Next Article in Journal
Inhaled Corticosteroids and the Lung Microbiome in COPD
Next Article in Special Issue
Multi-Omics Reveals the Immunological Role and Prognostic Potential of Mitochondrial Ubiquitin Ligase MARCH5 in Human Breast Cancer
Previous Article in Journal
Systems Analysis Reveals Ageing-Related Perturbations in Retinoids and Sex Hormones in Alzheimer’s and Parkinson’s Diseases
Previous Article in Special Issue
Therapeutic Potential of PARP Inhibitors in the Treatment of Gastrointestinal Cancers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

New Therapeutic Perspectives in the Treatment of Uveal Melanoma: A Systematic Review

1
Department of Ophthalmology, University of Zurich, 8091 Zurich, Switzerland
2
Department of General and Pediatric Ophthalmology, Medical University of Lublin, 20079 Lublin, Poland
3
Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
4
Clinical Pharmacology Unit, Regional Pharmacovigilance Centre, University Hospital of Catania, 95123 Catania, Italy
5
Plastic and Reconstructive Surgery Department, American Hospital Dubai, Dubai, United Arab Emirates
6
Department of Biomedical and Dental Sciences, Morphological and Functional Images, University of Messina, AOU ‘G. Martino’, 98124 Messina, Italy
7
Centre for Research and Consultancy in HTA and Drug Regulatory Affairs (CERD), University of Catania, 95123 Catania, Italy
8
Center of Research in Ocular Pharmacology—CERFO, University of Catania, 95123 Catania, Italy
9
Department of Ophthalmology, University of Catania, 95123 Catania, Italy
10
Department of Surgery/Ophthalmology, King Hussein Cancer Center, Amman 11941, Jordan
11
Shami Eye Center, Amman 11941, Jordan
*
Author to whom correspondence should be addressed.
Biomedicines 2021, 9(10), 1311; https://doi.org/10.3390/biomedicines9101311
Submission received: 11 August 2021 / Revised: 15 September 2021 / Accepted: 21 September 2021 / Published: 24 September 2021

Abstract

:
Uveal melanoma (UM) is a rare disease, but the most common primary intraocular cancer, mostly localized in the choroid. Currently, the first-line treatment options for UM are radiation therapy, resection, and enucleation. However, although these treatments could potentially be curative, half of all patients will develop metastatic disease, whose prognosis is still poor. Indeed, effective therapeutic options for patients with advanced or metastatic disease are still lacking. Recently, the development of new treatment modalities with a lower incidence of adverse events, a better disease control rate, and new therapeutic approaches, have merged as new potential and promising therapeutic strategies. Additionally, several clinical trials are ongoing to find new therapeutic options, mainly for those with metastatic disease. Many interventions are still in the preliminary phases of clinical development, being investigated in phase I trial or phase I/II. The success of these trials could be crucial for changing the prognosis of patients with advanced/metastatic UM. In this systematic review, we analyzed all emerging and available literature on the new perspectives in the treatment of UM and patient outcomes; furthermore, their current limitations and more common adverse events are summarized.

1. Introduction

Uveal melanoma (UM) is a rare disease (5% of all melanomas), but the most common primary intraocular cancer in adults (mean age of 60) [1,2]. It is more frequent in Caucasians, with an incidence of 0.69 per 100,000 person-year for males and 0.54 per 100,000 person-year for females. In Europe, UM incidence increases with latitude and range from 2/106 in Spain and Italy, 4–6/106 in Central Europe, and >8/106 in Denmark and Norway [1,3].
UM develops most often in individuals with fair skin, light eye color, ocular or oculodermal melanocytosis, cutaneous or iris, or choroidal nevus. Even if it is frequently associated with BAP1 or BRCA1 mutation carriers [1], its pathogenesis is not yet clearly understood [2]. The choroid is the most frequent localization (85–91%), whereas the ciliary body or the iris are affected in only 9–15% of cases [3]. However, iris melanomas is often detected early, resulting in the best prognosis [4], while ciliary body melanomas are associated with the worst prognosis [5]. Half of all patients with UM will develop metastatic disease, whose prognosis is still poor (6–12 months of survival) [6,7].
Based on the conclusions from the Collaborative Ocular Melanoma Study (COMS) [7], globe–vision-preserving radiation therapy is the primary treatment of choice for most UMs nowadays in the developed world [1]. Other globe-preserving therapies may include surgical or laser. In particular, radiation therapy modalities include brachytherapy, photon-based external-beam radiation, and charged-particle radiation [8,9,10,11]. Incidentally, glucocorticoids and anti-VEGF are used for different ocular diseases [12,13,14,15,16,17], including radiation maculopathy secondary to radiotherapy for UM [18,19,20,21].
Plaque brachytherapy involves an affixing dish-shaped source of radiation onto the sclera to cover the base of the intraocular tumor. During radioactive plaque therapy, radiation travels through and is sequentially absorbed by the sclera, the tumor, the retina, the vitreous, and normal ocular structures as it exits the eye [8,9,10,11].
Prior to introduction of plaque therapy, patients with the diagnosis of UM used to be treated by enucleation (surgical removal of the eye globe) as first-line treatment, despite the critical consequences regarding vision and quality of life [22,23,24]. Therefore, the introduction of plaque therapy significantly improved the management of these patients, allowing preservation of the globe, and saving vision in selected cases. For brachytherapy, reported local recurrence rates are 14.7%—for 106Ru treatment, 7%–10% for 125I, and 3.3%—for 103Pd. Brachytherapy does not lead to increased or decreased survival rates as compared to enucleation [3,7,22].
On the other hand, photodynamic laser photocoagulation and trans-pupillary thermal therapy (TTT) reduce local recurrences by the activation of light-sensitive compounds and free radicals, directly focusing energy to the damage tumor. TTT is effective, particularly in cases of limited lesions with few risk factors. To date, no adjuvant chemotherapy allows to prolong survival [3].
Although brachytherapy is the most common globe-preserving treatment option for small- and medium-sized UM patients, the treatment is associated with severe adverse reactions. Indeed, brachytherapy can lead to the onset of radiation-induced retinopathy, cataracts, neovascular glaucoma, and macular edema, with consequent impaired vision within 2 years [3,23,25,26].
Despite the progress in the development of new therapeutic strategies for ocular tumors, to date, all treatments are still unsatisfactory in terms of disease control, as the average treatment failure in all radiation therapies is 6.15%, 18.6% in surgical, and 20.8% in laser therapies [4,27,28]. Thus, the development of new treatment modalities with a lower incidence of adverse events and a better disease control rate are highly demanded. In this regard, new therapeutic approaches emerged in recent decades, highlighting interesting pharmacological targets, such as sigma receptors [29,30].
This systematic review analyzes the data available on the therapeutic perspectives in the treatment of UM and patient outcomes; furthermore, current limitations and more common adverse events are summarized.

2. Materials and Methods

2.1. Search Methods for Identification of Studies

This systematic review was conducted and reported in accordance with the preferred reporting items for systematic reviews and meta-analyses guidelines [31]. The review protocol was not recorded at study design, and no registration number is available for consultation.
The methodology used for this comprehensive review consisted of a systematic search of all available articles exploring the current available treatments or ongoing trials on UM treatment, localized or metastatic types, in adult subjects.
A comprehensive literature search of all original articles published up to December 2020 was performed in parallel by two authors (L.G. and L.T.) using the PubMed, Cochrane library, Embase, and Scopus databases. For the search strategy, we used the following keywords and Mesh terms: “uveal melanoma”, “pharmacological treatment”, “local treatment”, and “systemic therapy”. Furthermore, the reference lists of all identified articles were examined manually to identify any potential studies that were not captured by the electronic searches.
The same terms have been used to conduct a parallel analysis on clinicaltrial.gov, to identify ongoing clinical trials.
The search workflow was designed in adherence to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) statement [31].

2.2. Eligibility Criteria

All studies available in the literature, reporting on original data on UM treatments, localized or metastatic types, were included, without restriction for study design, sample size, and intervention performed. Review articles, protocols, and studies without efficacy data were excluded.

2.3. Data Collection

After preparation of the list of all electronic data captured, two reviewers (M.T. and G.L.R.) examined the titles and abstracts independently and identified relevant articles according to the eligibility criteria. Any disagreement was assessed by consensus and a third reviewer (Y.A.Y.) was consulted when necessary. The reference lists of the analyzed articles were also considered as potential sources of information.
The following data were analyzed for each article, using an Excel spreadsheet:
  • Study design: retrospective, prospective, comparative and non-comparative, randomized and non-randomized, open, and case report/case series;
  • Clinical outcomes: primary and secondary, efficacy, safety, PK;
  • Number of eyes studied, number of patients enrolled;
  • Primary treatment;
  • Follow-up (duration of the study);
  • Main results;
  • Side effects.
For unpublished data, no effort was made to contact the corresponding authors.

3. Results

The results of the search strategy are summarized in Figure 1. From 75 articles extracted from the initial research, 73 abstracts were identified for screening and 64 of these met the inclusion/exclusion criteria for full-text review. Moreover, 11 articles were excluded, including 4 studies without efficacy data, 3 protocols, 1 without full-text, 1 non-original article (Figure 1).
Figure 1. Preferred reporting items for systematic reviews and meta-analyses (PRISMA 2009) flowchart [18]. The 64 studies included in this systematic review were divided in the following groups: treatment of local complications (n = 3; Table 1), local treatment of primary tumor or metastases (n = 13; Table 2), and systemic therapy (n = 48; Table 3). Only one study was a randomized, double blind, placebo-controlled phase III trial; the other studies were phase I/pilot studies (n = 14), phase II studies (n = 34), other designs/design not available (n = 16).
Figure 1. Preferred reporting items for systematic reviews and meta-analyses (PRISMA 2009) flowchart [18]. The 64 studies included in this systematic review were divided in the following groups: treatment of local complications (n = 3; Table 1), local treatment of primary tumor or metastases (n = 13; Table 2), and systemic therapy (n = 48; Table 3). Only one study was a randomized, double blind, placebo-controlled phase III trial; the other studies were phase I/pilot studies (n = 14), phase II studies (n = 34), other designs/design not available (n = 16).
Biomedicines 09 01311 g001
Table 4, Table 5 and Table 6 summarize the main efficacy and safety results of these studies.
No data synthesis was possible for the heterogeneity of available data and the design of the available studies (i.e., case reports or case series). Thus, the current systematic review reports a qualitative analysis, detailed issue-by-issue below in narrative fashion.
Among the 13 studies about the local treatment of the primary tumor or metastases from UM, we found 4 clinical trials—3 phase II studies and 1 phase III study. One of these trials assessed the local tumor control of the intravitreal administration of ranibizumab [33], but all patients required enucleation, and the study was terminated early due to the lack of therapeutic advantages.
Olofsson R et al. [23] observed an overall radiological response of 68% in 34 patients with liver metastasis from UM treated with isolated hepatic perfusion (IHP) within a phase II trial. The time to local progression was 7 months and the median overall survival (OS) 24 months, with a significant survival advantage compared to the control group (National Patient Register; p = 0.029). All patients enrolled in the phase II study of Fiorentini G et al. [28] obtained an objective response of liver metastasis treated with hepatic transarterial chemoembolization (TACE) adopting irinotecan-loaded microspheres. Finally, the prospective, randomized, phase III trial of Leyvraz S et al. [24] compared the IV or intra-arterial hepatic (I.a.H.) fotemustine administration in 171 patients with liver metastases from UM followed for a median of 5 years. I.a.H. did not improve OS (median 14.6 months) compared to IV administration (median 13.8 months; p = 0.59). However, there was a benefit on progression free survival (PFS) for HIA (median PFS of 4.5 versus 3.5 months, respectively; 1-year PFS rate 24% versus 8%), and on response rate (10.5% versus 2.4%).
In regard to systemic therapy, 30 phase II studies and 1 phase III were conducted. Almost half of these studies evaluated the administration of various chemotherapy regimens in advanced or metastatic UM [43,52,54,66,68,69,71,72,74,76,77,78,79].
Overall, we found a low response rate and limited advantage in terms of survival.
The study by Lane AM et al. [62] did not demonstrate an advantage of adjuvant interferon treatment in terms of melanoma-related mortality compared to historical controls in 121 patients with choroidal or ciliary body melanoma during a long-term follow-up (approximately 9 years). However, Binkley E et al. [38] recently reported a survival benefit of adjuvant therapy based on sequential low-dose dacarbazine and interferon-alpha in 33 patients with high-risk UM (5-year median OS of 66% (45–80, median not observed) in treated patients and 37% (19–55, median 54 months) in control).
The trials that assessed the role of immunotherapy in patients with unresectable or metastatic UM showed a low rate of response, a median OS of 1 year with nivolumab and pembrolizumab [35,39], and 6 months with ipilimumab [51]. One patient treated with pembrolizumab reached a complete response (CR, ongoing at 25.5 months), but the drug was stopped after the first dose due to the onset of a severe form of diabetes (grade 4).
Target therapies obtained partial response (PR) or stable disease (SD) as the best objective outcome [36,40,44,48,49,57,58,82]. No difference in PFS or OS was observed versus chemotherapy or compared with the expected patient survival [36,48,50,58,82].
The only randomized, double blind, placebo-controlled phase III trial enrolled 129 patients with metastatic UM to receive either selumetinib or placebo plus dacarbazine [42]. The primary endpoint of PFS advantage was not met (median PFS was 2.8 months in the selumetinib + dacarbazine and 1.8 months in the placebo + dacarbazine group).
Finally, we found more than 140 interventional clinical trials (49 ongoing, 79 completed, 1 suspended, 4 withdrawn, and 9 with unknown status) on uveal melanoma listed on the clinicaltrials.gov database. Among the ongoing trials, 11 foresee the enrollment of patients with local disease and 38 patients with metastatic or unresectable uveal melanoma (Table 7a,b).

4. Discussion

To date, no drugs have been specifically approved for the treatment of non-metastatic uveal melanoma.
Pharmacological treatments result ineffective, likely due to the incapacity to reach enough concentration into the tumor area in the eye, as result of the characteristics of the posterior segment and the blood–retinal barrier [96]. It is possible that local delivery at the ocular site will obtain better results, in terms of efficacy and safety. Therefore, researchers are developing new drug delivery systems for uveal melanoma and other ophthalmological diseases, thanks in part to nanotechnology [97,98,99].
However, the possibility of using effective drug delivery still represents a big challenge, and further studies are needed to establish whether this new technology could help in the fight against uveal melanoma [100,101,102,103].
Despite advances in diagnosis and local treatment, the overall survival (OS) of patients with uveal melanoma remains poor because of the progression into metastatic disease. Indeed, up to 50% of cases develop metastasis, especially in the liver, at approximately 5 years after treatment of the primary tumor [104,105,106]. This time is shorter in patients with larger neoplasm, especially in those with a higher grade of malignancy [107,108].
Metastatic disease is particularly difficult to treat; available systemic therapy rarely produces durable responses or significant survival benefits. Actually, the reported median survival after detection of metastatic disease is less than 1 year [109].
Moreover, no adjuvant therapy, which may be more active in treating microscopic metastatic tumor, was shown to reduce the risk of disease spread or survival improvement, and would need further studies [104].
The treatment of metastatic uveal melanoma includes systemic chemotherapy, immunotherapy, and molecular targeted therapy. Moreover, local therapies for liver disease (resection, chemoembolization, immunoembolization, radioembolization, isolated hepatic perfusion, percutaneous hepatic perfusion) are also recommended [110].
The most updated clinical practice guidelines [111,112] recommend the enrollment of patients with metastatic disease in clinical trials, if possible. Otherwise, systemic therapies used to treat cutaneous melanoma can be considered, although no regimens demonstrated improved overall survival in uveal disease.
Chemotherapy regimens for cutaneous melanoma (dacarbazine, temozolomide, cisplatin, paclitaxel, treosulfan, fotemustine) have been used in uveal melanoma although with unsatisfactory results (response rate 0–15%, and no survival benefit) [113,114,115].
Immunotherapy has dramatically improved outcomes for patients with advanced cutaneous melanoma, but this clinical benefit has not been observed in metastatic uveal melanoma, probably due to a low mutational burden and low PD-L1 expression [116,117,118].
The randomized phase III trial that led to the approval of ipilimumab did not include patients with uveal melanoma, and subsequent smaller studies found a low response rate (0–5%) and an OS of less than 10 months [64,119,120,121,122].
Even the phase III CheckMate-067 trial, comparing the concomitant use of nivolumab plus ipilimumab versus the monotherapy alone, excluded uveal melanoma patients [123].
A large series of patients with metastatic uveal melanoma treated with PD-1 and PD-L1 antibodies (pembrolizumab N = 38; nivolumab N = 16; atezolizumab N = 2) showed a partial response rate of 3.6%, a median progression free survival (PFS) of 2.8 months, and an OS of 7.6 months [124].
Recently, the results of a single arm phase II trial demonstrated an overall response rate (ORR) of 18%, a median PFS of 5.5 months, and a median OS of 19.1 months in 33 patients with metastatic uveal melanoma treated with nivolumab plus ipilimumab [125]. Considering these results, the usefulness of immunotherapy in uveal melanoma requires additional investigation and many clinical trials are currently ongoing.
A novel bispecific molecule targeting T-cells (tebentafusp, IMCgp100) showed clinical benefit in patients with metastatic uveal melanoma in phase II, and recently, a phase III study [126].
The mechanism of action consists in the redirection of T cells to target the gp100 protein, highly expressed in melanocytes and melanoma cells. The phase III trial assessed OS as the primary endpoint in 378 naïve patients with metastatic uveal melanoma, randomized 2:1 to receive tebentafusp or the investigator’s choice among dacarbazine, ipilimumab, or pembrolizumab.
OS was statistically significantly improved in patients randomized in the experimental group compared to the control group in the first pre-planned interim analysis (OS hazard ratio of 0.51, and estimated 1-year OS rate of 73% for the study drug versus 58% with the investigator’s choice) [127,128]. These data confirm the positive survival benefit of the phase II clinical trial, and might likely support the use of this drug as a potential new treatment for cancer patients with this highly unmet need. Moreover, the drug was granted the fast-track and orphan drug designation by the FDA for uveal melanoma [129,130] and Promising Innovative Medicine designation under the UK Early Access to Medicines Scheme.
In regard to target therapies, BRAF and KIT inhibitors are not included among treatment options, as uveal melanomas usually lack BRAF and KIT mutations. Conversely, the typical mutations in GNAQ and GNA11 genes lead to constitutive activation of the MAPK and PI3K/Akt pathways and therapies that target downstream effectors, such as MEK, Akt, and protein kinase C (PKC) are under investigation, even with disappointing results so far [113].
For example, selumetinib, a potent and highly selective inhibitor of MEK, associated with dacarbazine, showed no significant improvement in terms of PFS compared to dacarbazine alone (2.8 versus 1.8 months, p = 0.32) in the phase III SUMIT trial [55]. Similarly, there was no significant difference in ORR (3.1 versus 0%, p = 0.36).
According to the underlying molecular mechanisms, target therapies could probably be improved by combinatory strategies [131].
To date, several clinical trials are ongoing to find new therapeutic options, mainly for those with metastatic disease [110]. Many interventions are still in the preliminary phases of clinical development, being investigated in phase I trial or phase I/II (Table 7).
Additionally, the possibility to exploit a possible ocular pharmaceutical RNA-based treatment against differentially expressed miRNAs in different ocular diseases [132,133], including UM, together with the success of these trials, could be crucial for changing the prognosis of patients with advanced/metastatic UM.

5. Conclusions

This systematic review shows the lack of well-designed randomized clinical trials so far and confirms the limited advantages, in terms of response and survival of treatment options for UM. Despite the progress in the development of new effective therapeutic strategies, to date, all treatments for UM are still unsatisfactory and patients have a poor long-term prognosis. The future success of ongoing trials could hopefully change the outcome of patients with advanced/metastatic UM.

Author Contributions

Conceptualization, G.L.R., M.D.T.; methodology, G.L.R., M.D.T.; formal analysis, L.G., L.T., G.L.R., M.D.T.; investigation, L.G., L.T., G.L.R., M.D.T.; resources, L.G., L.T.; data curation, L.G., L.T., M.C.; writing—original draft preparation, L.G., L.T., M.C., G.L.R., M.D.T.; writing—review and editing, Y.A.Y., G.L.R., M.D.T., C.B., F.D., T.A., R.R., S.Z., Y.A.Y., R.N., K.N.; supervision, G.L.R., M.D.T.; project administration, G.L.R., M.D.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding. G.L.R. was supported by the PON AIM R&I 2014-2020-E66C18001260007. M.D.T. was supported by the “Foundation to support the development of Ophthalmology” in Poland. The Foundation had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Publicly available datasets were analyzed in this study. These data can be found here: https://pubmed.ncbi.nlm.nih.gov/, (accessed on 1 February 2021); https://clinicaltrials.gov/, (accessed on 15 February 2021).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pt, F. Intraocular melanoma. In Cancer: Principles and Practice of Oncology, 10th ed.; DeVita, J.V.T., Lawrence, T.S., Rosenberg, S.A., Eds.; Wolter Kluwer, Lippincott, Williams and Wilkins: Philadelphia, PA, USA, 2014; pp. 1770–1779. [Google Scholar]
  2. Ragusa, M.; Barbagallo, C.; Statello, L.; Caltabiano, R.; Russo, A.; Puzzo, L.; Avitabile, T.; Longo, A.; Toro, M.D.; Barbagallo, D.; et al. miRNA profiling in vitreous humor, vitreal exosomes and serum from uveal melanoma patients: Pathological and diagnostic implications. Cancer Biol. Ther. 2015, 16, 1387–1396. [Google Scholar] [CrossRef]
  3. Shields, C.L.; Kaliki, S.; Furuta, M.; Mashayekhi, A.; Shields, J.A. Clinical spectrum and prognosis of uveal melanoma based on age at presentation in 8,033 cases. Retina 2012, 32, 1363–1372. [Google Scholar] [CrossRef]
  4. Shields, C.L.; Kaliki, S.; Shah, S.U.; Luo, W.; Furuta, M.; Shields, J.A. Iris melanoma: Features and prognosis in 317 children and adults. J. AAPOS 2012, 16, 10–16. [Google Scholar] [CrossRef] [PubMed]
  5. Oittinen, H.A.; O’Shaughnessy, M.; Cullinane, A.B.; Keohane, C. Malignant melanoma of the ciliary body presenting as extraocular metastasis in the temporalis muscle. J. Clin. Pathol. 2007, 60, 834–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Kujala, E.; Makitie, T.; Kivela, T. Very long-term prognosis of patients with malignant uveal melanoma. Investig. Ophthalmol. Vis. Sci. 2003, 44, 4651–4659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Diener-West, M.; Earle, J.D.; Fine, S.L.; Hawkins, B.S.; Moy, C.S.; Reynolds, S.M.; Schachat, A.P.; Straatsma, B.R.; Collaborative Ocular Melanoma Study Group. The COMS randomized trial of iodine 125 brachytherapy for choroidal melanoma, III: Initial mortality findings. COMS Report No. 18. Arch. Ophthalmol. 2001, 119, 969–982. [Google Scholar] [CrossRef] [PubMed]
  8. American Brachytherapy Society—Ophthalmic Oncology Task Force; ABS—OOTF Committee. The American Brachytherapy Society consensus guidelines for plaque brachytherapy of uveal melanoma and retinoblastoma. Brachytherapy 2014, 13, 1–14. [Google Scholar] [CrossRef] [Green Version]
  9. Gragoudas, E.S.; Goitein, M.; Verhey, L.; Munzenreider, J.; Urie, M.; Suit, H.; Koehler, A. Proton beam irradiation of uveal melanomas. Results of 5 1/2-year study. Arch. Ophthalmol. 1982, 100, 928–934. [Google Scholar] [CrossRef]
  10. Chiu-Tsao, S.T.; Astrahan, M.A.; Finger, P.T.; Followill, D.S.; Meigooni, A.S.; Melhus, C.S.; Mourtada, F.; Napolitano, M.E.; Nath, R.; Rivard, M.J.; et al. Dosimetry of (125)I and (103)Pd COMS eye plaques for intraocular tumors: Report of Task Group 129 by the AAPM and ABS. Med. Phys. 2012, 39, 6161–6184. [Google Scholar] [CrossRef] [Green Version]
  11. Finger, P.T. Radiation therapy for choroidal melanoma. Surv. Ophthalmol. 1997, 42, 215–232. [Google Scholar] [CrossRef]
  12. Ceravolo, I.; Oliverio, G.W.; Alibrandi, A.; Bhatti, A.; Trombetta, L.; Rejdak, R.; Toro, M.D.; Trombetta, C.J. The Application of Structural Retinal Biomarkers to Evaluate the Effect of Intravitreal Ranibizumab and Dexamethasone Intravitreal Implant on Treatment of Diabetic Macular Edema. Diagnostics 2020, 10, 413. [Google Scholar] [CrossRef]
  13. Yousef, Y.A.; ElRimawi, A.H.; Nazzal, R.M.; Qaroot, A.F.; AlAref, A.H.; Mohammad, M.; Abureesh, O.; Rejdak, R.; Nowomiejska, K.; Avitabile, T.; et al. Coats’ disease: Characteristics, management, outcome, and scleral external drainage with anterior chamber maintainer for stage 3b disease. Medicine 2020, 99, e19623. [Google Scholar] [CrossRef]
  14. Gagliano, C.; Toro, M.D.; Avitabile, T.; Stella, S.; Uva, M.G. Intravitreal Steroids for the Prevention of PVR After Surgery for Retinal Detachment. Curr. Pharm. Des. 2015, 21, 4698–4702. [Google Scholar] [CrossRef] [PubMed]
  15. Reibaldi, M.; Russo, A.; Avitabile, T.; Uva, M.G.; Franco, L.; Longo, A.; Toro, M.D.; Cennamo, G.; Mariotti, C.; Neri, P.; et al. Treatment of persistent serous retinal detachment in Vogt-Koyanagi-Harada syndrome with intravitreal bevacizumab during the systemic steroid treatment. Retina (Phila. Pa.) 2014, 34, 490–496. [Google Scholar] [CrossRef] [PubMed]
  16. Reibaldi, M.; Russo, A.; Longo, A.; Bonfiglio, V.; Uva, M.G.; Gagliano, C.; Toro, M.D.; Avitabile, T. Rhegmatogenous Retinal Detachment with a High Risk of Proliferative Vitreoretinopathy Treated with Episcleral Surgery and an Intravitreal Dexamethasone 0.7-mg Implant. Case Rep. Ophthalmol. 2013, 4, 79–83. [Google Scholar] [CrossRef]
  17. Reibaldi, M.; Russo, A.; Zagari, M.; Toro, M.; Grande De, V.; Cifalinò, V.; Rametta, S.; Faro, S.; Longo, A. Resolution of Persistent Cystoid Macular Edema due to Central Retinal Vein Occlusion in a Vitrectomized Eye following Intravitreal Implant of Dexamethasone 0.7 mg. Case Rep. Ophthalmol. 2012, 3, 30–34. [Google Scholar] [CrossRef] [Green Version]
  18. Russo, A.; Avitabile, T.; Uva, M.; Faro, S.; Franco, L.; Sanfilippo, M.; Gulisano, S.; Toro, M.; De Grande, V.; Rametta, S.; et al. Radiation Macular Edema after Ru-106 Plaque Brachytherapy for Choroidal Melanoma Resolved by an Intravitreal Dexamethasone 0.7-mg Implant. Case Rep. Ophthalmol. 2012, 3, 71–76. [Google Scholar] [CrossRef] [PubMed]
  19. Rayess, N.; Mruthyunjaya, P. Anti-Vascular Endothelial Growth Factor Therapy for Radiation Retinopathy. Ophthalmic Surg. Lasers Imaging Retin. 2020, 51, S44–S49. [Google Scholar] [CrossRef]
  20. Seibel, I.; Hager, A.; Riechardt, A.I.; Davids, A.M.; Böker, A.; Joussen, A.M. Antiangiogenic or Corticosteroid Treatment in Patients With Radiation Maculopathy After Proton Beam Therapy for Uveal Melanoma. Am. J. Ophthalmol. 2016, 168, 31–39. [Google Scholar] [CrossRef]
  21. Shafiee, A.; Bucolo, C.; Budzynski, E.; Ward, K.W.; López, F.J. In Vivo Ocular Efficacy Profile of Mapracorat, a Novel Selective Glucocorticoid Receptor Agonist, in Rabbit Models of Ocular Disease. Investig. Ophthalmol. Vis. Sci. 2011, 52, 1422–1430. [Google Scholar] [CrossRef] [Green Version]
  22. Finger, P.T.; Chin, K.J.; Duvall, G.; Palladium-103 for Choroidal Melanoma Study Group. Palladium-103 ophthalmic plaque radiation therapy for choroidal melanoma: 400 treated patients. Ophthalmology 2009, 116, 790–796.e1. [Google Scholar] [CrossRef]
  23. 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, 1684–1693. [Google Scholar] [CrossRef]
  24. Straatsma, B.R. The Jules Francois Memorial lecture. The collaborative ocular melanoma study and management of choroidal melanoma. Bull. Soc. Belg. Ophtalmol. 2002, 5–11. [Google Scholar]
  25. Jampol, L.M.; Moy, C.S.; Murray, T.G.; Reynolds, S.M.; Albert, D.M.; Schachat, A.P.; Diddie, K.R.; Engstrom, R.E., Jr.; Finger, P.T.; Hovland, K.R.; 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, 2197–2206. [Google Scholar] [CrossRef]
  26. Hawkins, B.S.; Collaborative Ocular Melanoma Study, G. 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, 936–951. [Google Scholar] [CrossRef] [PubMed]
  27. Yousef, Y.A.; Al Jboor, M.; Mohammad, M.; Mehyar, M.; Toro, M.D.; Nazzal, R.; Alzureikat, Q.H.; Rejdak, M.; Elfalah, M.; Sultan, I.; et al. Safety and Efficacy of Intravitreal Chemotherapy (Melphalan) to Treat Vitreous Seeds in Retinoblastoma. Front. Pharmacol. 2021, 12, 696787. [Google Scholar] [CrossRef] [PubMed]
  28. Amin, S.; AlJboor, M.; Toro, M.D.; Rejdak, R.; Nowomiejska, K.; Nazzal, R.; Mohammad, M.; Al-Hussaini, M.; Khzouz, J.; Banat, S.; et al. Management and Outcomes of Unilateral Group D Tumors in Retinoblastoma. Clin. Ophthalmol. (Auckl. N.Z.) 2021, 15, 65–72. [Google Scholar] [CrossRef]
  29. Bucolo, C.; Campana, G.; Di Toro, R.; Cacciaguerra, S.; Spampinato, S. Sigma1 recognition sites in rabbit iris-ciliary body: Topical sigma1-site agonists lower intraocular pressure. J. Pharm. Exp. 1999, 289, 1362–1369. [Google Scholar]
  30. Longhitano, L.; Castracani, C.C.; Tibullo, D.; Avola, R.; Viola, M.; Russo, G.; Prezzavento, O.; Marrazzo, A.; Amata, E.; Reibaldi, M.; et al. Sigma-1 and Sigma-2 receptor ligands induce apoptosis and autophagy but have opposite effect on cell proliferation in uveal melanoma. Oncotarget 2017, 8, 91099–91111. [Google Scholar] [CrossRef]
  31. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G.; Group, P. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA Statement. Open Med. 2009, 3, e123–e130. [Google Scholar]
  32. Schefler, A.C.; Fuller, D.; Anand, R.; Fuller, T.; Moore, C.; Munoz, J.; Kim, R.S.; RRR Study Group. Randomized Trial of Monthly Versus As-Needed Intravitreal Ranibizumab for Radiation Retinopathy-Related Macular Edema: 1-Year Outcomes. Am. J. Ophthalmol. 2020, 216, 165–173. [Google Scholar] [CrossRef] [PubMed]
  33. Murray, T.G.; Latiff, A.; Villegas, V.M.; Gold, A.S. Aflibercept for Radiation Maculopathy Study: A Prospective, Randomized Clinical Study. Ophthalmol. Retin. 2019, 3, 561–566. [Google Scholar] [CrossRef]
  34. Horgan, N.; Shields, C.L.; Mashayekhi, A.; Salazar, P.F.; Materin, M.A.; O’Regan, M.; Shields, J.A. Periocular triamcinolone for prevention of macular edema after plaque radiotherapy of uveal melanoma: A randomized controlled trial. Ophthalmology 2009, 116, 1383–1390. [Google Scholar] [CrossRef] [PubMed]
  35. Venturini, M.; Pilla, L.; Agostini, G.; Cappio, S.; Losio, C.; Orsi, M.; Ratti, F.; Aldrighetti, L.; De Cobelli, F.; Del Maschio, A. Transarterial chemoembolization with drug-eluting beads preloaded with irinotecan as a first-line approach in uveal melanoma liver metastases: Tumor response and predictive value of diffusion-weighted MR imaging in five patients. J. Vasc. Interv. Radiol. 2012, 23, 937–941. [Google Scholar] [CrossRef] [PubMed]
  36. Olofsson, R.; Cahlin, C.; All-Ericsson, C.; Hashimi, F.; Mattsson, J.; Rizell, M.; Lindner, P. Isolated hepatic perfusion for ocular melanoma metastasis: Registry data suggests a survival benefit. Ann. Surg. Oncol. 2014, 21, 466–472. [Google Scholar] [CrossRef] [Green Version]
  37. Leyvraz, S.; Piperno-Neumann, S.; Suciu, S.; Baurain, J.F.; Zdzienicki, M.; Testori, A.; Marshall, E.; Scheulen, M.; Jouary, T.; Negrier, S.; et al. Hepatic intra-arterial versus intravenous fotemustine in patients with liver metastases from uveal melanoma (EORTC 18021): A multicentric randomized trial. Ann. Oncol. 2014, 25, 742–746. [Google Scholar] [CrossRef]
  38. van Iersel, L.B.; de Leede, E.M.; Vahrmeijer, A.L.; Tijl, F.G.; den Hartigh, J.; Kuppen, P.J.; Hartgrink, H.H.; Gelderblom, H.; Nortier, J.W.; Tollenaar, R.A.; et al. Isolated hepatic perfusion with oxaliplatin combined with 100 mg melphalan in patients with metastases confined to the liver: A phase I study. Eur. J. Surg. Oncol. 2014, 40, 1557–1563. [Google Scholar] [CrossRef]
  39. Yamamoto, A.; Chervoneva, I.; Sullivan, K.L.; Eschelman, D.J.; Gonsalves, C.F.; Mastrangelo, M.J.; Berd, D.; Shields, J.A.; Shields, C.L.; Terai, M.; et al. High-dose immunoembolization: Survival benefit in patients with hepatic metastases from uveal melanoma. Radiology 2009, 252, 290–298. [Google Scholar] [CrossRef] [Green Version]
  40. Huppert, P.E.; Fierlbeck, G.; Pereira, P.; Schanz, S.; Duda, S.H.; Wietholtz, H.; Rozeik, C.; Claussen, C.D. Transarterial chemoembolization of liver metastases in patients with uveal melanoma. Eur. J. Radiol. 2010, 74, e38–e44. [Google Scholar] [CrossRef] [PubMed]
  41. Fiorentini, G.; Aliberti, C.; Del Conte, A.; Tilli, M.; Rossi, S.; Ballardini, P.; Turrisi, G.; Benea, G. Intra-arterial hepatic chemoembolization (TACE) of liver metastases from ocular melanoma with slow-release irinotecan-eluting beads. Early results of a phase II clinical study. Vivo 2009, 23, 131–137. [Google Scholar]
  42. Voelter, V.; Schalenbourg, A.; Pampallona, S.; Peters, S.; Halkic, N.; Denys, A.; Goitein, G.; Zografos, L.; Leyvraz, S. Adjuvant intra-arterial hepatic fotemustine for high-risk uveal melanoma patients. Melanoma Res. 2008, 18, 220–224. [Google Scholar] [CrossRef]
  43. van Iersel, L.B.; Hoekman, E.J.; Gelderblom, H.; Vahrmeijer, A.L.; van Persijn van Meerten, E.L.; Tijl, F.G.; Hartgrink, H.H.; Kuppen, P.J.; Nortier, J.W.; Tollenaar, R.A.; et al. Isolated hepatic perfusion with 200 mg melphalan for advanced noncolorectal liver metastases. Ann. Surg. Oncol. 2008, 15, 1891–1898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Noter, S.L.; Rothbarth, J.; Pijl, M.E.; Keunen, J.E.; Hartgrink, H.H.; Tijl, F.G.; Kuppen, P.J.; van de Velde, C.J.; Tollenaar, R.A. Isolated hepatic perfusion with high-dose melphalan for the treatment of uveal melanoma metastases confined to the liver. Melanoma Res. 2004, 14, 67–72. [Google Scholar] [CrossRef]
  45. Egerer, G.; Lehnert, T.; Max, R.; Naeher, H.; Keilholz, U.; Ho, A.D. Pilot study of hepatic intraarterial fotemustine chemotherapy for liver metastases from uveal melanoma: A single-center experience with seven patients. Int. J. Clin. Oncol. 2001, 6, 25–28. [Google Scholar] [CrossRef] [PubMed]
  46. Hussain, R.N.; Heimann, H.; Damato, B. Neoadjuvant intravitreal ranibizumab treatment in high-risk ocular melanoma patients: A two-stage single-centre phase II single-arm study. Melanoma Res. 2020, 30, 102–106. [Google Scholar] [CrossRef] [PubMed]
  47. Favilla, I.; Favilla, M.L.; Gosbell, A.D.; Barry, W.R.; Ellims, P.; Hill, J.S.; Byrne, J.R. Photodynamic therapy: A 5-year study of its effectiveness in the treatment of posterior uveal melanoma, and evaluation of haematoporphyrin uptake and photocytotoxicity of melanoma cells in tissue culture. Melanoma Res. 1995, 5, 355–364. [Google Scholar] [CrossRef] [PubMed]
  48. Nomura, M.; Oze, I.; Masuishi, T.; Yokota, T.; Satake, H.; Iwasawa, S.; Kato, K.; Andoh, M. Multicenter prospective phase II trial of nivolumab in patients with unresectable or metastatic mucosal melanoma. Int. J. Clin. Oncol. 2020, 25, 972–977. [Google Scholar] [CrossRef]
  49. Luke, J.J.; Olson, D.J.; Allred, J.B.; Strand, C.A.; Bao, R.; Zha, Y.; Carll, T.; Labadie, B.W.; Bastos, B.R.; Butler, M.O.; et al. Randomized Phase II Trial and Tumor Mutational Spectrum Analysis from Cabozantinib versus Chemotherapy in Metastatic Uveal Melanoma (Alliance A091201). Clin. Cancer Res. 2020, 26, 804–811. [Google Scholar] [CrossRef] [Green Version]
  50. Piha-Paul, S.A.; Sachdev, J.C.; Barve, M.; LoRusso, P.; Szmulewitz, R.; Patel, S.P.; Lara, P.N., Jr.; Chen, X.; Hu, B.; Freise, K.J.; et al. First-in-Human Study of Mivebresib (ABBV-075), an Oral Pan-Inhibitor of Bromodomain and Extra Terminal Proteins, in Patients with Relapsed/Refractory Solid Tumors. Clin. Cancer Res. 2019, 25, 6309–6319. [Google Scholar] [CrossRef] [Green Version]
  51. Binkley, E.; Triozzi, P.L.; Rybicki, L.; Achberger, S.; Aldrich, W.; Singh, A. A prospective trial of adjuvant therapy for high-risk uveal melanoma: Assessing 5-year survival outcomes. Br. J. Ophthalmol. 2020, 104, 524–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Johnson, D.B.; Bao, R.; Ancell, K.K.; Daniels, A.B.; Wallace, D.; Sosman, J.A.; Luke, J.J. Response to Anti-PD-1 in Uveal Melanoma Without High-Volume Liver Metastasis. J. Natl. Compr. Cancer Netw. 2019, 17, 114–117. [Google Scholar] [CrossRef] [Green Version]
  53. Shah, S.; Luke, J.J.; Jacene, H.A.; Chen, T.; Giobbie-Hurder, A.; Ibrahim, N.; Buchbinder, E.L.; McDermott, D.F.; Flaherty, K.T.; Sullivan, R.J.; et al. Results from phase II trial of HSP90 inhibitor, STA-9090 (ganetespib), in metastatic uveal melanoma. Melanoma Res. 2018, 28, 605–610. [Google Scholar] [CrossRef]
  54. Garcia, M.; Moreno, R.; Gil-Martin, M.; Cascallo, M.; de Olza, M.O.; Cuadra, C.; Piulats, J.M.; Navarro, V.; Domenech, M.; Alemany, R.; et al. A Phase 1 Trial of Oncolytic Adenovirus ICOVIR-5 Administered Intravenously to Cutaneous and Uveal Melanoma Patients. Hum. Gene 2019, 30, 352–364. [Google Scholar] [CrossRef]
  55. Carvajal, R.D.; Piperno-Neumann, S.; Kapiteijn, E.; Chapman, P.B.; Frank, S.; Joshua, A.M.; Piulats, J.M.; Wolter, P.; Cocquyt, V.; Chmielowski, B.; et al. Selumetinib in Combination With Dacarbazine in Patients With Metastatic Uveal Melanoma: A Phase III, Multicenter, Randomized Trial (SUMIT). J. Clin. Oncol. 2018, 36, 1232–1239. [Google Scholar] [CrossRef] [Green Version]
  56. Schinzari, G.; Rossi, E.; Cassano, A.; Dadduzio, V.; Quirino, M.; Pagliara, M.; Blasi, M.A.; Barone, C. Cisplatin, dacarbazine and vinblastine as first line chemotherapy for liver metastatic uveal melanoma in the era of immunotherapy: A single institution phase II study. Melanoma Res. 2017, 27, 591–595. [Google Scholar] [CrossRef] [PubMed]
  57. Daud, A.; Kluger, H.M.; Kurzrock, R.; Schimmoller, F.; Weitzman, A.L.; Samuel, T.A.; Moussa, A.H.; Gordon, M.S.; Shapiro, G.I. Phase II randomised discontinuation trial of the MET/VEGF receptor inhibitor cabozantinib in metastatic melanoma. Br. J. Cancer 2017, 116, 432–440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Naing, A.; Papadopoulos, K.P.; Autio, K.A.; Ott, P.A.; Patel, M.R.; Wong, D.J.; Falchook, G.S.; Pant, S.; Whiteside, M.; Rasco, D.R.; et al. Safety, Antitumor Activity, and Immune Activation of Pegylated Recombinant Human Interleukin-10 (AM0010) in Patients With Advanced Solid Tumors. J. Clin. Oncol. 2016, 34, 3562–3569. [Google Scholar] [CrossRef] [PubMed]
  59. Carvajal, R.D.; Sosman, J.A.; Quevedo, J.F.; Milhem, M.M.; Joshua, A.M.; Kudchadkar, R.R.; Linette, G.P.; Gajewski, T.F.; Lutzky, J.; Lawson, D.H.; et al. Effect of selumetinib vs chemotherapy on progression-free survival in uveal melanoma: A randomized clinical trial. JAMA 2014, 311, 2397–2405. [Google Scholar] [CrossRef]
  60. Adjei, A.A.; LoRusso, P.; Ribas, A.; Sosman, J.A.; Pavlick, A.; Dy, G.K.; Zhou, X.; Gangolli, E.; Kneissl, M.; Faucette, S.; et al. A phase I dose-escalation study of TAK-733, an investigational oral MEK inhibitor, in patients with advanced solid tumors. Investig. New Drugs 2017, 35, 47–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Mouriaux, F.; Servois, V.; Parienti, J.J.; Lesimple, T.; Thyss, A.; Dutriaux, C.; Neidhart-Berard, E.M.; Penel, N.; Delcambre, C.; Peyro Saint Paul, L.; et al. Sorafenib in metastatic uveal melanoma: Efficacy, toxicity and health-related quality of life in a multicentre phase II study. Br. J. Cancer 2016, 115, 20–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Shoushtari, A.N.; Ong, L.T.; Schoder, H.; Singh-Kandah, S.; Abbate, K.T.; Postow, M.A.; Callahan, M.K.; Wolchok, J.; Chapman, P.B.; Panageas, K.S.; et al. A phase 2 trial of everolimus and pasireotide long-acting release in patients with metastatic uveal melanoma. Melanoma Res. 2016, 26, 272–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Joshua, A.M.; Monzon, J.G.; Mihalcioiu, C.; Hogg, D.; Smylie, M.; Cheng, T. A phase 2 study of tremelimumab in patients with advanced uveal melanoma. Melanoma Res. 2015, 25, 342–347. [Google Scholar] [CrossRef]
  64. Zimmer, L.; Vaubel, J.; Mohr, P.; Hauschild, A.; Utikal, J.; Simon, J.; Garbe, C.; Herbst, R.; Enk, A.; Kampgen, E.; et al. Phase II DeCOG-study of ipilimumab in pretreated and treatment-naive patients with metastatic uveal melanoma. PLoS ONE 2015, 10, e0118564. [Google Scholar] [CrossRef]
  65. Lee, C.K.; Jung, M.; Choi, H.J.; Kim, H.R.; Kim, H.S.; Roh, M.R.; Ahn, J.B.; Chung, H.C.; Heo, S.J.; Rha, S.Y.; et al. Results of a Phase II Study to Evaluate the Efficacy of Docetaxel and Carboplatin in Metastatic Malignant Melanoma Patients Who Failed First-Line Therapy Containing Dacarbazine. Cancer Res. Treat. 2015, 47, 781–789. [Google Scholar] [CrossRef]
  66. Dickson, M.A.; Gordon, M.S.; Edelman, G.; Bendell, J.C.; Kudchadkar, R.R.; LoRusso, P.M.; Johnston, S.H.; Clary, D.O.; Schwartz, G.K. Phase I study of XL281 (BMS-908662), a potent oral RAF kinase inhibitor, in patients with advanced solid tumors. Investig. New Drugs 2015, 33, 349–356. [Google Scholar] [CrossRef]
  67. Homsi, J.; Bedikian, A.Y.; Papadopoulos, N.E.; Kim, K.B.; Hwu, W.J.; Mahoney, S.L.; Hwu, P. Phase 2 open-label study of weekly docosahexaenoic acid-paclitaxel in patients with metastatic uveal melanoma. Melanoma Res. 2010, 20, 507–510. [Google Scholar] [CrossRef]
  68. Borden, E.C.; Jacobs, B.; Hollovary, E.; Rybicki, L.; Elson, P.; Olencki, T.; Triozzi, P. Gene regulatory and clinical effects of interferon beta in patients with metastatic melanoma: A phase II trial. J. Interferon Cytokine Res. 2011, 31, 433–440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Danielli, R.; Ridolfi, R.; Chiarion-Sileni, V.; Queirolo, P.; Testori, A.; Plummer, R.; Boitano, M.; Calabro, L.; Rossi, C.D.; Giacomo, A.M.; et al. Ipilimumab in pretreated patients with metastatic uveal melanoma: Safety and clinical efficacy. Cancer Immunol. Immunother. 2012, 61, 41–48. [Google Scholar] [CrossRef]
  70. Tarhini, A.A.; Frankel, P.; Margolin, K.A.; Christensen, S.; Ruel, C.; Shipe-Spotloe, J.; Gandara, D.R.; Chen, A.; Kirkwood, J.M. Aflibercept (VEGF Trap) in inoperable stage III or stage iv melanoma of cutaneous or uveal origin. Clin. Cancer Res. 2011, 17, 6574–6581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Bhatia, S.; Moon, J.; Margolin, K.A.; Weber, J.S.; Lao, C.D.; Othus, M.; Aparicio, A.M.; Ribas, A.; Sondak, V.K. Phase II trial of sorafenib in combination with carboplatin and paclitaxel in patients with metastatic uveal melanoma: SWOG S0512. PLoS ONE 2012, 7, e48787. [Google Scholar] [CrossRef]
  72. Falchook, G.S.; Lewis, K.D.; Infante, J.R.; Gordon, M.S.; Vogelzang, N.J.; DeMarini, D.J.; Sun, P.; Moy, C.; Szabo, S.A.; Roadcap, L.T.; et al. Activity of the oral MEK inhibitor trametinib in patients with advanced melanoma: A phase 1 dose-escalation trial. Lancet Oncol. 2012, 13, 782–789. [Google Scholar] [CrossRef] [Green Version]
  73. Ott, P.A.; Carvajal, R.D.; Pandit-Taskar, N.; Jungbluth, A.A.; Hoffman, E.W.; Wu, B.W.; Bomalaski, J.S.; Venhaus, R.; Pan, L.; Old, L.J.; et al. Phase I/II study of pegylated arginine deiminase (ADI-PEG 20) in patients with advanced melanoma. Investig. New Drugs 2013, 31, 425–434. [Google Scholar] [CrossRef] [Green Version]
  74. Mahipal, A.; Tijani, L.; Chan, K.; Laudadio, M.; Mastrangelo, M.J.; Sato, T. A pilot study of sunitinib malate in patients with metastatic uveal melanoma. Melanoma Res. 2012, 22, 440–446. [Google Scholar] [CrossRef]
  75. Lane, A.M.; Egan, K.M.; Harmon, D.; Holbrook, A.; Munzenrider, J.E.; Gragoudas, E.S. Adjuvant interferon therapy for patients with uveal melanoma at high risk of metastasis. Ophthalmology 2009, 116, 2206–2212. [Google Scholar] [CrossRef] [PubMed]
  76. Hofmann, U.B.; Kauczok-Vetter, C.S.; Houben, R.; Becker, J.C. Overexpression of the KIT/SCF in uveal melanoma does not translate into clinical efficacy of imatinib mesylate. Clin. Cancer Res. 2009, 15, 324–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Bedikian, A.Y.; Papadopoulos, N.E.; Kim, K.B.; Vardeleon, A.; Smith, T.; Lu, B.; Deitcher, S.R. A pilot study with vincristine sulfate liposome infusion in patients with metastatic melanoma. Melanoma Res. 2008, 18, 400–404. [Google Scholar] [CrossRef]
  78. Adjei, A.A.; Cohen, R.B.; Franklin, W.; Morris, C.; Wilson, D.; Molina, J.R.; Hanson, L.J.; Gore, L.; Chow, L.; Leong, S.; et al. Phase I pharmacokinetic and pharmacodynamic study of the oral, small-molecule mitogen-activated protein kinase kinase 1/2 inhibitor AZD6244 (ARRY-142886) in patients with advanced cancers. J. Clin. Oncol. 2008, 26, 2139–2146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Schmittel, A.; Schmidt-Hieber, M.; Martus, P.; Bechrakis, N.E.; Schuster, R.; Siehl, J.M.; Foerster, M.H.; Thiel, E.; Keilholz, U. A randomized phase II trial of gemcitabine plus treosulfan versus treosulfan alone in patients with metastatic uveal melanoma. Ann. Oncol. 2006, 17, 1826–1829. [Google Scholar] [CrossRef]
  80. Richtig, E.; Langmann, G.; Schlemmer, G.; Mullner, K.; Papaefthymiou, G.; Bergthaler, P.; Smolle, J. Safety and efficacy of interferon alfa-2b in the adjuvant treatment of uveal melanoma. Ophthalmologe 2006, 103, 506–511. [Google Scholar] [CrossRef] [PubMed]
  81. O’Neill, P.A.; Butt, M.; Eswar, C.V.; Gillis, P.; Marshall, E. A prospective single arm phase II study of dacarbazine and treosulfan as first-line therapy in metastatic uveal melanoma. Melanoma Res. 2006, 16, 245–248. [Google Scholar] [CrossRef]
  82. Schmittel, A.; Schuster, R.; Bechrakis, N.E.; Siehl, J.M.; Foerster, M.H.; Thiel, E.; Keilholz, U. A two-cohort phase II clinical trial of gemcitabine plus treosulfan in patients with metastatic uveal melanoma. Melanoma Res. 2005, 15, 447–451. [Google Scholar] [CrossRef]
  83. Corrie, P.G.; Shaw, J.; Spanswick, V.J.; Sehmbi, R.; Jonson, A.; Mayer, A.; Bulusu, R.; Hartley, J.A.; Cree, I.A. Phase I trial combining gemcitabine and treosulfan in advanced cutaneous and uveal melanoma patients. Br. J. Cancer 2005, 92, 1997–2003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Schmittel, A.; Scheulen, M.E.; Bechrakis, N.E.; Strumberg, D.; Baumgart, J.; Bornfeld, N.; Foerster, M.H.; Thiel, E.; Keilholz, U. Phase II trial of cisplatin, gemcitabine and treosulfan in patients with metastatic uveal melanoma. Melanoma Res. 2005, 15, 205–207. [Google Scholar] [CrossRef] [PubMed]
  85. Schmidt-Hieber, M.; Schmittel, A.; Thiel, E.; Keilholz, U. A phase II study of bendamustine chemotherapy as second-line treatment in metastatic uveal melanoma. Melanoma Res. 2004, 14, 439–442. [Google Scholar] [CrossRef] [PubMed]
  86. Keilholz, U.; Schuster, R.; Schmittel, A.; Bechrakis, N.; Siehl, J.; Foerster, M.H.; Thiel, E. A clinical phase I trial of gemcitabine and treosulfan in uveal melanoma and other solid tumours. Eur. J. Cancer 2004, 40, 2047–2052. [Google Scholar] [CrossRef]
  87. Terheyden, P.; Brocker, E.B.; Becker, J.C. Clinical evaluation of in vitro chemosensitivity testing: The example of uveal melanoma. J. Cancer Res. Clin. Oncol. 2004, 130, 395–399. [Google Scholar] [CrossRef]
  88. Pfohler, C.; Cree, I.A.; Ugurel, S.; Kuwert, C.; Haass, N.; Neuber, K.; Hengge, U.; Corrie, P.G.; Zutt, M.; Tilgen, W.; et al. Treosulfan and gemcitabine in metastatic uveal melanoma patients: Results of a multicenter feasibility study. Anticancer Drugs 2003, 14, 337–340. [Google Scholar] [CrossRef]
  89. Bedikian, A.Y.; Papadopoulos, N.; Plager, C.; Eton, O.; Ring, S. Phase II evaluation of temozolomide in metastatic choroidal melanoma. Melanoma Res. 2003, 13, 303–306. [Google Scholar] [CrossRef]
  90. Kivela, T.; Suciu, S.; Hansson, J.; Kruit, W.H.; Vuoristo, M.S.; Kloke, O.; Gore, M.; Hahka-Kemppinen, M.; Parvinen, L.M.; Kumpulainen, E.; et al. Bleomycin, vincristine, lomustine and dacarbazine (BOLD) in combination with recombinant interferon alpha-2b for metastatic uveal melanoma. Eur. J. Cancer 2003, 39, 1115–1120. [Google Scholar] [CrossRef]
  91. Pyrhonen, S.; Hahka-Kemppinen, M.; Muhonen, T.; Nikkanen, V.; Eskelin, S.; Summanen, P.; Tarkkanen, A.; Kivela, T. Chemoimmunotherapy with bleomycin, vincristine, lomustine, dacarbazine (BOLD), and human leukocyte interferon for metastatic uveal melanoma. Cancer 2002, 95, 2366–2372. [Google Scholar] [CrossRef]
  92. Becker, J.C.; Terheyden, P.; Kampgen, E.; Wagner, S.; Neumann, C.; Schadendorf, D.; Steinmann, A.; Wittenberg, G.; Lieb, W.; Brocker, E.B. Treatment of disseminated ocular melanoma with sequential fotemustine, interferon alpha, and interleukin 2. Br. J. Cancer 2002, 87, 840–845. [Google Scholar] [CrossRef]
  93. Ellerhorst, J.A.; Bedikian, A.Y.; Smith, T.M.; Papadopoulos, N.E.; Plager, C.; Eton, O. Phase II trial of 9-nitrocamptothecin (RFS 2000) for patients with metastatic cutaneous or uveal melanoma. Anticancer Drugs 2002, 13, 169–172. [Google Scholar] [CrossRef]
  94. Mertens, W.C.; Lohmann, R.C. Oral indomethacin and ranitidine in advanced melanoma: A phase II study. Clin. Oncol. (R. Coll. Radiol.) 1996, 8, 112–115. [Google Scholar] [CrossRef]
  95. Penel, N.; Delcambre, C.; Durando, X.; Clisant, S.; Hebbar, M.; Negrier, S.; Fournier, C.; Isambert, N.; Mascarelli, F.; Mouriaux, F. O-Mel-Inib: A Cancero-pole Nord-Ouest multicenter phase II trial of high-dose imatinib mesylate in metastatic uveal melanoma. Investig. New Drugs 2008, 26, 561–565. [Google Scholar] [CrossRef]
  96. Kompella, U.B.; Amrite, A.C.; Pacha Ravi, R.; Durazo, S.A. Nanomedicines for back of the eye drug delivery, gene delivery, and imaging. Prog. Retin. Eye Res. 2013, 36, 172–198. [Google Scholar] [CrossRef] [Green Version]
  97. You, S.; Luo, J.; Grossniklaus, H.E.; Gou, M.L.; Meng, K.; Zhang, Q. Nanomedicine in the application of uveal melanoma. Int. J. Ophthalmol. 2016, 9, 1215–1225. [Google Scholar] [CrossRef]
  98. Nayak, K.; Misra, M. A review on recent drug delivery systems for posterior segment of eye. Biomed. Pharm. 2018, 107, 1564–1582. [Google Scholar] [CrossRef] [PubMed]
  99. Conti, B.; Bucolo, C.; Giannavola, C.; Puglisi, G.; Giunchedi, P.; Conte, U. Biodegradable microspheres for the intravitreal administration of acyclovir: In vitro/in vivo evaluation. Eur. J. Pharm. Sci. 1997, 5, 287–293. [Google Scholar] [CrossRef]
  100. Kompella, U.B.; Kadam, R.S.; Lee, V.H. Recent advances in ophthalmic drug delivery. Ther. Deliv. 2010, 1, 435–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Rowe-Rendleman, C.L.; Durazo, S.A.; Kompella, U.B.; Rittenhouse, K.D.; Di Polo, A.; Weiner, A.L.; Grossniklaus, H.E.; Naash, M.I.; Lewin, A.S.; Horsager, A.; et al. Drug and gene delivery to the back of the eye: From bench to bedside. Investig. Ophthalmol. Vis. Sci. 2014, 55, 2714–2730. [Google Scholar] [CrossRef] [Green Version]
  102. Beiu, C.; Giurcaneanu, C.; Grumezescu, A.M.; Holban, A.M.; Popa, L.G.; Mihai, M.M. Nanosystems for Improved Targeted Therapies in Melanoma. J. Clin. Med. 2020, 9, 318. [Google Scholar] [CrossRef] [Green Version]
  103. Fallico, M.; Maugeri, A.; Romano, G.L.; Bucolo, C.; Longo, A.; Bonfiglio, V.; Russo, A.; Avitabile, T.; Barchitta, M.; Agodi, A.; et al. Epiretinal Membrane Vitrectomy With and Without Intraoperative Intravitreal Dexamethasone Implant: A Systematic Review With Meta-Analysis. Front. Pharmacol. 2021, 12, 635101. [Google Scholar] [CrossRef]
  104. Triozzi, P.L.; Singh, A.D. Adjuvant Therapy of Uveal Melanoma: Current Status. Ocul. Oncol. Pathol. 2014, 1, 54–62. [Google Scholar] [CrossRef] [Green Version]
  105. Singh, A.D.; Bergman, L.; Seregard, S. Uveal melanoma: Epidemiologic aspects. Ophthalmol. Clin. N. Am. 2005, 18, 75–84. [Google Scholar] [CrossRef] [PubMed]
  106. Krantz, B.A.; Dave, N.; Komatsubara, K.M.; Marr, B.P.; Carvajal, R.D. Uveal melanoma: Epidemiology, etiology, and treatment of primary disease. Clin. Ophthalmol. 2017, 11, 279–289. [Google Scholar] [CrossRef] [Green Version]
  107. Damato, B.; Duke, C.; Coupland, S.E.; Hiscott, P.; Smith, P.A.; Campbell, I.; Douglas, A.; Howard, P. Cytogenetics of uveal melanoma: A 7-year clinical experience. Ophthalmology 2007, 114, 1925–1931. [Google Scholar] [CrossRef] [PubMed]
  108. Eleuteri, A.; Taktak, A.F.G.; Coupland, S.E.; Heimann, H.; Kalirai, H.; Damato, B. Prognostication of metastatic death in uveal melanoma patients: A Markov multi-state model. Comput. Biol. Med. 2018, 102, 151–156. [Google Scholar] [CrossRef] [PubMed]
  109. Lorenzo, D.; Piulats, J.M.; Ochoa, M.; Arias, L.; Gutierrez, C.; Catala, J.; Cobos, E.; Garcia-Bru, P.; Dias, B.; Padron-Perez, N.; et al. Clinical predictors of survival in metastatic uveal melanoma. Jpn. J. Ophthalmol. 2019, 63, 197–209. [Google Scholar] [CrossRef]
  110. Souto, E.B.; Zielinska, A.; Luis, M.; Carbone, C.; Martins-Gomes, C.; Souto, S.B.; Silva, A.M. Uveal melanoma: Physiopathology and new in situ-specific therapies. Cancer Chemother. Pharm. 2019, 84, 15–32. [Google Scholar] [CrossRef] [Green Version]
  111. NCCN. N.C.C.N. Uveal Melanoma. Version 2. 2021. Available online: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1488 (accessed on 1 August 2021).
  112. Seth, R.; Messersmith, H.; Kaur, V.; Kirkwood, J.M.; Kudchadkar, R.; McQuade, J.L.; Provenzano, A.; Swami, U.; Weber, J.; Alluri, K.C.; et al. Systemic Therapy for Melanoma: ASCO Guideline. J. Clin. Oncol. 2020, 38, 3947–3970. [Google Scholar] [CrossRef]
  113. Yang, J.; Manson, D.K.; Marr, B.P.; Carvajal, R.D. Treatment of uveal melanoma: Where are we now? Adv. Med. Oncol. 2018, 10, 1758834018757175. [Google Scholar] [CrossRef]
  114. Spagnolo, F.; Caltabiano, G.; Queirolo, P. Uveal melanoma. Cancer Treat. Rev. 2012, 38, 549–553. [Google Scholar] [CrossRef]
  115. Spagnolo, F.; Grosso, M.; Picasso, V.; Tornari, E.; Pesce, M.; Queirolo, P. Treatment of metastatic uveal melanoma with intravenous fotemustine. Melanoma Res. 2013, 23, 196–198. [Google Scholar] [CrossRef]
  116. Jonsson, B.; Hampson, G.; Michaels, J.; Towse, A.; von der Schulenburg, J.G.; Wong, O. Advanced therapy medicinal products and health technology assessment principles and practices for value-based and sustainable healthcare. Eur. J. Health Econ. HEPAC Health Econ. Prev. Care 2019, 20, 427–438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Helgadottir, H.; Hoiom, V. The genetics of uveal melanoma: Current insights. Appl. Clin. Genet. 2016, 9, 147–155. [Google Scholar] [CrossRef] [Green Version]
  118. Javed, A.; Arguello, D.; Johnston, C.; Gatalica, Z.; Terai, M.; Weight, R.M.; Orloff, M.; Mastrangelo, M.J.; Sato, T. PD-L1 expression in tumor metastasis is different between uveal melanoma and cutaneous melanoma. Immunotherapy 2017, 9, 1323–1330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Maio, M.; Danielli, R.; Chiarion-Sileni, V.; Pigozzo, J.; Parmiani, G.; Ridolfi, R.; De Rosa, F.; Del Vecchio, M.; Di Guardo, L.; Queirolo, P.; et al. Efficacy and safety of ipilimumab in patients with pre-treated, uveal melanoma. Ann. Oncol. 2013, 24, 2911–2915. [Google Scholar] [CrossRef] [PubMed]
  120. Kelderman, S.; van der Kooij, M.K.; van den Eertwegh, A.J.; Soetekouw, P.M.; Jansen, R.L.; van den Brom, R.R.; Hospers, G.A.; Haanen, J.B.; Kapiteijn, E.; Blank, C.U. Ipilimumab in pretreated metastastic uveal melanoma patients. Results of the Dutch Working group on Immunotherapy of Oncology (WIN-O). Acta Oncol. 2013, 52, 1786–1788. [Google Scholar] [CrossRef] [Green Version]
  121. Luke, J.J.; Callahan, M.K.; Postow, M.A.; Romano, E.; Ramaiya, N.; Bluth, M.; Giobbie-Hurder, A.; Lawrence, D.P.; Ibrahim, N.; Ott, P.A.; et al. Clinical activity of ipilimumab for metastatic uveal melanoma: A retrospective review of the Dana-Farber Cancer Institute, Massachusetts General Hospital, Memorial Sloan-Kettering Cancer Center, and University Hospital of Lausanne experience. Cancer 2013, 119, 3687–3695. [Google Scholar] [CrossRef]
  122. Piulats Rodriguez, J.M.; Ochoa de Olza, M.; Codes, M.; Lopez-Martin, J.A.; Berrocal, A.; García, M.; Gurpide, A.; Homet, B.; Martin-Algarra, S. Phase II study evaluating ipilimumab as a single agent in the first-line treatment of adult patients (Pts) with metastatic uveal melanoma (MUM): The GEM-1 trial. J. Clin. Oncol. 2014, 32, 9033. [Google Scholar] [CrossRef]
  123. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Algazi, A.P.; Tsai, K.K.; Shoushtari, A.N.; Munhoz, R.R.; Eroglu, Z.; Piulats, J.M.; Ott, P.A.; Johnson, D.B.; Hwang, J.; Daud, A.I.; et al. Clinical outcomes in metastatic uveal melanoma treated with PD-1 and PD-L1 antibodies. Cancer 2016, 122, 3344–3353. [Google Scholar] [CrossRef]
  125. Pelster, M.S.; Gruschkus, S.K.; Bassett, R.; Gombos, D.S.; Shephard, M.; Posada, L.; Glover, M.S.; Simien, R.; Diab, A.; Hwu, P.; et al. Nivolumab and Ipilimumab in Metastatic Uveal Melanoma: Results From a Single-Arm Phase II Study. J. Clin. Oncol. 2021, 39, 599–607. [Google Scholar] [CrossRef] [PubMed]
  126. Damato, B.E.; Dukes, J.; Goodall, H.; Carvajal, R.D. Tebentafusp: T Cell Redirection for the Treatment of Metastatic Uveal Melanoma. Cancers 2019, 11, 971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. ESMO. Promising Results are Provided by Tebentafusp in Metastatic Uveal Melanoma. Available online: https://www.esmo.org/oncology-news/promising-results-are-provided-by-tebentafusp-in-metastatic-uveal-melanoma (accessed on 15 January 2021).
  128. Sacco, J.J.; Carvajal, R.; Butler, M.O.; Shoushtari, A.N.; Hassel, J.C.; Ikeguchi, A.; Hernandez-Aya, L.; Nathan, P.; Hamid, O.; Piulats Rodriguez, J.M.; et al. A Phase (ph) II, Multi-Center Study of the Safety and Efficacy of Tebentafusp (Tebe) (IMCgp100) in Patients (pts) with Metastatic Uveal Melanoma (mUM). Available online: https://oncologypro.esmo.org/meeting-resources/esmo-immuno-oncology-virtual-congress-2020/a-phase-ph-ii-multi-center-study-of-the-safety-and-efficacy-of-tebentafusp-tebe-imcgp100-in-patients-pts-with-metastatic-uveal-melanoma-mum (accessed on 15 January 2021).
  129. Immunocore. Novel Immunotherapy Tebentafusp Granted Fast Track Designation for Metastatic Uveal Melanoma. Available online: https://www.targetedonc.com/view/novel-immunotherapy-tebentafusp-granted-fast-track-designation-for-metastatic-uveal-melanoma (accessed on 15 January 2021).
  130. Immunocore. Immunocore’s IMCgp100 Receives Promising Innovative Medicine (PIM) Designation under UK Early Access to Medicines Scheme (EAMS) for the Treatment of Patients with Uveal Melanoma. 11 December 2017. Available online: https://bit.ly/2UUPKNU (accessed on 15 January 2021).
  131. Vivet-Noguer, R.; Tarin, M.; Roman-Roman, S.; Alsafadi, S. Emerging Therapeutic Opportunities Based on Current Knowledge of Uveal Melanoma Biology. Cancers 2019, 11, 1019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Russo, A.; Ragusa, M.; Barbagallo, C.; Longo, A.; Avitabile, T.; Uva, M.G.; Bonfiglio, V.; Toro, M.D.; Caltabiano, R.; Mariotti, C.; et al. miRNAs in the vitreous humor of patients affected by idiopathic epiretinal membrane and macular hole. PLoS ONE 2017, 12, e0174297. [Google Scholar] [CrossRef]
  133. Toro, M.D.; Reibaldi, M.; Avitabile, T.; Bucolo, C.; Salomone, S.; Rejdak, R.; Nowomiejska, K.; Tripodi, S.; Posarelli, C.; Ragusa, M.; et al. MicroRNAs in the Vitreous Humor of Patients with Retinal Detachment and a Different Grading of Proliferative Vitreoretinopathy: A Pilot Study. Transl. Vis. Sci. Technol. 2020, 9, 23. [Google Scholar] [CrossRef]
Table 1. Studies about management of local complications in patients with uveal melanoma treated with radiation-therapy.
Table 1. Studies about management of local complications in patients with uveal melanoma treated with radiation-therapy.
NAuthor, YearIDStudy DesignClinical OutcomesEyesPatientsPrimary TreatmentFollow-Up
1Schefler AC et al., 2020 [32]NCT02222610Phase IIb, multicenter, prospective, RCTETDRS BCVA change from baseline4040 patients with radiation-induced macular edema and a resulting decrease in visual acuityIVT 0.5 mg ranibizumab monthly (Cohort A), monthly ranibizumab with TRP (Cohort B), or 3 monthly ranibizumab followed by as needed (PRN) injections, and TRP (Cohort C)2 years
2Murray TG et al., 2019 [33]/Randomized, prospective clinical trialBCVA and SD OCT central retinal thickness at 1 year/39 patients with visually compromising radiation maculopathy confirmed by a decline in BCVA and SD OCT documentation of radiation maculopathyAflibercept treatment via 1 of 2 regimens: (1) fixed, every-6-weeks treatment or (2) variable, treat-and-adjust treatment centered around 6 weeks1 year
3Horgan N et al., 2009 [34]NCT00441662Prospective, RCT.Development of macular edema; visual acuity/163 patients (55 patients randomized to the control group and 108 to the triamcinolone group)All patients were treated with iodine 125 plaque radiotherapy (8000 cGy to the tumor apex); adjuvant TTT applied to the tumor in most cases. Patients in the triamcinolone group received 3 periocular injections of triamcinolone (at the completion of plaque application, at 4- and 8-month follow-up)18 months
BCVA = best-corrected visual acuity; IVT = intravitreal; PRN = pro re nata; RCT = randomized clinical, trial; SD OCT = spectral-domain optical coherence tomography; TRP = targeted panretinal photocoagulation; TTP = time to progression; TTT = transpupillary thermotherapy.
Table 2. Studies about local treatment of primary tumor or metastases from uveal melanoma.
Table 2. Studies about local treatment of primary tumor or metastases from uveal melanoma.
NAuthor, YearIDStudy DesignClinical OutcomesEyesPatientsPrimary TreatmentFollow-Up
1Venturini M et al., 2012 [35]//Eventual complications and lesion devascularization, and tumor response/5 patients with UM metastatic to the liverTransarterial chemoembolization with DEBIRI as a first-line therapyFrom 8 to 13 months (mean, 10.6 months) after the first chemoembolization procedure
2Olofsson R et al., 2014 [36]NCT01785316Phase II trialOS comparison, made using data retrieved from the National Patient Register/34 patients with isolated liver metastasis from UMIHP2 patients still alive with CR after 23 and 69 months, respectively
3Leyvraz S et al., 2014 [37]2004-002245-12 and NCT00110123Prospective, randomized, phase III trialOS, RR, PFS, and safety/171 patients with liver metastases from UMIV or I.a.H. fotemustine at 100 mg/m2 on days 1, 8, 15 (and 22 in I.a.H. arm only), and after 5-week rest period every 3 weeks as maintenance5.6 years (range 0.25–6 years)
4van Iersel LB et al., 2014 [38]2006-005088-25Phase I dose-escalation studyOptimal oxaliplatin dose in combination with a fixed melphalan dose. Pharmacokinetic analysis, toxicity, response, and survival/11 patients (8 colorectal cancer and 3 UM patients, all with isolated liver metastases)One hour IHP with escalating doses of oxaliplatin combined with 100 mg melphalan71 months
5Yamamoto A et al., 2009 [39]/Retrospective evaluation of patients treated with either CE with BCNU (phase II study) or IE with GM-CSF (phase I/IIa study)Prognostic factors associated with OS and PFS in the liver and extrahepatic organs/53 consecutive patients with UM CE with BCNU or IE with GM-CSF for hepatic metastases/
6Huppert PE et al., 2010 [40]/Pilot trialRadiographic response of the liver metastases, time to progression and OS/14 patients with hepatic metastases from UMTACE repeated with a mean interval of 8 weeks (range: 4–24 weeks)From 5 to 58 months
7Fiorentini G et al., 2009 [41]/Phase II studySafety, feasibility, and tolerance of TACE adopting irinotecan-loaded microspheres, RR, QoL, and survival/10 patients with UM metastatic to the liverTACE with irinotecan (IRI; 100 mg) The median follow-up time was 6.5 months (range 4–9 months)
8Voelter V et al., 2008 [42]//OS/22 patients presenting with nonmetastatic UM at high risk of liver relapseI.a.h. fotemustine (100 mg/m2)Median follow-up was 4.6 years for the experimental group and 8.5 years for the control group
9van Iersel LB et al., 2008 [43]//Systemic and regional toxicity, tumor response, disease-free survival, OS/19 patients with isolated unresectable liver metastases from a variety of tumors (13 UM)IHP using 200 mg melphalan.Median follow-up was 74 months (range 4–137 months)
10Noter SL et al., 2004 [44]//Tumor response, progression and survival, toxicity/8 patients with UM hepatic metastasesIHP with 200 mg melphalan/
11Egerer G et al., 2001 [45]/Single-center experienceSafety and efficacy/7 patients with isolated hepatic metastases from UMFotemustine 100 mg/m2 administered IAH over a 4-h period(1 administration per week for 4 weeks, followed by a 5-week rest period, then every 3 weeks until progression or toxicity)All patients were followed until death or the last visit to the clinic
12Hussain RN et al., 2020 [46]/Phase II, non-randomized, single center trialLocal tumor control/7 patients with primary UM that otherwise required radical surgery because of tumor size.0.5 mg in 0.05 mL of ranibizumab via six IVT injections over 6 months/
13Favilla I et al., 1995 [47]//Response to treatment/36 patients with posterior UMHPD and the photocytotoxicity of PDT5 years
CE = chemoembolization; IE = immunoembolization; DEBIRI = drug-eluting beads preloaded with irinotecan; GM-CSF granulocyte-macrophage colony-stimulating factor; HPD = hematoporphyrin derivative; I.a.h. = Intra-arterial hepatic; IHP = isolated hepatic perfusion; IV = intravenous; IVT = intravitreal; ORR = overall response rate; OS = overall survival; PDT = photodynamic therapy; PFS = progression free survival; PD = progressive disease; PR = partial response; RR = response rate; SD = stable disease; T = TACE = Hepatic transarterial chemoembolization; UM = uveal melanoma.
Table 3. Studies about systemic therapy of uveal melanoma.
Table 3. Studies about systemic therapy of uveal melanoma.
NAuthor, YearIDStudy DesignClinical OutcomesEyesPatientsPrimary TreatmentFollow-Up
1Nomura M et al., 2020 [48]/Phase II prospective, multicenter trialRR; OS, PFS, disease control rate, and toxicity/20 unresectable or metastatic mucosal melanomaNivolumab monotherapy1.6 years (range 1.1–2.8 years)
2Luke JJ et al., 2020 [49]NCT01835145Randomized phase II trialImprovement of the 4-month PFS4 from 15%; PFS, OS, RR, and safety/46 UM that is metastatic or unresectableCabozantinib versus ChemotherapyMedian follow-up time of 2.1 years (range 1.9–2.3 years)
3Piha-Paul SA et al., 2019 [50]/Phase I, open-label, dose escalation studySafety analyses, PK, best response, PFS, and duration of OR/72 patients with relapsed, refractory advanced solid tumors (10 with UM)Mivebresib administered in 28 day-cycles, 3 + 3 dose escalation/
4Binkley E et al., 2020 [51]/Open label phase II studyMFS rate/33 patients with high-risk cytogeneticsAdjuvant therapy (sequential low-dose dacarbazine and interferon-alpha)5 years
5Johnson DB et al., 2019 [52]NCT02359851Single-arm phase II studyORR, PFS, OS, response duration, incidence of AEs/5 patients with metastatic UM naïve to PD-1–directed agentsPembrolizumab Median follow-up 11.1 months
6Shah S et al., 2018 [53]/Phase II trialRR/17 Patients with stage IV UM, and no previous chemotherapyGanetespib 200 mg weekly (cohort A) or 150 mg twice a week (cohort B)/
7García M et al., 2019 [54]/Dose-escalation phase I trialTolerability, efficacy, pharmacokinetics/12 patients with uveal (6) or cutaneous (6) metastatic melanomaOncolytic adenovirus ICOVIR5 administered as a single infusion/
8Carvajal RD et al., 2018 [55]NCT01974752Randomized, double-blind, placebo-controlled, phase III trialPFS, ORR, duration of response, change in tumor size at week 6, OS, safety and tolerability, and QoL/129 Patients metastatic UMSelumetinib or matched placebo, plus dacarbazine until disease progression, intolerable toxicity, or another discontinuation criterionMedian follow-up for PFS in the selumetinib plus dacarbazine and placebo plus dacarbazine groups was 2.7 and 1.5 months, respectively
9Schinzari G et al., 2017 [56]/Phase II studyORR, OS, PFS, and toxicity /25 patients with unresectable metastases of UM and BRAF wild typeCisplatin (80 mg/me, day 1), dacarbazine (250 mg/m2/day, days 1–3), vinblastine (2 mg maximum, day 1) every 21 days /
10Daud A et al., 2017 [57]/Phase II randomized discontinuation trialORR, PFS, safety and tolerability/77 patients with histologically confirmed melanoma (30% UM)Cabozantinib treatment during a 12-week lead-in stage. At week 12, patients with evidence of response remained on open-label cabozantinib/
11Naing A et al., 2016 [58]/Phase I study Safety and tolerability and MTD and PK properties of AM0010; antitumor activity/51 patients (33 patients with CRC, RCC, pancreatic ductal adenocarcinoma, ovarian cancer, prostate cancer, non–small-cell lung cancer, or melanoma were enrolled in the dose-escalation cohorts)6 dose escalation cohorts from 1 to 40 mg/kg/
12Carvajal RD 2014 [59]NCT01143402Phase II trialPFS, OS, RR, and safety/toxicity/101 patients with metastatic UM who had not received prior therapy with temozolomide or DTICPatients were randomized on a 1:1 ratio to selumetinib 75 mg orally twice daily (n = 50) or chemotherapy (temozolomide 150 mg/m2 orally daily for 5 of every 28 days or DTIC 1000 mg/m2 intravenously every 21 days; n = 51) until disease progression, death, intolerable toxicity12 months
13Adjei AA et al., 2017 [60]NCT00948467First in-human, multicenter, open-label, phase I, dose-escalation studySafety profile and DLTs, MTD, and recommended phase II dose (RP2D) of TAK-733, and PK of TAK-733;antitumor activity./51 patients with advanced solid tumors, 12 with UMPatients received oral TAK-733 once daily on days 1–21 of 28-day treatment cyclesBlood samples obtained at the following timepoints: pre-dose, and at 0.25, 0.5, 1, 2, 3, 4, 6, 8, 10, and 24 h post-dose on days 1 and 21 of cycle 1; pre-dose on days 8 and 15 of cycle 1; 48, 72, 96, and 120 h post-dose on day 21 of cycle 1; and pre-dose on day 1 of cycle 2. AEs were monitored throughout the trial and for 30 days after the last dose
14Mouriaux F et al., 2016 [61]EudraCT: 2010-022527-29Single-arm phase II trialNon-progression rate at 24 weeks, PFS, OS, toxicity, QoL/32 patients with metastatic UM400 mg twice a day (800 mg daily) of sorafenib until disease progression or unacceptably severe toxicity or an individual decision was made24 weeks
15Shoushtari AN et al., 2016 [62]NCT01252251Open-label, single-arm, phase II trialCBR, PFS, and OS/14 patients with metastatic UMEverolimus 10 mg orally daily plus pasireotide LAR 60 mg intramuscularly (IM) once every 28 days until progression or unacceptable toxicityMedian time on treatment was 8 weeks (range: 1–23 weeks)
16Joshua AM et al., 2015 [63]/Phase II, multicenter studyPFS at 6 months, ORR, DOR, DCR, and OS/11 patients with metastatic UM15 mg/kg tremelimumab, administered on day 1 of every 90-day cycle for up to 4 cycles or until progression or intolerance of toxicityThe median follow-up was 11 months (range 2–36 months)
17Zimmer L et al., 2015 [64]EudraCT-Number: 2010-021946-22Multicenter, open-label, phase II studyOS rate at 12 months/53 patients with metastatic UMIpilimumab was administered intravenously over 90 min at a dose of 3 mg/kg every 3 weeks for a total of 4 infusions/
18Lee CK et al., 2015 [65]NCT02223884Phase II, open-label, single-arm studyORR, DCR, PFS, OS, and safety/30 malignant melanoma patients who failed chemotherapy containing dacarbazine were enrolled (10 UM)Intravenous docetaxel (35 mg/m2 on days 1 and 8 of each cycle) and carboplatin (area under the curve 3 on days 1 and 8 of each cycle) administered every 21 daysThe median follow-up duration was 19.8 months.
19Dickson MA et al., 2015 [66]NCT00451880Phase I studySafety and tolerability of once daily and twice daily oral administration of XL281 and the MTD; PK and pharmacodynamic effects/160 patients with solid tumors were enrolled in the study (6 UM)XL281 administered orally daily for 28 days (1 cycle). In the absence of progression) or unacceptable toxicity, patients continued on treatment/
20Homsi J et al., 2010 [67]/Phase II open-label studySafety and efficacy/22 patients Docosahexaenoic acid (DHA)-paclitaxel (500 mg/m2/week) was administered by IV infusion for 5 consecutive weeks in a 6-weeks cycle/
21Borden EC et al., 2011 [68]/Phase I trialSafety and efficacy/21 patients with metastatic melanoma (cutaneous metastatic melanoma n = 17, and UM n = 4)IFN-b1a 12 × 106 IU/m2 subcutaneously daily with dose escalation after 14 days and if no adverse events >grade 2 to 18x106 IU/m2, until disease progression or a dose-limiting toxicity/
22Danielli R et al., 2012 [69]/Multicenter expanded access program (EAP)Tumor assessment. Safety./13 pretreated patients with metastatic UMInduction treatment with ipilimumab 10 mg/kg at weeks 1, 4, 7, and 10; maintenance doses in patients with clinical benefit or at physicians’ discretion at week 24/
23Tarhini AA et al., 2011 [70]/Multicenter phase II, single arm study Safety and efficacy./41 patients with stage III or IV melanoma and no prior chemotherapy (10 with primary UM)Aflibercept 4 mg/kg intravenously over at least 1 h on day 1 of each 14-day cycle/
24Bhatia S et al., 2012 [71]NCT00329641Phase II studyORR, PFS, and OS./25 patients with stage IV UM who had received 0–1 prior systemic therapyUp to 6 cycles of carboplatin (AUC = 6) and paclitaxel (225 mg/m2) on day 1 + sorafenib (400 mg twice daily), followed by sorafenib monotherapy until disease progression/
25Falchook GS et al., 2012 [72]NCT00687622Phase I, Dose-escalation TrialDose escalation, cohort expansion, and pharmacodynamic evaluation/97 melanoma patients, including 81 with cutaneous or unknown primary melanoma and 16 UMTrametinib doses ranged from 0.125 mg to 4·.0 mg, administered orally once daily/
26Ott PA et al., 2013 [73]/Phase I/II, open-label, dose-escalation studyToxicity and tumor response/31 previously treated patients with advanced melanoma (six patients with UM)40, 80, or 160 IU/m2 arginine deiminase (ADI)-polyethylene glycol (PEG) 20 i.m. weekly/
27Mahipal A et al., 2012 [74]/Open-label, single-institution pilot studyEvaluation of response carried out every 8 weeks. OS and PFS /20 patients with metastatic UM expressing c-kit, 17 of whom failed previous treatmentsSunitinib malate 37.5 mg daily continuously in 4-week cycles/
28Lane AM et al., 2009 [75]/Non-randomized trialMelanoma-related mortality compared to historical controls/121 patients with choroidal or ciliary body melanoma Adjuvant IFN treatment protocol consisted of 3 MIU IFN administered 3 times per week by subcutaneous injection over a 2-year periodApproximately 9 years
29Hofmann UB et al., 2009 [76]/A clinical study using Simon’s two-stage designORR, OS, time to progression, and toxicity/12 patients with metastatic uveal melanomaImatinib mesylate at a dose of 300 mg p.o. b.i.d. (600 mg daily) until progression or intolerable side effectsPatients were followed for at least 2-month intervals.
30Bedikian AY et al., 2008 [77]/Patients enrolled in two PK studiesSafety, tumor response, and survival/27 adult patients with malignant melanoma with surgically unresectable stage III or stage IV diseaseVincristine sulfate liposome infusion (VSLI) at a dose of2.0 mg/m2 every 2 weeks (one cycle)/
31Penel N et al., 20082005-003685-41Non-randomized phase II trialNon-progression rate at 3 months/13 patients.Imatinib at dose of 400 mg twice per day orally/
32Adjei AA et al., 2008 [78]/Phase I, open-label, multiple-dose studySafety, tolerability, PK, and pharmacodynamics of AZD6244/57 patients with advanced solid malignancies (35% malignant melanoma) Doses of 50, 100, 200, and 300 mg bid. MTD (200 mg bid) or 50% of the MTD dose (100 mg bid) to evaluate the dose that provided the best balance of safety/tolerabilityFor patients carrying RAS and BRAF mutations = median, 3.5 months; range, 1 to 6 months, greater than for those without a mutation (median, 2 months; range, 1 to 4 months)
33Schmittel A et al., 2006 [79]/Randomized phase II trialRate of responses and disease stabilizations, toxic effect, PFS and OS/48 patients1000 mg/m2 of gemcitabine plus 3500 mg/m2 of treosulfan (GeT) or 3500 mg/m2 of treosulfan alone (T)/
34Richtig E et al., 2006 [80]//Relapse-free survival, safety/39 patients with uveal melanoma Adjuvant IFN alfa 2b treatment 3 million units 3 times a week subcutaneously for 1 year after therapy of the primary tumor /
35O’Neill PA et al., 2006 [81]/Prospective single arm phase II studyORR, toxicity/15 previously untreated patients with metastatic UMDacarbazine (850 mg/m2) as an IV infusion and treosulfan (8 g/m2) every 21 days as an outpatient procedure up to a maximum of 6 cycles/
36Schmittel A et al., 2005 [82]/Two-cohort phase II clinical trialSafety and efficacy/33 patients were treated: 14 in cohort 1 and 19 in cohort 2.1000 mg/m2 of gemcitabine and 2500 or 3000 mg/m2 of treosulfan in cohort 1 and 3500 or 4000 mg/m2 in cohort 2, on days 1 and 8 every 4 weeks/
37Corrie PG et al., 2005 [83]/Phase I trial (1) MTD of gemcitabine combined with a fixed dose of treosulfan (2) safety, toxicity, and efficacy/27 advanced melanoma patients were enrolled, of whom 5 (19%) had UM.Chemotherapy on day 1 of a 21-day cycle. Fixed dose of 5 g m2 treosulfan, preceded by escalating doses of gemcitabine/
38Schmittel A et al., 2005 [84]/Phase II trialEfficacy and toxicity/19 patients with metastatic UM30 or 40 mg/m2 of cisplatin, 1000 mg/m2 of gemcitabine, and 3000 mg/m2 of treosulfan on days 1 and 8, repeated on day 29 (maximum of 6 cycles)/
39Schmidt-Hieber M et al., 2004 [85]/Phase II trialNumber of patients achieving an OR or SD, toxicity/11 patients with metastatic UMBendamustine, at a dose of 120 mg/m2 on days 1 and 2, repeated on day 22/
40Keilholz U et al., 2004 [86]/Phase I trialToxicity, clinical response/39 patients with advanced malignancies (33 with UM and 6 with other histologies)Gemcitabine (1 g/m2, followed by treosulfan on days 1 and 8). Treosulfan dose ranging from 2.5 to 4 g/m2; subsequent cohorts received either 3 or 3.5 g/m2 of treosulfan/
41Terheyden P et al., 2014 [87]/Non-randomized phase II trialResponse to treatment/20 patients with metastatic UM, chemo-naïve (8 patients) and pre-treatedTreosulfan 3500 mg/m2 followed by gemcitabine 1000 mg/m2/
42Pföhler C et al., 2003 [88]/Case seriesRR, PFS and OS, and toxicity/14 patients with metastatic UM, 13 previously untreated and one pretreated with chemoimmunotherapyTreosulfan + gemcitabine in four different dose regimens/
43Bedikian AY et al., 2003 [89]/Phase II clinical trialResponse to treatment, safety/14 patients with uveal melanoma metastatic to the liverTemozolomide at a starting dose of 75 mg/m2 per day for 21 days every 4 weeks/
44Kivelä T et al., 2003 [90]/Prospective, multicenter, nonrandomized phase II studyEfficacy and tolerability/24 patients with metastatic UMBleomycin 15 mg, vincristine 1 mg/m2, lomustine 80 mg, and dacarbazine 200 mg/m(2), given every 4 weeks for a minimum of 2 cycles. IFN alpha-2b at a dose of 3 × 10(6) IU on cycle 1, and continued at 6 × 10(6) IU 3 times per week/
45Pyrhönen S et al., 2002 [91]/Open, two-center non-randomized phase II trial Activity of bleomycin, vincristine, lomustine, and dacarbazine (BOLD) chemotherapy with human leukocyte interferon, as well as the PFS and OS/22 patients with histologically proven metastatic UM15 mg of bleomycin, 1 mg/m2 vincristine, 200 mg/m2 dacarbazine, and 80 mg lomustine every 4 weeks + interferon (3 × 106 IU daily for 6 weeks followed by 6 × 106 IU three times per week)/
46Becker JC et al., 2002 [92]/Prospective phase II trialActivity of a combination of chemotherapy with fotemustine followed by immune-modulation with IL-2 and IFN alfa/48 patients with metastatic UMFotemustine 100 mg m−2 IHA or IV, depending on the metastatic sites involved, subcutaneous IL-2 and IFN alpha(2)/
47Ellerhorst JA et al., 2002 [93]/Phase II trialRR and toxicity/28 patients, including14 with UMThe starting dose (level 0) of 9-NC was 1.5 mg/m2/day taken orally for 5 consecutive days of each week/
48Mertens WC et al., 1996 [94]/Phase II studyAntitumor activity/17 patients with advanced malignant melanoma (cutaneous, mucosal or UM)Indomethacin 50 mg orally every 8 h, and ranitidine 150 mg orally every 12 h, and reviewed after 2 weeks/
ADRs = adverse drug reactions; AEs = adverse events; AUC = areas under the concentration-time curves; CBR = clinical benefit rate; CR = complete response; DCR = disease control rate; DoR = Duration of Response; DTL = dose-limiting toxicity; IrAEs = Immune-related AEs; MFS = Metastasis-free survival; MTD = maximum tolerated dose; PK = pharmacokinetic; ORR = overall response rate; OS = overall survival; QD = once daily; PFS = progression free survival; PD = progressive disease; PR = partial response; RR = response rate; SAEs = serious adverse events; SD = stable disease; TTP = time to progression; UM = uveal melanoma.
Table 4. Main efficacy and safety results (management of local complications).
Table 4. Main efficacy and safety results (management of local complications).
NAuthor, YearMain ResultsSide Effects
1Schefler AC et al., 2020 [32]Mean ETDRS BCVA gains over first 48 weeks: 4.0, 1.9, and 0.9 letters for Cohort A, Cohort B, and Cohort C, respectively (statistically significant difference in mean BCVA values among 3 cohorts, p < 0.001; statistically significant difference in the mean change of BCVA from baseline among the 3 cohorts, Cohort A vs. B, p < 0.0001; B vs. C, p < 0.0001; A vs. C, p = 0.008).No serious ocular AEs. No cases of endophthalmitis or IO inflammation.
2Murray TG et al., 2019 [33]42.5% (showed better than 20/50 BCVA, and only 5% showed a BCVA worse than 20/200). No difference was found between a fixed 6-week treatment and a variable treat-and-adjust interval.No patients demonstrated endophthalmitis or metastatic disease or died during the study.
3Horgan N et al., 2009 [34]The triamcinolone group had a significantly lower risk of developing macular edema (hazard estimate, 0.35; 95% confidence interval, 0.11– 0.58; p = 0.004). Other factors predictive of development of macular edema were largest tumor base (p = 0.001) and tumor thickness (p = 0.018). By multivariate analysis, triamcinolone treatment was the most significant factor associated with a lower risk of macular edema (hazard estimate, 0.45; 95% confidence interval, 0.19–0.70; p = 0.001). At 18-month follow-up, moderate vision loss occurred significantly less frequently in the triamcinolone group than in the control group (31% vs. 48%; chi-square, 4.25; 1 df; p = 0.039).In 8 patients (7%), raised IOP developed after the 1st or 2nd triamcinolone injection. Elevated IOP occurred in 15% of the triamcinolone group and in 7% of the control group (chi-square, 1.93; 1 df; p = 0.165). All cases were controlled with topical treatment. Rates of cataract progression were similar in both groups. No case of globe perforation associated with periocular injection.
AEs = adverse events; BCVA = best-corrected visual acuity; IO= intraocular; IOP = intraocular pressure.
Table 5. Main efficacy and safety results (local treatment of primary tumor or metastases from uveal melanoma).
Table 5. Main efficacy and safety results (local treatment of primary tumor or metastases from uveal melanoma).
NAuthor, YearMain ResultsSide Effects
1Venturini M et al., 2012 [35]Tumor responses: 1 CR, 2 PRs, 1 SD, and 1 PD.Well tolerated in all 5 patients.
2Olofsson R et al., 2014 [36]Overall radiological response: 68% of patients (12% CR, 56% PR, 18% SD, and 15% PD); time to local progression: 7 months; 68% of patients developed extrahepatic metastases after a median of 13 months, and the median OS was 24 months. Significant survival advantage of 14 months (p = 0.029) when comparing these patients with a control group.No postoperative mortality was observed. There were 3 major complications: 1 patient developed a systemic inflammatory response syndrome with respiratory insufficiency as well as renal and cardiovascular failure; 1 patient also developed respiratory insufficiency with pneumonia and pleural effusion; 1 patient perforated duodenal ulcer at the fourth postoperative day.
3Leyvraz S et al., 2014 [37]HIA did not improve OS (median 14.6 months) when compared with the IV arm (median 13.8 months) [hazard ratio (HR) 1.09; 95% confidence interval (CI) 0.79–1.50, log-rank p = 0.59]. However, there was a significant benefit on PFS for HIA with a median of 4.5 versus 3.5 months, respectively (HR 0.62; 95% CI 0.45–0.84, log-rank p = 0.002). 1-year PFS rate was 24% in the HIA arm vs. 8% in the IV arm. Better RR in the IAH (10.5%) compared with IV (2.4%).In the IV arm, the most frequent grade ≥3 toxicity was thrombocytopenia (42.1%) and neutropenia (62.6%), compared with 21.2% and 28.7% in the IAH arm. The main grade ≥3 toxicity related to IAH was catheter complications (12%) and liver toxicity (4.5%).
4van Iersel LB et al., 2014 [38]The AUC of oxaliplatin at the MTD of 100 mg oxaliplatin ranged from 11.9 mg/L h to 16.5 mg/L h. All 4 patients treated at the MTD showed progressive disease 3 months after IHP. Only 8 patients were available for response evaluation of which 3 patients showed a PR, with a duration of response of 6.5–11.1 months. Median OS was 18.7 months.Dose limiting sinusoidal obstruction syndrome (SOS) occurred at 150 mg oxaliplatin.
5Yamamoto A et al., 2009 [39]High-dose IE resulted in significantly better OS (20.4 vs. 9.8 months, p = 0.005) and systemic PFS (12.4 vs. 4.8 months, p = 0.001). Patients who achieved regression of hepatic metastases after embolization lived much longer than did those who did not achieve regression (27.2 vs. 9.9 months, p = 0.001). At multivariate analysis, prolonged OS was confirmed for patients who underwent high-dose IE, and patients with regression of hepatic metastases./
6Huppert PE et al., 2010 [40]No patient showed CR, 8 patients (57%) showed PR, 4 patients (29%) had SD and 2 patients (14%) had PD. Time to progression ranged between 5 and 35 months (median 8.5 months). Median survival of all patients was 11.5 months (3–69 months) following first TACE and 18.5 months (5–75 months) following diagnosis of liver metastases. At the time of data analysis, 10 patients had died and 4 patients were alive. The survival rate was 86% at 6 months, 50% at 12 months, 28% at 18 months, and 14% at 24 months following first TACE.Symptoms of post-embolization syndrome were seen in all patients. In 2 patients, the application of additional morphine was necessary to overcome right upper quadrant pain. In 1 patient, acute renal insufficiency occurred after the second TACE.
7Fiorentini G et al., 2009 [41]All patients had an objective response; 3 patients had a major response with evidence of metastases reduction of 90%, 3 had a reduction of 80% and 4 presented a reduction between 70% and 60%; 8 patients are alive at the time of this analysis.The most important AE was abdominal pain during the procedure.
8Voelter V et al., 2008 [42]Median survival for patients treated with fotemustine was 9 years (95% confidence interval (CI) 2.2–12.7) compared with 7.4 years in the control group (95% CI 5.4–12.7). The corresponding 5-year survival rates were 75 and 56%, respectively (p = 0.539). The estimated hazard ratio for death at 5 years was estimated at 0.98, with a 95% CI of 0.38–2.61, p = 0.9815 patients (23%) received only the induction cycle (3–4 infusions), with early treatment cessation owing to hepatic toxicity (n = 4) and/or catheter-related complications (n = 2). The main side effect of the adjuvant treatment was drug-induced hepatitis; 3 patients experienced grade 3 gastric toxicity; 2 patients presented with grade 3 neutropenia and 1 with grade 3 thrombocytopenia.
9van Iersel LB et al., 2008 [43]Of the 12 UM patients perfused, 4 (33%) had a PR, 6 (50%) patients had SD, and 2 (17%) patients were immediately progressive. Median DFS was 6.6 months with a median OS of 10.0 months. 50% of other primary tumors showed at least PR.Reversible grade 3 or 4 hepatotoxicity occurred in 10 (56%) patients, while VOD occurred in 4 (22%) patients.
10Noter SL et al., 2004 [44]No CRs were observed. 4/8 patients (50%) showed a PR. 2 patients showed SD and 2 patients showed progression after 3 months of follow-up. The median TTP was 6.7 months (range, 1.7–16.9 months). The median survival was 9.9 months (range, 4.7–34.6 months). The 1-year survival rate was 50% and the 2-year survival rate was 37.5% with 1 patient still in follow-up 15 months after treatment.1 patient developed grade 3–4 leukopenia. 3 patients experienced grade 3–4 toxicity of 1 or more liver enzymes. Major complications occurred in 3 patients in the week following the perfusion procedure: VOD in 2 patients and lung embolism in 1 patient.
11Egerer G et al., 2001 [45]2 patients achieved a PR, 3 had SD, and in 2 patients, the tumor progressed in the liver. Extrahepatic progression to lung was diagnosed in 1 patient. The median survival was 18 months (range, 3–43 months) with a median TTP of 16 months (range, 0 to 43 months) in 6 patients. The median survival from diagnosis of the primary tumor was 54 months (range, 31–122 months); 2 patients survived for more than 2 years, and 2 patients are alive at present with residual liver metastasis, at 3 and at 43 months.The toxicity of the locoregional chemotherapy was minimal in the 7 patients
12Hussain RN et al., 2020 [46]No patients achieved CR or PR at any visit. All required enucleation. The study was terminated early, as alternative treatments were clearly superior for local tumor control./
13Favilla I et al., 1995 [47]The longest duration of tumor control = 6.5 years. 76% of melanomas were not growing at the end of the first year, 62% at the end of the second year, 38% with no signs of growth at the end of the fifth year. Three patients died during the 5-year period: 1 from melanoma metastases after 2 years, the second from a concurrent malignancy, and the third from heart failure after 1.7 years.The post-treatment visual function was worse in 26 patients (76%). However, in 24 (67%), the tumor was located posteriorly and involved the macula. No eyes were enucleated because of complications of PDT.
AEs = adverse events; AUC = areas under the concentration-time curves; CE = Chemoembolization; IE = immunoembolization; CP = carboplatinum/paclitaxel; CR = complete response; DCR = disease control rate; DoR = Duration of Response; DTL = dose-limiting toxicity; I.a.h. = Intra-arterial hepatic; IHP = Isolated hepatic perfusion; IrAEs = Immune-related AEs; MFS = Metastasis-free survival; MTD = maximum tolerated dose; ORR = overall response rate; OS = overall survival; QD = once daily; PDT = photodynamic therapy; PFS = progression free survival; PD = progressive disease; PR = partial response; RR = response rate; SAEs = serious adverse events; SD = stable disease; T = treosulfan alone; TEAEs = Treatment Emergent Adverse Events; TACE = Hepatic transarterial chemoembolization; TTP = time to progression; UM = uveal melanoma; VOD = veno-occlusive disease.
Table 6. Main efficacy and safety results (systemic therapy for uveal melanoma).
Table 6. Main efficacy and safety results (systemic therapy for uveal melanoma).
NAuthor, YearMain ResultsSide Effects
1Nomura M et al., 2020 [48]Best ORR = 23.5% (95% confidence interval (CI) 6.8–49.9%)); median PFS 1.4 months (95% CI 1.2–2.8); median OS 12.0 months (95% CI 3.5 to not reached); 1-year OS 50.0% (95% CI 25.9–70.0%).Treatment-related AEs were mostly grade 1 (pruritus in particular). Treatment-related AEs of grade 3 occurred in 15% (colitis, anemia, adrenal insufficiency, diarrhea), and no grade 4 or 5 AEs were observed.
2Luke JJ et al., 2020 [49]10/31 met the primary endpoint of PFS4 (32.3%) compared to 4/15 randomized to arm 2 (26.7%; p = 0.350). No difference in PFS was observed (95% CI: 56–162 days) compared with arm 2 (95% CI: 56–152 days; p = 0.964, hazard ratio (HR) 0.99 (95% CI: 0.51–1.86)). The median OS in arm 1 was 191 days (6.4 months; 95% CI: 168–314) versus 218 days (7.3 months; 95% CI: 170-NA days) in arm 2 with no difference (p = 0.580, HR = 1.21 (95% CI: 0.62–2.34)). The trial was terminated for futility.Grade 3–4 AEs were 71.0% and 66.7% in arms 1 and 2,respectively. Common attributable grade 3–4 events included fatigue, increased AST or ALT, and thromboembolic events.
3Piha-Paul SA et al., 2019 [50]Of 61 evaluable patients from dose escalation, 26 (43%) had SD and 35 (57%) had PD. Median PFS was 1.8 months (95% confidence interval, 1.8–1.9). All solid tumor patients (N = 84) discontinued mivebresib. Primary reasons for discontinuation were radiologic PD (63%), clinical PD (13%), withdrew consent (8%), AE related to progression (3%), AE not related to progression (3%), lost to follow-up (3%), and other (8%).Most common TEAE were dysgeusia (49%), thrombocytopenia (48%), fatigue (26%), and nausea (25%). Most common grade 3/4 TEAEs were thrombocytopenia (35%) and anemia (6%). Dose-limiting toxicities included thrombocytopenia, gastrointestinal bleed, hypertension, fatigue, decreased appetite, and aspartate aminotransferase elevation.
4Binkley E et al., 2020 [51]5-year and median MFS were 64% (44–78) and 79 months in treated patients and 33% (15–52) and 29 months in observed patients. 5-year median OS was 66% (45–80, median not observed) in treated patients and 37% (19–55, median 54 months) in observed patients.Grade 1 or 2 fatigue was reported by 33 patients (87%). Grade 1 or 2 elevations in transaminases were observed in 14 patients (37%), and grade 1 or 2 depression, in 5 patients (13%). Grade-3 hematological toxicity in 6 patients. No grade 4 AEs were reported.
5Johnson DB et al., 2019 [52]1 patient had a CR (ongoing at 25.5 months), and no PRs were observed (RR 20%). 1 patient experienced prolonged SD (ongoing at 11 months), and another had SD lasting 11 months before experiencing progression (clinical benefit rate 60%). The remaining 2 patients experienced rapid PD. Median PFS was 11.0 months and median OS was not reached (median follow-up, 11.1 months; range, 0.4–25.5 months). The patient who experienced the CR had fulminant type 1 diabetes (grade 4) that arose after the first dose of pembrolizumab, and stopped treatment. Other toxicities included grade 1 hypothyroidism and rash; 3 patients had no side effects.
6Shah S et al., 2018 [53]Response outcomes included 1 PR, 4 SD, 11 PD. Overall RR = 5.9%; DCR = 29.4%. PFS = 1.6 months cohort A and 1.8 months cohort B. OS = 8.5 months cohort A and 4.9 months cohort B.An overall 31% of AEs were grade 3–4 and were mostly related to gastrointestinal toxicities.
7García M et al., 2019 [54]No objective responses were observed. At the lower dose levels, 2 patients had SD, at the highest dose level SD was observed in 5/6 patients. The survival probability was 3.7 times longer for the UM patients. Median survival = 271 days for uveal versus 73 days for cutaneous (hazard ratio 0.15; 95% confidence interval 0.026–0.85).At dose levels 1a–3a, no relevant toxicity was observed. Acute toxicity was mainly a flu-like syndrome with fever, chills, arthromyalgia, headache, nausea, and vomiting, and diarrhea. The first patient at the level 5a experienced transaminitis grade 3 and grade 3 thrombocytopenia.
8Carvajal RD et al., 2018 [55]The primary endpoint of PFS was not met. In the selumetinib + dacarbazine group, there were 82 events (85%) compared with 24 (75%) for placebo + dacarbazine; the HR for PFS was 0.78 (95% CI: 0.48 to 1.27; p value = 0.32), with no significant benefit of selumetinib. Median PFS was 2.8 months in the selumetinib + dacarbazine and 1.8 months in the placebo + dacarbazine group. Incidence of AEs of special interest was more frequent with selumetinib plus dacarbazine; however, these were generally grade 1/2.
9Schinzari G et al., 2017 [56]PRs were observed in 5 (20%) patients, SD in 12 (48%); DCR was 68%. Median OS of all the patients was 13 months, median PFS 5.5 months. OS of responding patients was 21 months; OS of patients with disease control was 18 months, significantly longer than survival of progressing patients (7 months, p = 0.0003).5 (20%) patients experienced grade 3–4 toxicity.
10Daud A et al., 2017 [57]In the UM cohort, 61% of patients (14/23) had SD at week 12, and no patient had a PR, resulting in an overall DCR of 61%. The median PFS for the 23 patients with UM was 4.8 months (41% PFS rate at 6 months) and median OS was 12.6 months. Most patients with UM stayed on study treatment for 44 months, and 6 patients stayed on treatment for >10 months.The most common grade 3/4 events were fatigue (14%), hypertension (10%), abdominal pain (8%), hand-foot syndrome (5%), asthenia (5%), back pain (5%), and hypokalemia (5%); 6 patients (8%) discontinued study treatment because of AEs; 1 died from peritonitis due to diverticular perforation (deemed related), and 1 from an unknown cause (deemed unrelated).
11Naing A et al., 2016 [58]41 (80.4%) of 51 patients were evaluable. 1 patient with UM (40 mg/kg) had a PR, with histologically confirmed reduction of multiple gastric metastases.AM0010 was tolerated well, with manageable AEs. Most frequently observed AEs were anemia (51%), fatigue (45%), thrombocytopenia (42%), injection site reactions (36%), and fever (30%). Grade 3 to 4 non-hematopoietic AEs were observed in 15%. Grade 3 to 4 anemia or thrombocytopenia was observed in 18%. Only 1 patient discontinued treatment because of a DLT (recurring anemia). A grade 2 rash was observed in 3 patients, and a grade 3 rash was observed in 1.
12Carvajal RD 2014 [59]The median PFS was 7 (95% confidence interval (CI), 4.3–8.4) and 15.9 weeks (95% CI, 8.4–21.1) for chemotherapy (n = 49) and selumetinib (n = 47), respectively. The HR for PFS was 0.46 (95% CI, 0.30–0.71; p < 0.001) in favor of selumetinib. The median OS was 9.4 (95% CI, 6.0–11.4) and 10.8 months (95% CI, 7.5–12.9) for chemotherapy and selumetinib, respectively, with a HR of 0.79 (95% CI, 0.46–1.37; p = 0.40). Tumor regression was uncommon with chemotherapy, with no responses observed. 49% of patients with selumetinib achieved tumor regression.TEAEs were observed in 97% patients treated with selumetinib, with the most common being acneiform rash (75%), CPK elevation (60%), fatigue (57%), AST elevation (48%), and ALT elevation (42%). Blurred vision (6%) and other visual changes (7%) were observed. 37% experienced grade 3–4 TEAEs, including CPK elevation (13%), AST elevation (7%), and ALT elevation (6%).
13Adjei AA et al., 2017 [60]No DLTs were observed in patients who received TAK-733 in the first 8 dose cohorts (0.2–8.4 mg).Subsequently, 4 patients experienced DLTs in cycle 1. Based on the observed DLTs in cycle 1, the MTD of TAK- 733 was determined to be 16 mg once daily on days 1–21 in 28-day treatment cycles. All patients experienced at least 1 AE of any grade, and 88% reported drug-related AEs. Grade ≥ 3 AEs were reported in 53%. Overall, 67% experienced rash. 14% experienced ophthalmic AEs (visual impairment, photopsia, blurred vision, photophobia, periorbital, and retinal edema); 27% experienced at least 1 SAE.
14Mouriaux F et al., 2016 [61]There were no confirmed objective tumor responses. The estimated 24-week PFS was 31.2% (95% CI: 14.8%–47.6%) and the estimated 24-week OS was 62.5% (95% CI: 45.4%–79.6%). The OS rate among patients who received at least 2 months of treatment did not significantly differ compared with the expected patient survival rate (p > 0.05).281 ADRs were reported, including 20 grade 3 or 4 reported in 10 patients. 12 patients (41.4%) required dose modifications due to toxicity.
15Shoushtari AN et al., 2016 [62]23% had SD for at least 16 weeks and were considered to have clinical benefits. Overall, SD was the best objective outcome. Median duration of SD in these patients was 8 weeks (range: 8–16 weeks). 6/13 patients had PD on first assessment. Median PFS from first date of treatment was 16 weeks (range: 7–23 weeks); median OS from first day of study treatment was 11 months (range: 4.5–28.5 months).All patients experienced at least 1 possibly related AE. The most frequently AEs were metabolic (hyperglycemia, hypertriglyceridemia, hypercholesterolemia), gastrointestinal (diarrhea, oral mucositis), or hematologic (leukopenia, thrombocytopenia, neutropenia). The grade 3 AEs were hyperglycemia (n = 7), oral mucositis (n = 2), diarrhea (n = 1), hypophosphatemia (n = 1), and anemia (n = 1). There were no grade 4 or 5 AEs.
16Joshua AM et al., 2015 [63]2 of 11 patients received 4 cycles of treatment, whereas the rest only received 1 cycle. No responses were observed; 10 of the 11 evaluable patients had progressed at 90 days. The median PFS was 2.9 months (95% confidence interval (CI) 2.8–3.0) and the 6-month PFS was 9.1%. The median OS was 12.8 months (95% CI 3.8–19.7).Grade 3 or 4 rash, nausea, and diarrhea was observed in 1 (9.1%), 2 (18.2%), and 3 (27.3%) patients, respectively; 2 patients experienced SAEs secondary to grade 3 and 4 diarrhea; 2 patients developed hyperthyroidism.
17Zimmer L et al., 2015 [64]45 pretreated (85%) and 8 treatment-naïve (15%) patients received at least 1 dose of ipilimumab; 1-year and 2-year OS rates were 22% and 7%, respectively. Median OS was 6.8 months (95% CI 3.7–8.1), median PFS 2.8 months (95% CI 2.5–2.9). The DCR at weeks 12 and 24 was 47% and 21%, respectively; 16 patients had SD (47%), none experienced PR or CR.TEAEs were observed in 66%, including 36% of grade 3–4 events; 1 drug-related death due to pancytopenia was observed. Most common irAEs were gastrointestinal disorders, skin-related toxic effects, and hepatic disorders. The most frequent grade 3 or 4 irAEs were diarrhea (13%) and colitis (11%).
18Lee CK et al., 2015 [65]In the intent-to-treat population (n = 30), the median PFS was 3.7 months, and the median OS was 9.4 months. In the per-protocol population, the median PFS was 4.3 months (22 PFS events among 25 patients), and the median OS was 9.6 months (16 OS events among 25 patients). The ORR was 12% (n = 3; 95% confidence interval (CI), 0% to 24.74%), and DCR was 80.0% (n = 20; 95% CI, 64.32% to 95.68%). UM patients (n = 9) had the best prognosis when treated with docetaxel + carboplatin compared to those with other subtypes (median PFS 7.6 months; OS 9.9 months). All patients with UM had the best response as SD.Most patients (n = 29, 96.7%) reported at least 1 AE related to treatment, and the total incidence of grade 3 or 4 AE was 66.7%. The most common AE was neutropenia (67.7%), with half of the patients experiencing grade 3 or 4. Non-hematologic toxicity was less common and less severe.
19Dickson MA et al., 2015 [66]The MTD was 150 mg. 2 patients had PR (1 papillary thyroid cancer with NRAS mutation, lasting 72 weeks, and 1 with UM with unknown mutation status, lasting 44 weeks). Another 9 patients had tumor decrease of at least 10% but did not meet criteria for PR. Several patients had prolonged SD on study suggesting possible clinical benefit.Approximately 13% of patients had 1 or more dose reductions during their treatment due to AEs. Approximately 51% of patients experienced a dose delay during treatment, mostly due to AEs.
20Homsi J et al., 2010 [67]The median number of treatment cycles was 1 (range 1–7 cycles). 7 patients (32%) had SD with a median duration of 3 months (range: 3–7 months). The median OS was 9.8 months.Neutropenia (23%) and musculoskeletal pain (10%) were the most common grade 3 and grade 4 toxicities.
21Borden EC et al., 2011 [68]Although a single patient had a sustained regression, overall IFN-b1a did not have clinical benefit (response rate <10%; median PFS 1.8 months).Reversible drug-related severe (grade 3) AEs in 13/21 patients; anorexia and fatigue were mostly of mild or moderate severity.
22Danielli R et al., 2012 [69]No objective responses were observed; however, 2 patients had SD, a third patient had SD after initial PD. Median OS was 36 weeks (range 2–172 weeks).No grade 3/4 AEs of non-immune origin were reported; 23% experienced grade 3 irAEs that resolved with steroid therapy.
23Tarhini AA et al., 2011 [70]Among 40 patients evaluable for efficacy, 7.5% had a confirmed PR, and 50% had PFS of ≥4 months. The 1 year survival rate is 56.4% (95% CI 43%–74%), p < 0.005. Median OS in this trial is 16.3 months, (95% CI 9.2 months–not reached).Grade 3/4 toxicities included hypertension in 22% and proteinuria in 15%. AEs leading to treatment discontinuation included recurrent grade 3 proteinuria, grade 4 cerebrovascular ischemia, grade 3 left ventricular diastolic dysfunction and osteonecrosis of the mandibular bone.
24Bhatia S et al., 2012 [71]No confirmed objective responses occurred among the 24 evaluable patients (95% CI: 0–14%) and the study was terminated. The median PFS was 4 months (95% CI: 1–6 months) and the 6-month PFS was 29% (95% CI: 13%–48%). The median OS was 11 months (95% CI: 7–14 months).29% experienced grade 4 AEs, all hematologic; 3 patients discontinued due to toxicity (myelosuppression or neuropathy); 75% required dose modifications due to toxicity.
25Falchook GS et al., 2012 [72]Among the 36 BRAF-mutant patients, 30 were BRAF-inhibitor naïve. Among these patients, 2 confirmed CRs and 8 confirmed PRs (RR = 33%); the median PFS was 5.7 months (95% CI, 4·0–7·4). Among the 6 BRAF-mutant patients who received prior BRAF inhibitor therapy, 1 unconfirmed PR was observed. Among 39 patients with BRAF wild-type melanoma, 4 PRs were observed (RR = 10%). Among the 16 patients with UM, 13% achieved a 24% tumor reduction. SD for ≥16 weeks was observed in 25%, including 2 who received treatment for >40 weeks.The most common TEAEs were rash/dermatitis acneiform (80 out of 97; 82%) and diarrhea (n = 44; 45%), most of which were grade 2 or lower.
26Ott PA et al., 2013 [73]No objective responses were seen in any of the three patient cohorts. The best overall response was SD (31%); 71% had progressive disease at the 1 response assessment. In the phase II patient cohort, 23.5% patients had SD. The longest duration of SD in the cohort receiving the highest dose was 169 days. SDs >6 months occurred in patients with UM. The median duration of SD in the UM patients was 141 days (range: 57–337 days). The median TTP for the 29 evaluable patients was 57 days, whereas 113 days for the patients with UM.No DLTs were seen in the first cohort (40 IU/m2). In cohort 2 (80 IU/m2), 1 grade 3 episode of arthralgia. No DLTs were observed in cohort 3 and enrollment onto the phase II part of the protocol was continued. MTD was not reached. Overall, the treatment was well tolerated; 6 grade 3 toxicities were observed in total.
27Mahipal A et al., 2012 [74]1 patient achieved a PR and 12 had a SD, with the duration of SD ranging from 2.1 to 29.2 months (median = 5.5 months). The patient who achieved a PR remained on treatment for 13 months. The median OS and PFS were 8.2 and 4.2 months, respectively; 3 patients had SD for more than 12 months with sunitinib after failing previous treatments.The most common AEs were fatigue (90%), diarrhea (60%), hemorrhage (55%), anorexia (45%), hand-foot syndrome (25%), hypothyroidism (25%), and rash (25%); 11 patients required dose reduction due to grade 3 AEs.
28Lane AM et al., 2009 [75]Among IFN-treated patients, 34.7% developed metastasis compared with 27.5% in the control group; 31% developed metastasis after completing the 2-year course, and 13 died of the disease. The proportion of patients who died of metastatic disease was similar in the 2 groups: 30.6% in the IFN-group and 26.9% in the proton-therapy or enucleation only group. The 5-year melanoma-related death rates were 16.9% (95% confidence interval, 12.7%–22.4%) in the radiation or enucleation only group and 23.8% (95% confidence interval, 17.1%–32.6%) in the adjuvant therapy group. No differences were observed with longer follow-up. Symptoms or onset of new illnesses (37.9%) and abnormal laboratory values (34.8%), were the most common reasons for discontinuing therapy. DLTs occurred in 28 patients and included thrombocytopenia (10.7%), elevated liver enzymes (42.9%), and thyroid function alterations (35.7%). All patients reported flu-like symptoms after the first few injections. Most AEs were mild to moderate in severity and resolved with continued IFN-therapy.
29Hofmann UB et al., 2009 [76]A total of 9 patients (75%) received imatinib for 8 weeks; 25% discontinued because of disease progression. No patient achieved an objective response; the best clinical response was a SD in 1 patient, which lasted for 52 weeks. Thus, the median PFS was not calculated. The median OS of all patients was 6.8 months. For the 8 patients who received imatinib as 1 line therapy, the median OS was 7.8 months. The 4 patients treated in a 2-line setting had a median OS of 4.9 months. Abdominal pain and vomiting was the most common toxicity, resulting in a dose reduction in 2 cases (17%); 1 patient had facial edema. There was no significant hematologic toxicity.
30Bedikian AY et al., 2008 [77]1 CR, 2 PRs, and 5 SDs were observed in the 26 evaluable patients, resulting in a DCR of 31%. The median TTP was 1.9 months (95% CI: 1.8–2.2 months). The median survival was 9.6 months; (95% CI: 7.3–32 months) with 30% of the patients alive at 1 year. The median duration of SD was 4.2 months (range: 3.2–4.7 months).Most of the side effects were grade 1–2 neurological, gastrointestinal, or constitutional. Nausea was the most common side effect. It occurred in about 75% of the patients and was usually mild. Neurologic AEs were constipation, hypoesthesia, anxiety, paresthesia, and peripheral neuropathy. The hematologic side effects were mild (mostly grade 1/2 neutropenia). None developed grade 3/4 thrombocytopenia.
31Penel N et al., 2008 [95]No objective response and only 1 SD with duration of 5 months were noted. No patient was found to be free of disease progression 6 months after the initiation of treatment. The overall survival was 10.8 months.5 and 1 out of 13 enrolled patients experienced grade 3 and grade 4 toxicities, respectively. The most common severe AEs were abdominal pain.
32Adjei AA et al., 2008 [78]19 patients (33%) had SD at the end of cycle 2, and 9 patients (16%) had SD for ≥ 5 months; 1 patient with medullary thyroid cancer experienced SD for 19 cycles, whereas 1 patient with both UM and renal cell carcinoma had SD for 22 cycles.Rash was the most frequent toxicity and DLT, occurring in 74% of all patients, and precluded dose escalation greater than 300 mg bid. Of the 43 episodes of skin rash, 34 were of maximum grade 1–2, and 9 were grade 3–4. Mild to moderate diarrhea was the principal gastrointestinal toxicity (56% of patients). Mild-moderate reversible ALT and AST elevation occurred in 14%; 14% experienced SAEs, including hypoxia, pneumonitis, bradycardia, renal insufficiency, and exfoliative dermatitis.
33Schmittel A et al., 2006 [79]7 confirmed SDs and 1 PR were observed in 24 patients treated with the GeT regimen, whereas no PR and only 3 SDs were observed in the T arm (p = 0.08). Median PFS was 3 months (95% CI 1.1–4.9) and 2 months (95% CI 1.7–2.3) in the GeT and T arm (p = 0.008, log-rank); 6 and 12 months PFS was 34.8% and 17.9% and 16.7% and 0% always favoring the GeT arm.Grades 3 and 4 leukopenia only occurred in the GeT arm (17%; p = 0.001). 8% experienced a febrile neutropenia. Frequencies of anemia, nausea, vomiting, and infections were not significantly different in both treatment arms.
34Richtig E et al., 2006 [80]In 3 patients, therapy had to be withdrawn because of the appearance of metastases. Neither a univariate approach nor a multivariate approach could show a protective effect of interferon treatment on survival.For 46% the initial dose had to be reduced due to leukopenia, thrombocytopenia, cardiac symptoms, elevated of liver function, or vertigo. In 5 patients, therapy had to be withdrawn because of serious side effects.
35O’Neill PA et al., 2006 [81]14 patients are evaluable for response. 4 patients completed all 6 cycles of chemotherapy. Of these, all 4 achieved SD after 3 cycles but 2 patients had progressed at re-assessment after cycle 6. The other 10 patients all had PD. Median PFS from the first cycle of chemotherapy was 12 weeks (2–26 weeks) and median OS was 30 weeks (2–64 weeks).The treosulfan/dacarbazine combination was generally tolerated well. The major toxicities were hematological. Grade 1 or 2 thrombocytopenia was seen in 8 patients and grade 3 thrombocytopenia was seen in 3 patients. Grade 4 thrombocytopenia was also seen in 1 patient and Grade 4 neutropenia occurred in 1 patient. Non-hematological toxicity was generally mild with 2 patients experiencing grade 3 nausea and vomiting while 1 patient experienced grade 3 lethargy.
36Schmittel A et al., 2005a [82]In cohort 1 with a treosulfan dose of < or = 3000 mg/m2, no objective response was observed. Of the patients treated with > or = 3500 mg/m2 in cohort 2, 1 had PR (5%), 10 showed SD and 8 PD. An increased survival was observed in the second cohort with higher treosulfan doses, with median survival times of 6.0 versus 9.0 months (p = 0.03) in cohort 1 and 2, respectively, and a 1-year survival of 7.1% versus 47.3%, respectively.Grade 3 thrombocytopenia was observed in 5/14 patients treated within cohort 1 and in 5/19 patients treated within cohort 2. Grade 3–4 leukopenia occurred in 2 patients in cohort 1 and 4 patients in cohort 2. 1 patient in cohort 2 had grade 3 anemia. No non-hematological AEs >grade 2 was observed.
37Corrie PG et al., 2005 [83]No objective CRs or PRs were documented. Best responses were 8% minor responses (both UM), 46% SD (10 cutaneous, 2 uveal) and 46% PD (10 cutaneous, 1 uveal). DCR was 54%. Median survival was 36 weeks (range 5–121). Median overall TTP was 14 weeks (range 3–74). Median survival and TTP for the UM patients were 53 weeks (range 20–103) and 27 weeks (range 7–38), respectively.DLT was reached at 3.0 g m2 gemcitabine, when 2 of 6 patients experienced grade 3 myelosuppression. At the lower gemcitabine dose of 2.5 g m2, only 1 episode of grade 3 neutropenia occurred. Other common toxicities were nausea and vomiting, fatigue, skin rash and constipation.
38Schmittel A et al., 2005b [84]No objective response was observed; 7 patients (41%) had SD and 10 (59%) progressed. The median PFS of all 19 patients was 3.0 months (95% confidence interval (CI), 1.8–3.1); the median OS was 7.7 months (95% CI, 1.9–13.8). The 1-year survival was 31%.Grade 3 and 4 leukopenia was observed in 9/19 patients. Grade 3 and 4 thrombocytopenia and leukopenia occurred in 8 and 9 patients, respectively. Grade 3 nausea, vomiting, and mucositis occurred in 1 patient each.
39Schmidt-Hieber M et al., 2004 [85]All 9 evaluable patients had progressive disease. 1 patient with progression experienced clinically significant symptom relief and therefore received 6 cycles. 2 patients died after the first cycle because of progressive disease.Grade 3 and 4 toxicity consisted of anemia, thrombocytopenia, and leukocytopenia in 2, 1, and 2 patients, respectively. 3 patients showed grade 2 nausea, 1 patient grade 2 diarrhea and 1 patient grade 2 to 3 drug fever.
40Keilholz U et al., 2004 [86]For cases treated at dose levels 1/2, no objective responses were observed, whereas 2 patients (UM, renal cancer) on dose level 3 and 1 patient on dose level 4 (ovarian cancer) had a PR. Furthermore, we observed a stabilization of disease for more than 3 months in 15 patients with UM. A significant trend for improved OS with higher treosulfan doses was recorded.Chemotherapy was generally well tolerated. Acute toxicity consisted of mild nausea in 9 patients. The predominant delayed toxicity was myelotoxicity. Grade 3 or 4 thrombocytopenia was observed in 3 and 1 patient, respectively. On dose levels 3 and 4, 1 and 2 patients, respectively, developed thrombocytopenia requiring a dose reduction. Grade 3 leukopenia was observed in 1 patient on dose level 3. Non-hematological side effects > grade 2 were alopecia and neutropenic fever.
41Terheyden P et al., 2014 [87]No patient achieved an objective response, 25% of patients (95% confidence interval, 8.6–49.1%) had stabilization of disease. The median time to progression for the patients achieving a SD was 187 days (range 182–316 days), with a prolonged median OS of 17 months compared with 7 months for the patients with PD. The median OS was worst in patients receiving treosulfan/gemcitabine as first-line therapy, i.e., 206.5 days (range 25–491 days).The combination therapy of treosulfan/gemcitabine was well tolerated with no common toxicity criteria grade 2–4 non-hematological AEs.
42Pföhler C et al., 2003 [88]The analysis revealed 1 CR, 3 PR, and a SD in 8 cases. The objective response rate was 28.6%, the median OS was 61 weeks (95% confidence interval (CI) 54–133 weeks), the PFS was 28.5 weeks (95% CI 13–62 weeks), and the 1-year survival rate was 80%.The drugs were well tolerated. The most common side effects were leukocytopenia and thrombocytopenia. 3 patients were withdrawn because of toxicity (thrombocytopenia grade 4). Grade 4 neutropenia occurred in only 1 patient.
43Bedikian AY et al., 2003 [89]No CR or PR were observed. SD was achieved in two patients. The median survival of the group was 6.7 months, with a range of 1–12.7 months. The median TTP was 1.84 months, with a range of 0.7–3.8 months.Hematological toxicity was moderate; 3 patients developed grade 4 neutropenia, and 2 of these also developed grade 4 thrombocytopenia; 1 patient had grade 4 thrombocytopenia. Gastrointestinal side effects were the most common non-hematological toxic effects.
44Kivelä T et al., 2003 [90]None achieved an objective response (95% Confidence Interval (CI): 0–14), 8.3% remained stable, 20 showed progression. The median PFS was 1.9 months (95% CI: 1.8–3.4) and OS 10.6 months (95% CI: 6.9–16.4). OS improved with increasingly favorable pretreatment characteristics (median, 14.7 versus 6.9 versus 6.0 months for stages IVBa, IVBb, and IVBc, respectively; p = 0.018).Grade 1–2 nausea, fever, flu-like syndrome, alopecia, hepatic toxicity, and neurotoxicity occurred in more than 30% of patients, and more than 10% experienced grade 3 alopecia and neurotoxicity.
45Pyrhönen S et al., 2002 [91]15% (95% confidence interval [CI] 0–38) obtained a partial objective response in hepatic and extrahepatic sites and 55% (95% CI 32–77) showed SD. The median PFS was 4 months (95% CI 2–10) and the median OS was 12 months (95% CI 8–22); 11 patients with Stage IVBa survived a median of 17 months (95% CI 4–37) whereas 10 patients with Stage IVBb survived a median of 11 months (95% CI 1–23).Grade 1–2 malaise, fatigue, and fever were common. Almost 40% experienced grade 3–4 hematologic toxicity, and 35% developed grade 2–3 constipation. 2 treatment-associated deaths occurred (due to sepsis and myocardial infarction).
46Becker JC et al., 2002 [92]Only 1 patient (2%) achieved a CR and 6 (12.5%) a PR, for an ORR of 14.5% (95% confidence interval, 6.1 to 28.4%); 5 of these objective responses were observed in the cohort of patients receiving fotemustine HIA, while only 2 (1 CR and 1 PR) in the IV group experienced an objective response (21.7 vs. 8%). This difference did not translate into a significant benefit in OS, i.e., 369 and 349 days, respectively.The most prominent side effect due to fotemustine was thrombocytopenia but never exceeded grade 3. A more prominent systemic toxicity of IV infusion was the more common occurrence of leukocytopenia. Thrombocytopenia was observed in 12 patients within the IV group in contrast to 4 patients in the HIA group (p = 0.028); 2 patients receiving intra-arterial fotemustine developed gastroenteric complications.
47Ellerhorst JA et al., 2002 [93]No CR or PR were observed. SD was achieved in 4 individuals (15%) for durations of 3, 4, 6, and 8 months; 2 of these patients had UM and 2 had cutaneous primaries. Disease progressed in spite of treatment in 22 individuals (85%).17.9% developed grade 4 neutropenia and 7.1% grade 4 thrombocytopenia. 43% experienced grade 3 or 4 diarrhea and 18% grade 3 or 4 vomiting. Dehydration secondary to gastrointestinal toxicity lead to 4 hospitalizations. Myalgia and fatigue were also common, but were usually described as mild to moderate in intensity.
48Mertens WC et al., 1996 [94]1 PR was achieved for an ORR of 6% (95% CI 0–29) in a patient with UM metastatic to the liver, after 4 months of therapy, and lasted a further 8 months; 7 patients had SD (range 2–8 months), and lasting 3 months; 9 patients were found to have PD 6 weeks after initiation of treatment.10 patients were able to escalate to 75 mg 3 times daily, but, of those, 3 required dose reductions because of toxicity. Of the 7 other patients, 5 could not escalate to a higher dose; 2 other patients required decrease in dosage of indomethacin.
ADRs = adverse drug reactions; AEs = adverse events; CP = carboplatinum/paclitaxel; CR = complete response; DCR = disease control rate; DoR = duration of response; DTL = dose-limiting toxicity; GeT = gemcitabine plus treosulfan; IrAEs = immune-related AEs; MFS = metastasis-free survival; MTD = maximum tolerated dose; ORR = overall response rate; OS = overall survival; QD = once daily; PFS = progression free survival; PD = progressive disease; PR = partial response; RR = response rate; SAEs = serious adverse events; SD = stable disease; T = treosulfan alone; TEAEs = treatment emergent adverse events; TTP = time to progression; UM = uveal melanoma.
Table 7. Ongoing clinical trials in patients with uveal melanoma.
Table 7. Ongoing clinical trials in patients with uveal melanoma.
(a) Local treatment or prevention of relapse and of metastatic disease or prevention of local complications in locally treated uveal melanoma.
NLocal treatment or Prevention of Relapse and of Metastatic Disease or Prevention of Local Complications in Locally Treated Uveal MelanomaIDPhaseStatus
1Dexamethasone Implant for Retinal Detachment in Uveal MelanomaNCT040829621Recruiting
2Crizotinib in High-Risk Uveal Melanoma Following Definitive TherapyNCT022238192Active, not recruiting
3Stereotactic Body Radiation Therapy and Aflibercept in Treating Patients with Uveal MelanomaNCT037129042Recruiting
4Adjuvant Sunitinib or Valproic Acid in High-Risk Patients with Uveal MelanomaNCT020685862Recruiting
5Phase II Trial to Evaluate Safety and Efficacy of AU-011 Via Suprachoroidal Administration in Subjects with Primary Indeterminate Lesions and Small Choroidal MelanomaNCT044175302Recruiting
6Neoadjuvant and Adjuvant Checkpoint BlockadeNCT025193222Recruiting
7Dendritic Cells Plus Autologous Tumor RNA in Uveal MelanomaNCT019837483Recruiting
8Study in Subjects with Small Primary Choroidal MelanomaNCT030521271/2Active, not recruiting
9Follow-up of Patients with Uveal Melanoma Adapted to the Risk of Relapse (SALOME)NCT04424719Not applicableRecruiting
10Influence of Oral Treatment with Citicoline for the Prevention of Radiation Optic Neuropathy in Patients Treated for Uveal Melanomas with Proton Beam TherapyNCT01338389Not applicableActive, not recruiting
11Endoresection of the Tumor Scar or Transpupillary Thermotherapy for the Treatment of Large Uveal Melanomas (Endoresection-Laser)NCT02874040Not applicableRecruiting
(b) Metastatic or unresectable uveal melanoma.
NTreatment of Metastatic or Unresectable Uveal MelanomaIDPhaseStatus
1Intermittent Selumetinib for Uveal MelanomaNCT027687661Recruiting
2A Phase I Study of LXS196 in Patients with Metastatic Uveal Melanoma.NCT026013781Active, not recruiting
3Study of Immunotherapy Plus ADI-PEG 20 for the Treatment of Advanced Uveal MelanomaNCT039228801Active, not recruiting
4Isolated Hepatic Perfusion in Combination with Ipilimumab and Nivolumab in Patients with Uveal Melanoma Metastases (SCANDIUM II)NCT044633681Recruiting
5Autologous CD8+ SLC45A2-Specific T Lymphocytes with Cyclophosphamide, Aldesleukin, and Ipilimumab in Treating Patients with Metastatic Uveal MelanomaNCT030686241Active, not recruiting
6IKKb-matured, RNA-loaded Dendritic Cells for Metastasized Uveal MelanomaNCT043358901Recruiting
7A Study of RO7293583 in Participants with Unresectable Metastatic Tyrosinase Related Protein 1 (TYRP1)-Positive MelanomasNCT045513521Recruiting
8C7R-GD2.CART Cells for Patients with Relapsed or Refractory Neuroblastoma and Other GD2 Positive Cancers (GAIL-N)NCT036356321Recruiting
9Study of Safety and Tolerability of BCA101 Alone and in Combination with Pembrolizumab in Patients with EGFR-driven Advanced Solid TumorsNCT044295421Recruiting
10A Safety and Tolerability Study of INCAGN02390 in Select Advanced MalignanciesNCT036520771Active, not recruiting
11Modified Virus VSV-IFNbetaTYRP1 in Treating Patients with Stage III-IV MelanomaNCT038652121Recruiting
12Intravenous and Intrathecal Nivolumab in Treating Patients with Leptomeningeal DiseaseNCT030252561Recruiting
13A Study to Assess PV-10 Chemoablation of Cancer of the LiverNCT009866611Recruiting
14IN10018 Monotherapy and Combination Therapy for Metastatic MelanomaNCT041094561Recruiting
15A Phase II Study of BVD-523 in Metastatic Uveal MelanomaNCT034177392Active, not recruiting
16Nivolumab Plus Relatlimab in Patients with Metastatic Uveal MelanomaNCT045522232Recruiting
17Transarterial Radioembolization in Comparison to Transarterial Chemoembolization in Uveal Melanoma Liver Metastasis (SirTac)NCT029363882Recruiting
18Safety and Efficacy of IMCgp100 Versus Investigator Choice in Advanced Uveal MelanomaNCT030703922Active, not recruiting
19Trial of Nivolumab in Combination with Ipilimumab in Subjects with Previously Untreated Metastatic Uveal Melanoma (GEM1402)NCT026269622Active, not recruiting
20SIR-Spheres®® 90Y Microspheres Treatment of Uveal Melanoma Metastasized to LiverNCT014730042Active, not recruiting
21Nivolumab and Ipilimumab in Treating Patients with Metastatic Uveal MelanomaNCT015851942Active, not recruiting
22Efficacy Study of Pembrolizumab with Entinostat to Treat Metastatic Melanoma of the Eye (PEMDAC)NCT026976302Active, not recruiting
23Adoptive Transfer of Tumor Infiltrating Lymphocytes for Metastatic Uveal MelanomaNCT034675162Recruiting
24Ipilimumab and Nivolumab with Immunoembolization in Treating Participants with Metastatic Uveal Melanoma in the LiverNCT034725862Recruiting
25A Trial of Niraparib in BAP1 and Other DNA Damage Response (DDR) Deficient Neoplasms (UF-STO-ETI-001)NCT032073472Recruiting
26Cabozantinib-S-Malate Compared with Temozolomide or Dacarbazine in Treating Patients with Metastatic Melanoma of the Eye That Cannot Be Removed by SurgeryNCT018351452Active, not recruiting
27Iodine I 131 Monoclonal Antibody 3F8 in Treating Patients with Central Nervous System Cancer or Leptomeningeal CancerNCT004459652Active, not recruiting
28The Scandinavian Randomized Controlled Trial of Isolated Hepatic Perfusion for Uveal Melanoma Liver Metastases (SCANDIUM)NCT017853163Recruiting
29Percutaneous Hepatic Perfusion in Patients with Hepatic-dominant Ocular Melanoma (FOCUS)NCT026785723Active, not recruiting
30Study of PAC-1 and Entrectinib for Patients with Metastatic Uveal MelanomaNCT045898321/2Not yet recruiting
31A Study of the Intra-Patient Escalation Dosing Regimen with IMCgp100 in Patients with Advanced Uveal MelanomaNCT025703081/2Active, not recruiting
32Yttrium90, Ipilimumab, & Nivolumab for Uveal Melanoma with Liver MetastasesNCT029134171/2Recruiting
33PHP and Immunotherapy in Metastasized UM (CHOPIN)NCT042838901/2Recruiting
34Safety & Activity of Controllable PRAME-TCR Therapy in Previously Treated AML/MDS or Metastatic Uveal MelanomaNCT027436111/2Active, not recruiting
35Study of IDE196 in Patients with Solid Tumors Harboring GNAQ/11 Mutations or PRKC FusionsNCT039473851/2Recruiting
36A Study of PLX2853 in Advanced Malignancies.NCT032974241/2Recruiting
37Hypofractionated Stereotactic Linear Accelerator Radiotherapy of Uveal MelanomaNCT00872391Not applicableRecruiting
38Communicating with Patients on Cancer Resistance to Treatment: the Development of a Communication Tool (HECTOR)NCT04118062Not applicableNot yet recruiting
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Toro, M.D.; Gozzo, L.; Tracia, L.; Cicciù, M.; Drago, F.; Bucolo, C.; Avitabile, T.; Rejdak, R.; Nowomiejska, K.; Zweifel, S.; et al. New Therapeutic Perspectives in the Treatment of Uveal Melanoma: A Systematic Review. Biomedicines 2021, 9, 1311. https://doi.org/10.3390/biomedicines9101311

AMA Style

Toro MD, Gozzo L, Tracia L, Cicciù M, Drago F, Bucolo C, Avitabile T, Rejdak R, Nowomiejska K, Zweifel S, et al. New Therapeutic Perspectives in the Treatment of Uveal Melanoma: A Systematic Review. Biomedicines. 2021; 9(10):1311. https://doi.org/10.3390/biomedicines9101311

Chicago/Turabian Style

Toro, Mario Damiano, Lucia Gozzo, Luciano Tracia, Marco Cicciù, Filippo Drago, Claudio Bucolo, Teresio Avitabile, Robert Rejdak, Katarzyna Nowomiejska, Sandrine Zweifel, and et al. 2021. "New Therapeutic Perspectives in the Treatment of Uveal Melanoma: A Systematic Review" Biomedicines 9, no. 10: 1311. https://doi.org/10.3390/biomedicines9101311

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

Article Metrics

Back to TopTop