ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Initial management of uveal and conjunctival melanomas

Initial management of uveal and conjunctival melanomas
Literature review current through: Jan 2024.
This topic last updated: Aug 31, 2022.

INTRODUCTION — Approximately 85 percent of ocular melanomas arise from the uveal tract (including the iris, ciliary body, and choroid), and the remainder arise in the conjunctiva or (rarely) the orbit. There are significant clinical and genomic differences between melanomas of the uvea, conjunctiva, skin, and other sites.

The initial management of uveal and conjunctival melanoma is discussed in this section. The management of metastatic uveal melanoma is discussed separately. (See "Metastatic uveal melanoma".)

UVEAL MELANOMA — Uveal melanomas can arise in any part of the uveal tract, including the iris, ciliary body, and choroid (figure 1). Approximately 95 percent of uveal melanomas arise from the ciliary body and/or choroid, and approximately 5 percent arise in the iris.

Natural history of uveal melanocytic tumors

Choroidal and ciliary body tumors — Most choroidal and ciliary body melanomas are thought to arise de novo, but they can occasionally develop from preexisting nevi. Research has identified factors that can predict which nevi are most likely to progress to melanoma. Most of these risk factors for choroidal nevus growth were first described in the 1970s [1]. Subsequent studies have validated these factors and have suggested additional potential risk factors [2-5].

The following clinical features are the most commonly used to predict which choroidal nevi are most likely to grow:

Tumor thickness greater than 2 mm by ultrasound

Serous subretinal fluid overlying and/or surrounding the tumor

Orange lipofuscin pigment clumps overlying the tumor

Tumor location less than 3 mm from the optic disc

Low internal reflectivity on ultrasound, or acoustic "hollowness"

Absence of chronic features such as retinal pigment epithelial drusen, fibrosis, and atrophy

Symptoms attributable to the tumor (eg, visual blurring, distortion, flashes, and/or floaters)

Importantly, the presence of these features and documented tumor growth is not always indicative of malignant transformation [6,7] or of high-risk genetic features [8]. Thus, these features should only be used as management guidelines, and caution is recommended when making clinical decisions based on these features alone, especially if growth is minimal or slow.

Iris tumors — Melanoma of the iris is rare, representing only approximately 5 percent of uveal melanomas [9]. By contrast, iris nevi are relatively common. However, the rate of transformation of iris nevus into melanoma is very low, with an actuarial risk of only approximately 5 percent at 10 years [10]. Risks factors associated with malignancy include larger tumor size, intratumoral vascularity, invasion through the anterior chamber angle to the ciliary body, increased intraocular pressure, and secondary glaucoma [10,11].

Prognosis is much better for melanomas of the iris than for melanomas of the ciliary body and choroid. Earlier diagnosis due to their readily visible location may contribute to this improved prognosis. Because of the less aggressive nature of iris melanomas, these tumors are often managed with close monitoring, although large or fast-growing tumors may require treatment with brachytherapy, proton beam radiation therapy (RT), local excision, or enucleation.

Risk factors — Epidemiologic factors that may increase the risk of uveal melanoma include:

Host pigmentation factors – Light eye color, fair skin color, and propensity to sunburn may confer increased risk for uveal melanoma [12].

Cutaneous and iris nevi – Cutaneous nevi, cutaneous freckles, and iris nevi have been associated with an increased risk of uveal melanoma [13,14].

Ultraviolet light exposure – The role of ultraviolet exposure is unclear. If ultraviolet light is a risk factor for uveal melanoma, its contribution is much weaker than for cutaneous melanoma.

Epidemiologic studies investigating a potential association between ultraviolet light and uveal melanoma have yielded mixed results [15,16]. A meta-analysis of 12 studies found a borderline or nonsignificant association between uveal melanoma and leisure sun exposure, occupational sun exposure, and latitude of birth [17]. Arc welding was found to be a significant risk factor, but it is unclear whether this association can be attributed to ultraviolet light exposure. Next-generation sequencing of primary uveal melanomas does not reveal an ultraviolet-light-associated mutation signature [18].

Ocular/oculodermal melanocytosis – Ocular/oculodermal melanocytosis is the condition that most strongly predisposes to uveal melanoma, with a lifetime risk of approximately 1 in 400 of developing uveal melanoma [19]. Patients with this condition should undergo periodic ophthalmic examination to rule out uveal melanoma.

Clinical presentation — Uveal melanomas often present without symptoms, being discovered on a routine eye exam. Approximately one-half of patients will present with visual symptoms such as flashes, floaters, or visual field defects.

Diagnosis of the primary tumor — The diagnosis of uveal melanoma is based upon funduscopic examination by an experienced clinician, which is followed by further characterization with specialized noninvasive testing techniques, such as ultrasound, optical coherence tomography, and fluorescein angiography. The most common simulating lesion in the differential diagnosis of uveal melanoma is a uveal nevus, and the two cannot always be distinguished with certainty on clinical examination due to an overlap in size between small melanomas and large nevi [20]. The differential diagnosis of uveal melanoma also includes metastasis to the uvea, especially from lung and breast cancers, which can be the first manifestation of an occult primary tumor [21].

The funduscopic and ultrasonographic features of uveal tumors usually allow the diagnosis to be determined noninvasively. Magnetic resonance imaging (MRI) of the orbit may occasionally be needed to confirm the diagnosis and may also be indicated in patients with tumors that are large, in proximity to nerves, or are suspicious for extraocular involvement. In approximately 5 percent of cases, however, a diagnostic biopsy is needed to distinguish an atypical uveal melanoma from a metastatic tumor, hemorrhagic lesion, or other simulating lesion.

Fine needle aspiration biopsy is now performed in the vast majority of patients with uveal melanoma for molecular prognostic testing, which can be used to customize metastatic surveillance testing and to stratify high-risk patients into clinical trials. The molecular characteristics of the tumor may influence the choice of initial management in selected cases. (See "The molecular biology of melanoma", section on 'Uveal melanomas' and 'Posttreatment systemic surveillance' below.)

Staging of distant (extraocular) disease — The American Joint Committee on Cancer (AJCC) eighth edition tumor, node, metastasis (TNM) staging system is used to stage patients with uveal melanoma (table 1 and table 2).

It is rare for patients diagnosed with uveal melanoma to have detectable metastatic disease at the time of primary tumor diagnosis. For most patients, we offer baseline imaging to assess for distant (extraocular) metastatic disease prior to treating the primary tumor. However, patients with small tumors at low risk for distant metastases may be offered systemic imaging after treatment of the primary lesion. We image the liver using MRI, given the propensity of patients with uveal melanoma to develop hepatic metastases. Alternative options include ultrasound of the liver or computed tomography (CT). However, in patients without evidence of liver metastases, extrahepatic disease is uncommon. Therefore, initial staging for extrahepatic disease using CT of the chest, abdomen, and pelvis may be omitted unless liver metastases are confirmed or the patient is experiencing relevant symptoms. (See "Metastatic uveal melanoma", section on 'Clinical presentation'.)

Prognosis — Local treatment for primary uveal melanoma is effective in preventing local recurrence in over 95 percent of cases, yet up to 50 percent of patients are at risk for metastatic disease. The high risk of metastatic disease is thought to be due to a propensity for early micrometastasis followed by a variable latency period prior to the emergence of overt metastatic disease [22].

The clinical, histopathologic, and molecular features that determine prognosis in uveal melanoma are as follows:

Clinical and histopathologic features — In patients with uveal melanoma, the clinical and histopathologic features of the primary tumor that are associated with poor prognosis include increased patient age, largest basal diameter of the tumor, ciliary body involvement, extrascleral tumor extension, epithelioid cell type, and vasculogenic mimicry patterns [23,24]. The AJCC utilizes several of these prognostic factors for its TNM staging system (table 1 and table 2) [25]. However, the eighth edition TNM system for uveal melanoma is cumbersome and appears to be prognostically equivalent to simply using the largest basal diameter of the tumor [26]. Future simplifications in the system may render it more practical for routine clinical use.

Molecular features — Advances in understanding the molecular pathogenesis of uveal melanoma are providing important information regarding prognosis. Additional studies are required to validate these observations. (See "The molecular biology of melanoma", section on 'Uveal melanomas'.)

Chromosomal markers – Tumor chromosomal markers (eg, monosomy 3 and 8q gain) have some utility for predicting prognosis [27], but these markers are subject to sampling error due to tumor heterogeneity [28,29], and methods to detect them vary in their accuracy [30].

Gene expression profiling – Gene expression profiling (GEP) has been shown to be superior to chromosomal markers, as well as clinical and histopathological prognostic factors, for defining groups at high risk for the development of metastatic disease [31-33]. Using primary uveal melanoma samples obtained by fine needle biopsy, GEP classifies tumors as having low (class 1) or high (class 2) metastatic potential depending on the expression of 12 discriminating genes and three control genes [34]. This profile test has been validated in 459 patients in a prospective multicenter study that showed it to be a better prognostic marker than monosomy 3 and the TNM staging system [31].

The use of gene expression profiling to determine posttreatment systemic surveillance in patients with treated uveal melanoma is discussed below. (See 'Posttreatment systemic surveillance' below.)

Circulating tumor cells/DNA – Detection of circulating tumor cells or circulating tumor DNA may be a risk factor for metastasis and shortened survival in patients with uveal melanoma [35,36].

Management — Management of primary uveal melanoma is guided by many factors, including the size and location of the tumor, presence of extraocular extension, visual potential, patient age and preference, and presence or absence of metastases.

Observation — For asymptomatic patients with small uveal melanocytic tumors (<12 mm in diameter and <2 to 3 mm in height), initial management is often observation for evidence of growth, rather than immediate intervention [37]. (See 'Natural history of uveal melanocytic tumors' above.)

When observation is chosen, initial follow-up at two- to four-month intervals is typical. Imaging modalities commonly used in monitoring a small suspicious uveal melanocytic tumor include fundus photography, ultrasonography, optical coherence tomography, and fundus autofluorescence to identify evidence of tumor growth, subretinal fluid, orange lipofuscin pigmentation, and other risk factors for malignant transformation [8,38].

Radiation therapy — RT is the most common treatment for primary uveal melanoma [3]. Since uveal melanomas are relatively radioresistant, they must be treated with high-dose radiation, usually in the form of plaque brachytherapy or charged-particle RT.

Plaque brachytherapy — Plaque brachytherapy is the most common form of RT used in the treatment of uveal melanoma worldwide. Several radioisotopes have been used, including cobalt-60 (60Co), iodine-125 (125I), iridium-192 (192Ir), palladium-103 (103Pd), and ruthenium-106/rhodium-106 (106Ru/106Rh). The most commonly used isotope is 125I because of its favorable dosimetric characteristics across a broad range of tumor sizes (picture 1) [39] and because of its greater manufacturing accessibility with regards to application in the United States. 106Ru/106Rh is favored by some centers in the treatment of small tumors [40], but this isotope is associated with an increased rate of local treatment failure in larger tumors [41].

Earlier studies reported higher local recurrence rates with brachytherapy compared with charged-particle RT [42], but with the use of intraoperative ultrasonography for plaque localization [43-45], the local recurrence rates with plaque brachytherapy have decreased substantially and are now similar to charged-particle RT [46,47]. These findings suggest that most local recurrences after plaque brachytherapy are due to suboptimal plaque localization and undetected plaque tilting, which can be detected and corrected with intraoperative ultrasonography at plaque insertion and removal [48]. This technique is particularly important for tumors located posteriorly in the eye, where surgical access can be challenging and where the plaque is most prone to tilt [48]. Local tumor recurrence following primary RT is a risk factor for metastasis [49,50]. The importance of local control was illustrated by a study in which cause-specific survival at 10 years was 72 percent for patients with local control following proton beam RT versus 48 percent for those with local recurrence [51].

Consensus opinion guidelines for the use of radioactive plaque therapy have been published by the American Brachytherapy Society (ABS) [52], but research is ongoing into the optimal dosimetric parameters for uveal melanoma [53].

Charged-particle radiation therapy — Charged-particle RT (protons, carbon ions, helium ions) is the second most common form of RT used to treat uveal melanoma [42,54-56]. The physical properties of charged particles allow increased dose targeting at the end of the beam range and a sharp decrease in the dose of the radiation beam beyond the targeted area (the Bragg peak effect) (figure 2) [57]. Nevertheless, charged-particle RT can result in collateral damage to ocular structures such as the lashes, lacrimal gland, cornea, iris, lens, retina, and optic nerve. (See 'Ocular complications of radiation therapy' below.)

In most circumstances, plaque brachytherapy and charged-particle RT render very similar local control rates [58]. Associated ocular radiation complications are slightly different, with greater anterior eye complications with charged-particle RT and with greater visual acuity loss and immediate procedural discomfort with plaque brachytherapy. One circumstance in which charged-particle RT has an advantage over plaque brachytherapy is when the melanoma is encircling the optic nerve in a so-called "circumpapillary" configuration, where it is not possible to place a plaque completely around the tumor. Charged-particle RT also has dosimetric advantages when the tumor basal dimensions are too large to accommodate a plaque. Normally, such large tumors are treated with enucleation, but eye preservation may be desired in occasional situations, especially for patients with poor vision in the contralateral eye.

Photon stereotactic radiation therapy — Linear-accelerator-adapted stereotactic RT has also been used to treat uveal melanomas [59-61]. The total dose is typically between 50 and 70 Gy, delivered in five daily fractions.

While long-term follow-up data for photon stereotactic RT are more limited than for other radiation modalities, the local control and distant metastatic disease rates appear to be similar. However, the complication rate may be higher than for plaque brachytherapy, with approximately two-thirds of patients developing ocular complications within five years after treatment [62,63].

Ocular complications of radiation therapy — All types of RT for uveal melanoma can be associated with ocular complications, including dry eye, cataracts, neovascular glaucoma, vitreous hemorrhage, exudative retinal detachment, uveitis, scleral necrosis, radiation retinopathy, and optic neuropathy [64]. Some complications are more specific to charged-particle and photon stereotactic RT, including lash loss and lacrimal gland dysfunction [65,66].

The severity of ocular radiation damage is related to the increased total radiation dose and dose per fraction, and it may be increased in patients with diabetes or hypertension, or with previous chemotherapy [67]. The threshold dose for developing ocular radiation damage varies by tissue and is approximately 30 to 35 Gy for the retina and optic disc.

Early clinical signs of radiation retinopathy include macular edema and ischemia, which may be demonstrable using optical coherence tomography prior to becoming funduscopically detectable [68,69]. Later manifestations include microaneurysms, telangiectasia, hard (lipid) exudates, and cotton wool spots. Retinal neovascularization and vitreous hemorrhage are more severe but less common manifestations of radiation retinopathy. Radiation optic neuropathy presents initially with optic disc swelling and hemorrhages, and later evolves to optic atrophy with disc pallor.

Risk factors for visually significant radiation retinopathy and optic neuropathy include proximity of the tumor to the optic disc and fovea, increased tumor thickness, tumor-associated retinal detachment, and diabetes [70,71].

The radiation complication most commonly resulting in secondary enucleation is neovascular glaucoma, wherein neovascularization of the iris is caused by the elaboration of angiogenic factors by an ischemic and/or chronically detached retina, leading to closure of the anterior chamber angle and increased intraocular pressure. One study of patients treated with charged-particle RT reported five-year rates of neovascular glaucoma and neovascular-glaucoma-associated enucleation of 12.7 and 4.9 percent, respectively [72]. Risk factors for neovascular glaucoma include larger tumor diameter and thickness, increased patient age, chronic retinal detachment, and greater tumor vascularity [72-74].

Treatment of ocular radiation complications — Radiation-induced cataracts and dry eye can be managed by conventional ophthalmic means [75].

The management of radiation retinopathy, optic neuropathy, and neovascular glaucoma has been more challenging. Radiation causes endothelial cell loss and capillary closure, leading to retinal ischemia and the elaboration of vascular endothelial growth factor (VEGF) and other pro-angiogenic factors [76,77]. Attempted treatments have included photodynamic therapy, laser photocoagulation, oral pentoxifylline, hyperbaric oxygen, periocular or intravitreal injection of corticosteroids, and intravitreal injection of anti-VEGF agents such as bevacizumab, ranibizumab, and aflibercept [78-80].

While none of these treatments is curative or preventative, promising results have been observed for intravitreal anti-VEGF therapy in maintaining or improving visual function in some patients with radiation maculopathy [81]. Intraocular anti-VEGF therapy, often combined with panretinal laser photocoagulation to reduce the production of pro-angiogenic factors from an ischemic retina, has also improved the management of neovascular glaucoma and decreased the rate of secondary enucleation [82]. The risk of neovascular glaucoma can also be reduced by prompt surgical repair of postradiation retinal detachment using vitrectomy techniques [83].

Other noninvasive techniques

Transpupillary thermotherapy — Transpupillary thermotherapy (TTT), also called diode laser hyperthermia, combines three features to induce deep-penetrating tumor necrosis: a modified infrared diode laser, typically at a wavelength of 810 nanometers, that allows deep tissue penetration; a large spot size, which decreases the risk of acute burns to the retina and tumor; and a low-energy, long-duration technique that allows the entire tumor to gradually heat up to approximately 45 to 60°C [84]. Despite these improvements over laser photocoagulation, TTT is only used as a primary treatment in tumors up to approximately 3 mm in thickness, and it is associated with significant complications, such as retinal vascular occlusions, macular edema, epiretinal membrane, vitreous hemorrhage, and retinal detachment [85]. Further, the local recurrence rate is around 30 percent after three years [86].

Consequently, TTT is used today only in highly selected patients for primary treatment of uveal melanoma. By contrast, TTT is often valuable in selected patients as an adjunct after RT to reduce the risk of local tumor recurrence [46]. Further, TTT is often useful at lower energy levels as a nonablative therapy in small borderline uveal melanocytic tumors producing symptomatic subretinal fluid [87].

Photodynamic therapy — Photodynamic therapy comprises intravenous injection of a photosensitive compound followed by light application at a wavelength that activates the photosensitizer, thereby forming singlet oxygen and free radicals that damage endothelial cells and lead to vascular occlusion and tumor necrosis [88]. Photodynamic therapy is not effective in larger tumors and is not generally used as an ablative primary treatment for uveal melanoma. However, it can be useful for drying up subretinal fluid in small, symptomatic macular tumors [89].

Laser photocoagulation — Laser photocoagulation uses high-temperature thermal energy to damage the tumor and its vasculature. However, photocoagulation is associated with visually significant adverse effects, including retinal vascular occlusions, vitreous hemorrhage, and retinal detachment. Furthermore, local recurrences are common due to the inability of this method to destroy tumor cells below the surface. Consequently, photocoagulation is not a common treatment for uveal melanoma in most centers.

Surgery

Local tumor resection — External or internal resection of a uveal melanoma without removing the entire eye is technically challenging, may require risky hypotensive anesthesia, and often results in serious and immediate postoperative complications, such as vitreous hemorrhage, rhegmatogenous retinal detachment, and proliferative vitreoretinopathy [90]. Additionally, local tumor recurrence in the eye and/or orbit is more common with these techniques than with more conventional forms of treatment.

For these reasons, and because modern RT techniques offer a reasonable, if not superior, alternative, enthusiasm for local resection has waned in recent years. Surgeons continuing to use external or internal resection of a uveal melanoma will now often use adjuvant plaque brachytherapy or charged-particle RT in an effort to reduce the risk of recurrence.

Enucleation — Until the 1970s, enucleation was the standard of care for uveal melanoma. However, retrospective studies comparing enucleation with RT did not show a survival advantage for enucleation [91-93].

In a large prospective clinical trial, the Collaborative Ocular Melanoma Study (COMS) compared 1317 patients with medium-sized choroidal melanoma randomly assigned to enucleation versus 125I brachytherapy with overall survival as the primary outcome measure [94]. Five-, 10-, and 12-year rates of death with a histopathologically confirmed melanoma metastasis were 10, 18, and 21 percent, respectively, in the 125I brachytherapy arm and 11, 17, and 17 percent, respectively, in the enucleation arm, and there was no statistically significant difference in survival between the two groups. No comparable study has been performed for charged-particle RT, but it is generally assumed that the outcome would be similar.

Enucleation is now generally reserved for patients who would not be expected to have a favorable outcome with RT, such as those with large tumors, marked extrascleral extension, neovascular glaucoma, extensive retinal detachment, or poor visual potential, or for select patients who prefer enucleation. Fortunately, the quality of life for patients undergoing enucleation appears to be similar to that for those treated with RT [95]. The COMS also confirmed that there is no demonstrable benefit to pre-enucleation RT to the eye and orbit for patients with large uveal melanomas [96].

Posttreatment systemic surveillance — Despite effective treatment of the primary site, many patients with uveal melanoma remain at risk for developing metastatic disease. Therefore, long-term surveillance is necessary to detect late recurrences [54]. (See "Metastatic uveal melanoma", section on 'Clinical presentation'.)

The standard of care in most centers is use molecular prognostic testing to risk-stratify patients and guide surveillance for metastatic disease. Molecular testing is usually obtained at the time of primary tumor treatment based on a fine needle biopsy, among other clinical tumor characteristics [97]. Patients are stratified based on low, medium, and high risk of metastatic disease based on the following criteria. (See "The molecular biology of melanoma", section on 'Uveal melanomas' and 'Prognosis' above.)

Low risk – Class 1A metastatic potential [31], disomy 3, gain of chromosome 6p, EIF1AX mutation, AJCC T1 disease (table 1 and table 2).

Medium risk – Class 1B metastatic potential, SF3B1 mutation, AJCC T2 and T3 disease.

High risk – Class 2 metastatic potential, monosomy 3, gain chromosome 8q, BAP1 mutation, PRAME expression [98], AJCC T4 disease, extraocular extension, ciliary body involvement.

Systemic surveillance imaging for metastasis typically focuses on the liver, the most common site of metastatic disease [99]. While we prefer surveillance with MRI of the liver, other options include abdominal ultrasound for low-risk patients or CT. For all patients, we offer such systemic imaging to evaluate any signs or symptoms of metastatic disease as clinically indicated.

For patients with low-risk tumors, we offer systemic imaging annually.

For those with medium-risk tumors, we offer systemic imaging every 6 to 12 months for 10 years, then as clinically indicated.

For those with high-risk tumors, we offer systemic imaging every 3 to 6 months for years 1 through 5; every 6 to 12 months for years 6 through 10; then as clinically indicated. Clinical trial participation is strongly encouraged in patients with high-risk disease.

The clinical presentation, diagnosis, and treatment of patients who develop metastatic disease while on surveillance is discussed separately. (See "Metastatic uveal melanoma".)

CONJUNCTIVAL MELANOMA

Clinical presentation — Conjunctival melanomas arise from melanocytes located in the basal layer of the conjunctival epithelium, and they represent approximately 5 percent of all ocular melanomas (figure 3) [100]. Risk factors include primary acquired melanosis (PAM) and preexisting conjunctival nevi. Approximately two-thirds of conjunctival melanomas arise from PAM, 16 to 25 percent de novo, and 1 to 6 percent from nevi [101,102]. The incidence of conjunctival melanoma is increasing, suggesting a possible association with ultraviolet light exposure [101,103]. Consistent with this possibility, an ultraviolet mutation signature was identified in conjunctival melanoma samples by whole-exome sequencing [104].

Molecular pathogenesis — The molecular pathogenesis of conjunctival melanoma is distinct from that of uveal melanoma and is more similar to cutaneous and mucosal melanoma. (See "The molecular biology of melanoma", section on 'MAPK pathway'.)

The most common mutations in cutaneous melanoma include BRAF, NRAS, NF1, EGFR, ALK, TERT, and APC, which are rare in uveal melanoma [104]. Common mutations in uveal melanoma, such as GNAQ, GNA11, BAP1, SF3B1, and EIF1AX, are lacking in most conjunctival melanomas. BRAF mutations are found in approximately 35 percent of conjunctival melanomas and are associated with sun-exposed tumor location (bulbar conjunctiva or caruncle), preexisting nevus, and absence of PAM [105]. Gain of chromosome 6p appears to be the most common chromosomal change [104].

Primary treatment — Conjunctival melanoma is typically treated with wide local surgical excision, followed by double freeze-thaw cryotherapy to the margins and possibly alcohol application [100]. Adjunctive plaque radiation therapy after resection has been used successfully [106]. Incisional biopsy and direct manipulation of the tumor should be avoided to prevent tumor cell seeding. Posterior tumor extension may require orbital exenteration [107].

Metastatic disease — The most common sites of metastasis include the regional lymph nodes, lung, liver, skin, and brain [102,108]. In one large series from a single country, tumor-related survival was 86.3 percent at five years, 72 percent at 10 years, and 67 percent at 15 years [102]. Some reports suggest that disseminated conjunctival melanoma may be responsive to targeted molecular therapies, such as BRAF and MEK inhibitors in BRAF-mutant tumors [109], and checkpoint inhibitor immunotherapeutic agents, such as pembrolizumab [110]. Further research is necessary to confirm these findings.

Prognosis — Clinical prognostic factors associated with poor prognosis include extrabulbar location (palpebral conjunctiva, caruncle, plica, or fornices), tumor thickness exceeding 2 mm, origin of the tumor (PAM and de novo versus nevus), involvement of adjacent tissue structures, and local tumor recurrence [102,105,111]. Histopathologic risk factors for tumor-related mortality include tumor infiltration beyond the substantia propria, incomplete surgical excision, and nodular or mixed (nodular and superficial) growth pattern [112]. Conjunctival melanoma can be staged according to the tumor, node, metastasis (TNM) system of the American Joint Committee on Cancer (AJCC) (table 3), which includes most of these risk factors [25].

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Melanoma screening, prevention, diagnosis, and management".)

SUMMARY AND RECOMMENDATIONS

Management of small, asymptomatic uveal melanoma – For asymptomatic patients with small uveal melanocytic tumors (<12 mm in diameter and <2 to 3 mm in thickness), an initial period of observation for evidence of growth is generally recommended until evidence of growth is documented. (See 'Natural history of uveal melanocytic tumors' above.)

Management of larger, symptomatic uveal melanoma – For patients with larger tumors and for those with symptoms, initial treatment is usually indicated:

For symptomatic patients and those with medium or large tumors, treatment with radiation therapy (RT) is generally recommended. RT can be administered using plaque brachytherapy, the most widely available form of RT, or with charged-particle RT. (See 'Radiation therapy' above and 'Enucleation' above.)

Enucleation is generally reserved for patients in whom RT is unlikely to achieve adequate local tumor control or is likely to result in unacceptable ocular radiation complications due to large tumor size, extrascleral extension, or risk of neovascular glaucoma. (See 'Enucleation' above.)

Posttreatment surveillance for uveal melanoma – After treatment of the primary tumor is completed, most centers use molecular prognostic testing to risk-stratify patients and guide surveillance for metastatic disease. (See 'Posttreatment systemic surveillance' above.)

Management of metastatic uveal melanoma – For patients with metastatic disease, treatment options include liver-directed regional therapies, systemic targeted therapies such as tebentafusp, and immunotherapy. Clinical trial participation is strongly encouraged. (See "Metastatic uveal melanoma".)

Initial management of conjunctival melanoma – For patients with conjunctival melanoma, initial management focuses upon wide local surgical excision, supplemented by cryotherapy and possibly alcohol application. (See 'Conjunctival melanoma' above.)

Management of metastatic conjunctival melanoma – The management of metastatic conjunctival melanoma differs from that of uveal melanoma, is based upon molecular pathogenesis, and may include targeted molecular therapy and immunotherapy. (See "The molecular biology of melanoma", section on 'Conjunctival melanoma' and "Overview of the management of advanced cutaneous melanoma".)

ACKNOWLEDGMENTS — The UpToDate editorial staff acknowledges Evangelos Gragoudas, MD; Anne Marie Lane, MPH; and Richard D Carvajal, MD, who contributed to earlier versions of this topic review.

  1. Gass JD. Problems in the differential diagnosis of choroidal nevi and malignant melanomas. The XXXIII Edward Jackson Memorial Lecture. Am J Ophthalmol 1977; 83:299.
  2. Augsburger JJ, Schroeder RP, Territo C, et al. Clinical parameters predictive of enlargement of melanocytic choroidal lesions. Br J Ophthalmol 1989; 73:911.
  3. Ramaiya KJ, Harbour JW. Current management of uveal melanoma. Expert Rev Ophthalmol 2007; 2:939.
  4. Factors predictive of growth and treatment of small choroidal melanoma: COMS Report No. 5. The Collaborative Ocular Melanoma Study Group. Arch Ophthalmol 1997; 115:1537.
  5. Shields CL, Furuta M, Berman EL, et al. Choroidal nevus transformation into melanoma: analysis of 2514 consecutive cases. Arch Ophthalmol 2009; 127:981.
  6. Abramson DH. Growing melanocytic tumor is not always cancer. Arch Ophthalmol 2005; 123:1457.
  7. Elner VM, Flint A, Vine AK. Histopathology of documented growth in small melanocytic choroidal tumors. Arch Ophthalmol 2004; 122:1876.
  8. Espinoza G, Rosenblatt B, Harbour JW. Optical coherence tomography in the evaluation of retinal changes associated with suspicious choroidal melanocytic tumors. Am J Ophthalmol 2004; 137:90.
  9. Henderson E, Margo CE. Iris melanoma. Arch Pathol Lab Med 2008; 132:268.
  10. Shields CL, Shields JA, Materin M, et al. Iris melanoma: risk factors for metastasis in 169 consecutive patients. Ophthalmology 2001; 108:172.
  11. Harbour JW, Augsburger JJ, Eagle RC Jr. Initial management and follow-up of melanocytic iris tumors. Ophthalmology 1995; 102:1987.
  12. Egan KM, Seddon JM, Glynn RJ, et al. Epidemiologic aspects of uveal melanoma. Surv Ophthalmol 1988; 32:239.
  13. Bataille V, Sasieni P, Cuzick J, et al. Risk of ocular melanoma in relation to cutaneous and iris naevi. Int J Cancer 1995; 60:622.
  14. van Hees CL, de Boer A, Jager MJ, et al. Are atypical nevi a risk factor for uveal melanoma? A case-control study. J Invest Dermatol 1994; 103:202.
  15. Tucker MA, Shields JA, Hartge P, et al. Sunlight exposure as risk factor for intraocular malignant melanoma. N Engl J Med 1985; 313:789.
  16. Seddon JM, Gragoudas ES, Glynn RJ, et al. Host factors, UV radiation, and risk of uveal melanoma. A case-control study. Arch Ophthalmol 1990; 108:1274.
  17. Shah CP, Weis E, Lajous M, et al. Intermittent and chronic ultraviolet light exposure and uveal melanoma: a meta-analysis. Ophthalmology 2005; 112:1599.
  18. Johansson P, Aoude LG, Wadt K, et al. Deep sequencing of uveal melanoma identifies a recurrent mutation in PLCB4. Oncotarget 2016; 7:4624.
  19. Singh AD, De Potter P, Fijal BA, et al. Lifetime prevalence of uveal melanoma in white patients with oculo(dermal) melanocytosis. Ophthalmology 1998; 105:195.
  20. Augsburger JJ, Corrêa ZM, Trichopoulos N, Shaikh A. Size overlap between benign melanocytic choroidal nevi and choroidal malignant melanomas. Invest Ophthalmol Vis Sci 2008; 49:2823.
  21. Freedman MI, Folk JC. Metastatic tumors to the eye and orbit. Patient survival and clinical characteristics. Arch Ophthalmol 1987; 105:1215.
  22. Eskelin S, Pyrhönen S, Summanen P, et al. Tumor doubling times in metastatic malignant melanoma of the uvea: tumor progression before and after treatment. Ophthalmology 2000; 107:1443.
  23. Augsburger JJ, Gamel JW. Clinical prognostic factors in patients with posterior uveal malignant melanoma. Cancer 1990; 66:1596.
  24. Seddon 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.
  25. AJCC Cancer Staging Manual, 8th ed, Springer Verlag, New York 2016.
  26. Skinner CC, Augsburger JJ, Augsburger BD, Correa ZM. Comparison of Alternative Tumor Size Classifications for Posterior Uveal Melanomas. Invest Ophthalmol Vis Sci 2017; 58:3335.
  27. Trolet J, Hupé P, Huon I, et al. Genomic profiling and identification of high-risk uveal melanoma by array CGH analysis of primary tumors and liver metastases. Invest Ophthalmol Vis Sci 2009; 50:2572.
  28. Schoenfield L, Pettay J, Tubbs RR, Singh AD. Variation of monosomy 3 status within uveal melanoma. Arch Pathol Lab Med 2009; 133:1219.
  29. Mensink HW, Vaarwater J, Kiliç E, et al. Chromosome 3 intratumor heterogeneity in uveal melanoma. Invest Ophthalmol Vis Sci 2009; 50:500.
  30. Tschentscher F, Prescher G, Zeschnigk M, et al. Identification of chromosomes 3, 6, and 8 aberrations in uveal melanoma by microsatellite analysis in comparison to comparative genomic hybridization. Cancer Genet Cytogenet 2000; 122:13.
  31. Onken MD, Worley LA, Char DH, et al. Collaborative Ocular Oncology Group report number 1: prospective validation of a multi-gene prognostic assay in uveal melanoma. Ophthalmology 2012; 119:1596.
  32. Worley LA, Onken MD, Person E, et al. Transcriptomic versus chromosomal prognostic markers and clinical outcome in uveal melanoma. Clin Cancer Res 2007; 13:1466.
  33. Petrausch U, Martus P, Tönnies H, 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.
  34. Harbour JW, Chen R. The DecisionDx-UM Gene Expression Profile Test Provides Risk Stratification and Individualized Patient Care in Uveal Melanoma. PLoS Curr 2013; 5.
  35. Bidard FC, Madic J, Mariani P, et al. Detection rate and prognostic value of circulating tumor cells and circulating tumor DNA in metastatic uveal melanoma. Int J Cancer 2014; 134:1207.
  36. Schuster R, Bechrakis NE, Stroux A, et al. Prognostic relevance of circulating tumor cells in metastatic uveal melanoma. Oncology 2011; 80:57.
  37. Lane AM, Egan KM, Kim IK, Gragoudas ES. Mortality after diagnosis of small melanocytic lesions of the choroid. Arch Ophthalmol 2010; 128:996.
  38. Gündüz K, Pulido JS, Bakri SJ, Petit-Fond E. Fundus autofluorescence in choroidal melanocytic lesions. Retina 2007; 27:681.
  39. Earle J, Kline RW, Robertson DM. Selection of iodine 125 for the Collaborative Ocular Melanoma Study. Arch Ophthalmol 1987; 105:763.
  40. Marinkovic M, Horeweg N, Laman MS, et al. Ruthenium-106 brachytherapy for iris and iridociliary melanomas. Br J Ophthalmol 2018; 102:1154.
  41. Lommatzsch PK, Werschnik C, Schuster E. Long-term follow-up of Ru-106/Rh-106 brachytherapy for posterior uveal melanoma. Graefes Arch Clin Exp Ophthalmol 2000; 238:129.
  42. Char DH, Quivey JM, Castro JR, et al. Helium ions versus iodine 125 brachytherapy in the management of uveal melanoma. A prospective, randomized, dynamically balanced trial. Ophthalmology 1993; 100:1547.
  43. Williams DF, Mieler WF, Lewandowski M, Greenberg M. Echographic verification of radioactive plaque position in the treatment of melanomas. Arch Ophthalmol 1988; 106:1623.
  44. Pavlin CJ, Japp B, Simpson ER, et al. Ultrasound determination of the relationship of radioactive plaques to the base of choroidal melanomas. Ophthalmology 1989; 96:538.
  45. Harbour JW, Murray TG, Byrne SF, et al. Intraoperative echographic localization of iodine 125 episcleral radioactive plaques for posterior uveal melanoma. Retina 1996; 16:129.
  46. Badiyan SN, Rao RC, Apicelli AJ, et al. Outcomes of iodine-125 plaque brachytherapy for uveal melanoma with intraoperative ultrasonography and supplemental transpupillary thermotherapy. Int J Radiat Oncol Biol Phys 2014; 88:801.
  47. Tabandeh H, Chaudhry NA, Murray TG, et al. Intraoperative echographic localization of iodine-125 episcleral plaque for brachytherapy of choroidal melanoma. Am J Ophthalmol 2000; 129:199.
  48. Almony A, Breit S, Zhao H, et al. Tilting of radioactive plaques after initial accurate placement for treatment of uveal melanoma. Arch Ophthalmol 2008; 126:65.
  49. Vrabec TR, Augsburger JJ, Gamel JW, et al. Impact of local tumor relapse on patient survival after cobalt 60 plaque radiotherapy. Ophthalmology 1991; 98:984.
  50. Harbour JW, Char DH, Kroll S, et al. Metastatic risk for distinct patterns of postirradiation local recurrence of posterior uveal melanoma. Ophthalmology 1997; 104:1785.
  51. Egger E, Schalenbourg A, Zografos L, et al. Maximizing local tumor control and survival after proton beam radiotherapy of uveal melanoma. Int J Radiat Oncol Biol Phys 2001; 51:138.
  52. American Brachytherapy Society - Ophthalmic Oncology Task Force. Electronic address: [email protected], ABS – OOTF Committee. The American Brachytherapy Society consensus guidelines for plaque brachytherapy of uveal melanoma and retinoblastoma. Brachytherapy 2014; 13:1.
  53. Oellers P, Mowery YM, Perez BA, et al. Efficacy and Safety of Low-Dose Iodine Plaque Brachytherapy for Juxtapapillary Choroidal Melanoma. Am J Ophthalmol 2018; 186:32.
  54. Lane AM, Kim IK, Gragoudas ES. Long-term Risk of Melanoma-Related Mortality for Patients With Uveal Melanoma Treated With Proton Beam Therapy. JAMA Ophthalmol 2015; 133:792.
  55. Caujolle JP, Mammar H, Chamorey E, et al. Proton beam radiotherapy for uveal melanomas at nice teaching hospital: 16 years' experience. Int J Radiat Oncol Biol Phys 2010; 78:98.
  56. Tsuji H, Ishikawa H, Yanagi T, et al. Carbon-ion radiotherapy for locally advanced or unfavorably located choroidal melanoma: a Phase I/II dose-escalation study. Int J Radiat Oncol Biol Phys 2007; 67:857.
  57. Saunders WM, Char DH, Quivey JM, et al. Precision, high dose radiotherapy: helium ion treatment of uveal melanoma. Int J Radiat Oncol Biol Phys 1985; 11:227.
  58. Sikuade MJ, Salvi S, Rundle PA, et al. Outcomes of treatment with stereotactic radiosurgery or proton beam therapy for choroidal melanoma. Eye (Lond) 2015; 29:1194.
  59. Modorati G, Miserocchi E, Galli L, et al. Gamma knife radiosurgery for uveal melanoma: 12 years of experience. Br J Ophthalmol 2009; 93:40.
  60. Muller K, Naus N, Nowak PJ, et al. Fractionated stereotactic radiotherapy for uveal melanoma, late clinical results. Radiother Oncol 2012; 102:219.
  61. Dieckmann K, Georg D, Zehetmayer M, et al. Stereotactic photon beam irradiation of uveal melanoma: indications and experience at the University of Vienna since 1997. Strahlenther Onkol 2007; 183 Spec No 2:11.
  62. Dunavoelgyi R, Dieckmann K, Gleiss A, et al. Radiogenic side effects after hypofractionated stereotactic photon radiotherapy of choroidal melanoma in 212 patients treated between 1997 and 2007. Int J Radiat Oncol Biol Phys 2012; 83:121.
  63. Krema H, Heydarian M, Beiki-Ardakani A, et al. A comparison between ¹²⁵Iodine brachytherapy and stereotactic radiotherapy in the management of juxtapapillary choroidal melanoma. Br J Ophthalmol 2013; 97:327.
  64. Jensen AW, Petersen IA, Kline RW, et al. Radiation complications and tumor control after 125I plaque brachytherapy for ocular melanoma. Int J Radiat Oncol Biol Phys 2005; 63:101.
  65. Conway RM, Poothullil AM, Daftari IK, et al. Estimates of ocular and visual retention following treatment of extra-large uveal melanomas by proton beam radiotherapy. Arch Ophthalmol 2006; 124:838.
  66. Thariat J, Maschi C, Lanteri S, et al. Dry Eye Syndrome After Proton Therapy of Ocular Melanomas. Int J Radiat Oncol Biol Phys 2017; 98:142.
  67. Archer DB, Gardiner TA. Ionizing radiation and the retina. Curr Opin Ophthalmol 1994; 5:59.
  68. Matet A, Daruich A, Zografos L. Radiation Maculopathy After Proton Beam Therapy for Uveal Melanoma: Optical Coherence Tomography Angiography Alterations Influencing Visual Acuity. Invest Ophthalmol Vis Sci 2017; 58:3851.
  69. Horgan N, Shields CL, Mashayekhi A, et al. Early macular morphological changes following plaque radiotherapy for uveal melanoma. Retina 2008; 28:263.
  70. Seibel I, Cordini D, Hager A, et al. Predictive risk factors for radiation retinopathy and optic neuropathy after proton beam therapy for uveal melanoma. Graefes Arch Clin Exp Ophthalmol 2016; 254:1787.
  71. Melia BM, Abramson DH, Albert DM, et al. Collaborative ocular melanoma study (COMS) randomized trial of I-125 brachytherapy for medium choroidal melanoma. I. Visual acuity after 3 years COMS report no. 16. Ophthalmology 2001; 108:348.
  72. Mishra KK, Daftari IK, Weinberg V, et al. Risk factors for neovascular glaucoma after proton beam therapy of uveal melanoma: a detailed analysis of tumor and dose-volume parameters. Int J Radiat Oncol Biol Phys 2013; 87:330.
  73. Mills MD, Harbour JW. Lipid exudation following plaque radiotherapy for posterior uveal melanoma. Am J Ophthalmol 2006; 141:594.
  74. Detorakis ET, Engstrom RE Jr, Wallace R, Straatsma BR. Iris and anterior chamber angle neovascularization after iodine 125 brachytherapy for uveal melanoma. Ophthalmology 2005; 112:505.
  75. Wachtlin J, Bechrakis NE, Schueler AO, et al. Phacoemulsification following treatment of choroidal melanoma. Graefes Arch Clin Exp Ophthalmol 2000; 238:942.
  76. Archer DB, Amoaku WM, Gardiner TA. Radiation retinopathy--clinical, histopathological, ultrastructural and experimental correlations. Eye (Lond) 1991; 5 ( Pt 2):239.
  77. Levin LA, Gragoudas ES, Lessell S. Endothelial cell loss in irradiated optic nerves. Ophthalmology 2000; 107:370.
  78. Giuliari GP, Sadaka A, Hinkle DM, Simpson ER. Current treatments for radiation retinopathy. Acta Oncol 2011; 50:6.
  79. Seibel I, Hager A, Riechardt AI, et al. Antiangiogenic or Corticosteroid Treatment in Patients With Radiation Maculopathy After Proton Beam Therapy for Uveal Melanoma. Am J Ophthalmol 2016; 168:31.
  80. Tarmann L, Langmann G, Mayer C, et al. Ozurdex(®) reduces the retinal thickness in radiation maculopathy refractory to bevacizumab. Acta Ophthalmol 2014; 92:e694.
  81. Murray TG, Latiff A, Villegas VM, Gold AS. Aflibercept for Radiation Maculopathy Study: A Prospective, Randomized Clinical Study. Ophthalmol Retina 2019; 3:561.
  82. Olmos LC, Lee RK. Medical and surgical treatment of neovascular glaucoma. Int Ophthalmol Clin 2011; 51:27.
  83. Tran BK, Schalenbourg A, Bovey E, et al. Role of vitreoretinal surgery in maximizing treatment outcome following complications after proton therapy for uveal melanoma. Retina 2013; 33:1777.
  84. Oosterhuis JA, Journée-de Korver HG, Kakebeeke-Kemme HM, Bleeker JC. Transpupillary thermotherapy in choroidal melanomas. Arch Ophthalmol 1995; 113:315.
  85. Godfrey DG, Waldron RG, Capone A Jr. Transpupillary thermotherapy for small choroidal melanoma. Am J Ophthalmol 1999; 128:88.
  86. Harbour JW, Meredith TA, Thompson PA, Gordon ME. Transpupillary thermotherapy versus plaque radiotherapy for suspected choroidal melanomas. Ophthalmology 2003; 110:2207.
  87. Caminal JM, Mejia-Castillo KA, Arias L, et al. Subthreshold transpupillary thermotherapy in management of foveal subretinal fluid in small pigmented choroidal lesions. Retina 2013; 33:194.
  88. Young LH, Howard MA, Hu LK, et al. Photodynamic therapy of pigmented choroidal melanomas using a liposomal preparation of benzoporphyrin derivative. Arch Ophthalmol 1996; 114:186.
  89. Fabian ID, Stacey AW, Papastefanou V, et al. Primary photodynamic therapy with verteporfin for small pigmented posterior pole choroidal melanoma. Eye (Lond) 2017; 31:519.
  90. Damato BE. Local resection of uveal melanoma. Dev Ophthalmol 2012; 49:66.
  91. Adams KS, Abramson DH, Ellsworth RM, et al. Cobalt plaque versus enucleation for uveal melanoma: comparison of survival rates. Br J Ophthalmol 1988; 72:494.
  92. Seddon JM, Gragoudas ES, Egan KM, et al. Relative survival rates after alternative therapies for uveal melanoma. Ophthalmology 1990; 97:769.
  93. Augsburger JJ, Corrêa ZM, Freire J, Brady LW. Long-term survival in choroidal and ciliary body melanoma after enucleation versus plaque radiation therapy. Ophthalmology 1998; 105:1670.
  94. Collaborative 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.
  95. Cruickshanks KJ, Fryback DG, Nondahl DM, et al. Treatment choice and quality of life in patients with choroidal melanoma. Arch Ophthalmol 1999; 117:461.
  96. Hawkins 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.
  97. Aaberg TM Jr, Cook RW, Oelschlager K, et al. Current clinical practice: differential management of uveal melanoma in the era of molecular tumor analyses. Clin Ophthalmol 2014; 8:2449.
  98. Gezgin G, Luk SJ, Cao J, et al. PRAME as a Potential Target for Immunotherapy in Metastatic Uveal Melanoma. JAMA Ophthalmol 2017; 135:541.
  99. 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.
  100. Wong JR, Nanji AA, Galor A, Karp CL. Management of conjunctival malignant melanoma: a review and update. Expert Rev Ophthalmol 2014; 9:185.
  101. Tuomaala S, Eskelin S, Tarkkanen A, Kivelä T. Population-based assessment of clinical characteristics predicting outcome of conjunctival melanoma in whites. Invest Ophthalmol Vis Sci 2002; 43:3399.
  102. Missotten GS, Keijser S, De Keizer RJ, De Wolff-Rouendaal D. Conjunctival melanoma in the Netherlands: a nationwide study. Invest Ophthalmol Vis Sci 2005; 46:75.
  103. Triay E, Bergman L, Nilsson B, et al. Time trends in the incidence of conjunctival melanoma in Sweden. Br J Ophthalmol 2009; 93:1524.
  104. Swaminathan SS, Field MG, Sant D, et al. Molecular Characteristics of Conjunctival Melanoma Using Whole-Exome Sequencing. JAMA Ophthalmol 2017; 135:1434.
  105. Larsen AC. Conjunctival malignant melanoma in Denmark: epidemiology, treatment and prognosis with special emphasis on tumorigenesis and genetic profile. Acta Ophthalmol 2016; 94 Thesis 1:1.
  106. Karim R, Conway RM. Conservative resection and adjuvant plaque brachytherapy for early-stage conjunctival melanoma. Clin Exp Ophthalmol 2011; 39:293.
  107. Paridaens AD, McCartney AC, Minassian DC, Hungerford JL. Orbital exenteration in 95 cases of primary conjunctival malignant melanoma. Br J Ophthalmol 1994; 78:520.
  108. Esmaeli B, Wang X, Youssef A, Gershenwald JE. Patterns of regional and distant metastasis in patients with conjunctival melanoma: experience at a cancer center over four decades. Ophthalmology 2001; 108:2101.
  109. Dagi Glass LR, Lawrence DP, Jakobiec FA, Freitag SK. Conjunctival Melanoma Responsive to Combined Systemic BRAF/MEK Inhibitors. Ophthalmic Plast Reconstr Surg 2017; 33:e114.
  110. Kini A, Fu R, Compton C, et al. Pembrolizumab for Recurrent Conjunctival Melanoma. JAMA Ophthalmol 2017; 135:891.
  111. Aziz HA, Gastman BR, Singh AD. Management of Conjunctival Melanoma: Critical Assessment of Sentinel Lymph Node Biopsy. Ocul Oncol Pathol 2015; 1:266.
  112. Seregard S, Kock E. Conjunctival malignant melanoma in Sweden 1969-91. Acta Ophthalmol (Copenh) 1992; 70:289.
Topic 7617 Version 69.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟