Open access peer-reviewed chapter

Uveal Melanoma: Factors Determining Metastatic Process, Epidemiology, Diagnosis, and Treatment

Written By

Darina Lysková, Paulína Plesníková, Viera Horvathova Kajabova, Lucia Demkova, Božena Smolková and Jela Valášková

Submitted: 18 July 2022 Reviewed: 02 September 2022 Published: 18 October 2022

DOI: 10.5772/intechopen.107683

From the Edited Volume

Melanoma - Standard of Care, Challenges, and Updates in Clinical Research

Edited by Sonia Maciá and Eduardo Castañón

Chapter metrics overview

99 Chapter Downloads

View Full Metrics

Abstract

Uveal melanoma (UM) is an ocular tumor with a dismal prognosis. It is the most frequent primary intraocular tumor in adults. The primary goal of treatment for uveal melanomas is to prevent metastasis. Despite outstanding advances in the diagnosis and treatment of primary UM, nearly 50% of patients develop metastases via hematogenous dissemination. Estimation of prognosis for patients with UM can be achieved by detecting genetic alterations or epigenetic changes in the tumor tissues. However, these techniques are not always available. The clinicopathological characteristics with limited accuracy are widely used instead to predict metastatic potential. Identifying novel markers with prognostic potential can help refine the prognosis of UM patients. As we know, no existing therapy has a significantly better impact on preventing metastasis. Based on published theories, the key role is existing micrometastasis before therapy starts. Researchers are focusing on developing adjuvant systemic therapy for metastatic UM. Getting to know the cause of metastatic uveal melanoma is crucial in it.

Keywords

  • uveal melanoma
  • metastases
  • genetic changes in UM
  • epigenetic changes in UM
  • epidemiology of UM
  • diagnosis and treatment UM

1. Introduction

Uveal melanoma is a rare form of melanoma, but the most frequent intraocular tumor in adults [1]. Comprising approximately 83% of ocular and 3% of all melanomas. It arises from melanocytes along the uveal layer of the eye, including the iris, ciliary body, and most often the choroid [2].

Primary UM is treated with either surgery or radiation with a low local recurrence rate. However, almost half of UM patients develop metastases, which may be caused by a virtually undetectable neoplasm already present at the time of the primary tumor diagnosis [3]. Most UM patients survive less than 12 months after metastases diagnosis due to the lack of effective therapies [4]. UM spreads through the blood. The liver is the preferred metastatic site, followed by the lungs and bones [5].

Various clinical, pathological, molecular, and cytogenetic markers assessed in tumors, such as specific chromosome copy number alterations [6], gene expression profiles [7], and the mutation status of known UM driver genes [8], can predict the risk of metastases and survival.

Advertisement

2. Genetic changes in uveal melanoma

2.1 Chromosomal rearrangements

The most frequent UM-specific aberrations include monosomy of chromosome 3 (M3), a gain in the short arm of chromosome 6 (6p), or a gain in the long arm of chromosome 8 (8q). Similar to the loss of the short arm of chromosome 8 (8p), the long arm of chromosome 6 (6q), and the short arm of chromosome 1 (1p) pose a high metastatic risk and present a poor prognosis [9, 10, 11].

Conversely, the presence of 6p amplification represents a protective factor due to its association with a good prognosis and lowered metastatic risk [12]. Although their prognostic value has been proven, and their sensitivity and specificity are limited in clinical use [13]. The problem seems to be that results differ based on laboratory methods used for detecting the amount of chromosomal copies, and they are not accurate.

2.2 Change in gene expression

Another way to predict the risk of metastasis is via gene expression analysis. A commercially available expression panel of 15 genes developed by Castle Biosciences categorizes patients as Class 1 (low metastatic risk) or Class 2 transcriptional subtype (high metastatic risk) [7, 14]. Four molecular subsets were proposed recently, based on a more complex classification [15, 16].

2.3 Mutation of genes

UM occurs mostly sporadically, however, rarely it occurs in families with an inherited predisposition for this malignancy. Mutations in gene BAP 1 are segregated in an autosomal dominant manner in the hereditary tumor syndrome. It is characterized by the occurrence of tumor disease in a family member at a young age, by the presence of numerous primary tumors, often bilaterally when the steam organs are affected. BAP 1 mutation is associated with cutaneous melanoma, mesothelioma, meningioma, and many others. The clinical phenotype includes UM in patients with oculodermal melanocytosis, skin melanoma, neurofibromatosis type 1, and Li-Fraumeni syndrome. In the case of a familiar form, the combination of clinical signs and genetic information can be used for early diagnosis in patients [17, 18, 19].

Advertisement

3. Epigenetics in uveal melanoma

The term epigenetics includes changes in gene expression and chromatin structure that are not related to a change in primary genetic information, that is, changes not encoded in the sequence of bases in the DNA chain [20]. In the broadest sense of the word, epigenetics can be understood as a bridge between the genotype and the phenotype of a cell [21].

The basic epigenetic mechanisms of gene expression regulation include DNA methylation, histone modification with subsequent chromatin remodeling, and non-coding RNA [22]. These mechanisms are essential for the normal development and homeostasis of the organism, and their disruption can lead to changes in gene function and malignant transformation, and can have an impact on individual signaling pathways involved in metastasis [23].

Epigenetic inactivation plays a role in genes located on chromosomes 1, 3, 6, or 8, that is, in chromosomes with proven abnormalities in UM. Monosomy 3 is present in approximately half of patients with UM. Genes that play a key role in hematogenous dissemination are located on this chromosome, for example, BAP1, RASSF1A, FHIT, CTNNB1, and SRY.

3.1 Methylation

It is the binding of a methyl group (-CH3) to the fifth carbon of cytosine by a covalent bond. Compared to normal cells, tumor cells have a disturbed DNA methylation pattern either by decreasing (hypomethylation) or increasing (hypermethylation) the number of methyl groups. During the onset of oncological diseases, these are significant processes that lead to an increase in chromosome instability. Primarily hypermethylation of promoters of tumor suppressor genes, hypomethylation of proto-oncogenes, and global hypomethylation [24].

In UM patients, DNA methylation was identified as the cause of inactivation of several genes. Aberrant hypomethylation of the PRAME gene, leading to its transcriptional inactivation, was associated with an increased metastatic risk [25]. The majority of hypermethylated genes in UM are p16, TIMP3, RASSF1A, RASEF, hTERT, and ES genes. They participate in the regulation of the cell cycle. Only the RASSF1A and p16 genes are also methylated in skin melanoma. In comparison, genes methylated in cutaneous melanoma, such as pTEN, TNFSF10D, COL1A2, MAGE, or CLDN11, were not methylated in UM [26].

Decreased levels of E-cadherin, a key protein that is inhibited in the epithelial-mesenchymal transition process, were identified in 56.2% of UM. They were indirectly correlated with the methylation of the CDH1 promoter gene, which encodes it [27, 28].

The researchers induced an increase in the expression of E-cadherin, which affected the phenotypic change in UM cells from spindle cell to epithelial type. Reactivation of the expression of aberrantly methylated genes by DNMTs inhibitors may represent a promising therapeutic strategy [23].

3.2 Histones modifications

Histones are basic proteins abundant in lysine and arginine residues that are found in nuclei of eukaryotic cells. They create structural units called nucleosomes. We know five families of histones H1/H5 (linker histones), H2, H3, and H4 (core histones). The nucleosome core is formed of two H2A–H2B dimers and a H3–H4 tetramer. Nucleosomes are wrapped into fibers of tightly packed chromatin. That means DNA winds around them. Histones prevent DNA from becoming tangled and protect it from DNA damage. They play important roles in DNA replication and gene regulation [29].

Post-translational covalent changes occur at the N-terminal ends of histones in mammalian cells through the action of histone-modifying enzymes. The most common modifications of histones, which play a key role in the regulation of gene expression are methylation, acetylation, phosphorylation, and ubiquitination. They affect the mobility and stability of chromatin and regulate its transcription [23].

Most UM Class 2 transcriptional subtype (high metastatic risk) contains inactivating mutations of the tumor suppressor gene BAP1. It encodes bap 1, which has a role in the progression of UM. It modifies histones by catalyzing the removal of ubiquitin from histone H2A. Its depletion leads to hyperubiquitination of H2A in melanocytes and melanoma cells and subsequent loss of differentiation and acquisition of tumor stem cell properties [30].

Histone deacetylase inhibitors (HDAC), therefore enable the restoration of the expression of epigenetically inactivated genes, necessary, for example, to control the cell cycle. In UM cell lines, primocultures created from patient tumor cells, and HDAC inhibitors, such as valproic acid, trichostatin A, panobinostat LBH-589, and suberoylanilide hydroxamic acid-induced proliferation inhibition, cell cycle arrest, increased tumor cell apoptosis, morphological and transcriptional changes consistent with melanocyte differentiation. HDAC inhibitors are in preclinical studies for the treatment of UM with the aim of prolonging the dormancy of micrometastatic disease [31, 32].

3.3 Non-coding mRNA

MicroRNA (miRNA) is mainly considered non-coding mRNA. These are short nucleotide single-stranded RNA molecules that participate in the post-transcriptional regulation of the expression of mediator RNAs (mRNA). It has been proven that miRNA functions as an oncogene or tumor suppressor gene in carcinogenesis. It binds to complementary mRNA and thereby inhibits mRNA translation and inactivates target genes [33].

Changes in the expression of many miRNAs have been described in cell lines of tumor structures and peripheral blood from patients with UM [34]. They play an important role in the deregulation of oncogenic pathways in UM and may promote metastatic spread. In addition to the fact that miRNAs can be interesting diagnostic and prognostic biomarkers, they offer us new therapeutic targets [35].

Epigenetic changes play an important role in the pathogenesis of oncological diseases. They are reversible; therefore, they are a good therapeutic target. In many preclinical studies, it has been proven that epigenetic drugs enable the restoration of aberrantly inactivated tumor-suppressor genes, and increase the sensitivity of resistant tumor cells to treatment.

The prerequisite for the discovery of effective drugs for the adjuvant therapy of UM and the treatment of metastatic UM is to necessarily accept the importance of epigenetic changes and understand their role in the pathogenesis of this disease.

Advertisement

4. Epidemiology

The most common primary intraocular malignancy in adults is uveal melanoma. It arises from melanocytes in the choroid, ciliary body, or iris. The incidence is 5.1 per million and has remained stable since at least 1970s. UM is the most common in Caucasians during the fifth to sixth decade of life [1]. Approximately 85% of UM is localized in the choroid [36], about 4–7% in the ciliary body, and 2–4% in iris, which is associated with early diagnosis and the best prognosis [37]. Associated with the worst prognosis is UM in the ciliary body.

Advertisement

5. Clinical diagnosis

Physical examination and health history are used to help diagnose intraocular melanoma, as well as eye exam with the dilated pupil (by ophthalmoscopy or slit-lamp biomicroscopy). Diagnosing uveal melanoma often requires serial fundus photography. Fluorescein angiography or indocyanine green angiography is used in the screening and follow-up of suspicious lesions. Other critical tools in the diagnosis of uveal melanoma are A and B scan ultrasonography and optical coherence tomography.

Advertisement

6. Management

The primary goal of treatment for uveal melanomas is to prevent metastasis. However, treatment of small lesions (less than 3 mm in thickness) is controversial, and it is not proven whether it prevents metastasis. Observation is generally recommended whenever it is possible.

Biopsy of the lesion is the only way to definitively identify uveal melanoma. It can be done after enucleation or by fine needle aspiration biopsy. The collected material is used for histological examination and cytopathological analysis.

Historically, enucleation (eyeball removal) was the standard treatment for primary UM, and it is still used when large tumors are present. However, it has been largely replaced by radiation therapy (i.e., brachytherapy or proton beam therapy) to spare the affected eye.

The results of the Collaborative Ocular Melanoma Study (COMS) in 2001, a large multicenter randomized control trial with 1317 patients confirmed that there was no significant difference in mortality after brachytherapy in comparison to enucleation for malignant UM [38]. Later other publications reported similar positive findings [39, 40, 41]. The decision to use brachytherapy vs. proton beam therapy is now largely made in regard to the size and location of the tumor and patient preference [42, 43, 44, 45]. Secondary complication can be present as glaucoma, serous retinal detachment, or cataract. The only effective treatment for cataracts is surgery with precise intra ocular lens power calculation [46, 47]. The serous retinal detachment can be present as complication in whole scale of eye disease, for example, uveal effusion syndrome [48].

For small tumors, the less commonly available treatment options can be used. These include transpupillary thermotherapy, photocoagulation, photodynamic therapy, and local resection.

Advertisement

Acknowledgments

Our publication was supported by APVV-17-0369 and VEGA 1/0395/21 projects.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Appendices and nomenclature

UMUveal melanoma
BAP 1BRCA1 associated protein 1
RASSF1Ras association domain family member 1
FHIT 2Fragile Histidine Triad Diadenosine Triphosphatase 2
CTNNB 1Catenin Cadherin-Associated Protein Beta 1
SRY, SOX2Sex determining region Y-box 2
PRAMENuclear Receptor Transcriptional Regulator
p16, CDKN2ACyclin-dependent kinase inhibitor 2A
TIMP3TIMP metallopeptidase inhibitor 3
RASSF1Ras association domain family member 1
RASEFRAS And EF-Hand Domain Containing
hTERTTelomerase reverse transcriptase in humans
PTENPhosphatase and tensin homolog
TNFSF10DTumor necrosis factor receptor super family member 10D
COL1A2Collagen Type I Alpha 2 Chain
MAGEThe Melanoma Antigen Gene
CLDN11Claudin 11
DNMTsDNA methyltransferases
CDH1Cadherin 1
HDACHistone deacetylase inhibitors

References

  1. 1. Singh AD, Turell ME, Topham AK. Uveal melanoma: Trends in incidence, treatment, and survival. Ophthalmology. 2011;118(9):1881-1885. DOI: 10.1016/j.ophtha.2011.01.040
  2. 2. McLaughlin CC, Wu XC, Jemal A, Martin HJ, Roche LM, Chen VW. Incidence of noncutaneous melanomas in the U.S. Cancer. 2005;103(5):1000-1007. DOI: 10.1002/cncr.20866
  3. 3. Eskelin S, Pyrhönen S, Summanen P, Hahka-Kemppinen M, Kivelä T. Tumor doubling times in metastatic malignant melanoma of the uvea: Tumor progression before and after treatment. Ophthalmology. 2000;107:1443-1449. DOI: 10.1016/S0161-6420(00)00182-2
  4. 4. Blum ES, Yang J, Komatsubara KM, Carvajal RD. Clinical management of uveal and conjunctival melanoma. Oncology (Williston Park). 2016;30(1):29-48
  5. 5. Diener-West M, Reynolds SM, Agugliaro DJ, et al. Development of metastatic disease after enrollment in the COMS trials for treatment of choroidal melanoma: Collaborative Ocular Melanoma Study Group Report No. 26. Archives of Ophthalmology. 2005;123(12):1639-1643. DOI: 10.1001/archopht.123.12.1639
  6. 6. Damato B, Coupland SE. Translating uveal melanoma cytogenetics into clinical care. Archives of Ophthalmology. 2009;127(4):423-429. DOI: 10.1001/archophthalmol.2009.40
  7. 7. Onken MD, Worley LA, Ehlers JP, Harbour JW. Gene expression profiling in uveal melanoma reveals two molecular classes and predicts metastatic death. Cancer Research. 2004;64(20):7205-7209. DOI: 10.1158/0008-5472.CAN-04-1750
  8. 8. Johansson PA, Brooks K, Newell F, et al. Whole genome landscapes of uveal melanoma show an ultraviolet radiation signature in iris tumours. Nature Communications. 2020;11:2408. DOI: 10.1038/s41467-020-16276-8
  9. 9. Ehlers JP, Worley L, Onken MD, Harbour JW. Integrative genomic analysis of aneuploidy in uveal melanoma. Clinical Cancer Research. 2008;14(1):115-122. DOI: 10.1158/1078-0432.CCR-07-1825
  10. 10. Ewens KG, Kanetsky PA, Richards-Yutz J, et al. Genomic profile of 320 uveal melanoma cases: Chromosome 8p-loss and metastatic outcome. Investigative Ophthalmology & Visual Science. 2013;54(8):5721-5729. DOI: 10.1167/iovs.13-12195
  11. 11. Versluis M, de Lange MJ, van Pelt SI, Ruivenkamp CAL, Kroes WGM, et al. Digital PCR validates 8q dosage as prognostic tool in uveal melanoma. PLoS One. 2015;10(3):e0116371. DOI: 10.1371/journal.pone.0116371
  12. 12. Amaro A, Gangemi R, Piaggio F, et al. The biology of uveal melanoma. Cancer Metastasis Reviews. 2017;36:109-140. DOI: 10.1007/s10555-017-9663-3
  13. 13. Kaliki S, Shields CL, Shields J. Uveal melanoma. Indian Journal of Ophthalmology. 2015;63(2):93-102. DOI: 10.4103/0301-4738.154367
  14. 14. Onken MD, Worley LA, Char DH, Augsburger JJ, Correa ZM, Nudleman E, et al. Collaborative ocular oncology group report number 1: Prospective validation of a multi-gene prognostic assay in uveal melanoma. Ophthalmology. 2012;119:1596-1603. DOI: 10.1016/j.ophtha.2012.02.017
  15. 15. Robertson AG, Shih J, Yau C, et al. Integrative analysis identifies four molecular and clinical subsets in uveal melanoma. Cancer Cell. 2017;32(2):204, e15-220. DOI: 10.1016/j.ccell.2017.07.003
  16. 16. Jager MJ, Shields CL, Cebulla CM, et al. Uveal melanoma. Nature Reviews. Disease Primers. 2020;6(1):24. DOI: 10.1038/s41572-020-0158-0
  17. 17. Houlston RS, Damato BE. Genetic predisposition to ocular melanoma. Eye (London, England). 1999;13(Pt 1):43-46. DOI: 10.1038/eye.1999.9
  18. 18. Jay M, McCartney AC. Familial malignant melanoma of the uvea and p53: A Victorian detective story. Survey of Ophthalmology. 1993;37(6):457-462. DOI: 10.1016/0039-6257(93)90142-t
  19. 19. Easton DF, Steele L, Fields P, et al. Cancer risks in two large breast cancer families linked to BRCA2 on chromosome 13q12-13. American Journal of Human Genetics. 1997;61(1):120-128. DOI: 10.1086/513891
  20. 20. Herceg Z, Hainaut P. Genetic and epigenetic alterations as biomarkers for cancer detection, diagnosis and prognosis. Molecular Oncology. 2007;1(1):26-41. DOI: 10.1016/j.molonc.2007.01.004
  21. 21. Bird A. DNA methylation patterns and epigenetic memory. Genes & Development. 2002;16(1):6-21. DOI: 10.1101/gad.947102
  22. 22. Chokhachi Baradaran P, Kozovska Z, Furdova A, Smolkova B. Targeting epigenetic modifications in uveal melanoma. International Journal of Molecular Sciences. 2020;21(15):5314
  23. 23. Sharma S, Kelly TK, Jones PA. Epigenetics in cancer. Carcinogenesis. 2010;31(1):27-36. DOI: 0.1093/carcin/bgp220
  24. 24. Greenberg MVC, Bourc'his D. The diverse roles of DNA methylation in mammalian development and disease. Nature Reviews. Molecular Cell Biology. 2019;20(10):590-607. DOI: 10.1038/s41580-019-0159-6
  25. 25. Field MG, Durante MA, Decatur CL, et al. Epigenetic reprogramming and aberrant expression of PRAME are associated with increased metastatic risk in class 1 and class 2 uveal melanomas. Oncotarget. 2016;7(37):59209-59219. DOI: 10.18632/oncotarget.10962
  26. 26. Fu S, Wu H, Zhang H, Lian CG, Lu Q. DNA methylation/hydroxymethylation in melanoma. Oncotarget. 2017;8(44):78163-78173. DOI: 10.18632/oncotarget.18293
  27. 27. Venza M, Visalli M, Catalano T, et al. DNA methylation-induced E-cadherin silencing is correlated with the clinicopathological features of melanoma. Oncology Reportsa. 2016;35(4):2451-2460. DOI: 10.3892/or.2016.4618
  28. 28. Versluis M, De Lange MJ, Van Pelt S, Luzten GPM, Jager MJ. Epigenetic regulation of the epithelial phenotype in uveal melanoma. Acta Ophtalmology. 2013;2:5-8. DOI: 10.1111/j.1755-3768.2013.1773.x
  29. 29. Kanwal R, Gupta S. Epigenetic modifications in cancer. Clinical Genetics. 2012;81(4):303-311. DOI: 10.1111/j.1399-0004.2011.01809.x
  30. 30. Matatall KA, Agapova OA, Onken MD, Worley LA, Bowcock AM, Harbour JW. BAP1 deficiency causes loss of melanocytic cell identity in uveal melanoma. BMC Cancer. 2013;13:371. DOI: 10.1186/1471-2407-13-371
  31. 31. Landreville S, Agapova OA, Matatall KA, et al. Histone deacetylase inhibitors induce growth arrest and differentiation in uveal melanoma. Clinical Cancer Research. 2012;18(2):408-416. DOI: 10.1158/1078-0432.CCR-11-0946
  32. 32. Moschos SJ, Mantzoros CS. The role of the IGF system in cancer: From basic to clinical studies and clinical applications. Oncology. 2002;63(4):317-332. DOI: 10.1159/000066230
  33. 33. Piva R, Spandidos DA, Gambari R. From microRNA functions to microRNA therapeutics: Novel targets and novel drugs in breast cancer research and treatment (review). International Journal of Oncology. 2013;43(4):985-994. DOI: 10.3892/ijo.2013.2059
  34. 34. Horvathova Kajabova V, Soltysova A, Demkova L, Plesnikova P, Lyskova D, Furdova A, et al. KIT expression is regulated by DNA methylation in uveal melanoma tumors. International Journal of Molecular Sciences. 2021;22(19):10748
  35. 35. Smit KN, Chang J, Derks K, et al. Aberrant MicroRNA expression and its implications for uveal melanoma metastasis. Cancers (Basel). 2019;11(6):815. DOI: 10.3390/cancers11060815
  36. 36. Shields CL, Kaliki S, Furuta M, Mashayekhi A, Shields JA. Clinical spectrum and prognosis of uveal melanoma based on age at presentation in 8,033 cases. Retina. 2012;32(7):1363-1372
  37. 37. Shields CL, Kaliki S, Shah SU, Luo W, Furuta M, Shields JA. Iris melanoma: Features and prognosis in 317 children and adults. Journal of AAPOS. 2012;16(1):10-16
  38. 38. Singh AD, Kalyani P, Topham A. Estimating the risk of malignant transformation of a choroidal nevus. Ophthalmology. 2005;112(10):1784-1789
  39. 39. Furdova A, Sramka M, Chorvath M, Kralik G, Krasnik V, Krcova I, Waczulikova I. Stereotactic radiosurgery in intraocular malignant melanoma-retrospective study. Neuro Endocrinology Letters. Jan 2014;35(1):28-36
  40. 40. Furdova A, Sramka M, Chorvath M, Kralik G, Furda R, Gregus M. Clinical experience of stereotactic radiosurgery at a linear accelerator for intraocular melanoma. Melanoma Research. Oct 2017;27(5):463-468
  41. 41. Furdova A, Babal P, Kobzova D, Zahorjanova P, Kapitanova K, Sramka M, et al. Uveal melanoma survival rates after single dose stereotactic radiosurgery. Neoplasma. 15 Nov 2018;65(6):965-971
  42. 42. Furdova A, Furda R, Sramka M, et al. Stereotactic irradiation on linear accelerator - ultrasound versus MRI in choroidal melanoma volume calculation. BMC Ophthalmology. 2022;22:333
  43. 43. Zahorjanová P, Sekáč J, Babál P, Štubňa M. Enucleation after stereotactic radiosurgery in patients with uveal melanoma. Čes. a slov. Oftal. 2020;76(1):46-51
  44. 44. Furdova A, Sramka M. Uveal Malignant Melanoma and Stereotactic Radiosurgery: Intraocular Uveal Melanoma and One-Day Session Stereotactic Radiosurgery at Linear Accelerator. Saarbrücken: LAP LAMBERT Academic Publishing; 2014. p. 188
  45. 45. Horkovicova K, Furdova A. Uveal melanoma. In: Lasfar A, Cohen-Solal K, editors. Melanoma. London, UK: IntechOpen; 2020
  46. 46. Popov I, Waczulikova I, Stefanickova J, et al. Analysis of biometric parameters of 2340 eyes measured with optical biometer Lenstar LS900 in a Caucasian population. European Journal of Ophthalmology. 2022;32(1):213-220
  47. 47. Popov I, Popova V, Sekac J, Krasnik V. Does every calculation formula fit for all types of intraocular lenses? Optimization of constants for Tecnis ZA9003 and ZCB00 is necessary. Medicina. 2021;57(4):319
  48. 48. Popov I, Popova V, Krasnik V. Comparing the results of vitrectomy and sclerectomy in a patient with nanophthalmic uveal effusion syndrome. Medicina. 2021;57(2):120

Written By

Darina Lysková, Paulína Plesníková, Viera Horvathova Kajabova, Lucia Demkova, Božena Smolková and Jela Valášková

Submitted: 18 July 2022 Reviewed: 02 September 2022 Published: 18 October 2022