma is the most significant vascular pattern associated with death from metastatic melanoma. [92] The detection of at least one closed loop within a tumor is associated with the presence of epithelioid cells and at least one mitotic figure. [92] In a study by Al-Jamal et al., the Kaplan-Meier estimate for 10-year melanoma-specific survival was estimated at 80% if no loops, 48% if loops were present without networks, and 40% if loops forming networks were present. [79]

Tumor infiltrating lymphocytes

Increased infiltration of uveal melanoma by lymphocytes suggests poor prognosis. [95],[96],[97],[98] An association between monosomy 3 and influx of tumor-infiltrating lymphocytes has been established. [97] Uveal melanoma cells which have lost one copy of chromosome 3 may produce inflammatory mediators, which recruit and activate CD8 + , CD4 + , and Foxp3 + T cells, as well as CD68 + , and CD68 + and CD163 + macrophages. Activation of these infiltrating cells will result in production of more inflammatory mediators generating a tumor-promoting inflammatory microenvironment, resulting in poor prognosis. [98]

In a study of 1193 cases by de la Cruz et al., 134 (12%) tumors contained 100 or more lymphocytes per 20 HPF and were classified as high lymphocytic group. [95] An equivalent number of cases with fewer lymphocytes comprised the low lymphocytic group. The survival rate at 15 years was 37% for patients in the high lymphocytic group and 70% for patients in the low lymphocytic group. Despite control of other risk factors, there was a significant association between increased number of lymphocytes per 20 HPF and survival. [95]

Tumor infiltrating macrophages

High numbers of tumor-infiltrating macrophages in uveal melanoma are associated with an unfavorable prognosis. [99],[100],[101],[102],[103],[104] These tumors are associated with the presence of epithelioid cells, increased MVD, and monosomy 3. [99],[100],[101],[102],[103] Macrophages of the M2 phenotype promotes phagocytic activity, tissue remodeling, tumor progression, and angiogenesis. [104]

In a study of 43 uveal melanomas by immunohistochemistry, the infiltrating macrophages were predominantly CD68 + CD163 + (M2 phenotype). [104] Kaplan-Meier survival analysis revealed that a low CD68 + or CD68 + CD163 + macrophages were associated with a significantly better survival. However, the significance of tumor-infiltrating macrophages as a predictor of melanoma-related mortality could not be established on multivariable analysis. [104]

Insulin-like growth factor-1 receptor

High expression levels of IGF-1R in primary tumors correlates significantly with lower survival rates. [105] The significant association between high IGF-1R expression and death due to metastatic disease may be related to the fact that IGF-1 is mainly produced in the liver, which is the preferential site for uveal melanoma metastases. [105] Based on this finding, IGF-1R blockage is a possible new treatment modality for metastases that may also play a role as adjuvant therapy in preventing the development of metastatic disease; [106] and serum IGF-1 level may be used as a predictive biomarker for metastatic uveal melanoma when measured repeatedly. [107]

In a study of 36 cases of uveal melanoma with more than 15 years follow-up, Kaplan-Meier analysis showed a significant association between a high IGF-1R (expression in >50% melanoma cells) and melanoma-related mortality. [105] Over a period of 15 years follow-up, 56% of patients with high IGF-1R, and 42% with low IGF-1R (expression in 15 to 50% cells), and 0% with very low IGF-1R (expression in <15% cells) died due to metastasis. [105]

Human leukocyte antigen expression

Unlike the general rule of higher mortality with lower expression of HLA Class I determinants, [108] uveal melanoma exhibits an opposite association. This could be related to natural killer cells playing an essential role in immune responses directed against uveal melanoma metastases rather than cytotoxic T-lymphocytes. [109],[110] Higher HLA Class I and II expression is associated with higher melanoma-related mortality. [109],[110],[111] However, some studies have found no association between HLA expression and melanoma-related mortality. [112]

In a study by Ericsson et al., a significant correlation between the expression of HLA Class I antigens, β2 -microglobulin, and HLA Class II antigens and the development of metastases was noted. [111] Jager et al. demonstrated that the tumors expressing HLA-A exhibited higher melanoma-related mortality (75%) as compared to those not staining for HLA-A (20%). [113] HLA-A was found to be the strongest independent predictor of tumor-related mortality, whereas HLA-B expression was not an independent predictor of survival. [113] Blom et al. have reported that a high expression of HLA-B significantly correlated with the presence of epitheloid cells, a cell type that carries a bad prognosis. [110]

 Cytogenetic Features



Recent studies underscore the importance of cytogenetic features in the prognosis of uveal melanoma. Tumor sample for genetic testing is either obtained from enucleation specimen or intraoperatively by fine needle aspiration biopsy. Aberrations in chromosome 1, 3, 6, and 8 determine the survival in patients with uveal melanoma. [39],[114],[115],[116] Chromosome 3 loss, 8q gain, 1p loss, and 6q loss are associated with poor prognosis. [39],[41],[114],[115],[116],[117],[118],[119],[120],[121],[122],[123],[124],[125],[126],[127],[128],[129],[130],[131],[132],[133]

Chromosome 3

In majority of cases with chromosome 3 aberrations in uveal melanoma, monosomy 3 (complete loss of one copy of chromosome 3) is more common and is the most important prognostic factor. [39],[41],[118],[119],[122] Partial aberrations on chromosome 3 (partial deletion of one copy of chromosome 3) and isodisomy (loss of one copy of chromosome 3 and then duplication of the remaining defective copy) have also been reported, both of which have a metastatic potential. [123],[124],[125] A variation of monosomy 3 status can occur within the same tumor. In a study of uveal melanomas by Schoenfield et al., monosomy 3 was noted at the base and disomy 3 at apex of the tumor. [124]

In a landmark publication by Prescher et al., monosomy 3 was established as a significant prognostic factor for uveal melanoma. [39] Of 54 patients with uveal melanoma, monosomy 3 was identified in 30 (56%) patients. Three-year mortality rate in patients with tumors harboring monosomy 3 was 50%, and those with no monosomy 3 was 0%. [39] Subsequent studies have shown that monosomy 3 occurs in 21 to 56% cases and is associated with melanoma-related mortality in 42-54% over a follow-up period ranging from 2 to 8 years. [39],[41],[118],[120],[122] Presence of monosomy 3 indicates high-risk melanoma, with an increased risk for metastasis. Monosomy 3 is associated with clinical and histopathological risk factors including larger tumor diameter, ciliary body tumor location, epithelioid cell type, high mitotic rate, vascular loops, and extraocular extension. [39],[41],[118],[120] Recently, the tumor suppressor gene BRCA1-associated protein 1 (BAP1) has been mapped on chromosome 3p21.1. Its somatic mutation has been associated with metastatic uveal melanoma. [127]

Chromosome 8

In majority of cases with aberrations in chromosome 8, 8q gain is more common occurring in 41 to 53% cases of uveal melanoma, while 8p loss occurs rarely. [128],[129],[130] The most common forms of 8q gain are trisomy 8, isochromosome 8q, and amplification of the c-myc gene. [123]

Chromosome 8q gain is an important prognostic factor for uveal melanoma either when it presents alone or co-exists with monosomy 3. [117],[119] Chromosome 8q gain most commonly co-exists with monosomy 3 and is associated with poor prognosis than 8q gain alone or monosomy 3 alone. In a study of 356 patients with uveal melanoma by Damato et al., the tumors showed no cytogenetic abnormalities of chromosomes 3 or 8 in 42%, 8q gain in 11%, monosomy 3 in 21%, and combined 8q gain and monosomy 3 in 27%. [118] Five-year disease specific mortality rates were 6% in the absence of chromosomal abnormality, 31% with only 8q gain, 40% with only monosomy 3, and 66% with combined 8q gain and monosomy 3. [118] 8q gain was associated with clinical and histopathologic risk factors, including larger tumor diameter, ciliary body tumor location, epithelioid cell type, high mitotic rate, and vascular loops. [118]

Chromosome 1

Loss of part or all of chromosome 1p is associated with poor prognosis either when it presents alone or co-exists with monosomy 3. [120],[121] Chromosome 1p loss occurs more frequently in tumors with monosomy 3 (40%) than those with disomy 3 (10%). [121]

Concomitant loss of chromosomes 1p and 3 has a stronger correlation with melanoma-related metastasis than either one of them separately. In a study 0f 120 patients with uveal melanoma by Kilic et al., it was noted that the effect of monosomy 3 on survival was largely modified by changes in 1p36. [120] After correcting for confounding variables, it was found that patients harboring tumors with concurrent loss of chromosomes 1p36 and 3 have 7.8 times higher chance of developing metastases than do those without these losses or with loss of either chromosomes 1p36 or 3. [120]

Chromosome 6

Chromosome 6 gain has an inverse relationship with melanoma-related metastasis, and is a strong indicator of good prognosis of uveal melanoma. [114],[119] Chromosome 6p gain is usually mutually exclusive with monosomy 3. [114],[131] These tumors with chromosome 6p gain have been proposed to represent a separate group of uveal melanomas with an alternative genetic pathway in carcinogenesis compared with those with monosomy 3. [131],[132] The coexistence of 6p gain and monosomy 3 occurs in only 4% cases of uveal melanoma. [132]

Chromosome 6q loss is associated with poor prognosis. In a study of 35 tumors by Aalto et al., 6q loss was noted in 40% metastasizing tumors when compared to 7% non-metastasizing tumors. [133]

 Transcriptomic Features



Based on analysis of mRNA by GEP of all chromosomes, 2 tumor classes of uveal melanoma were described by Tschentscher et al. [134] All tumors in Class I had disomy 3, and all tumors in Class II had monosomy 3. [134] Subsequently, a relationship between GEP and melanoma-related survival in patients with uveal melanoma was studied by Onken et al. [135] Based on the comparison of molecular classification to cytogenetic changes in 10 tumor samples, Class I tumors were considered as low-risk tumors and Class II tumors as high-risk tumors. [135] Chromosome 6p gain occurred in 80% Class I tumors, and monosomy 3 was noted in 80% Class II tumors and no Class I tumors. [135] Survival analysis of 50 patients revealed 95% survival probability in Class I versus 31% in class 2 at 7.7 years follow-up. [135] Subsequent studies noted similar findings. Class I lesions were unlikely to undergo metastasis, whereas Class II lesions predicted a greater rate of metastasis and melanoma-related mortality. [135],[136],[137],[138],[139],[140] Patients with Class II tumors tend to be older, and are associated with thicker tumors, epithelioid cytology, higher proliferative rate (higher Ki-67 positivity), and mutations in BAP1 tumor suppressor gene. [127],[138],[139]

A recent study of 459 patients with uveal melanoma by a multicenter trial from 12 oncology centers, revealed a strong association between GEP and prognosis of uveal melanoma. [140] At a median of 17 months follow-up, metastasis was detected in 1% Class I cases and 26% Class II cases. [140] GEP class had a strong independent association with metastasis and chromosome 3 status did not provide additional prognostic information that was independent of GEP. [140]

 Conclusion



The long-term prognosis of uveal melanoma is poor with death occurring in more than 50% cases. The prognosis of uveal melanoma can be estimated by clinical, histopathological, cytogenetic, and transcriptomic markers. Improved prognostication for uveal melanoma allows identification of patients at high risk for metastasis, thereby facilitating targeted screening, and probable adjunctive/adjuvant systemic treatment. Currently, the most effective measure to minimize poor prognosis is early detection of melanoma at a time when the tumor is small and at least risk for metastatic disease.

References

1Singh AD, Turell ME, Topham AK. Uveal melanoma: Trends in incidence, treatment, and survival. Ophthalmology 2011;118:1881-5.
2Virgili G, Gatta G, Ciccolallo L, Capocaccia R, Biggeri A, Crocetti E, et al. Incidence of uveal melanoma in Europe. Ophthalmology 2007;114:2309-15.
3Chang AE, Karnell LH, Menck HR. The National Cancer Data Base report on cutaneous and noncutaneous melanoma: A summary of 84,836 cases from the past decade. The American College of Surgeons Commission on Cancer and the American Cancer Society. Cancer 1998;83:1664-78.
4Jampol LM, Moy CS, Murray TG, Reynolds SM, Albert DM, Schachat AP, et al. The COMS randomized trial of iodine 125 brachytherapy for choroidal melanoma: IV. Local treatment failure and enucleation in the first 5 years after brachytherapy. COMS report no 19. Ophthalmology 2002;109:2197-206.
5Virgili G, Gatta G, Ciccolallo L, Capocaccia R, Biggeri A, Crocetti E, et al. Survival in patients with uveal melanoma in Europe. Arch Ophthalmol 2008;126:1413-8.
6Collaborative Ocular Melanoma Study Group. The COMS randomized trial of iodine 125 brachytherapy for choroidal melanoma: V. Twelve-year mortality rates and prognostic factors: COMS report No 28. Arch Ophthalmol 2006;124:1684-93.
7Diener-West M, Hawkins BS, Markowitz JA, Schachat AP. A review of mortality from choroidal melanoma. II. A meta-analysis of 5-year mortality rates following enucleation, 1966 through 1988. Arch Ophthalmol 1992;110:245-50.
8Diener-West M, Reynolds SM, Agugliaro DJ, Caldwell R, Cumming K, Earle JD, et al. Development of metastatic disease after enrollment in the COMS trials for treatment of choroidal melanoma: Collaborative Ocular Melanoma Study Group Report No 26. Arch Ophthalmol 2005;123:1639-43.
9Kujala E, Mäkitie T, Kivelä T. Very long-term prognosis of patients with malignant uveal melanoma. Invest Ophthalmol Vis Sci 2003;44:4651-9.
10Gamel JW, McLean IW, McCurdy JB. Biologic distinctions between cure and time to death in 2892 patients with intraocular melanoma. Cancer 1993;71:2299-305.
11Duh EJ, Schachat AP, Albert DM, Patel SM. Long-term survival in a patient with uveal melanoma and liver metastasis. Arch Ophthalmol 2004;122:285-7.
12Gragoudas ES, Egan KM, Seddon JM, Glynn RJ, Walsh SM, Finn SM, et al. Survival of patients with metastases from uveal melanoma. Ophthalmology 1991;98:383-9.
13Eschelman DJ, Gonsalves CF, Sato T. Transhepatic therapies for metastatic uveal melanoma. Semin Intervent Radiol 2013;30:39-48.
14Buder K, Gesierich A, Gelbrich G, Goebeler M. Systemic treatment of metastatic uveal melanoma: Review of literature and future perspectives. Cancer Med 2013;2:674-86.
15Gomez D, Wetherill C, Cheong J, Jones L, Marshall E, Damato B, et al. The Liverpool uveal melanoma liver metastases pathway: Outcome following liver resection. J Surg Oncol 2014;109:542-7.
16Yamamoto A, Chervoneva I, Sullivan KL, Eschelman DJ, Gonsalves CF, Mastrangelo MJ, et al. High-dose immunoembolization: Survival benefit in patients with hepatic metastases from uveal melanoma. Radiology 2009;252:290-8.
17Apt L. Uveal melanomas in children and adolescents. Int Ophthalmol Clin 1962;2:403-10.
18Paul EV, Parnell BL, Fraker M. Prognosis of malignant mela-nomas of the choroid and ciliary body. Int Ophthalmol Clin 1962;2:387-402.
19Barr CC, McLean IW, Zimmerman LE. Uveal melanoma in children and adolescents. Arch Ophthalmol 1981;99:2133-6.
20Shields CL, Shields JA, Milite J, De Potter P, Sabbagh R, Menduke H. Uveal melanoma in teenagers and children. A report of 40 cases. Ophthalmology 1991;98:1662-6.
21Pogrzebielski A, Or³owska-Heitzman J, Romanowska-Dixon B. Uveal melanoma in young patients. Graefes Arch Clin Exp Ophthalmol 2006;244:1646-9.
22Singh AD, Shields CL, Shields JA, Sato T. Uveal melanoma in young patients. Arch Ophthalmol 2000;118:918-23.
23Leonard BC, Shields JA, McDonald PR. Malignant melanomas of the uveal tract in children and young adults. Can J Ophthalmol 1975;10:441-9.
24Jensen OA. Malignant melanomas of the human uvea: 25-year follow-up of cases in Denmark, 1943--1952. Acta Ophthalmol (Copenh) 1982;60:161-82.
25Verdaguer J Jr. Prepuberal and puberal melanomas in ophthalmology. Am J Ophthalmol 1965;60:1002-11.
26Shields CL, Kaliki S, Furuta M, Mashayekhi A, Shields JA. Clinical spectrum and prognosis of uveal melanoma based on age at presentation in 8,033 cases. Retina 2012;32:1363-72.
27Kaliki S, Shields CL, Mashayekhi A, Ganesh A, Furuta M, Shields JA. Influence of age on prognosis of young patients with uveal melanoma: A matched retrospective cohort study. Eur J Ophthalmol 2013;23:208-16.
28Bergman L, Seregard S, Nilsson B, Lundell G, Ringborg U, Ragnarsson-Olding B. Uveal melanoma survival in Sweden from 1960 to 1998. Invest Ophthalmol Vis Sci 2003;44:3282-7.
29Zloto O, Pe'er J, Frenkel S. Gender differences in clinical presentation and prognosis of uveal melanoma. Invest Ophthalmol Vis Sci 2013;54:652-6.
30Rietschel P, Panageas KS, Hanlon C, Patel A, Abramson DH, Chapman PB. Variates of survival in metastatic uveal melanoma. J Clin Oncol 2005;23:8076-80.
31Holly EA, Aston DA, Ahn DK, Kristiansen JJ, Char DH. Uveal melanoma, hormonal and reproductive factors in women. Cancer Res 1991;51:1370-2.
32The Collaborative Ocular Melanoma Study (COMS) randomized trial of pre-enucleation radiation of large choroidal melanoma II: Initial mortality findings. COMS report no 10. Am J Ophthalmol 1998;125:779-96.
33Hawkins BS, Collaborative Ocular Melanoma Study Group. The Collaborative Ocular Melanoma Study (COMS) randomized trial of pre-enucleation radiation of large choroidal melanoma: IV. Ten-year mortality findings and prognostic factors. COMS report number 24. Am J Ophthalmol 2004;138:936-51.
34Shields CL, Furuta M, Thangappan A, Nagori S, Mashayekhi A, Lally DR, et al. Metastasis of uveal melanoma millimeter-by-millimeter in 8033 consecutive eyes. Arch Ophthalmol 2009;127:989-98.
35Damato B, Coupland SE. A reappraisal of the significance of largest basal diameter of posterior uveal melanoma. Eye (Lond) 2009;23:2152-60.
36Augsburger JJ, Gamel JW. Clinical prognostic factors in patients with posterior uveal malignant melanoma. Cancer 1990;66:1596-600.
37McLean IW, Foster WD, Zimmerman LE. Uveal melanoma: Location, size, cell type, and enucleation as risk factors in metastasis. Hum Pathol 1982;13:123-32.
38Shields CL, Kaliki S, Shah SU, Luo W, Furuta M, Shields JA. Iris melanoma: Features and prognosis in 317 children and adults. J AAPOS 2012;16:10-6.
39Prescher G, Bornfeld N, Hirche H, Horsthemke B, Jöckel KH, Becher R. Prognostic implications of monosomy 3 in uveal melanoma. Lancet 1996;347:1222-5.
40Seddon JM, Albert DM, Lavin PT, Robinson N. A prognostic factor study of disease-free interval and survival following enucleation for uveal melanoma. Arch Ophthalmol 1983;101:1894-9.
41Damato B, Duke C, Coupland SE, Hiscott P, Smith PA, Campbell I, et al. Cytogenetics of uveal melanoma: A 7-year clinical experience. Ophthalmology 2007;114:1925-31.
42Kivelä T. Iris melanomas in children. Arch Ophthalmol 2001;119:925-6.
43Gündüz K, Shields CL, Shields JA, Cater J, Freire JE, Brady LW. Plaque radiotherapy of uveal melanoma with predominant ciliary body involvement. Arch Ophthalmol 1999;117:170-7.
44Li W, Gragoudas ES, Egan KM. Metastatic melanoma death rates by anatomic site after proton beam irradiation for uveal melanoma. Arch Ophthalmol 2000;118:1066-70.
45Rummelt V, Folberg R, Woolson RF, Hwang T, Pe'er J. Relation between the microcirculation architecture and the aggressive behavior of ciliary body melanomas. Ophthalmology 1995;102:844-51.
46Font RL, Spaulding AG, Zimmerman LE. Diffuse malignant melanoma of the uveal tract: A clinicopathologic report of 54 cases. Trans Am Acad Ophthalmol Otolaryngol 1968;72:877-95.
47Rones B, Zimmerman LE. The production of heterochromia and glaucoma by diffuse malignant melanoma of the iris. Trans Am Acad Ophthalmol Otolaryngol 1957;61:447-63.
48Demirci H, Shields CL, Shields JA, Eagle RC Jr, Honavar SG. Diffuse iris melanoma: A report of 25 cases. Ophthalmology 2002;109:1553-60.
49Demirci H, Shields CL, Shields JA, Honavar SG, Eagle RC Jr. Ring melanoma of the ciliary body: Report on twenty-three patients. Retina 2002;22:698-706.
50Shields CL, Kaliki S, Furuta M, Shields JA. Diffuse versus nondiffuse small (=3 MM thickness) choroidal melanoma: Comparative analysis in 1,751 cases. The 2012 F. Phinizy Calhoun lecture. Retina 2013;33:1763-76.
51Parsons JH. Diffuse sarcomata of the uveal tract. Arch Ophthalmol 1904;33:101-12.
52Rones B, Zimmerman LE. The prognosis of primary tumors of the iris treated by iridectomy. AMA Arch Ophthalmol 1958;60:193-205.
53Shields CL, Shields JA, Materin M, Gershenbaum E, Singh AD, Smith A. Iris melanoma: Risk factors for metastasis in 169 consecutive patients. Ophthalmology 2001;108:172-8.
54Singh AD, De Potter P, Fijal BA, Shields CL, Shields JA, Elston RC. Lifetime prevalence of uveal melanoma in white patients with oculo (dermal) melanocytosis. Ophthalmology 1998;105:195-8.
55Shields CL, Kaliki S, Livesey M, Walker B, Garoon R, Bucci M, et al. Association of ocular and oculodermal melanocytosis with the rate of uveal melanoma metastasis: Analysis of 7872 consecutive eyes. JAMA Ophthalmol 2013;131:993-1003.
56Mashayekhi A, Kaliki S, Walker B, Park C, Sinha N, Kremer FZ, et al. Metastasis from uveal melanoma associated with congenital ocular melanocytosis: A matched study. Ophthalmology 2013;120:1465-8.
57Affeldt JC, Minckler DS, Azen SP, Yeh L. Prognosis in uveal melanoma with extrascleral extension. Arch Ophthalmol 1980;98:1975-9.
58Pach JM, Robertson DM, Taney BS, Martin JA, Campbell RJ, O'Brien PC. Prognostic factors in choroidal and ciliary body melanomas with extrascleral extension. Am J Ophthalmol 1986;101:325-31.
59Gündüz K, Shields CL, Shields JA, Cater J, Brady L. Plaque radiotherapy for management of ciliary body and choroidal melanoma with extraocular extension. Am J Ophthalmol 2000;130:97-102.
60Histopathologic characteristics of uveal melanomas in eyes enucleated from the Collaborative Ocular Melanoma Study. COMS report no 6. Am J Ophthalmol 1998;125:745-66.
61Coupland SE, Campbell I, Damato B. Routes of extraocular extension of uveal melanoma: Risk factors and influence on survival probability. Ophthalmology 2008;115:1778-85.
62Edge SB, Fritz AG, Byrd DR, Greene FL, Trotti III A, Compton CC, editors. AJCC Cancer Staging Manual. 7 th ed. New York: Springer; 2010. p. 547-59.
63Shields CL, Kaliki S, Peshtani A, Alarcon C, Fulco E, Komati R, et al. American Joint Committee on Cancer (AJCC) classification of iris melanoma is predictive of patient prognosis. Analysis of 452 patients. Submitted for publication.
64Shields CL, Kaliki S, Furuta M, Fulco E, Alarcon C, Shields JA. American Joint Committee on Cancer classification of posterior uveal melanoma (tumor size category) predicts prognosis in 7731 patients. Ophthalmology 2013;120:2066-71.
65Callender GR. Malignant melanotic tumors of the eye. A study of histologic types in 111 cases. Trans Am Acad Ophthalmol Otolaryngol 1931;36:131-40.
66McLean IW, Foster WD, Zimmerman LE, Gamel JW. Modifications of Callender's classification of uveal melanoma at the Armed Forces Institute of Pathology. Am J Ophthalmol 1983;96:502-9.
67Paul EV, Parnell BI, Fraker M. Prognosis of malignant melanomas of the choroid and ciliary body. Int Ophthalmol Clin 1962;2:387-402.
68Seregard S, Kock E. Prognostic indicators following enucleation for posterior uveal melanoma. A multivariate analysis of long-term survival with minimized loss to follow-up. Acta Ophthalmol Scand 1995;73:340-4.
69Seddon JM, Polivogianis L, Hsieh CC, Albert DM, Gamel JW, Gragoudas ES. Death from uveal melanoma. Number of epithelioid cells and inverse SD of nucleolar area as prognostic factors. Arch Ophthalmol 1987;105:801-6.
70Gamel JW, McLean IW, Foster WD, Zimmerman LE. Uveal melanomas: Correlation of cytologic features with prognosis. Cancer 1978;41:1897-901.
71McLean MJ, Foster WD, Zimmerman LE. Prognostic factors in small malignant melanomas of choroid and ciliary body. Arch Ophthalmol 1977;95:48-58.
72Pe'er J, Gnessin H, Shargal Y, Livni N. PC-10 immunostaining of proliferating cell nuclear antigen in posterior uveal melanoma. Enucleation versus enucleation postirradiation groups. Ophthalmology 1994;101:56-62.
73Seregard S, Lundell G, Lax I, af Trampe E, Kock E. Tumour cell proliferation after failed ruthenium plaque radiotherapy for posterior uveal melanoma. Acta Ophthalmol Scand 1997;75:148-54.
74Schilling H, Sehu KW, Lee WR. A histologic study (including DNA quantification and Ki-67 labeling index) in uveal melanomas after brachytherapy with ruthenium plaques. Invest Ophthalmol Vis Sci 1997;38:2081-92.
75Chiquet C, Grange JD, Ayzac L, Chauvel P, Patricot LM, Devouassoux-Shisheboran M. Effects of proton beam irradiation on uveal melanomas: A comparative study of Ki-67 expression in irradiated versus non-irradiated melanomas. Br J Ophthalmol 2000;84:98-102.
76Pe'er J, Stefani FH, Seregard S, Kivela T, Lommatzsch P, Prause JU, et al. Cell proliferation activity in posterior uveal melanoma after Ru-106 brachytherapy: An EORTC ocular oncology group study. Br J Ophthalmol 2001;85:1208-12.
77Karlsson M, Boeryd B, Carstensen J, Frånlund B, Gustafsson B, Kågedal B, et al. Correlations of Ki-67 and PCNA to DNA ploidy, S-phase fraction and survival in uveal melanoma. Eur J Cancer 1996;32A: 357-62.
78Mooy CM, Luyten GP, de Jong PT, Luider TM, Stijnen T, van de Ham F, et al. Immunohistochemical and prognostic analysis of apoptosis and proliferation in uveal melanoma. Am J Pathol 1995;147:1097-104.
79Al-Jamal RT, Mäkitie T, Kivelä T. Nucleolar diameter and microvascular factors as independent predictors of mortality from malignant melanoma of the choroid and ciliary body. Invest Ophthalmol Vis Sci 2003;44:2381-9.
80Huntington A, Haugan P, Gamel J, McLean I. A simple cytologic method for predicting the malignant potential of intraocular melanoma. Pathol Res Pract 1989;185:631-4.
81Moshari A, McLean IW. Uveal melanoma: Mean of the longest nucleoli measured on silver-stained sections. Invest Ophthalmol Vis Sci 2001;42:1160-3.
82McCurdy J, Gamel J, McLean I. A simple, efficient, and reproducible method for estimating the malignant potential of uveal melanoma from routine H and E slides. Pathol Res Pract 1991;187:1025-7.
83Sørensen FB, Gamel JW, McCurdy J. Stereologic estimation of nucleolar volume in ocular melanoma: A comparative study of size estimators with prognostic impact. Hum Pathol 1993;24:513-8.
84McLean IW, Sibug ME, Becker RL, McCurdy JB. Uveal melanoma: The importance of large nucleoli in predicting patient outcome - an automated image analysis study. Cancer 1997;79:982-8.
85Pe'er J, Rummelt V, Mawn L, Hwang T, Woolson RF, Folberg R. Mean of the ten largest nucleoli, microcirculation architecture, and prognosis of ciliochoroidal melanomas. Ophthalmology 1994;101:1227-35.
86Foss AJ, Alexander RA, Jefferies LW, Hungerford JL, Harris AL, Lightman S. Microvessel count predicts survival in uveal melanoma. Cancer Res 1996;56:2900-3.
87Mäkitie T, Summanen P, Tarkkanen A, Kivelä T. Microvascular density in predicting survival of patients with choroidal and ciliary body melanoma. Invest Ophthalmol Vis Sci 1999;40:2471-80.
88Silverman RH, Folberg R, Boldt HC, Lloyd HO, Rondeau MJ, Mehaffey MG, et al. Correlation of ultrasound parameter imaging with microcirculatory patterns in uveal melanomas. Ultrasound Med Biol 1997;23:573-81.
89Coleman DJ, Silverman RH, Rondeau MJ, Boldt HC, Lloyd HO, Lizzi FL, et al. Noninvasive in vivo detection of prognostic indicators for high-risk uveal melanoma: Ultrasound parameter imaging. Ophthalmology 2004;111:558-64.
90Mueller AJ, Bartsch DU, Folberg R, Mehaffey MG, Boldt HC, Meyer M, et al. Imaging the microvasculature of choroidal melanomas with confocal indocyanine green scanning laser ophthalmoscopy. Arch Ophthalmol 1998;116:31-9.
91Chen X, Maniotis AJ, Majumdar D, Pe'er J, Folberg R. Uveal melanoma cell staining for CD34 and assessment of tumor vascularity. Invest Ophthalmol Vis Sci 2002;43:2533-9.
92Folberg R, Pe'er J, Gruman LM, Woolson RF, Jeng G, Montague PR, et al. The morphologic characteristics of tumor blood vessels as a marker of tumor progression in primary human uveal melanoma: A matched case-control study. Hum Pathol 1992;23:1298-305.
93Mäkitie T, Summanen P, Tarkkanen A, Kivelä T. Microvascular loops and networks as prognostic indicators in choroidal and ciliary body melanomas. J Natl Cancer Inst 1999;91:359-67.
94Kivelä T, Mäkitie T, Al-Jamal RT, Toivonen P. Microvascular loops and networks in uveal melanoma. Can J Ophthalmol 2004;39:409-21.
95De la Cruz PO Jr, Specht CS, McLean IW. Lymphocytic infiltration in uveal malignant melanoma. Cancer 1990;65:112-5.
96Whelchel JC, Farah SE, McLean IW, Burnier MN. Immunohistochemistry of infiltrating lymphocytes in uveal malignant melanoma. Invest Ophthalmol Vis Sci 1993;34:2603-6.
97Bronkhorst IH, Vu TH, Jordanova ES, Luyten GP, Burg SH, Jager MJ. Different subsets of tumor-infiltrating lymphocytes correlate with macrophage influx and monosomy 3 in uveal melanoma. Invest Ophthalmol Vis Sci 2012;53:5370-8.
98Bronkhorst IH, Jager MJ. Uveal melanoma: The inflammatory microenvironment. J Innate Immun 2012;4:454-62.
99Mäkitie T, Summanen P, Tarkkanen A, Kivelä T. Tumor-infiltrating macrophages (CD68(+) cells) and prognosis in malignant uveal melanoma. Invest Ophthalmol Vis Sci 2001;42:1414-21.
100Toivonen P, Mäkitie T, Kujala E, Kivelä T. Macrophages and microcirculation in regressed and partially regressed irradiated choroidal and ciliary body melanomas. Curr Eye Res 2003;27:237-45.
101Bronkhorst IH, Ly LV, Jordanova ES, Vrolijk J, Versluis M, Luyten GP, et al. Detection of M2-macrophages in uveal melanoma and relation with survival. Invest Ophthalmol Vis Sci 2011;52:643-50.
102Maat W, Ly LV, Jordanova ES, de Wolff-Rouendaal D, Schalij-Delfos NE, Jager MJ. Monosomy of chromosome 3 and an inflammatory phenotype occur together in uveal melanoma. Invest Ophthalmol Vis Sci 2008;49:505-10.
103Vu TH, Bronkhorst IH, Versluis M, Marinkovic M, van Duinen SG, Vrolijk J, et al. Analysis of inflammatory cells in uveal melanoma after prior irradiation. Invest Ophthalmol Vis Sci 2013;54:360-9.
104Herwig MC, Grossniklaus HE. Role of macrophages in uveal melanoma. Expert Rev Ophthalmol 2011;6:405-07.
105All-Ericsson C, Girnita L, Seregard S, Bartolazzi A, Jager MJ, Larsson O. Insulin-like growth factor-1 receptor in uveal melanoma: A predictor for metastatic disease and a potential therapeutic target. Invest Ophthalmol Vis Sci 2002;43:1-8.
106Girnita A, All-Ericsson C, Economou MA, Aström K, Axelson M, Seregard S, et al. The insulin-like growth factor-I receptor inhibitor picropodophyllin causes tumor regression and attenuates mechanisms involved in invasion of uveal melanoma cells. Clin Cancer Res 2006;12:1383-91.
107Frenkel S, Zloto O, Pe'er J, Barak V. Insulin-like growth factor-1 as a predictive biomarker for metastatic uveal melanoma in humans. Invest Ophthalmol Vis Sci 2013;54:490-3.
108Cohen EP, Kim TS. Neoplastic cells that express low levels of MHC class I determinants escape host immunity. Semin Cancer Biol 1994;5:419-28.
109Jager MJ, Hurks HM, Levitskaya J, Kiessling R. HLA expression in uveal melanoma: There is no rule without some exception. Hum Immunol 2002;63:444-51.
110Blom DJ, Luyten GP, Mooy C, Kerkvliet S, Zwinderman AH, Jager MJ. Human leukocyte antigen class I expression. Marker of poor prognosis in uveal melanoma. Invest Ophthalmol Vis Sci 1997;38:1865-72.
111Ericsson C, Seregard S, Bartolazzi A, Levitskaya E, Ferrone S, Kiessling R, et al. Association of HLA class I and class II antigen expression and mortality in uveal melanoma. Invest Ophthalmol Vis Sci 2001;42:2153-6.
112Krishnakumar S, Abhyankar D, Lakshmi SA, Pushparaj V, Shanmugam MP, Biswas J. HLA expression in choroidal melanomas: Correlation with clinicopathological features. Curr Eye Res 2004;28:409-16.
113Jager MJ, de Wolff-Rouendaal D, Breebaart AC, Ruiter DJ. Expression of HLA antigens in paraffin sections of uveal melanomas. Doc Ophthalmol 1986;64:69-76.
114White VA, Chambers JD, Courtright PD, Chang WY, Horsman DE. Correlation of cytogenetic abnormalities with the outcome of patients with uveal melanoma. Cancer 1998;83:354-9.
115Horsman DE, White VA. Cytogenetic analysis of uveal melanoma. Consistent occurrence of monosomy 3 and trisomy 8q. Cancer 1993;71:811-9.
116Sisley K, Rennie IG, Cottam DW, Potter AM, Potter CW, Rees RC. Cytogenetic findings in six posterior uveal melanomas: Involvement of chromosomes 3, 6, and 8. Genes Chromosomes Cancer 1990;2:205-9.
117Sisley K, Rennie IG, Parsons MA, Jacques R, Hammond DW, Bell SM, et al. Abnormalities of chromosomes 3 and 8 in posterior uveal melanoma correlate with prognosis. Genes Chromosomes Cancer 1997;19:22-8.
118Scholes AG, Damato BE, Nunn J, Hiscott P, Grierson I, Field JK. Monosomy 3 in uveal melanoma: Correlation with clinical and histologic predictors of survival. Invest Ophthalmol Vis Sci 2003;44:1008-11.
119Damato B, Dopierala J, Klaasen A, van Dijk M, Sibbring J, Coupland SE. Multiplex ligation-dependent probe amplification of uveal melanoma: Correlation with metastatic death. Invest Ophthalmol Vis Sci 2009;50:3048-55.
120Kilic E, Naus NC, van Gils W, Klaver CC, van Til ME, Verbiest MM, et al. Concurrent loss of chromosome arm 1p and chromosome 3 predicts a decreased disease-free survival in uveal melanoma patients. Invest Ophthalmol Vis Sci 2005;46:2253-7.
121Häusler T, Stang A, Anastassiou G, Jöckel KH, Mrzyk S, Horsthemke B, et al. Loss of heterozygosity of 1p in uveal melanomas with monosomy 3. Int J Cancer 2005;116:909-13.
122Shields CL, Ganguly A, Bianciotto CG, Turaka K, Tavallali A, Shields JA. Prognosis of uveal melanoma in 500 cases using genetic testing of fine-needle aspiration biopsy specimens. Ophthalmology 2011;118:396-401.
123Werdich XQ, Jakobiec FA, Singh AD, Kim IK. A review of advanced genetic testing for clinical prognostication in uveal melanoma. Semin Ophthalmol 2013;28:361-71.
124Tschentscher F, Prescher G, Horsman DE, White VA, Rieder H, Anastassiou G, et al. Partial deletions of the long and short arm of chromosome 3 point to two tumor suppressor genes in uveal melanoma. Cancer Res 2001;61:3439-42.
125White VA, McNeil BK, Thiberville L, Horsman DE. Acquired homozygosity (isodisomy) of chromosome 3 during clonal evolution of a uveal melanoma: Association with morphologic heterogeneity. Genes Chromosomes Cancer 1996;15:138-43.
126Schoenfield L, Pettay J, Tubbs RR, Singh AD. Variation of monosomy 3 status within uveal melanoma. Arch Pathol Lab Med 2009;133:1219-22.
127Harbour JW, Onken MD, Roberson ED, Duan S, Cao L, Worley LA, et al. Frequent mutation of BAP1 in metastasizing uveal melanomas. Science 2010;330:1410-3.
128Kilic E, van Gils W, Lodder E, Beverloo HB, van Til ME, Mooy CM, et al. Clinical and cytogenetic analyses in uveal melanoma. Invest Ophthalmol Vis Sci 2006;47:3703-7.
129Damato B, Dopierala JA, Coupland SE. Genotypic profiling of 452 choroidal melanomas with multiplex ligation-dependent probe amplification. Clin Cancer Res 2010;16:6083-92.
130Onken MD, Worley LA, Harbour JW. A metastasis modifier locus on human chromosome 8p in uveal melanoma identified by integrative genomic analysis. Clin Cancer Res 2008;14:3737-45.
131Parrella P, Sidransky D, Merbs SL. Allelotype of posterior uveal melanoma: Implications for a bifurcated tumor progression pathway. Cancer Res 1999;59:3032-7.
132Ehlers JP, Worley L, Onken MD, Harbour JW. Integrative genomic analysis of aneuploidy in uveal melanoma. Clin Cancer Res 2008;14:115-22.
133Aalto Y, Eriksson L, Seregard S, Larsson O, Knuutila S. Concomitant loss of chromosome 3 and whole arm losses and gains of chromosome 1, 6, or 8 in metastasizing primary uveal melanoma. Invest Ophthalmol Vis Sci 2001;42:313-7.
134Tschentscher F, Hüsing J, Hölter T, Kruse E, Dresen IG, Jöckel KH, et al. Tumor classification based on gene expression profiling shows that uveal melanomas with and without monosomy 3 represent two distinct entities. Cancer Res 2003;63:2578-84.
135Onken MD, Worley LA, Ehlers JP, Harbour JW. Gene expression profiling in uveal melanoma reveals two molecular classes and predicts metastatic death. Cancer Res 2004;64:7205-9.
136van Gils W, Lodder EM, Mensink HW, Kiliç E, Naus NC, Brüggenwirth HT, et al. Gene expression profiling in uveal melanoma: Two regions on 3p related to prognosis. Invest Ophthalmol Vis Sci 2008;49:4254-62.
137Petrausch U, Martus P, Tönnies H, Bechrakis NE, Lenze D, Wansel S, et al. Significance of gene expression analysis in uveal melanoma in comparison to standard risk factors for risk assessment of subsequent metastases. Eye (Lond) 2008;22:997-1007.
138Onken MD, Worley LA, Tuscan MD, Harbour JW. An accurate, clinically feasible multi-gene expression assay for predicting metastasis in uveal melanoma. J Mol Diagn 2010;12:461-8.
139Chappell MC, Char DH, Cole TB, Harbour JW, Mishra K, Weinberg VK, et al. Uveal melanoma: Molecular pattern, clinical features, and radiation response. Am J Ophthalmol 2012;154:227-232.e2.
140Onken MD, Worley LA, Char DH, Augsburger JJ, Correa ZM, Nudleman E, et al. Collaborative Ocular Oncology Group report number 1: Prospective validation of a multi-gene prognostic assay in uveal melanoma. Ophthalmology 2012;119:1596-603.