663
Views
2
CrossRef citations to date
0
Altmetric
Editorial

Retinoblastoma beyond RB1: recent advances in genetic biomarkers

, &
Pages 717-721 | Published online: 09 Jan 2014

Retinoblastoma is an eye cancer that accounts for approximately 4% of all pediatric malignancies and is the most common malignant ocular tumor in children Citation[1]. The disease can be unilateral (when it affects only one eye, as seen in two-thirds of patients) or bilateral (when it affects both eyes). Although most cases of retinoblastoma do not show family history, the importance of hereditary factors for this cancer is well established Citation[2]. The familial disease is transmitted as a typical Mendelian autosomal dominant trait, with high penetrance. The heritable form affects all patients with bilateral retinoblastoma and approximately 15% of patients with the unilateral form, is generally multifocal and is associated with increased risk of second primary cancers. Approximately 40% of retinoblastoma cases are heritable, meaning that patients are at risk of transmitting the disease to their offspring, although most heritable cases arise as sporadic disease. Nonheritable retinoblastoma, which occurs in approximately 60% of cases, is always unilateral, unifocal and is not associated with increased risk of other cancers.

Clinical studies on retinoblastoma led to the identification of the first tumor-suppressor gene – retinoblastoma (RB1), located at 13q14 – and the development of the ‘two-hit’ model Citation[3]. According to Knudson, in fact, two mutational events (M1 and M2) or ‘hits’ are required for tumor onset. Thus, an individual with a germline mutation – M1 – in a tumor-suppressor gene (inherited or occurring de novo) is predisposed to cancer because only another somatic mutation in the same gene – M2 – will be enough to knock out gene function and trigger tumor development Citation[3].

Indeed, biallelic mutations of RB1 have long been recognized as the causative genetic alteration underlying retinoblastoma Citation[4–8]. RB1 functions as the gatekeeper of the G1–S transition of the cell cycle by binding to members of the E2F family of transcription factors and repressing genes that are required for cell proliferation. Consistent with its role as tumor suppressor, mutations in the RB1 pathway are found in most human cancers. However, in the past decades, a myriad of studies focusing on the role of RB1 in cancer development implicated RB1 in many other cellular processes, which could all contribute to its tumor-suppressor function, suggesting that the role of RB1 in cancer is much more complex than previously thought. Moreover, the other members of the RB family, retinoblastoma-like 1 (RBL1 or p107) and retinoblastoma-like 2 (RBL2 or p130), both have overlapping and distinct functions compared with RB1, making it more difficult to dissect specific gene functions.

Thus, despite over two decades having passed since RB1 loss was found to be a crucial event for retinoblastoma development, many questions remain open. For example, why is the high susceptibility of individuals bearing RB1 mutations to specifically develop retinoblastoma not recapitulated in mouse models? In mice, retinal tumors only develop when RB1 is lost together with at least one other member of the RB family Citation[9,10], suggesting that, at least in mice, other family members can compensate for RB1 loss. What makes the human retina so extraordinarily susceptible to RB1 loss? Retinal development is a complex process that leads to the generation of at least seven different specialized cell types. Understanding what is – among all these cell types – the origin of retinoblastoma, could explain how the cellular context affects the requirement of specific mutations for cancer initiation and progression Citation[11–13].

Although RB1 mutations are crucial for retinoblastoma development, some authors argued that the two-hit model oversimplifies the disease process, and suggest that other molecular mechanisms of tumorigenesis must be involved Citation[14,15]. Beyond disputes, the importance of mutational events other than RB1 mutations is almost universally recognized. Many cytogenetic and comparative genomic hybridization (CGH) studies, which have been comprehensively reviewed by Gallie et al., have shown that mutational events affecting RB1 are not the only genomic changes in retinoblastoma Citation[16]. The same authors recently showed that, in humans, loss of both copies of RB1 does not lead directly to retinoblastoma, but to the benign precursor lesion retinoma, which commonly progresses to retinoblastoma after further mutational events – defined M3 to Mn in keeping with Knudson nomenclature Citation[17]. Further studies seem to support this model Citation[18,19].

A detailed understanding of the sequence of molecular events that underlie retinoblastoma pathogenesis is crucial to identify new possible therapeutic targets and markers for early diagnosis and prevention. In fact, although in developed countries the survival rate among children with retinoblastoma is as high as 95% or more, children who survive this disease suffer serious morbidity. Most children with unilateral disease are treated by enucleation, while those with bilateral disease are treated with globe salvage therapies, which are associated with serious toxicities Citation[20]. Furthermore, in less economically developed countries the survival rates are dramatically worse, and retinoblastoma still poses a serious life-threatening problem for children, mostly owing to late diagnosis. A tumor spreading beyond the confines of the eye is still impossible to cure worldwide Citation[21,22]. Therefore, there is currently a great need for the identification of new molecular markers that could serve to open novel therapeutic avenues for more advanced disease, or that could minimize the adverse effects of the current strategies.

Alterations of many other genes beyond RB1 have been found in retinoblastoma. However, for most of these it remains to be established whether they are drivers or passenger mutations. This will be necessary to understand whether such genes have a causal role in tumor development or progression and thus if they can be exploited as cancer-predictive/prognostic biomarkers or, ideally, as therapeutic targets. Moreover, RB1 deficiency itself induces genomic instability, not only by causing defects in the DNA replication process but also by causing chromosome gains, losses and breakage during mitosis Citation[23–27]. Therefore, it needs to be determined whether mutations in other genes are necessary for tumor progression or, rather, are a by-product of cancer cell development.

Several studies implicate RBL2 as one of the genes involved in retinoblastoma development. These are consistent with data from mouse models, as mentioned before, in which at least another RB family member has to be ablated along with RB1 to develop retinoblastoma Citation[28]. We reported that loss of RBL2 expression in sporadic retinoblastomas correlates with a minor degree of cellular differentiation and a lower apoptotic index Citation[29]. Moreover, we identified sequence variants in exon 1 of the RBL2 gene that seem to predispose to promoter hypermethylation and a decreased protein expression in retinoblastomas Citation[30]. We also showed that the treatment of the retinoblastoma cell line Weri-RB1 with the demethylating agent 5-aza-2´-deoxycytidine induced an increase in the expression levels of RBL2 and a significant growth rate reduction. Recently, another study showed that RBL2 is also inactivated by loss of heterozygosity in a high percentage of retinoblastomas Citation[31]. As for the role of RBL2 in retinal tumorigenesis, Dimaras and colleagues suggest that it might be involved in the progression from retinoma to retinoblastoma Citation[17].

Other genes affecting the RB pathway seem to be relevant for retinoblastoma development. For example, p16INK4A – the cell cycle inhibitor that indirectly regulates RB phosphorylation – was proposed to block the transformation of RB1-/- retina cells at the stage of the nonproliferative precursor lesion retinoma, whereas its inactivation seemed to promote retinoblastoma development Citation[17]. We consistently found a significant downregulation of p16INK4A in approximately half of the tumor and blood samples from the analyzed retinoblastoma patients, and in most parents of the patients showing p16INK4A deregulation. Therefore, p16INK4A alterations might be novel inheritable susceptibility markers to retinoblastoma Citation[32].

Among other cell cycle-related genes, the mitotic kinesin KIF14, which is essential for proper spindle formation and cytokinesis, is often found to be overexpressed in retinoblastomas compared with normal retina, owing in part to gene copy number alteration Citation[33–35]. An increase in KIF14 copy number and mRNA expression was also found in retinomas, although its expression was much higher in the adjacent retinoblastoma tissue, suggesting that it might play a part in tumor progression Citation[17]. Despite no significant correlation being observed between KIF14 expression and laterality, invasion, differentiation status and duration of the disease, an older median age at diagnosis was significantly correlated with a higher KIF14 expression, suggesting that different mutational pathways probably underlie early- or late-onset tumors Citation[34]. Interestingly, KIF14 is also overexpressed in breast tumors and non-small-cell lung cancer, which correlates with a poor prognosis, and in medulloblastoma cell lines Citation[16,33,36].

Madhavan and colleagues also described, in association with KIF14 overexpression, the upregulation of the RB-regulated transcription factor E2F3Citation[35]. E2F3 maps on the 6p22 locus, and 6p gains are among the most common changes identified by CGH studies in retinoblastoma Citation[16]. On the same locus lies the DNA-binding protein DEK, which exerts a pleiotropic role in cancer, affecting both chromatin remodeling and RNA processing Citation[37]. Thus, E2F3 and DEK are both candidate oncogenes for retinoblastoma. Moreover, E2F3 has recently been shown to regulate DEK transcription, and Gallie and colleagues suggest that they might cooperate when both are upregulated in retinoblastoma Citation[16].

MYCN amplification is another feature of retinoblastomas, both in humans and in mouse models. MYCN is a member of a small family of transcription factors (MYC, MYCL and MYCN), which regulate many cellular processes, including cell growth and proliferation, differentiation and apoptosis. MYCN expression is normally restricted to the central and peripheral nervous systems, kidney, lung and spleen during embryonic development Citation[38], whereas MYCN amplification occurs in a variety of tumors, most notably in neuroblastoma Citation[39]. In retinoblastoma, MYCN amplification is associated with a high proliferation index Citation[40] and seems to correlate with distant metastases Citation[41,42], suggesting a role in tumor progression. Interestingly, in neuroblastoma, MYCN has shown a paradoxical function because it is able to induce cell proliferation and also to sensitize cells to apoptosis Citation[43]. This apparent inconsistency for a tumor as aggressive and therapy resistant as neuroblastoma might be explained by the high levels of the inhibitor of apoptosis, survivin. High levels of survivin have in fact been associated with MYCN amplification and a poor prognosis Citation[43]. Remarkably, retinoblastomas also show high levels of survivin, which, on this basis, warrants further investigation and might represent a useful therapeutic target Citation[44].

Considering the invasive nature of retinoblastoma, genes involved in extracellular matrix degradation and cell adhesion must also be relevant to tumor development and progression. Two studies have addressed this issue thus far Citation[45,46]. Most notably, the gene encoding CDH11, a type II classical cadherin from the cadherin superfamily that mediates cell–cell adhesion, is frequently lost in both human retinoblastomas and in retinoblastomas arising in mouse models Citation[47,48]. Recent data show that CDH11 exerts tumor-suppressor functions in retinoblastoma through the promotion of cell death Citation[49].

Similarly to the RB pathway, the p53 pathway is deregulated in most human tumors. In retinoblastoma, despite an intact p53 molecule, the p53 pathway is altered by amplification of p53-negative regulators such as MDM4 (also known as MDMX; amplified in 65% of retinoblastomas) and MDM2 (amplified in 10% of retinoblastomas) Citation[50]. Interestingly, targeting the p53–MDM2/4 interaction with the small molecule nutlin-3 has proven effective against retinoblastoma both in vitro and in vivoCitation[50,51]. Other studies are investigating the potential role of other p53 family members in retinoblastoma Citation[52].

Recently, epigenetic modifications have also been increasingly recognized as one of the most common molecular alterations in cancer, associated both with the inactivation of tumor-suppressor genes and chromosomal instability Citation[53]. Several aberrantly methylated genes have also been identified in retinoblastoma tumors, including RB1 itself Citation[54], RBL2Citation[32], CASP8 and RASSF1ACitation[55–57], MGMT and MLH1Citation[57,58], and NEUROG1Citation[59]. Because these alterations might have an important role in retinoblastoma progression, the potential anticancer properties of epigenetic drugs should be further investigated Citation[60].

The list of other candidate genes, beyond RB1, which are crucial to retinoblastoma development and progression, is rapidly growing owing to powerful new techniques and high-throughput screenings. Although for many of these genes the functional role in retinal tumorigenesis still needs to be clarified, it is likely that some will serve to identify new possible therapeutic strategies. Furthermore, the recently developed animal models of retinoblastoma will be an invaluable tool to analyze novel therapeutic strategies at the preclinical level, and to help move the clinical translation of these findings forward.

Financial & competing interests disclosure

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

References

  • ShieldsCL. Forget-me-nots in the care of children with retinoblastoma. Semin. Ophthalmol.23(5), 324–334 (2008).
  • Vogel F. Genetics of retinoblastoma. Hum. Genet.52(1), 1–54 (1979).
  • Knudson AG Jr. Mutation and cancer: statistical study of retinoblastoma. Proc. Natl Acad. Sci. USA68(4), 820–823 (1971).
  • Cavenee WK, Dryja TP, Phillips RA et al. Expression of recessive alleles by chromosomal mechanisms in retinoblastoma. Nature305(5937), 779–784 (1983).
  • Friend SH, Bernards R, Rogelj S et al. A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma. Nature323(6089), 643–646 (1986).
  • Fung YK, Murphree AL, T’Ang A, Qian J, Hinrichs SH, Benedict WF. Structural evidence for the authenticity of the human retinoblastoma gene. Science236(4809), 1657–1661 (1987).
  • Lee WH, Bookstein R, Hong F, Young LJ, Shew JY, Lee EY. Human retinoblastoma susceptibility gene: cloning, identification, and sequence. Science235(4794), 1394–1399 (1987).
  • Dunn JM, Phillips RA, Becker AJ, Gallie BL. Identification of germline and somatic mutations affecting the retinoblastoma gene. Science241(4874), 1797–1800 (1988).
  • Robanus-Maandag E, Dekker M, van der Valk M et al. p107 is a suppressor of retinoblastoma development in pRB-deficient mice. Genes Dev.12(11), 1599–1609 (1998).
  • MacPherson D, Sage J, Kim T, Ho D, McLaughlin ME, Jacks T. Cell type-specific effects of RB deletion in the murine retina. Genes Dev.18(14), 1681–1694 (2004).
  • Dyer MA, Bremner R. The search for the retinoblastoma cell of origin. Nat. Rev. Cancer5(2), 91–101 (2005).
  • Ajioka I, Martins RA, Bayazitov IT et al. Differentiated horizontal interneurons clonally expand to form metastatic retinoblastoma in mice. Cell131(2), 378–390 (2007).
  • Ajioka I, Dyer MA. A new model of tumor susceptibility following tumor suppressor gene inactivation. Cell Cycle7(6), 735–740 (2008).
  • Mastrangelo D, De Francesco S, Di Leonardo A, Lentini L, Hadjistilianou T. Does the evidence matter in medicine? The retinoblastoma paradigm. Int. J. Cancer121(11), 2501–2505 (2007).
  • Prigoda-Lee NL, Rushlow D, Piovesan B et al. Retinoblastoma: the evidence does matter. Cell. Oncol.31(3), 249–250 (2009).
  • Corson TW, Gallie BL. One hit, two hits, three hits, more? Genomic changes in the development of retinoblastoma. Genes Chromosomes Cancer46(7), 617–634 (2007).
  • Dimaras H, Khetan V, Halliday W et al. Loss of RB1 induces non-proliferative retinoma: increasing genomic instability correlates with progression to retinoblastoma. Hum. Mol. Genet.17(10), 1363–1372 (2008).
  • Sampieri K, Mencarelli MA, Epistolato MC et al. Genomic differences between retinoma and retinoblastoma. Acta Oncol.47(8), 1483–1492 (2008).
  • Sampieri K, Amenduni M, Papa FT et al. Array comparative genomic hybridization in retinoma and retinoblastoma tissues. Cancer Sci.100(3), 465–471 (2009).
  • Shields CL, Shields JA. Retinoblastoma management: advances in enucleation, intravenous chemoreduction, and intra-arterial chemotherapy. Curr. Opin. Ophthalmol.21(3), 203–212 (2010).
  • Dimaras H, Dimba EA, Gallie BL. Challenging the global retinoblastoma survival disparity through a collaborative research effort. Br. J. Ophthalmol.94(11), 1415–1416 (2010).
  • Kivela T. The epidemiological challenge of the most frequent eye cancer: retinoblastoma, an issue of birth and death. Br. J. Ophthalmol.93(9), 1129–1131 (2009).
  • Burkhart DL, Sage J. Cellular mechanisms of tumour suppression by the retinoblastoma gene. Nat. Rev. Cancer8(9), 671–682 (2008).
  • Hernando E, Nahle Z, Juan G et al. RB inactivation promotes genomic instability by uncoupling cell cycle progression from mitotic control. Nature430(7001), 797–802 (2004).
  • Coschi CH, Martens AL, Ritchie K et al. Mitotic chromosome condensation mediated by the retinoblastoma protein is tumor-suppressive. Genes Dev.24(13), 1351–1363 (2010).
  • Manning AL, Longworth MS, Dyson NJ. Loss of pRB causes centromere dysfunction and chromosomal instability. Genes Dev.24(13), 1364–1376 (2010).
  • van Harn T, Foijer F, van Vugt M et al. Loss of RB proteins causes genomic instability in the absence of mitogenic signaling. Genes Dev.24(13), 1377–1388 (2010).
  • MacPherson D, Dyer MA. Retinoblastoma: from the two-hit hypothesis to targeted chemotherapy. Cancer Res.67(16), 7547–7550 (2007).
  • Bellan C, De Falco G, Tosi GM et al. Missing expression of pRB2/p130 in human retinoblastomas is associated with reduced apoptosis and lesser differentiation. Invest. Ophthalmol. Vis. Sci.43(12), 3602–3608 (2002).
  • Tosi GM, Trimarchi C, Macaluso M et al. Genetic and epigenetic alterations of RB2/p130 tumor suppressor gene in human sporadic retinoblastoma: implications for pathogenesis and therapeutic approach. Oncogene24(38), 5827–5836 (2005).
  • Priya K, Jada SR, Quah BL, Quah TC, Lai PS. High incidence of allelic loss at 16q12.2 region spanning RBL2/p130 gene in retinoblastoma. Cancer Biol. Ther.8(8), 714–717 (2009).
  • Indovina P, Acquaviva A, De Falco G et al. Downregulation and aberrant promoter methylation of p16INK4A: a possible novel heritable susceptibility marker to retinoblastoma. J. Cell. Physiol.223(1), 143–150 (2010).
  • Corson TW, Huang A, Tsao MS, Gallie BL. KIF14 is a candidate oncogene in the 1q minimal region of genomic gain in multiple cancers. Oncogene24(30), 4741–4753 (2005).
  • Madhavan J, Coral K, Mallikarjuna K et al. High expression of KIF14 in retinoblastoma: association with older age at diagnosis. Invest. Ophthalmol. Vis. Sci.48(11), 4901–4906 (2007).
  • Madhavan J, Mitra M, Mallikarjuna K et al.KIF14 and E2F3 mRNA expression in human retinoblastoma and its phenotype association. Mol. Vis.15, 235–240 (2009).
  • Corson TW, Gallie BL. KIF14 mRNA expression is a predictor of grade and outcome in breast cancer. Int. J. Cancer119(5), 1088–1094 (2006).
  • Riveiro-Falkenbach E, Soengas MS. Control of tumorigenesis and chemoresistance by the DEK oncogene. Clin. Cancer Res.16(11), 2932–2938 (2010).
  • Galderisi U, Di Bernardo G, Cipollaro M et al. Differentiation and apoptosis of neuroblastoma cells: role of N-myc gene product. J. Cell. Biochem.73(1), 97–105 (1999).
  • Cheng JM, Hiemstra JL, Schneider SS et al. Preferential amplification of the paternal allele of the N-myc gene in human neuroblastomas. Nat. Genet.4(2), 191–194 (1993).
  • Kim CJ, Chi JG, Choi HS et al. Proliferation not apoptosis as a prognostic indicator in retinoblastoma. Virchows Arch.434(4), 301–305 (1999).
  • Seshadri R, Matthews C, Norris MD, Brian MJ. N-myc amplified in retinoblastoma cell line FMC-RB1. Cancer Genet. Cytogenet.33(1), 25–27 (1988).
  • MacPherson D, Conkrite K, Tam M, Mukai S, Mu D, Jacks T. Murine bilateral retinoblastoma exhibiting rapid-onset, metastatic progression and N-myc gene amplification. EMBO J.26(3), 784–794 (2007).
  • Bell E, Chen L, Liu T, Marshall GM, Lunec J, Tweddle DA. MYCN oncoprotein targets and their therapeutic potential. Cancer Lett.293(2), 144–157 (2010).
  • Shehata HH, Abou Ghalia AH, Elsayed EK, Ziko OO, Mohamed SS. Detection of survivin protein in aqueous humor and serum of retinoblastoma patients and its clinical significance. Clin. Biochem.43(4–5), 362–366 (2010).
  • Adithi M, Nalini V, Kandalam M, Krishnakumar S. Expression of matrix metalloproteinases and their inhibitors in retinoblastoma. J. Pediatr. Hematol. Oncol.29(6), 399–405 (2007).
  • Mitra M, Kandalam M, Verma RS, UmaMaheswari K, Krishnakumar S. Genome-wide changes accompanying the knockdown of Ep-CAM in retinoblastoma. Mol. Vis.16, 828–842 (2010).
  • Marchong MN, Chen D, Corson TW et al. Minimal 16q genomic loss implicates cadherin-11 in retinoblastoma. Mol. Cancer Res.2(9), 495–503 (2004).
  • Laurie N, Mohan A, McEvoy J et al. Changes in retinoblastoma cell adhesion associated with optic nerve invasion. Mol. Cell. Biol.29(23), 6268–6282 (2009).
  • Marchong MN, Yurkowski C, Ma C, Spencer C, Pajovic S, Gallie BL. Cdh11 acts as a tumor suppressor in a murine retinoblastoma model by facilitating tumor cell death. PLoS Genet.6(4), e1000923 (2010).
  • Laurie NA, Donovan SL, Shih CS et al. Inactivation of the p53 pathway in retinoblastoma. Nature444(7115), 61–66 (2006).
  • Elison JR, Cobrinik D, Claros N, Abramson DH, Lee TC. Small molecule inhibition of HDM2 leads to p53-mediated cell death in retinoblastoma cells. Arch. Ophthalmol.124(9), 1269–1275 (2006).
  • Adithi M, Nalini V, Kandalam M, Krishnakumar S. Expression of p63 and p73 in retinoblastoma: a clinicopathological correlation study. Exp. Eye Res.87(4), 312–318 (2008).
  • Macaluso M, Paggi MG, Giordano A. Genetic and epigenetic alterations as hallmarks of the intricate road to cancer. Oncogene22(42), 6472–6478 (2003).
  • Richter S, Vandezande K, Chen N et al. Sensitive and efficient detection of RB1 gene mutations enhances care for families with retinoblastoma. Am. J. Hum. Genet.72(2), 253–269 (2003).
  • Harada K, Toyooka S, Shivapurkar N et al. Deregulation of caspase 8 and 10 expression in pediatric tumors and cell lines. Cancer Res.62(20), 5897–5901 (2002).
  • Harada K, Toyooka S, Maitra A et al. Aberrant promoter methylation and silencing of the RASSF1A gene in pediatric tumors and cell lines. Oncogene21(27), 4345–4349 (2002).
  • Choy KW, Lee TC, Cheung KF et al. Clinical implications of promoter hypermethylation in RASSF1A and MGMT in retinoblastoma. Neoplasia7(3), 200–206 (2005).
  • Choy KW, Pang CP, Fan DS et al. Microsatellite instability and MLH1 promoter methylation in human retinoblastoma. Invest. Ophthalmol. Vis. Sci.45(10), 3404–3409 (2004).
  • Cohen Y, Merhavi-Shoham E, Avraham RB, Frenkel S, Pe’er J, Goldenberg-Cohen N. Hypermethylation of CpG island loci of multiple tumor suppressor genes in retinoblastoma. Exp. Eye Res.86(2), 201–206 (2008).
  • Graham JS, Kaye SB, Brown R. The promises and pitfalls of epigenetic therapies in solid tumours. Eur. J. Cancer45(7), 1129–1136 (2009).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.