• No results found

Genomic and proteomic analysis in uveal melanoma Zuidervaart, W.

N/A
N/A
Protected

Academic year: 2021

Share "Genomic and proteomic analysis in uveal melanoma Zuidervaart, W."

Copied!
17
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Citation

Zuidervaart, W. (2005, May 25). Genomic and proteomic analysis in uveal melanoma. Retrieved from https://hdl.handle.net/1887/2696

Version: Corrected Publisher’s Version

License: Licence agreement concerning inclusion of doctoral thesis in theInstitutional Repository of the University of Leiden Downloaded from: https://hdl.handle.net/1887/2696

(2)
(3)

C

LINICAL ASPECTS OF UVEAL MELANOMA

Epidemiology

Uveal melanoma is the most common primary intraocular tumor in adults in the Western world and represents 3% of all melanoma (Egan et al., 1988; Singh et al., 2003). The age-adjusted incidence of uveal melanoma varies from 5-8 patients per 1 million per year and has remained stable over the past 25 years (Singh et al., 2003).Uveal melanoma spread almost without any exception hematogenously, predominantly to the liver. The tumor is of an aggressive type: almost 50% of the patients with uveal melanoma will ultimately die from

metastatic disease (Jensen et al., 1982; McLean et al., 1993), while no effective treatment for metastases is yet available. Once the primary tumor has been diagnosed in the patient, the median survival time is 6.5 years (McLean et al., 1993). Life expectancy after metastases have been detected is only 2-9 months (Seddon et al., 1983; Albert et al., 1992; Kath et al., 1993).

Most uveal melanomas are located in the ciliary body and/or the choroid (23% and 72%,

respectively), whereas iris melanomas account for only a small percentage of these tumors, i.e. 5% (Yanoff and Fine, 1989). Iris melanomas are less aggressive with a low incidence of

developing metastases and are therefore not a subject of study in this thesis.

At the time of diagnosis, less than 2% of the patients have clinically detectable metastases, but

many patients may have already subclinical metastases (Donoso et al., 1985). It may be that treatment at this stage with e.g. immunotherapy may be the only way to improve survival in these patients, but would only be used in very high risk individuals. This highlights the critical need to identify prognostic markers indicative of uveal melanoma invasive and metastatic potential.

Diagnosis and treatment

Like cutaneous melanoma, uveal melanoma is originally derived, from melanocytes of the neurectoderm (choroid and ciliary body, Figure 1.1). Two main cell types have been recognized, i.e. the spindle cell type and the epithelioid cell type. Since both cell types are present in many uveal melanomas, additionally, the mixed cell type classification has been introduced (Zimmerman 1986).

(4)

Figure 1.1 Schematic view of an eye. (A) Choroid and (B) Ciliary body

In order to reduce the risk to develop metastases it is essential to treat the primary tumor as effectively as possible. For large tumors (Largest Tumor Diameter (LTD) > 15 mm and/or

thickness > 5 mm), enucleation remains the first choice of treatment, as well as in case of

extensive involvement of the optic disc or extensive extrascleral extension (Shields et al., 1991; Seregard and Kock 1995). For small (LTD < 10 mm and thickness ≤ 3 mm) and

medium-sized (LTD between 10-15 mm and/or thickness between 3-5 mm) tumors, different treatment strategies are used (Shields et al., 1991). These include local resection of the tumor (Damato 1993), plaque radiotherapy (Lommatzsch and Kirsch 1988; Shields and Shields., 1993; Lommatzsch et al., 2000), stereotactic radiotherapy (Zehetmayer et al., 2000) and thermotherapy (Oosterhuis et al., 1995). A new trend in ocular oncology is combining different treatment modalities such as transpupillary thermotherapy (TTT) combined with plaque radiotherapy to obtain better tumor control (Journee-de Korver et al., 1997; Seregard and Landau 2001).

Predisposing factors

Several parameters that predispose to uveal melanoma have been described, including

phenotypic risk factors. Race is one of the most significant host factors, as uveal melanoma is about 150 times more common in Caucasian than in African individuals (Egan et al., 1988; Singh et al., 2003) and is less common in Asians (Kuo et al., 1982; Biswas et al., 2002). People with light-colored eyes appear to be at a higher risk than dark-eyed people (Gallagher et al., 1985; Tucker et al., 1985) and subjects with brown eyes seems to be more protected against sunlight compared to those with blue eyes (Tucker et al., 1985; Holly et al., 1990). Although there is ample evidence of sunlight exposure as a risk factor for cutaneous

melanoma (Elwood and Jopson 1997; Gilchrest et al., 1999), the evidence in uveal melanoma is contradictory (Dolin et al., 1994; Egan et al., 1988). In a report by Li et al. (2000), the distribution of tumor origin correlated with the dose distribution of solar radiation on the retinal hemisphere, but these findings contradicted another report, in which the distribution of tumors in the choroid was found to be randomly distributed (Schwartz et al., 1997). Although there is biologic evidence that ultraviolet radiation induces DNA damage in cutaneous

B

Melanoma

B

(5)

melanoma (Gilchrest et al., 1999), still, no conclusive judgement about the role of sunlight in uveal melanoma can be made (reviewed by Singh 2004).

Uveal melanoma usually occurs sporadically in the absence of obvious genetic predisposing factors. However, in some patients, there might be a genetic predisposition. Familial uveal melanoma most often (63%) affects first-degree relatives, but rarely affects more than two

persons in one family, and therefore it may be associated with a generalized inherited predisposition to cancer (Singh et al., 1996).

Furthermore, there are a few clinical conditions that can predispose to or are associated with uveal melanoma, including ocular melanocytosis, neurofibromatosis type I, and familial atypical multiple mole melanoma syndrome (FAMMM). Ocular melanocytosis is a congenital condition characterized by hyperpigmentation of the uveal tract, sclera and epissclera. When hyperpigmentation of periocular skin takes place along the trigeminal nerve distribution, it is known as oculodermal melanocytosis (Nevus of Ota) (Gonder et al., 1982). Approximately 1,4% of uveal melanoma patients have oculodermal melanocytosis (ODM), indicating that

ODM is about 35 times more common in the uveal melanoma population (Gonder et al., 1982). The lifetime risk to develop uveal melanoma is estimated to be one of about 400 ODM patients compared to one out of 13,000 in the general population (Singh et al., 1998).

Neurofibromatosis type 1 has also been linked to uveal melanoma. This combination of tumors serves to emphasize the common neuroectodermal origin of tumors in this autosomal dominant condition (Specht and Smith 1988; Antle et al., 1990). In the FAMMM or

Dysplastic Nevus Syndrome, increased numbers of cutaneous nevi, cutaneous melanoma, as well as conjunctival nevi and uveal melanoma have been reported. Patients with uveal melanoma are more at risk to develop dysplastic nevi and cutaneous melanoma (Bataille et al., 1995; Van Hees et al., 1998). The p16 gene (CDKN2A) which is localized on

chromosome 9p21, is inactivated in a significant number of sporadic cancers, including cutaneous melanoma. It has been demonstrated that approximately 50% of all FAMMM

families show linkage to this region (Goldstein et al., 1994; Gruis et al., 1995). Whereas

p16ink4a (CDKN2A) is the main target for inactivation in cutaneous melanoma, mutation

screening and deletion mapping in uveal melanoma did not reveal the same results (Merbs et al., 1999). However, Van der Velden et al. (2001) demonstrated that hypermethylation of

p16ink4a is the cause of inactivation of this gene in uveal melanoma, which interestingly was

found more frequently in tumors from patients who developed metastatic disease. Germline

BRCA2 gene mutations have also been described to have an association with ocular melanoma

(Easton et al., 1997; Sinilnikova et al., 1999): they occur in 3% of patients younger than 50

years (Scott et al., 2002).

Prognostic indicators, clinical, immunological and histopathological parameters

(6)

also considered to be significant factors. Other immunological parameters are also of prognostic relevance. The number of CD3+ (T-lymphocytes) and CD4+ lymphocytes are

positively correlated with HLA expression (De Waard-Siebinga et al., 1996). HLA phenotype alterations is a common feature in various tumors, including cutaneous and uveal melanoma. In uveal melanoma, lack of expression of HLA-A as well as HLA-B antigens was found to be correlated with better patient survival (Blom et al., 1997). Most tumors lack HLA-C and HLA-G as well, but the biological importance of this is not yet clear (Hurks et al., 2001). These findings may suggest that shedding of uveal melanoma micrometastases with downregulated HLA class I antigen expression into the systemic circulation may facilitate their removal and prevent the development of metastases. It also may suggest that Natural Killer-cell-based lysis may be more effective in destroying blood-borne uveal melanoma cells than T-cell-mediated cytotoxicity (Blom et al., 1997; Jager et al., 2002; Anastassiou et al., 2003).

The expression of Epidermal Growth Factor Receptor (EGFR) has been found to be correlated with death due to metastatic disease in uveal melanoma (Hurks et al., 2000), with another report concluding that tumor-associated macrophages can express this receptor (Scholes et al., 2001). However, Ma and Niederkorn (1998) reported specific EGFR expression on uveal melanoma cell lines, indicating that these findings may not be exclusive.

Differences in invasive behaviour between uveal melanomas could be one of the most important factors for their differences in clinical outcome. Molecular markers like integrins and the involvement of cadherin-catenin adhesion complexes in the invasive potential of uveal melanoma cells have been the subject of several studies and have provided more insight in these complicated processes (Elshaw et al., 2001; Woodward et al., 2002; Seftor et al., 2002; Conway et al., 2003).

Cytogenetic and genetic markers as prognostic indicators will be discussed in the next paragraph.

P

ROGNOSTIC INDICATORS

, (C

YTO

)G

ENETIC PARAMETERS

Chromosomal aberrations

Cytogenetic and comparative genomic hybridization analysis have revealed the involvement of several chromosomal aberrations in uveal melanoma. Sisley et al. (1997) identified a correlation with abnormalities in chromosomes 3, 6 and 8 and prognostic outcome.

Monosomy 3 in uveal melanoma has been proven to be an important determinant to predict metastatic potential (Prescher et al., 1996) and gain of chromosomal arm 8q, which includes the C-Myc oncogene locus, has been found in 65% of uveal melanomas (Speicher et al., 1994;

(7)

The subsequent paragraphs describe the potential involvement of genes and genetic pathways in uveal melanoma.

AKT and MAPK pathways

Another potential role in uveal melanogenesis involved the PTEN gene. The PTEN gene encodes a dual specific phosphatase, which plays a major role in the inhibition of cell migration and the formation of focal adhesions (Tamura et al., 1998). PTEN counteracts phosphatidylinositol 3-OH kinase (PI3-kinases) functions, which are associated with cell growth and survival (Di Cristofano and Pandolfi 2000; Vazquez and Sellers 2000). Somatic mutations of the PTEN gene have been detected in about 40% of cutaneous melanoma and

loss of this gene contributes to tumor development (Guldberg et al., 1997, Stahl et al., 2003). Furthermore, it has been recently found that there is a potential cooperation between the tumor suppressor gene PTEN and two other components of the RAS signaling network, NRAS and BRAF, suggesting that the MAPK (RAS) and the AKT pathways (PTEN) are frequently activated in parallel in melanogenesis (Tsao et al., 2004) However, no PTEN mutations were detected in a panel of uveal melanoma cell lines and cytogenetic

abnormalities involving chromosome 10q23 were not observed, as described in chapter 2 (Naus et al., 2000). The influence of the MAPK pathway in uveal melanoma will be discussed later in this chapter.

Rb and p53 pathways

Mutational deregulation of the cell-cycle is a hallmark of tumorigenesis (Hanahan and Weinberg 2000). The protein product of the Retinoblastoma gene (Rb) plays a central role as inhibitor of cellular proliferation (Bartek et al., 1997). The Rb gene is frequently mutated in certain cancers such as retinoblastoma, osteosarcoma, and small-cell lung cancer (Friend et al., 1986; Harbour et al., 1988), but no Rb mutations have been reported in uveal melanoma. This stimulated research into alterations in components of the interconnecting signaling pathways of the Rb gene and the p53 transcription factor (reviewed in Sherr and McCormick 2002). P53 is the most commonly mutated tumor suppressor in human cancer (Harris and Hollstein 1993), but no evidence exists for p53 mutations in either cutaneous or uveal melanoma (Chana et al., 1999; Kishore et al., 1996; Florenes et al., 1994). However, there is now evidence that functional abnormalities in both the Rb and p53 pathways are playing a role (Brantley and Harbour 2000a). HDM2 (human homologue of murine double minute MDM2)) is an inhibitor of p53 that targets p53 for degradation (Haupt et al., 1997). Although high expression of HDM2 in uveal melanoma cells has been related to poor clinical outcome (Coupland et al., 2000) and blocking experiments of HDM2 have led to rapid onset of

(8)

Recently, the mitogen-activated protein kinase (MAPK) pathway, has been found to be of great importance in the development of melanocytic neoplasia (Cohen et al., 2002;

Satyamoorthy et al., 2003). Of three RAS genes found to be activated by mutation in human tumors, NRAS (neuroblastoma RAS viral (v-ras) oncogene homolog) is most commonly mutated in cutaneous melanomas (Van Elsas et al., 1996). In the active GTP-bound state, RAS activates a number of downstream signalling cascades involved in controlling cell growth and behaviour. Initially, RAS interacts with and activates the serine/threonine protein kinase BRAF that acts in the MAPK pathway to transduce regulatory signals from RAS to MEK1/2. The signal tranducer mitogen-activated protein kinase/extracellular signal-related kinase kinase (MEK1/2) phosphorylates extracellular signal-regulated kinase (ERK1/2, p44/42), leading to the activation of these kinases, which in turn activate a variety of transcription factors, including ELK1, again through phosphorylation.

It has emerged that BRAF (v-raf murine sarcoma viral oncogene homolog B1) is very frequently activated by mutation in cutaneous nevi and melanomas (Davies et al., 2002; Pollock et al., 2003; Omholt et al., 2003; Cohen et al., 2004;Tsao et al., 2004). All BRAF mutations in cutaneous pigmented neoplasms occur within the kinase domain and most frequently consists of a 1796T A transversion in exon 15 (V599E) (Davies et al., 2002). Since cutaneous and uveal melanoma both arise from neural crest-derived melanocytes, the MAPK pathway came also to the attention in uveal melanoma research (see table III in chapter 3). Our data support the notion that activation of MAPK is indeed involved in development of uveal melanoma, but occurs via a different mechanism(s) to that in the majority of cutaneous melanomas (see chapter 3 in this thesis).

Genomics and Proteomics

The term genomics refers to the study of the human genome and proteomics refers to the analysis of the protein complement of the genome. With the introduction of high-throughput gene expression profiling techniques, and the enormous improvement in protein analysis technology, a major progress in the understanding of the involvement of multiple genes and gene products in association with uveal melanoma development, and cancer in general, have been established (Bittner et al., 2000 ; Tschentscher et al., 2003; Van der Velden et al., 2003; Zuidervaart et al., 2003).

This section describes the underlying methodology, as well as the present use of genomic and proteomic analysis, with special attention to its application in uveal melanoma research. Large scale gene expression analysis has proved to be an important strategy to identify gene

expression profiles, which are useful in classifying tissues according to pathological or prognostic subgroups. The unique quality of this technique lies in the ability to define

coordinate expression patterns, capable of representing a large number of relevant genes and a high degree of interaction, leading eventually to the detection of a potentially affected

(9)

expression data that may lead to a better understanding of the regulatory events involved in developmental processes in uveal melanoma.

G

ENOMICS

GENE EXPRESSION PROFILING

Techniques

Different types of microarrays have been developed. One of the mostly used arrays are the gene expression arrays with thousands of different gene transcripts hybridized on nylon filters, glass slides or chips. Hybridization of tumor RNA on the carrier will provide a profile of genes that are transcribed in the tumor. Two types of arrays are frequently used, the cDNA-array (see Figure 1.2) and the oligonucleotide-cDNA-array (Affymetrix). Oligonucleotide-cDNA-arrays determine the expression of a messenger by measuring hybridization to a perfect match oligo and an internal control mismatch oligo (Lockhart et al., 1996).

Another microarray application is the tissue microarray (TMA) (Kononen et al., 1998). With this method, sections of multiple small tissue cores are placed on a glass slide.

Immunohistochemistry and In Situ Hybridisation can be performed on these sections revealing a clear impression of the tumor under study.

Gene expression profiling in uveal melanoma research

Maniotis et al. (1999) used cDNA microarray analysis to investigate the relationship between aggressive melanoma cell phenotype and the mechanism responsible for the generation of patterned matrix-associated vascular channels characteristic for both aggressive uveal melanoma and cutaneous melanoma. They furthermore used cDNA microarray analysis to confirm a genetic reversion to a pluripotent embryonic-like gene expression phenotype in highly aggressive uveal melanoma cells.

Seftor and coworkers (2002), extended their previous observations of vasculogenic mimicry by comparing gene expression profiles of highly invasive versus poorly invasive uveal melanoma cells from a heterogeneous tumor comprised of cells that had metastasized from the original tumor to the liver.

(10)

Figure 1.2 Flowchart of a microarray experiment.

P

ROTEOMICS

Techniques

Two dimensional Poly-Acrylamide Gel Electrophoresis (2D-PAGE) is a powerful technique for protein separation, first according to pH (isoelectric point) and then acccording to size (molecular weight). The proteins can be visualized by staining with different agents, such as colloidal silver, fluorescent stains, or colloidal Coomassie Brilliant Blue G. Between 500-2000 spots can be reliably detected on large gels. Spots of interest are excized from the gel, digested, and analysed with Mass Spectrometry (MS). Proteins or peptides are ionized by electrospray ionization from liquid state (Fenn et al., 1989) or matrix-assisted laser desorption ionization (MALDI) from solid state (Karas and Hillenkamp 1988), and the mass of the ions is measured by various coupled analyzers (James 1997). The masses of the peptides can then be measured by MS, to produce a peptide mass fingerprint. This fingerprint will be compared with databases in order to identify the protein of interest (Figure 1.3). Protein microarrays have been developed recently as well, based on affinity (Haab et al., 2001; Schweitzer 2002). A great advantage is the decrease amount of material required, compared to the 2D method.

Test Reference RNA isolation Reverse transcriptase Labeling Cy5 (Red) R R R Cy3 (Green) G G G

Co-hybridization to microarray slide

(11)

Figure 1.3 Flowchart of a 2D-PAGE experiment.

Proteomics in cancer and uveal melanoma research

Proteomics is a powerful screening method for alterations in protein expression and posttranslational modifications. 2D-PAGE analysis in tumor research in general has been succesful in identifying proteins differentially expressed between normal liver cells and hepatocellular carcinoma cells (Yu et al., 2000) and in the identification of markers that differentiate between stages of bladder squamous cell carcinomas (Ostergaard et al., 1997). Although several examples of the use of proteomic research by 2D-PAGE have been

described for cutaneous melanoma (Bernard et al., 2003), the study by Missotten et al. (2003) is to our knowledge the only published project on proteomic analysis of uveal melanoma and outlines the use of proteomics in aqueous humor of uveal melanoma patients compared to a control group.

In chapter 8, two-dimensional electrophoresis (2D-PAGE) and mass spectrometry on a primary uveal melanoma cell line and two of its metastatic cell lines is used, in order to identify proteins associated with metastasis of uveal melanoma.

A

IMS AND SCOPE OF THIS THESIS

The prognosis of patients with uveal melanoma is poor. Therefore, the importance to identify indicators of metastatic potential of uveal melanomas is essential. First of all, biological mechanisms that determine the level of aggressiveness of uveal melanoma are still unclear and secondly development of metastases is a complex mechanism in which many interrelated, but yet to identify, steps are involved.

Gene expression profiling technology has improved tremendously during the last couple of years. It is only a couple of years ago that expression profiling by microarray analysis has gained interest in human cancer research. 5 years ago conventional profiling allowed to study only a few hundreds of genes, whereas only a few years later, technology evolved in more affordable, and very advanced expression profiling techniques, providing data on more than

Condition A Condition B

Protein extraction

First dimension IEF

Second dimension SDS-PAGE

2DE image analysis Excision protein spots

and protein digestion MS analysis

Protein identification and characterization

(12)

30.000 genes. The same technological improvement has taken place in the analysis of the proteome. By taking advantage of these recent technological in genomic and proteomic research, the main outline of this thesis is to define prognostic markers and/or profiles to categorize uveal melanoma in groups in order to identify the biological mechanism of metastatic disease of uveal melanomas.

D

ESCRIPTION CONTENTS PER CHAPTER

In contrast to cutaneous melanoma, genetic mutations and biological evidence of aberrations in signalling pathways are less well described in uveal melanoma. Involvement of PTEN and the MAPK pathway in uveal melanoma, both proven relevant in cutaneous melanoma, are investigated in chapters 2 and 3. We assess whether the MAPK pathway is similarly

activated in melanoma of the uvea and screen for activating mutations in the NRAS, HRAS,

KRAS and BRAF genes in uveal melanoma cell lines and in primary uveal melanomas. The

expression level of the downstream MAPK pathway members MEK, ERK and ELK are determined to reveal any additional potential alterations.

In the past recent years, cDNA microarray studies have become a powerful tool to detect differences in gene expression in different types of cancer and subsequently demonstrated that expression profiling can serve as a prognostic tool. In chapters 4 and 5 of this thesis, filter

(macro) arrays are used with more than thousand cDNAs related to cancer development. To identify molecules involved in dissemination, expression profiles are compared using cell lines derived from a primary uveal melanoma and two liver metastases from the same patient as a model for tumor progression. In addition, to reveal genes suppressed by

hypermethylation, a comparison of expression profiles is performed between the primary melanoma cell line and the same cell line after demethylation by 5-aza-2'deoxycytidine treatment.

In chapter 5, uveal melanoma cell lines are compared to normal human melanocytes of the eye in order to reveal genes that are differentially expressed. Potential progression markers are subsequently validated for their prognostic value in primary uveal melanoma specimens. In the next two chapters gene expression profiling is put to a higher level by using 19K cDNA microarrays. In chapter 6, we use microarray expression profiling combined with phenotypic

characterization of uveal melanoma and melanocyte cell lines, in order to define a gene signature associated with invasive potential. In light of the observation that Wnt5a expression is associated with invasive potential of cutaneous melanoma, we determine in chapter 7 if

activation of the Wnt signalling pathway, through increased expression of Wnt5a, -catenin and MMP7, contributes to an invasive phenotype in uveal melanoma.

In chapter 8, we focus on the differential protein expression of a primary uveal melanoma

cell line and two of its metastatic cell lines, using two-dimensional electrophoresis (2D-PAGE) and mass spectrometry in order to identify proteins that are associated with metastasis of uveal melanoma.

In chapter 9 the previous studies and future research perspectives are discussed. To conclude,

(13)

R

EFERENCES

Albert DM, Niffenegger AS, Willson JKV. Treatment of metastatic uveal melanoma: review and recommendations. Surv Ophthalmol 1992;36:429-438.

Anastassiou G, Rebmann V, Wagner S, Bornfeld N, Grosse-Wilde H. Expression of classic and nonclassic HLA class I antigens in uveal melanoma. Invest Ophthalmol Vis Sci. 2003;44:2016-9.

Antle CM, Damji KF, White VA, Rootman J. Uveal malignant melanoma and optic nerve glioma in von Recklinghausen's neurofibromatosis. Br J Ophthalmol. 1990;74:502-4.

Bartek J, Bartkova J, Lukas J. The retinoblastoma protein pathway in cell cycle control and cancer. Exp Cell Res. 1997;237:1-6.

Bataille V, Sasieni P, Cuzick J, Hungerford JL, Swerdlow A, Bishop JA. Risk of ocular melanoma in relation to cutaneous and iris naevi. Int J Cancer. 199560:622-6.

Bernard K, Litman E, Fitzpatrick JL, Shellman YG, Argast G, Polvinen K, Everett AD, Fukasawa K, Norris DA, Ahn NG, Resing KA. Functional proteomic analysis of melanoma progression. Cancer Res.

2003;63:6716-25.

Biswas J, Krishnakumar S, Shanmugam MP. Uveal melanoma in Asian Indians: a clinicopathological study. Arch Ophthalmol. 2002;120:522-3.

Bittner M, Meltzer P, Chen Y, Jiang Y, Seftor E, Hendrix M, Radmacher M, Simon R, Yakhini Z, Ben-Dor A, Sampas N, Dougherty E, Wang E, Marincola F, Gooden C, Lueders J, Glatfelter A, Pollock P, Carpten J, Gillanders E, Leja D, Dietrich K, Beaudry C, Berens M, Alberts D, Sondak V. Molecular classification of cutaneous malignant melanoma by gene expression profiling. Nature. 2000;406:536-40.

Blom DJ, Luyten GP, Mooy C, Kerkvliet S, Zwinderman AH, Jager MJ. Human leukocyte antigen class I expression. Marker of poor prognosis in uveal melanoma. Invest Ophthalmol Vis Sci. 1997;38:1865-72. Brantley MA Jr, Harbour JW. Deregulation of the Rb and p53 pathways in uveal melanoma. Am J Pathol.

2000(a);157:1795-801.

Brantley MA Jr, Harbour JW. Inactivation of retinoblastoma protein in uveal melanoma by phosphorylation of sites in the COOH-terminal region. Cancer Res. 2000(b);60:4320-3.

Chana JS, Wilson GD, Cree IA, Alexander RA, Myatt N, Neale M, Foss AJ, Hungerford JL. c-myc, p53, and Bcl-2 expression and clinical outcome in uveal melanoma. Br J Ophthalmol. 1999;83:110-4.

Cohen Y, Goldenberg-Cohen N, Parrella P, Chowers I, Merbs SL, Pe'er J, Sidransky D. Lack of BRAF mutation in primary uveal melanoma. Invest Ophthalmol Vis Sci. 2003;44:2876-8.

Cohen Y, Rosenbaum E, Begum S, Goldenberg D, Esche C, Lavie O, Sidransky D, Westra WH. Exon 15 BRAF mutations are uncommon in melanomas arising in nonsun-exposed sites. Clin Cancer Res. 2004;10:3444-3447.

Coleman K, Baak JP, Van Diest P, Mullaney J, Farrell M, Fenton M. Prognostic factors following enucleation of 111 uveal melanomas. Br J Ophthalmol. 1993;77:688-92.

Conway RM, Cursiefen C, Behrens J, Naumann GO, Holbach LM. Biomolecular markers of malignancy in human uveal melanoma: the role of the cadherin-catenin complex and gene expression profiling. Ophthalmologica. 2003;217:68-75.

Coupland SE, Anastassiou G, Stang A, Schilling H, Anagnostopoulos I, Bornfeld N, Stein H. The prognostic value of cyclin D1, p53, and MDM2 protein expression in uveal melanoma. J Pathol. 2000;191:120-6. Damato BE. Local resection of uveal melanoma.Bull Soc Belge Ophtalmol. 1993;248:11-7.

Shields CL, Shields JA, Gunduz K, Freire JE, Mercado G. Radiation therapy for uveal malignant melanoma. Ophthalmic Surg Lasers. 1998;29:397-409.

Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C, Shipley J, Hargrave D, Pritchard-Jones K, Maitland N, Chenevix-Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho JW, Leung SY, Yuen ST, Weber BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ, Wooster R, Stratton MR, Futreal PA. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949-54.

De Waard-Siebinga I, Hilders CG, Hansen BE, van Delft JL, Jager MJ. HLA expression and tumor-infiltrating immune cells in uveal melanoma. Graefes Arch Clin Exp Ophthalmol. 1996;234:34-42.

(14)

Dolin PJ, Foss AJ, Hungerford JL. Uveal melanoma: is solar ultraviolet radiation a risk factor? Ophthalmic Epidemiol. 1994;1:27-30.

Donoso LA, Shields JA, Augsburger JJ, Orth DH, Johnson P. Metastatic uveal melanoma: diffuse hepatic metastasis in a patient with concurrent normal serum liver enzyme levels and liver scan. Arch Ophthalmol. 1985;103:758.

Easton DF, Steele L, Fields P, Ormiston W, Averill D, Daly PA, McManus R, Neuhausen SL, Ford D, Wooster R, Cannon-Albright LA, Stratton MR, Goldgar DE. Cancer risks in two large breast cancer families linked to BRCA2 on chromosome 13q12-13. Am J Hum Genet. 1997;61:120-8.

Egan KM, Seddon JM, Glynn R, Gragoudas ES, Albert DM. epidemiologic aspects of uveal melanoma. Surv Ophthalmol 1988;32:239-251.

Elshaw SR, Sisley K, Cross N, Murray AK, MacNeil SM, Wagner M, Nichols CE, Rennie IG. A comparison of ocular melanocyte and uveal melanoma cell invasion and the implication of alpha1beta1, alpha4beta1 and alpha6beta1 integrins. Br J Ophthalmol. 2001;85:732-8.

Elwood JM, Jopson J. Melanoma and sun exposure: an overview of published studies. Int J Cancer. 1997;73:198-203.

Fenn JB, Mann M, Meng CK. Electrospray ionization for mass spectrometry of large biomolecules. Science 1989; 246: 64-71

Florenes VA, Oyjord T, Holm R, Skrede M, Borresen AL, Nesland JM, Fodstad O. TP53 allele loss, mutations and expression in malignant melanoma. Br J Cancer. 1994;69:253-9.

Folberg R, Rummelt V, Parys-Van Ginderdeuren R, Hwang T, Woolson RF, Pe'er J, Gruman LM. The prognostic value of tumor blood vessel morphology in primary uveal melanoma. Ophthalmology. 1993;100:1389-98.

Friend SH, Bernards R, Rogelj S, Weinberg RA, Rapaport JM, Albert DM, Dryja TP. A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma. Nature. 1986;323:643-6. Gallagher RP, Elwood JM, Rootman J, Spinelli JJ, Hill GB, Threlfall WJ, Birdsell JM. Risk factors for ocular

melanoma: Western Canada Melanoma Study. J Natl Cancer Inst. 1985;74:775-8.

Ghazvini S, Char DH, Kroll S, Waldman FM, Pinkel D. Comparative genomic hybridization analysis of archival formalin-fixed paraffin-embedded uveal melanomas. Cancer Genet Cytogenet. 1996;90:95-101.

Gilchrest BA, Eller MS, Geller AC, Yaar M. The pathogenesis of melanoma induced by ultraviolet radiation. N Engl J Med. 1999;340:1341-8.

Goldstein AM, Dracopoli NC, Engelstein M, Fraser MC, Clark WH Jr, Tucker MA. Linkage of cutaneous malignant melanoma/dysplastic nevi to chromosome 9p, and evidence for genetic heterogeneity. Am J Hum Genet. 1994;54:489-96.

Gonder JR, Shields JA, Albert DM, Augsburger JJ, Lavin PT. Uveal malignant melanoma associated with ocular and oculodermal melanocytosis. Ophthalmology. 1982;89:953-60.

Gruis NA, van der Velden PA, Sandkuijl LA, Prins DE, Weaver-Feldhaus J, Kamb A, Bergman W, Frants RR. Homozygotes for CDKN2 (p16) germline mutation in Dutch familial melanoma kindreds. Nat Genet. 1995;10:351-3.

Guldberg P, thor Straten P, Birck A, Ahrenkiel V, Kirkin AF, Zeuthen J. Disruption of the MMAC1/PTEN gene by deletion or mutation is a frequent event in malignant melanoma. Cancer Res. 1997;57:3660-3. Haab BB, Dunham MJ, Brown PO. Protein microarrays for highly parallel detection and quantitation of specific

proteins and antibodies in complex solutions. Genome Biol. 2001;2:research0004.1-0004.13. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57-70.

Harbour JW, Lai SL, Whang-Peng J, Gazdar AF, Minna JD, Kaye FJ. Abnormalities in structure and expression of the human retinoblastoma gene in SCLC. Science. 1988;241:353-7.

Harbour JW, Worley L, Ma D, Cohen M. Transducible peptide therapy for uveal melanoma and retinoblastoma. Arch Ophthalmol. 2002;120:1341-6.

Harris CC, Hollstein M. Clinical implications of the p53 tumor-suppressor gene. N Engl J Med. 1993;329:1318-27.

Haupt Y, Maya R, Kazaz A, Oren M. Mdm2 promotes the rapid degradation of p53. Nature. 1997;387:296-9. Holly EA, Aston DA, Char DH, Kristiansen JJ, Ahn DK. Uveal melanoma in relation to ultraviolet light

(15)

Hurks HM, Metzelaar-Blok JA, Barthen ER, Zwinderman AH, De Wolff-Rouendaal D, Keunen JE, Jager MJ. Expression of epidermal growth factor receptor: risk factor in uveal melanoma. Invest Ophthalmol Vis Sci. 2000;41:2023-7.

Hurks HM, Valter MM, Wilson L, Hilgert I, van den Elsen PJ, Jager MJ. Uveal melanoma: no expression of HLA-G. Invest Ophthalmol Vis Sci. 2001;42:3081-4.

Jager MJ, Hurks HM, Levitskaya J, Kiessling R. HLA expression in uveal melanoma: there is no rule without some exception. Hum Immunol. 2002;63:444-51.

James P. Protein identification in the post-genome-era: the rapid rise of proteomics. Q Rev Biophys. 1997;30:279-331.

Jensen OA. Malignant melanomas of the human uvea: 25-year follow-up of cases in Denmark, 1943-1952. Acta Ophthalmol. 1982;60:161-182.

Journee-de Korver JG, Oosterhuis JA, de Wolff-Rouendaal D. Histopathological findings in human choroidal melanomas after transpupillary thermotherapy. Br J Ophthalmol. 1997;81:234-239.

Karas M, Hillenkamp F. Laser desorption ionization of proteins with molecular masses exceeding 10,000 daltons. Anal Chem. 1988;60:2299-301.

Kath R, Hayungs J, Bornfeld N, Sauerwein W, Höffken K, Seeber S. Prognosis and treatment of disseminated uveal melanoma. Cancer. 1993;72:2219-2223.

Kishore K, Ghazvini S, Char DH, Kroll S, Selle J. p53 gene and cell cycling in uveal melanoma. Am J Ophthalmol. 1996;121:561-7.

Kononen J, Bubendorf L, Kallioniemi A, Barlund M, Schraml P, Leighton S, Torhorst J, Milhatsch MJ, Sauter G, Kallioniemi OP. Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med. 1998;4:844–847.

Kuo PK, Puliafito CA, Wang KM, Liu HS, Wu BF. Uveal melanoma in China. Int Ophthalmol Clin. 1982;22:57-71.

Li W, Judge H, Gragoudas ES, Seddon JM, Egan KM. Patterns of tumor initiation in choroidal melanoma. Cancer Res. 2000;60:3757-60.

Lockhart DJ, Dong H, Byrne MC, Follettie MT, Gallo MV, Chee MS, Mittmann M, Wang C, Kobayashi M, Horton H, Brown EL. Expression monitoring by hybridization to high-density oligonucleotide arrays. Nat. Biotechnol. 1996;14:1675-1680.

Lommatzsch PK, Correns HJ, Rudolph JM. The reliability of radioactive phosphorus P-32 in the diagnosis of intraocular tumors, experience with 912 patients. Doc Ophthalmol. 1984;56:353-361.

Lommatzsch PK, Kirsch IH. 106Ru/106Rh plaque radiotherapy for malignant melanomas of the choroid. With follow-up results more than 5 years. Doc Ophthalmol. 1988;68:225-38.

Lommatzsch PK, Werschnik C, Schuster E. Long-term follow-up of Ru-106/Rh-106 brachytherapy for posterior uveal melanoma. Graefes Arch Clin Exp Ophthalmol. 2000;238:129-37.

Ma D, Niederkorn JY. Role of epidermal growth factor receptor in the metastasis of intraocular melanomas. Invest Ophthalmol Vis Sci. 1998;39:1067-75.

Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LM, Pe'er J, Trent JM, Meltzer PS, Hendrix MJ. Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol. 1999;155:739-752.

McLean IW. The biology of haematogenous metastasis in human uveal malignant melanoma. Vir Arch Path Anat. 1993;422:433-437.

Merbs SL, Sidransky D. Analysis of p16 (CDKN2/MTS-1/INK4A) alterations in primary sporadic uveal melanoma. Invest Ophthalmol Vis Sci. 1999;40:779-83.

Missotten GS, Beijnen JH, Keunen JE, Bonfrer JM. Proteomics in uveal melanoma. Melanoma Res. 2003;13:627-9.

Mooy CM, de Jong PT. Prognostic parameters in uveal melanoma: a review. Surv Ophthalmol. 1996;41:215-228.

Naus NC, Zuidervaart W, Rayman N, Slater R, van Drunen E, Ksander B, Luyten GP, Klein A. Mutation analysis of the PTEN gene in uveal melanoma cell lines. Int J Cancer 2000;87:151-153.

Omholt K, Platz A, Kanter L, Ringborg U, Hansson J. NRAS and BRAF mutations arise early during melanoma pathogenesis and are preserved throughout tumor progression. Clin Cancer Res. 2003;9:6483-8.

(16)

Oosterhuis JA, Journee-de Korver HG, Kakebeeke-Kemme HM, Bleeker JC. Transpupillary thermotherapy in choroidal melanomas. Arch Ophthalmol. 1995;113:315-21.

Ostergaard M, Rasmussen HH, Nielsen HV, Vorum H, Orntoft TF, Wolf H, Celis JE. Proteome profiling of bladder squamous cell carcinomas: identification of markers that define their degree of differentiation. Cancer Res. 1997;57:4111-7.

Overkleeft ENM, Zuidervaart W, Hurks HMH, Eilers PHC, Wolff de-Rouendaal D, Keunen JEE, Jager MJ. The prognostic value of the 32P-uptake test in uveal melanoma: a long-term study. Arch Ophthalmol. 2003;121:1398-403.

Parrella P, Caballero OL, Sidransky D, Merbs SL. Detection of c-myc amplification in uveal melanoma by fluorescent in situ hybridization. Invest Ophthalmol Vis Sci. 2001;42:1679-84.

Pollock PM, Harper UL, Hansen KS, Yudt LM, Stark M, Robbins CM, Moses TY, Hostetter G, Wagner U, Kakareka J, Salem G, Pohida T, Heenan P, Duray P, Kallioniemi O, Hayward NK, Trent JM, Meltzer PS. High frequency of BRAF mutations in nevi. Nat Genet. 2003;33:19-20.

Prescher G, Bornfeld N, Hirche H, Horsthemke B, Jockel KH, Becher R. Prognostic implications of monosomy 3 in uveal melanoma. Lancet. 1996;347:1222-1225.

Royds JA, Sharrard RM, Parsons MA, Lawry J, Rees R, Cottam D, Wagner B, Rennie IG. C-myc oncogene expression in ocular melanomas. Graefes Arch Clin Exp Ophthalmol. 1992;230:366-71.

Satyamoorthy K, Li G, Gerrero MR, Brose MS, Volpe P, Weber BL, Van Belle P, Elder DE, Herlyn M. Constitutive mitogen-activated protein kinase activation in melanoma is mediated by both BRAF mutations and autocrine growth factor stimulation. Cancer Res. 2003;63:756-9.

Scholes AG, Hagan S, Hiscott P, Damato BE, Grierson I. Overexpression of epidermal growth factor receptor restricted to macrophages in uveal melanoma. Arch Ophthalmol. 2001;119:373-7.

Schwartz LH, Ferrand R, Boelle PY, Maylin C, D'Hermies F, Virmont J. Lack of correlation between the location of choroidal melanoma and ultraviolet-radiation dose distribution. Radiat Res. 1997;147:451-6. Schweitzer B. Multiplexed protein profiling on microarrays by rolling-circle amplification. Nat Biotechnol

2002;20:359-365.

Scott RJ, Vajdic CM, Armstrong BK, Ainsworth CJ, Meldrum CJ, Aitken JF, Kricker A. BRCA2 mutations in a population-based series of patients with ocular melanoma. Int J Cancer. 2002;102:188-91.

Seddon JM, Albert DM, Lavin PT, Robinson N. A prognostic factor study of disease-free interval and survival following enucleation for uveal melanoma. Arch Ophthalmol. 1983;101:1894-1899.

Seftor EA, Meltzer PS, Kirschmann DA, Pe'er J, Maniotis AJ, Trent JM, Folberg R, Hendrix MJ. Molecular determinants of human uveal melanoma invasion and metastasis. Clin Exp Metastasis. 2002;19:233-46. Seregard S, Kock E. Prognostic indicators following enucleation for posterior uveal melanoma. Acta Ophthalmol

Scand. 1995;73:340-344.

Seregard S, Landau I. Transpupillary thermothrerapy as an adjunct to ruthenium plaque radiotherapy for choroidal melanoma. Acta Ophthalmol Scand. 2001;79:19-22.

Sherr CJ, McCormick F. The RB and p53 pathways in cancer. Cancer Cell. 2002;2:103-12.

Shields JA. Accuracy and limitations of the 32P test in the diagnosis of ocular tumors: an analysis of 500 cases. Ophthalmology. 1978;85:950-966.

Shields JA, Shields CL, Donoso LA. Management of posterior uveal melanoma. Major review. Surv Ophthalmol. 1991;36:161-195.

Shields JA, Shields CL. Current management of posterior uveal melanoma. Mayo Clin Proc. 1993;68:1196-200. Sinilnikova OM, Egan KM, Quinn JL, Boutrand L, Lenoir GM, Stoppa-Lyonnet D, Desjardins L, Levy C,

Goldgar D, Gragoudas ES. Germline brca2 sequence variants in patients with ocular melanoma. Int J Cancer. 1999;82:325-8.

Singh AD, Shields CL, De Potter P, Shields JA, Trock B, Cater J, Pastore D. Familial uveal melanoma. Clinical observations on 56 patients. Arch Ophthalmol. 1996;114:392-9.

Singh AD, De Potter P, Fijal BA, Shields CL, Shields JA, Elston RC. Lifetime prevalence of uveal melanoma in white patients with oculo(dermal) melanocytosis. Ophthalmology. 1998;105:195-8.

Singh AD, Topham A. Incidence of Uveal Melanoma in the United States: 1973-1997. Ophthalmology. 2003;110:956-961.

(17)

Sisley K, Rennie IG, Parsons MA, Jacques R, Hammond DW, Bell SM, Potter AM, Rees RC. Abnormalities of chromosomes 3 and 8 in posterior uveal melanoma correlate with prognosis. Genes Chromosomes Cancer. 1997;19:22-28.

Specht CS, Smith TW. Uveal malignant melanoma and von Recklinghausen's neurofibromatosis. Cancer. 1988;62:812-7.

Speicher MR, Prescher G, du Manoir S, Jauch A, Horsthemke B, Bornfeld N, Becher R, Cremer T. Chromosomal gains and losses in uveal melanomas detected by comparative genomic hybridization. Cancer Res. 1994;54:3817-23.

Stahl JM, Cheung M, Sharma A, Trivedi NR, Shanmugam S, Robertson GP. Loss of PTEN promotes tumor development in malignant melanoma. Cancer Res. 2003;63:2881-90.

Tamura M, Gu J, Matsumoto K, Aota S, Parsons R, Yamada KM. Inhibition of cell migration, spreading, and focal adhesions by tumor suppressor PTEN. Science. 1998;280:1614-7.

Thomas CI, Krohmer JS, Storasli JP. Detection of intraocular tumors with radioactive phosphorus. Arch Ophthalmol. 1952;47:276-286.

Tsao H, Goel V, Wu H, Yang G, Haluska FG. Genetic interaction between NRAS and BRAF mutations and PTEN/MMAC1 inactivation in melanoma. J Invest Dermatol. 2004;122:337-41.

Tschentscher F, Hüsing J, Hölter T, Kruse E, Dresen IG, Jöckel KH, Anastassiou G, Schilling H, Bornfeld N, Horsthemke B, Lohnmann DR, Zeschnigk M. Tumor classification based on gene expression profiling shows that uveal melanomas with and without monosomy 3 represent two distinct entities. Cancer Res. 2003;63:2578-84.

Tucker MA, Shields JA, Hartge P, Augsburger J, Hoover RN, Fraumeni JF Jr. Sunlight exposure as risk factor for intraocular malignant melanoma. N Engl J Med. 1985;313:789-92.

Van der Velden PA, Metzelaar-Blok JA, Bergman W, Monique H, Hurks H, Frants RR, Gruis NA, Jager MJ. Promoter hypermethylation: a common cause of reduced p16(INK4a) expression in uveal melanoma. Cancer Res. 2001;61:5303-5306.

Van der Velden PA, Zuidervaart W, Hurks MH, Pavey S, Ksander BR, Krijgsman E, Frants RR, Tensen CP, Willemze R, Jager MJ, Gruis NA. Expression profiling reveals that methylation of TIMP3 is involved in uveal melanoma development. Int J Cancer. 2003;106:472-9.

Van Elsas A, Zerp SF, van der Flier S, Kruse KM, Aarnoudse C, Hayward NK, Ruiter DJ, Schrier PI. Relevance of ultraviolet-induced N-ras oncogene point mutations in development of primary human cutaneous melanoma. Am J Pathol. 1996;149:883-93.

Van Hees CL, Jager MJ, Bleeker JC, Kemme H, Bergman W. Occurrence of cutaneous and uveal melanoma in patients with uveal melanoma and their first degree relatives. Melanoma Res. 1998;8:175-80.

Vazquez F, Sellers WR. The PTEN tumor suppressor protein: an antagonist of phosphoinositide 3-kinase signaling. Biochim Biophys Acta. 2000;1470:M21-35.

Woodward JK, Nichols CE, Rennie IG, Parsons MA, Murray AK, Sisley K. An in vitro assay to assess uveal melanoma invasion across endothelial and basement membrane barriers. Invest Ophthalmol Vis Sci. 2002;43:1708-14.

Yanoff M, Fine BS. Ocular pathology, a text and atlas, Third ed. J.B. Lippincott Company, Philadelphia, 1989. Yu LR, Zeng R, Shao XX, Wang N, Xu YH, Xia QC. Identification of differentially expressed proteins between

human hepatoma and normal liver cell lines by two-dimensional electrophoresis and liquid chromatography-ion trap mass spectrometry. Electrophoresis. 2000;21:3058-68.

Zehetmayer M, Kitz K, Menapace R, Ertl A, Heinzl H, Ruhswurm I, Georgopoulos M, Dieckmann K, Potter R. Local tumor control and morbidity after one to three fractions of stereotactic external beam irradiation for uveal melanoma. Radiother Oncol. 2000;55:135-44.

Zimmerman LE. Malignant melanoma. In: Spencer WH, ed. Ophthalmic Pathology. Philadelphia: WB Saunders Company, 1986;2072-2139.

Referenties

GERELATEERDE DOCUMENTEN

The molecular data includes mRNA and miRNA expression levels (Illumina HiSeq data), protein levels (RPPA data), SCNA (derived from segmented Affymetrix SNP data as well as

Het onderzoek van dit proefschrift is gefinancierd door de Mozaiekbeurs (017.003.059) van De Nederlandse Organisatie voor Wetenschappelijk Onderzoek (NWO), Landelijke Stichting

CD11b+ macrophages have been attributed a role in T cell regulation, and McKenna and Kapp 59 showed that they are also involved in immune regulation against intraocular tumors

Endothelin 2 (ET2), Laminin Receptor 1 (LAMR1), Von Hippel Lindau Binding Protein 1 (VBP1) and Cullin 2 (CUL2) belong to the group of genes exhibiting the most discriminating

In order to identify proteins involved in metastasis development, we compared protein expression in a cell line obtained from a primary uveal melanoma with two cell lines

By means of this technique, we compared gene expression profiles in different uveal melanoma cell lines and identified potential genes, such as TIMP3, Endothelin 2 (ET2) and

In Chapter 3, we performed mutation analysis and expression analysis in uveal melanoma cell lines and tumor samples on members of the MAPK pathway.. Although we demonstrated

Op basis van genexpressieprofielen kunnen primaire uveamelanomen in twee clusters worden verdeeld, gebaseerd op genen die verlaagd tot expressie komen op chromosoom 3 en genen die