LEAD ARTICLE Genetics and Cytogenetics of Retinoblastoma John K. Cowell and Annette Hogg INTRODUCTION Cancer cells are referred to as " i m m o r t a l i z e d , " because, for the most part, they no longer r e s p o n d to intra- and extracellular signals preventing their proliferation or causing t h e m to differentiate. In some cases the b r e a k d o w n of ordered cell division m a y be due to either the inappropriate expression (in the wrong cell or at the wrong time) of a particular protein(s) or the overproduction or stable expression of a growth-promoting gene product. In other tumors, particularly those inherited as a d o m i n a n t trait, it is the absence of a functional gene product, at an important stage of cellular development, that is the rate-limiting step. Because it is the normal function of these genes that prevents tumorigenesis they have been referred to as " t u m o r suppressor genes" or "recessive oncogenes" or, perhaps inappropriately, "antioncogenes" [1]. The study of one particular tumor suppressor gene, responsible for the d e v e l o p m e n t of retinoblastoma, is allowing us to investigate and understand f u n d a m e n t a l processes required for m a n y aspects of tumorigenesis and is the subject of this review. Retinoblastoma Genetics Retinoblastoma is an intraocular t u m o r occurring almost exclusively in young c h i l d r e n and has both hereditary and sporadic forms. In the familial form, w h i c h represents 1 0 % - 1 2 % of all cases, the t u m o r p h e n o t y p e segregates as an autosomal d o m i n a n t trait with 90% penetrance. This means that inheritance of a single mutant gene will result in the tumor p h e n o t y p e in 9 of 10 cases. Transmitting individuals, therefore, are heterozygous for the mutation, their c h i l d r e n having a 50 : 50 chance of inheriting it. Knudson [2], in a mathematical treatise of the subject, showed that, in those i n d i v i d u a l s carrying a predisposing mutation, a single a d d i t i o n a l event (which was later proved to be at the homologous normal locus) is sufficient for tumorigenesis. Thus, in familial cases, it is a p r e d i s p o s i t i o n to cancer that is inherited, and only a single r a n d o m mutational event is required for tumor initiation. Because the chances of this single a d d i t i o n event occurring in the precursor retinal cell p o p u l a t i o n is relatively high, gene carriers develop multiple, bilateral tumors with an early age of onset. In contrast, because in sporadic cases, both mutations must occur in homologous genes in the same retinal precursor

From the ICRF Oncology Group, Institute of Child Health,

London. Address reprint requests to: Dr. John K. Cowell, ICRF Oncology Group, Institute of Child Health, 30 Guilford Street, London WCINIEH, UK. Received March 11, 1992; accepted June 12, 1992.

© 1992 Elsevier Science Publishing Co.. Inc. 655 Avenue of the Americas, New York, NY 10010

cell, they are u s u a l l y unilateral, unifocal, and have a later age of onset. The age of onset m a y be s o m e w h a t m i s l e a d i n g because, histopathologically, Rb consists of e m b r y o n i c - l i k e cells that have failed to differentiate. Because differentiation of the retina is c o m p l e t e soon after birth, it is likely that all Rb tumors have begun d e v e l o p i n g during embryogenesis, and presentation d e p e n d s on m a n y factors, including the stage of d e v e l o p m e n t at w h i c h the second mutation occurred, tumor growth rate, and identification of the symptoms. Because the second m u t a t i o n in hereditary cases occurs r a n d o m l y , the n u m b e r of tumors that develop follow a Poisson distribution that accounts for some of the heterogeneity seen in the p h e n o t y p e , but not all of it. Matsunaga [3] suggested in a series of papers that other factors m a y affect the occurrence of Rb, i n c l u d i n g host resistance. These genetic factors m a y also serve to m o d i f y the expression of the RB1 gene [4]. Although the vast majority of familial cases develop multifocal tumors (3-4, on average) in both eyes, occasional unilaterally affected i n d i v i d u a l s occur w i t h i n these families. In our own series, a p p r o x i m a t e l y 1 0 - 1 5 % of families have at least one unilaterally affected i n d i v i d u a l . However, there are other families where all affected i n d i v i d u a l s only develop either a single, unilateral t u m o r or no t u m o r at all [5]. In some families, a p p a r e n t l y unaffected i n d i v i d u a l s have been seen to have retinal scars that resemble successfully treated tumors. These have been described as benign t u m o r s - - r e t i n o m a s [ 6 ] - - o r as regressed tumors. In family linkage studies, it has been confirmed that these patients with scars carry the m u t a n t gene. Many of these various " m i l d " forms of the tumor can occur in different i n d i v i d u als in the same family [5]. It is not clear w h e t h e r these u n u s u a l p h e n o t y p e s are just part of a c o n t i n u o u s s p e c t r u m of the p h e n o t y p e or w h e t h e r specific m u t a t i o n s w i t h i n the gene are responsible (see later). Occasionally, several affected children can be born to unaffected parents w i t h no prior family history. W h i l e this m a y be due to the segregation of u n u s u a l insertional translocations (see later), it is also possible that it is related to tissue m o s a i c i s m (see ref. 7 for review). A major challenge in the genetic analysis of Rb will be to try to rationalize all this p h e n o t y p i c heterogeneity in terms of m o l e c u l a r events w i t h i n the retinoblastoma gene, RB1. Localization of the RB1 Gene The first steps toward the isolation of the RB1 gene came from cytogenetic analysis of rare Rb patients w i t h other congenital abnormalities, especially mental retardation [8]. These patients had constitutional deletions from one copy of c h r o m o s o m e 13 [9]. A l t h o u g h these deletions vary in size, part of c h r o m o s o m e band 13q14 was always lost, indi1 Cancer Genet Cytogenet 64:1-11 (1992) 0165-4608/92/$05.00

2 cating the location of the predisposition gene. Usually, the larger the deletion, the greater is the range of associated congenital abnormalities [10]. The esterase-D gene (ESD) is also located in 13q14, and those patients with constitutional deletions had reduced cellular ESD enzyme levels [11]. This assay allowed population-based studies demonstrating that approximately 3% of Rb patients carry 13q14 deletions [10, 12]. The position of the ESD gene within 13q14 was demonstrated by Mitchell and Cowell [13], who analyzed an Rb patient with a 13q14-q31 deletion [14] and who had normal ESD levels. When the deletion chromosome was isolated in a somatic cell hybrid, the ESD gene was present, but RB1 was not, indicating that ESD lay proximal to Rb. The suggested location of RB1, within 13q14, varies depending on the particular study. Sparkes et al. [15] favored a 13q14.1 location based on the analysis of the breakpoints in one deletion patient. Yunis and Ramsay [16], using high-resolution cytogenetics, suggested the 13 q14.2-14.3 region as the site of the Rb gene from studying a microdeletion, and we [14] and others [17] favor a more distal location in 13q14.3. Although not of fundamental importance in the cloning of the gene, it may be important in siting the position of other genes and chromosomal breakpoints within 13q14. The dominantly inherited form of Rb was also shown to be due to a gene located in 13q14 by virtue of its linkage to the ESD gene in Rb families [18, 19]. Finally, evidence was presented [20-22] that the same gene was likely involved in sporadic tumors because they frequently underwent loss of heterozygosity for the 13q14 region. The suggestion here was that in retinal precursor cells, an acquired mutation was duplicated at the expense of its normal homologue, rendering the cell homozygous for the initial loss of function RB1 mutation. The mechanisms most frequently involved were due to chromosome nondisjunction and mitotic recombination [21]. This assumption was confirmed because in hereditary Rb, the chromosome retained in the tumor was that transmitted by the affected parent [23]. It now appears that up to 70% of tumors experience this loss of heterozygosity (LOH) [21, 24, 25]. These kind of analyses allowed the origin of the parental mutation to be determined [26]. In sporadic cases there was no differential susceptibility to somatic mutation between the homologous copies of the gene. However, for new germline mutations, the heritable mutation arose on the paternally derived chromosome. These findings and those of Zhu et al. [24] argue against genomic imprinting being important in Rb tumorigenesis but point to new mutational events arising predominantly during spermatogenesis. It has not, however, been possible to attribute these to a paternal age effect [27].

Cytogenetic Analysis of Rb Tumors The mechanisms leading to LOH can clearly occur without structural chromosome rearrangements. It was hardly surprising, therefore, that chromosome 13 abnormalities were found infrequently in tumor cells, although several interesting observations have come out of these cytogenetic analyses. Occasionally, tumor cells from different histologic sites

J.K. Cowell and A. Hogg reveal structural chromosome abnormalities that are specific to that tumor and, therefore, potentially important in the malignant process. However, because this analysis is usually performed on advanced stage tumors, it is difficult to detemine whether these changes are causal in tumorigenesis or consquences of it. This is particularly true for Rb because, in countries where adequate treatment is available, tumors are only removed when too large to treat in situ. Other difficulties encountered in the cytogenetic analysis of Rb is their low mitotic index and the relatively poor quality of the chromosomes derived from them. Despite these limitations several groups have presented interesting observations. Much of the early work has been collated by Potluri et al. [28], who reviewed cytogenetic analysis from 82 cases prepared directly from tumor tissue or cultured cells and cell lines from patients with both sporadic and hereditary Rb. By far the most consistent finding was the presence of (usually two copies of) an isochromosome 6p [i(6p)] and trisomy for all, or part, of the long arm of chromosome 1 (lq+) in 45% and 44% of tumors, respectively. Chromosome abnormalities involving chromosome 13, usually resulting in monosomy 13, was only found in 20% of cases. Similar observations were reported independently by Squire et al. [29] from a single-center study, with i(6p) present in 56% of tumors and trisomy for 1q23-qter in 75%. In this study only 11% of tumors had abnormalities involving chromosome 13. Abnormalities involving lq are the most commonly quoted in all tumor cells. By contrast i(6p) is less frequently observed, being restricted largely to Rb and malignant melanomas [30]. The presence of i(6p) has also been described by others [31-33] but its exact significance in tumorigenesis is still unclear. One possibility is that duplication of certain genes on the short arm of chromosome 6 may be important in tumor progression. Trent and colleagues [34] presented evidence for a tumor suppressor gene on chromosome 6 by introducing this chromosome into melanoma cells and suppressing the malig~ nant phenotype. Other, less frequently reported chromosome abnormalities [28] in Rb tumors include monosomy 16, lp ÷ and the presence of double minutes (DM) and homogeneously staining regions (HSR). DM and HSR represent the cytologic manifestation of gene amplification [35]. In Rb, amplification of the N-myc oncogene has been reported in a few tumors [36-38]. Another cellular oncogene, INT-1, was also shown to the amplified in three tumors [39]. Both of these oncogenes appear to be expressed in cells of neurogenic origin, although, again, their role in tumorigenesis is unclear. Chromosome analysis of independent tumor foci from a bilaterally affected patient showed that each had distinct abnormalities, suggesting an independent origin for them [29]. Tien et al. [31] analyzed a large, apparently unilateral tumor and found cytogenetically distinct clones. This was interpreted to mean that the tumor probably arose as the result of fusion of several foci. This has important implications for counseling, because unilateral tumors are thought to be associated predominantly with sporadic events. Multifocal tumors, however, even in only one eye, probably identify that patient as a hereditary case, especially if these tumors have a relatively early age of onset.

Genetics and Cytogenetics of Retinoblastoma

Constitutional Chromosome Abnormalities In addition to deletions of 13q14, individuals predisposed to Rb may also carry constitutional chromosome translocations with breakpoints in 13q14. In some cases inheritance of an unbalanced form of the rearrangement results in deletion of the 13q14 region [40-45]. In other cases the translocation appears to be reciprocal, and molecular analysis of the breakpoint junctions, so far, has shown that the 13q14 breakpoint interrupts the RB1 gene )46-48]. In one case, at least, this rearrangement is associated with a small deletion within the RB1 gene [47]. A few cases have been reported where the translocation partner chromosome is the X [49-51]. Because random X-activation occurs in females, when the derivative chromosome is inactivated in retinal precursor cells, the position of the breakpoint on 13 is not important because the whole chromosome experiences "genetic silencing," thereby constituting the first hit. In two families in our series, with t(1;13)(q22;q14) and t(13;20)(q14;p12) translocations, the patient with Rb inherited the rearrangement from a parent who was not affected (B. Gibbons, personal communication). A similar situation was discussed by Dryja et al. [22] for 13q14 deletions, in that deletion carriers often only have unilateral, unifocal tumors. In our survey [10], only half of the 16 cases reported had unilateral tumors. There are also reports of deletion patients who have never developed tumors [52-54]. The age of onset of tumors from patients with chromosome 13 abnormalities also appears to be later than those with germinal mutations [55]. The explanation for the low penetrance in deletion patients is not clear, although one proposal is that these deletions expose lethal mutations that are deleterious to the rapidly growing tumor cells [22]. This does not explain the low penetrance af the reciprocal translacatian carriers, unless large deletions are associated with the rearrangement. In one case of a t(1;13) rearrangement [47], although a deletion was associated with the translocation, it was maximally 8 kb long and confined to the RB1 gene. Keith and Webb [56] described a patient with a 13q14 deletion with a single tumor in one eye and a retinoma in the other, both of which are considered to be "mild" forms of the disease. Isolation and Structure of the RB1 Gene The cloning of the RB 1 gene proceeded very rapidly following the construction of a chromosome 13 specific (DNA) library from flow-sorted chromosomes [57]. Of 12 probes isolated at random, one was shown to lie in the 13q14 region, and adjacent sequences identified a highly conserved region [58]. In fact, this probe lay within the RB1 gene and was used to isolate a 4.7-kb cDNA from a fetal retinal cell library [59]. Several other groups [60, 61] repeated this procedure and collectively showed that structural rearrangements of the RB1 gene were present in approximately 20% of tumors. Abnormal mRNAs were seen to a greater [60] or lesser [62] degree. Often the mRNA product was completely missing in tumors, reflecting the loss of function nature of many of the mutations. RB1 is a relatively large and complex gene, approximately 180 kb long and composed of 27 exons ranging in length from 31 bp to 1873 bp [63, 64]. The intervening

3

intron sequences vary in size from 80 bp to 70,500 bp. The exons are clustered into three groups, separated by the two largest introns [65]. In human cells, the gene encodes an mRNA transcript of 4.7 kb [59, 60], which is expressed in the majority of cell types studied. The RB 1 promoter region is unusual in that it lacks a TATA box or CCAAT motif [65], typically found in eukaryotic promoters and involved in the positioning of RNA polymerase II and binding of the CAT/enhancer binding protein (C/EBP), respectively. The lack of a typical TATA box element may account for the variable transcription start sites observed in $1 nuclease experiments [66]. The 5' promotor region is GC-rich and has characteristics of a CpG island [65, 67], whereas the 3' end of the gene, involved with transcription termination, contains a hexanucleotide with a consensus signal for polyadenylation. Several studies have defined regions of the RB1 promotor required for transcription. One such region, between 185 and 206 bp upstream of the initiating methionine residue, has been located by the analysis of deletion mutants and appears to be essential for transcription [64, 66, 68]. This area contains a sequence showing homology to the binding site for the SP1 transcription factor [64], suggesting it might contain the RB1 promotor, although SP1 itself shows only weak binding. This binding site associates with another nuclear factor, referred to as RBF1, whose function is not known [68]. A sequence showing homology to the binding sites for the ATF/CREB family of transcription factors is also located within the putative RB 1 promotor region and occurs adjacent to the site where RBF-1 and SP1 bind. Potential zinc-finger motifs, associated with DNA binding ability, were found in exons 17, 18, 20, and 21 and putative leucine zipper motif, associated with protein-protein interaction, was entirely encoded for by exon 20 [66]. Experimental evidence that these regions function as such is still lacking.

Family Linkage Analysis With the cloning of the RB1 gene, the expectation was that the cDNA would identify restriction fragment length polymorphisms (RFLPs) within the population that, in turn, could be used in linkage analyses. However, despite extensive studies in several laboratories, no useful RFLPs were discovered. To overcome this limitation, Wiggs et al. [69] isolated five unique DNA sequences from the introns of RB1 that, in combination, were useful in gene carrier detection. The principles of "gene tracking" have been presented elsewhere (see ref. 70), but essentially the transmitting individual must be heterozygous at the locus in question. Despite the theoretical probability of recombination within the RB1 gene, no recombinants have yet been reported in Rb families [25, 69, 71, 72]. The most useful single copy probe isolated by Wiggs et al. [69] is RS2.0, which identifies a variable number tandem repeat (VNTR) based on a 53-bp repeat with eight different alleles. In our own study [(72) and unpublished observations), 60% of patients were heterozygous--"informative"--at this locus, and 80-85% of all families were informative using a combination of all five probes. Using this type of linkage analysis, one can now determine which individuals in Rb families

4 have inherited the predisposing mutation and, equally important, which have not. For those who, unequivocally, have not inherited the mutant gene, repeat ophthalmologic screening of subsequent generations is not necessary. For those patients identified as gene carriers (affected and unaffected), prenatal screening is possible. The first report of this kind [73] used chorionic villus (CV) sampling after only 9 weeks of pregnancy and demonstrated the child would be unaffected. This patient is now 55 months old and tumor free. Since that report, we have performed nine more pre- and postnatal predictions, most of which have been followed for at least 2 years, and all of which have proved correct [74]. Since the first report by Wiggs et al. [69], it has become possible to analyze two of the polymorphic sites within RB1 using the polymerase chain reaction [75, 76]. This means that the analysis can be completed in 2 days compared with 5-7 days for probes requiring analysis by Southern blotting techniques [74]. In addition, several other RFLPs have been added to the armory of tests available for family studies, some of which are single base pair polymorphisms identified by sequencing [77], and another is a VNTR based on a small (1-4 bp) repeat unit, which must be resolved on sequencing gels. This latter RFLP, called Rbl.20, has approximately 28 different alleles and is informative in 85-95% of families [74, 77]. Thus, it is now possible to offer family linkage analysis and prenatal screening to virtually all Rb families with the realistic expectation that all gene carriers can be identified. In addition to the obvious advantages to the patients and their families, the screening load for ophthalmology departments will be greatly reduced in the future. The application of the polymerase chain reaction (PCR) to the analysis of polymorphic sites means that DNA isolated from formalin fixed, histopathologically processed materials can now also be used. We have been able to use this strategy to establish linkage phase in a family where the key member had died [78]. For the 85% of Rb patients without a family history, many will carry "new" germline mutations. The problem for them is that linkage analysis will not be available for their firstborn children. It is necessary, therefore, to identify the causative mutation in these cases that will demonstrate, unequivocally, who is at risk to tumor development. The characterization of the structure of the RB1 gene has made this possible. The RB Protein The RB1 gene is widely expressed in all human tissues examined to date [59, 60, 79]. A messenger ribonucleic acid (mRNA), 4.7 kb long, is transcribed in human tissues and encodes a protein of 928 amino acids. In all but a few exceptions, rodent and murine tissues also express a similar-sized transcript. A shorter 2.3-kb transcript has been reported in rat fetal brain [60], and a 2.8-kb transcript was found in adult mouse testes [80]. In the latter case, the onset of expression of the shorter mRNA coincided with the appearance of spermatids in the testes. The significance of these shorter transcripts has not yet been determined but may result from differential processing of the RB1 gene. The protein product of the RB1 gene (pRB) is generally thought to suppress cell growth because the absence of a

J.K. Cowell and A. Hogg normal functional product is associated with tumor formation [60, 81]. Reintroduction of a functional pRB into cell lines lacking the RB1 gene apparently suppresses tumorigenicity in immunosuppressed mice [82-85]. Others, however, have failed to repeat this observation and cast doubt on the infallability of the nude mouse assay systems used for this kind of analysis [86, 87]. Early analysis of pRB showed that it could be phosphorylated, mainly on serine and threonine residues [88, 89]. A number of species of pRB, ranging from approximately 110 to 115 kd, were detected using Western blotting techniques [88, 90]. The higher molecular weight species represented more highly phosphorylated proteins, those with lower molecular size being less phosphorylated [89, 91]. Subsequently, the phosphorylation status of pRB was shown to be cell cycle dependent [92-94]. The underphosphorylated forms of pRB were found in quiescent cells, whereas the phosphorylated forms appeared toward the onset of DNA synthesis [93, 94]. Moreover, in synchronized cells, the appearance of highly phosphorylated forms of pRB coincided with the transition from G1 --~ S-phase of the cell cycle [93, 94]. The more highly phosphorylated forms were still present in G2 and M, although the underphosphorylated forms began to predominate [91, 94, 95]. These observations provided strong circumstantial evidence that pRB was involved in the control of the cell cycle, pRB, however, does not have DNA sequence-specific binding motifs so must apparently exert any regulatory controls as a result of interactions with other cellular proteins. Association of pRB with Viral Oncoproteins Several DNA tumor viruses produce proteins that can override the normal growth regulation of the cell, driving infected cells through S-phase where viral DNA can be synthesized.This is apparently activated by binding to host cellular proteins. During studies aimed at characterizing these cellular proteins, pRB was found to interact with the Ela protein of adenovirus [90] as well as with the transforming proteins of two members of the papovavirus family; the Simian virus 40 large T (LT) antigen [95] and the E7 protein of human papilloma virus [96]. All of the viral transforming proteins share conserved regions that are necessary for their transforming function. Mutations in these conserved regions, which prevent cellular transformation, also prevent binding to pRB [97]. LT binds specifically to the underphosphorylated form of pRB [89], and LT-pRB complexes are found only in G1 when the underphosphorylated form of pRB is present [89]. These observations suggested that the underphosphorylated pRB is active in the suppression of cell growth and that phosphorylation, or sequestration by LT, allowed the cells to enter S-phase. Furthermore, entry of cells into S-phase could be blocked by microinjection, into cells lacking pRB function, of either the full length or a truncated protein lacking the LT binding domain [98]. By selective deletion of parts of the RB1 gene, two noncontiguous regions were identified that were necessary for Ela and LT binding [99-101). These protein binding domains comprised amino acids 393-572 and 646-772, respectively, which have collectively been called the RB

Genetics and Cytogenetics of Retinoblastoma "pocket." Deletion of the spacer region, located between these two noncontiguous sequences, prevents Ela/LT binding [99], but replacement with nonspecific sequences restored that function. It appears, therefore, that, by sequestering pRB from the cell during G1, the viral transforming proteins allow the cell to enter S-phase. In fact, it has emerged that pRB participates in the establishment of protein complexes that associate and dissociate during the cell cycle. Many of these processes are under tight control, and the whole system appears to be regulated by the biochemical modification of the participants in these complexes and their availability to join the complex.

Involvement of pRB in the Cell Cycle The Ela protein is thought to transform cells by altering the activity of cellular transcription factors. One such protein is E2F, which has been shown to be involved as a transcription regulator of several cellular genes [102]. Ordinarily E2F is complexed with specific cellular proteins that effectively suppress its function [103]. Ela, however, can dissociate E2F from its protein complexes, releasing free E2F. To exert its transcriptional regulation, E2F must form a stable complex with other proteins in order to bind to specific DNA sequences in the promotor regions of the genes it controls. The conserved regions of Ela facilitate E2F binding; the same regions are also responsible for binding pRB. It was not surprising, therefore, to find that pRB also complexes with E2F and that Ela can dissociate them [103-106]. E2F associates with the form of pRB found primarily in G1 [106]. The E2F/pRB complex ordinarily dissociates near the G1-S boundary, before S phase, releasing free E2F. The suggestion is that pRB can control the transcriptional activities of E2F by binding to it. Dissociation of this complex allows free E2F to activate responsive cellular promotors that contribute to the release of cells from their proliferative suppression. pRb has also been shown to complex with a developmentally regulated cellular transcription factor, DRTF-1, which binds to the same sequence motif as E2F, although the relationship between them is unclear [103, 105, 107, 108]. The DRTF-la complex associates with pRB [107], and the complex can be dissociated by Ela. The suggestion is that, by binding with a sequence-specific transcription factor, pRB can control transcription of target genes. Because DRTF is developmentally regulated and its expression is tissue dependent, this association suggests a role for pRB in differentiation [105, 107, 108]. Two other Rbbinding cellular proteins, called RBF-1 and RBF-2, have recently been cloned and sequenced. They contain amino acid sequences homologous to the Ela/LT/E7 Rb binding domains [109]. E2F binding sites are present in the promoters of a number of cellular genes, implicating pRB in transcriptional regulation. Sequences resembling the E2F binding site are present in the c - m y c promoter [110] and pRb can suppress c - m y c transcription. Robbins et al. [111] showed pRB could interact with the c-los promotor region to repress transcription as well as down regulating Apl, a transcription factor thought to be involved in regulating a set of early response genes required for cell growth. The m y c and los gene prod-

5 ucts are part of the early response genes following growth stimulation and the demonstration that pRB can bind to their promoters in vitro suggests a cooperation in the control of cell proliferation [110]. The same c/s-acting element is found in the promoter of the transforming growth factorfll (TGFB) gene [112]. TGFB molecules are potent growthinhibiting polypeptides and act in G1, although the exact mechanism is not known. Transcriptional initiation of growth related genes such as c - m y c is inhibited by TGFB. Treatment of cells with TGFB prevents phosphorylation of pRB and maintains it in its growth suppressive state [113]. Because TGFB also inhibits cells lacking pRB their interaction, however, is not obligatory for suppression of cell growth [114]. From the preceeding discussion, it is clear that pRB may be involved in a variety of cell-cycle-regulated activities. The cyclins constitute a family of related, cell-cycle-dependent, proteins [115] that associated with cellular protein kinases, including the protein products of cdc2 and cdk2 [116, 117], which are thought to be key regulators of cell cycle progression [115]. Both cyclin A and pRB locate to DNA by virtue of the ability to complex with DRTF and cyclin A functions to assemble pRB into the transcription factor complex. It is the underphosphorylated form of pRB that joins the complex [107]. Cyclin A, therefore, may serve to direct cdc2 kinase to dephosphorylate pRB releasing it from the complex. These observations further support that pRB is involved in the regulation of the cell cycle, pRB can be phosphorylated at all potential sites in vitro by cdc-2, at least [116]. Further evidence supporting an in vivo role for cdc 2 phosphorylation of pRB is currently lacking. Cyclin A also associates with the form of E2F that appears once cells have entered S-phase [118]. In constrast, E2F has been found to interact with the unphosphorylated form of pRB, implying that this complex occurs in G1 [106]. Another protein, p107, is associated with the cyclin A/ E2F/cdk2 complex [117, 119-121]. It has been proposed that the E2F/p107 containing S-phase complex and the E2F/ pRB containing Gl-phase complex may each serve specific functions relevant to the maintenance of normal cell growth. Both complexes were virtually absent in G2/M [120]. The p107 protein is highly homologous to pRB and was known to be a cellular target for Ela [90, 95-97,121]. p107 has been cloned and has two viral oncoprotein binding domains, homologous to those forming the Rb pocket in pRB, but separated by a much larger "spacer element" (121). It is the spacer element that binds cyclin A. The ability of cyclins to activate cellular kinases raises many possibilities for their role in these complexes. The p107 and pRB proteins may themselves be phosphorylated through these interactions, or the E2F protein may be modified as part of its cell cycle specific activity. It is thought that this may result in the release of free E2F, which can then activate transcription of other genes. It is possible, however, that the mechanism is more indirect and that other, as yet unknown, intermediary factors are involved. The p107 protein is found in cell lines with homozygous deletions of RB1 [121]. If one assumes p107 is normal in these cells, it clearly cannot compensate for the lack of pRB. The p107 protein is found in a cyclin A/E2F complex in S-phase

6 [117], which also contains cdk2, suggesting pRB and p107 cooperate in regulating E2F activity at different stages of the cell cycle. RB Inactivation in Other Tumors

Individuals carrying constitutional mutations in the RB1 gene are at significantly higher risk than the general population to the development of second, nonocular tumors later in life (122-124). These tumors are usually osteosarcomas (OS) and soft-tissue sarcomas. For some patients who have received radiation therapy for their retinoblastoma(s), these second tumors arise within the irradiated field, but a significant proportion occur in unirradiated tissues. These observations suggest that the RB1 gene also plays a role in tumorigenesis in other tissues, the frequency of which can be raised by 7 irradiation. Although the risk of the development of OS is highest in adolescence, the risk to other cancers persists throughout the lives of mutant gene carriers [125]. Structural rearrangements of the RB1 gene have been identified in osteosarcomas [81,126,127] and in soft-tissue sarcomas [63,128], supporting its role in the development of these tumors. Loss of heterozygosity (LOH) for RB 1 infers that a recessive mutation is exposed and has been demonstrated in a wide range of human malignancies. A combination of LOH studies, analysis of structural rearrangements by Southern blotting, and absence of pRB using immnnofluorescence and Western blotting has implicated the RB1 gene in tumorigenesis in breast cancer [129, 130], small cell lung carcinoma [131-133], bladder cancer (134-136), esophageal cancers (137), hepatocarcinomas (138), prostate cancer (83), and a range of leukemias (139). However, only a proportion of these tumors have RB1 abnormalities and it is unlikely that RB1 mutations are the rate limiting step in these tumors. Instead, these mutations probably contribute to a multistep process of tumorigenesis in these tissues. One interesting tumor associated with hereditary Rb is pineoblastoma. Because the pineal is thought to be a vestigial photoreceptor organ, patients with retinal and pineal tumors have been described as having "trilateral" retinoblastoma [140, 141]. The chances of two such rare tumors occurring coincidentally is negligible and suggests that the RB1 gene also contributes to the development of pineoblastoma. Mutations in RB1

Early data, derived mostly from Southern blot analysis, identified structural abnormalities of RB1 in 10%-40% of hereditary and sporadic Rb tumors. The majority of these involved deletions of all or part of the gene [59-61]. It appears that deletion breakpoints can occur throughout the length of the gene [48, 142,143]. In the study by Canning and Dryja [142] 2 of 12 deletions had breakpoints in the region containing exons 13-17, and 7 of 12 deletions included that region. Others also found this region was involved in deletions and rearrangements [47, 61] possibly indicating the location of a breakpoint cluster region. A clustering of dyad symmetrical elements was found in exons 16 and 17 and an inverted repeat sequence in exon 17 [64]. Whether this is related to the chromosome abnormali-

J.K. Cowell and A. Hogg ties is not known, but these DNA sequences can form stem and loop structures that possibly promote rearrangement. In 6 of 8 deletions reported by Canning and Dryja, there was a direct (sometimes imperfect) repeat at the breakpoint site, and one of these was always lost. We (Hogg, unpublished) and others [144] have also found short, direct repeats associated with small deletions within RB1. This observation is consistent with a slipped mispairing during DNA replication. In this model, pairing on one DNA strand occurs with the downstream sequence on the other strand creating a loop. When the replication loop is resolved, one copy of the repeat, plus the intervening sequence, is deleted. The majority of mutations, however, appear to be more subtle sequence changes and have been found in both Rb tumors and normal cells from predisposed individuals [145-148]. Analysis of mRNA from tumors is not often possible, because the majority of them are successfully treated in situ. Of those that are removed, some do not produce RB1 and mRNA, and from many others the samples are too small to analyze. Depsite this limitation, Dunn and colleagues [145,146] were able to use tumor cell lines to isolate RNA and, with the use of RNase protection, identify mutations in 50%-65% of cases. They found that small deletions were the most common abnormality but suggested this may reflect limitations in the RNase protection procedure. Small deletions were also found most frequently in a proportion of SCLC cell lines studied by Mori et al. [149]. Unfortunately, it appears that, in constitutional cells heterozygous for the causative RB 1 mutation, the normal transcript dominates over the abnormal one [146]. This effectively excludes the possibility of using RNase protection, or sequencing from mRNA, to identify mutations in constitutional cells from Rb patients. To overcome these problems, Yandell et al. [147] sequenced the RB1 gene, exon by exon, following amplification using (PCR). Although this approach proved useful in identifying mutations, it was very time-consuming. The introduction of methods to prescreen the amplified products, such as the single strand conformation polymorphism (SSCP) technique [150] and chemical cleavage mismatch methods such as the HOT technique [151], have made the analysis more rapid. In combination, the available data shows that there is no apparent hot spot for mutations within RB 1, the majority of which are insertions, deletions, or single base pair substitutions that result in the production of premature stop codons. These mutations would be predicted to result in a structurally grossly abnormal protein and are consistent with studies where Rb tumors mostly had no detectable pRB [81]. Approximately 10% of mutations affect splice junction sites. Bookstein et al. [152], analyzing mRNA from a prostate carcinoma cell line, identified an in-frame deletion of exon 21, but, surprisingly, the splice junction sequences were normal. A single-point mutation within exon 20 had resulted in removal of the exon. A similar situation was reported by Mori et al. [149], who showed that the mRNA from an SCLC cell line was missing exon 22. A two base pair mutation GC --~ TT, within the exon, apparently converted a glutamine to a stop codon but, in fact, resulted in the entire exon being spliced out. Presumably these muta-

Genetics and Cytogenetics of Retinoblastoma

tions generate cryptic splice sites that result in the endogenous splice sites being ignored. A n o t h e r e x a m p l e was described in an SCLC that occurred as a second tumor in a patient with hereditary Rb [151]. A single base pair substitution w i t h i n exon 21, b e y o n d the splice site, results in excision of that exon from the mRNA. In some cases, however, [134, 146], mutations actually occur in the splice junction and affect splicing accordingly. It is clear, therefore, that the consequences of p o i n t mutations found by sequencing the DNA m a y not be so obvious, and, without access to the mRNA, h o w processing will be affected cannot always be predicted. Missence mutations, s i m p l y substituting one amino acid for another, appear to be less common. In our o w n survey of hereditary Rb patients, one example was found in exon 20, w h i c h was associated with a "lowp e n e t r a n c e " p h e n o t y p e [153]. It is tempting to speculate that the substitution of a single amino acid only compromises the function of the protein, and, unless the second m u t a t i o n in the tumor precursor cell causes loss of RB1 function, d u p l i c a t i o n of the " w e a k " mutation allows sufficient functional pRB to be produced, so preventing tumorigenesis. This is consistent with our observation in this particular family, because m a n y of the mutant gene carriers were either unaffected or have regressed tumors. Sakai et al. [68] also investigated low-penetrance families and found mutations in recognition sequences for different transcription factors in the promoter region of RB1. Again, the suggestion was that, as a result, a quantitative decrease in transcription occurs rather than complete inactivity. Sufficient pRB is p r o d u c e d , however, and any p h e n o t y p i c consequences are mild. Whether single amino acid changes will generally be found in patients with m i l d phenotypes is still not clear. Against this, Yandell et al [147] reported a ser--~ leu substitution in exon 14 in a tumor, and we have found a met --~ arg change in one t u m o r in the same exon (Hogg, unpublished). A cys ~ phe amino acid substitution in exon 21 was reported that resulted in a protein unable to b i n d to the viral oncoproteins and unable to complex with the set of Rb-associated cellular proteins [104, 154]. CONCLUSIONS The d e v e l o p m e n t of techniques to analyze the RB1 gene exon by exon means that heterozygous mutations can n o w be detected in constitutional cells in Rb patients. This means that, for the 85% of cases without a family history, it will be possible to identify mutant gene carriers that will greatly i m p r o v e the clinical m a n a g e m e n t of the disease. Too few naturally occurring mutations in Rb patients have been identified, so far, to allow correlations to be d r a w n b e t w e e n genotype and phenotype. Depending on the patterns that might emerge, it m a y eventually be possible, however, to predict the course of the disease and perhaps even to determine who is susceptible to the d e v e l o p m e n t of second tumors. Analysis of mutations in RB1 has also h e l p e d our understanding of the function of the gene. In those tumor cells where a nonfunctional pRB is produced, the mutations u s u a l l y prevent Ela, LT, and E7 b i n d i n g [90, 95, 96]. The same is true for m a n y of the other proteins that b i n d to pRB

7

[104], i n c l u d i n g transcription factors DRTF1 a n d RBP-1, and early growth response genes such as c-myc a n d N-myc [110]. This analysis has d e m o n s t r a t e d i m p o r t a n t functional domains of pRB and highlighted w h i c h interactions are important for normal cell growth control. The attachment of pRB to nuclear structures w i t h i n the cell [155] is also affected by mutations in the Rb pocket. One issue that is still v i r t u a l l y u n a d d r e s s e d is h o w pRB controls the differentiation of i m m a t u r e retinal cells into mature photoreceptors. Presumably, there are other, as yet undiscovered, proteins (growth factors?) that m u s t bind pRB to carry out their function but are o n l y available in particular cells at a particular stage of development. These interactions, promoting terminal differentiation, m u s t be critical, because loss of function of RB1 alone is responsible for tumorigenesis. The challenge is still to find these proteins and resolve one of the f u n d a m e n t a l processes of embryonic development.

REFERENCES 1. Knudson AG (1985): Hereditary cancer, oncogenes, and antioncogenes. Cancer Res 45:1437-1443. 2. Knudson AG (1971): Mutation and cancer: Statistical study of retinoblastoma. Proc Natl Acad Sci USA 68:820-823. 3. Matsunaga E (1980): Retinoblastoma: host resistance and 13q- chromosomal deletion. Hum Genet 56:53-58. 4. Matsunaga E (1980): On estimating penetrance of the retinoblastoma gene. Hum Genet 56:127-128. 5. Onadim Z, Hykin PG, Hungerford JL, Cowell JK, (1991): Genetic counselling in retinoblastoma: Importance of ocular fundus examination of first degree relatives and linkage analysis. Br J Ophthalmol 75:147-150. 6. Gallie BL, Ellsworth RM, Abramson DH, Phillips RA (1982): Retinoma: Spontaneous regression of retinoblasoma or benign manifestation of mutation? Br J Cancer 45:513-521. 7. Ribeiro MCM, Andrade JA, Erwenne CM, Brunoni D (1988): Bilateral retinoblastoma associated with 13q- mosaicism: Possible manifestation of a germinal mutation. Cancer Genet Cytogenet 32:169-175. 8. Stallard HB (1962): The conservative treatment of retinoblastoma. Trans Ophthalmol Soc 83:473-535. 9. Vogel W (1979): The genetics of retinoblastoma. Hum Genet 52:1-54. 10. Cowell JK, Hungerford J, Rutland P, Jay M (1988): Genetic and cytogenetic analysis of patients showing reduced esterase-D levels and mental retardation from a survey of 500 individuals with retinoblastoma. Ophthal Paediat Genet 10:117-127. 11. Sparkes RS, Sparkes MC, Wilson MG, Towner JW, Benedict W, Murphree AL, Yunis JJ (1980): Regional assignment of genes for human esterase-D and retinoblastoma to chromosome band 13q14. Science 208:1042-1044. 12. Cowell JK, Rutland P, Jay M, Hungerford J (1986): Deletions of the esterase-D locus from a survey of 200 retinoblastoma patients. Hum Genet 72:164-167. 13. Mitchell CD, Cowell JK (1988): Molecular evidence that the esterase-D gene lies proximal to the retinoblastoma susceptibility locus in chromosome region 13q14. Hum Genet 81:57-60. 14. Cowell JK, Hungerford J, Rutland P, Jay M (1987): A chromosomal breakpoint which separates the esterase-D and retinoblastoma predisposition loci in a patient with del(13) (q14-q31). Cancer Genet Cytogenet 27:27-31.

8

J . K . C o w e l l a n d A. Hogg

15. Sparkes RS, Sparkes MC, Kalina RE, Pagon RA, Salk DJ, Distechne CM (1984): Separation of retinoblastoma and esterase D loci in a patient with sporadic retinoblastoma and del(13)(q14.1q22.3). Hum Genet 68:258-259. 16. Yunis JJ, Ramsay N (1978): Retinoblastoma and subband deletion of chromosome 13. Am J Dis Child 132:161-163. 17. Duncan AMV, Morgan C, Gallie BL, Phillips RA, Squire J (1987): Re-evaluation of the sublocalization of esterase-D and its relation to the retinoblastoma locus by in situ hybridization. Cytogenet Cell Genet 44:153-157. 18. Sparkes RS, Murphree AL, Lingua RW, Sparkes Mc, Field LL, Funderbuck S, Benedict WF (1983): Gene for hereditary retinoblastoma assigned to human chromosome 13 by linkage to esterase-D. Science 217:971-973. 19. Cowell JK, Jay M, Rutland P, Hungerford J (1987): An assessment of the usefulness of electrophoretic varients of esteraseD in antenatal diagnosis of retinoblastoma in the United Kingdom. Br J Cancer 55:661-664. 20. Godbout R, Dryja TP, Squire J, Gallie BL, Phillips RA (1983): Somatic inactivation of genes on chromosome 13 is a common event in retinoblastoma. Nature 304:451-453. 21. Cavenee WK, Dryja TP, Phillips RA, Benedict WF, Godbout R, Gallie BL, Murphree A, Strong LC, White R (1983): Expression of recessive alleles by chromosomal mechanisms in retinoblastoma. Nature 305:779-784. 22. Dryja TP, Cavenee WK, White R, Rapaport JM, Peterson R, Albert DM, Bruns GA (1984): Homozygosity of chromosome 13 in retinoblastoma. New Eng J Med 310:550-553. 23. Cavenee WK, Hansen MF, Nordenskjold M, Kock E, Maumenee I, Squire JA, Phillips RA, Gallie BL (1985): Genetic origin of mutations predisposing to retinoblastoma. Science 228:501-503. 24. Zhu X, Dunn JM, Phillips RA, Goddard AD, Paton KE, Becker A, Gallie BL (1989): Preferential germline mutation of the paternal allele in retinoblastoma. Nature 340:312-313. 25. Goddard AD, Phillips RA, Greger V, Passarge E, Hopping W, Zhu X, Gallie BL, Horsthemke B (1990): Use of the RB1 cDNA as a diagnostic probe in retinoblastoma families. Clin Genet 37:117-126. 26. Dryja TP, Mukai S, Petersen R, Rapaport JM, Walton D, Yondell DW (1989): Parental origin of mutations of the retinoblastoma gene. Nature 339:556-558. 27. Matsunaga E, Minoda K, Sasaki MS (1990): Parental age and seasonal variation in the births of children with sporadic retinoblastoma: A mutation-epidemiologic study. Hum Genet 84:155-158. 28. Potluri VR, Helson L, Ellsworth RM, Reid T, Gilbert F (1986): Chromosomal abnormalities in human retinoblastoma. Cancer 58:663-671. 29. Squire J, Gallie BL, Phillips RA (1985): A detailed analysis of chromosomal changes in heritable and non-heritable retinoblastoma. Hum Genet 70:291-301. 30. Becher R, Gibas Z, Sandberg AA (1983): Chromosome 6 in malignant melanoma. Cancer Genet Cytogenet 9:173-175. 31. Tien H-F, Chuang S-M, Chen M-S, Lee F-Y, Hou P-K (1989): Cytogenetic evidence of multifocal origin of a unilateral retinoblastoma. Cancer Genet Cytogenet 42:203-208. 32. Squire J, Phillips RA, Boyce S, Godbout R, Rogers B, Gallie BL (1984): Isochromosome 6p, a unique chromosomal abnormality in retinoblastoma: verification by standard staining techniques, new densitometric methods, and somatic cell hybridisation. Hum Genet 66:46-53. 33. Horsthemke B, Greger V, Becher R, Passarge E (1989): Mechanism of i(6p) formation in retinoblastoma tumor ceils. Cancer Genet Cytogenet 37:95-102. 34. Trent JM, Stanbridge EJ, McBride HL, Meese EU, Casey G,

35.

36.

37.

38.

39.

40.

41.

42.

43.

44.

45.

46.

47.

48.

49.

50.

51.

52.

Araujo DE, Witkowski CM, Nagle RB (1990): Tumorigenicity in human melanoma cell lines controlled by introduction of human chromosome 6. Science 247:568-571. Cowell JK (1982): Double minutes and homogeneously staining regions: Gene amplification in mammalian cells. Ann Rev Genet 16:21-59. Lee W-H, Murphree AL, Benedict WF (1984): Expression and amplification of the N-myc gene in primary retinoblastoma. Nature 309:458-460. Sakai K, Kanda N, Shiloh Y, Donlon T, Schreck R, Shipley J, Dryja T, Chaum E, Chaganti RSK (1985): Molecular and cytologic analysis of DNA amplification in retinoblastoma. Cancer Genet Cytogenet 17:95-112. Squire J, Goddard AD, Canton M, Becker A, Phillips RA, Gallie BL (1986): Tumour induction by the retinoblastoma mutation is independent of N-myc expression. Nature 322:555-557. Arheden K, Tommerup N, Mandahl N, Heim S, Winther J, Jensen OA, Prause JU, Mitelman F (1988): Amplification of the human putative oncogene INT1 in primary retinoblastoma tumors. Cytogenet Cell Genet 48:174-177. Strong LC, Riccardi VM, Ferrell RE, Sparkes RS (1981): Familial retinoblastoma and chromosome 13 deletion transmitted via an insertional translocation, Science 213:1501-1503. Riccardi VM, Hittner HM, Francke U, Pipperin S, Holmquist GP, Kretzer FL, Ferrell R (1979): Partial triplication and deletion of 13q: study of a family presenting with bilateral retinoblastomas. Clin Genet 15:332-345. Calzolari E, Palazzi P, Aiello V, Mazzeo E, Perri P, Minelli A, Senno Ld, Patracchini P, Bernardi F (1987): De novo complex autosomal translocation involving chromosomes 8, 13 and 15 in a girl with sporadic retinoblastoma. Hum Genet 77:51-54. Rivera H, Turleau L, De Grouchy J, Junien C, Despoisse S, Zucker JM (1981): Retinoblastoma deletion (13q-14); report of two patients, one with a trisomic sib due to maternal insertion. Gene dosage effect for esterase-D. Hum Genet 59:211-214. Turleau C, Grouchy Jd, Chavin-Colin F, Despoisses S, Leblanc A, (1983): Two cases of del(13q)-retinoblastoma and two cases of partial trisomy due to a familial insertion. Ann Genet 26:158-160. Turleau C, Grouchy J, Chavin-Colin F, Junien C, Seger J, Schlienger P, Leblanc A, Haye C 11985): Cytogenetic forms of retinoblastoma: their incidence in a survey of 66 patients. Cancer Genet Cytogenet 16:321-334. Higgins MJ, Hansen MF, Cavenee WK, Lalande M (1989): Molecular detection of chromosomal translocations that disrupt the putative retinoblastoma susceptibility locus. Mol Cell Biol 9:1-5. Mitchell CD, Cowell JK (1989): Predisposition to retinoblastoma due to a translocation within the 4.7R locus. Oncogene 4:253-257. Blanquet V, Turleau C, Grouchy J, Creau-Goldberg N (1991): Physical map around the retinoblastoma gene: Possible genomic imprinting suggested by Nrul digestion. Genomics 10:350-355. Hida T, Kinoshita Y, Matsumoto R, Suzuki N, Tanaka H (1980): Bilateral retinoblastoma with 13qXp translocation. J Paediat Ophthalmol Strab 17:144-146. Nichols WW, Miller RC, Sobel M, Hoffman E (1980): Further observations on a 13qXp translocation associated with retinoblastoma. Am J Ophthalmol 89:621-627. Ejima Y, Sasaki MS, Kaneko A, Tanooka H, Hara Y, Hida T, Kinoshita Y (1982): Possible inactivation of part of chromosome 13 due to 13q xp translocation associated with retinoblastoma. Clin Genet 21:357-361. Cowell JK, Rutland P, Hungerford J, Jay M (1988): Deletion

G e n e t i c s a n d C y t o g e n e t i c s of R e t i n o b l a s t o m a

of chromosome region 13q14 is transmissible and does not always predispose to retinoblastoma. Hum Genet 80:43-45. 53. Wilson WG, Campochiaro PA, Conway BO, Carter BT, Sudduth KW, Watson A, Sparkes RS (1987): Deletion (13) (q14.1q14.3) in two generations: Variability of ocular manifestations and definition of the phenotype. Am J Med Genet 28:675-683. 54. Fukushima Y, Kuroki Y, Ito T, Kondo I, Nishigaki I (1987): Familial retinoblastoma (mother and son) with 13q14 deletion. Hum Genet 77:104-107. 55. Ejima Y, Sasaki MS, Kaneko A, Tanooka H (1988): Types, rates, origin and expressivity of chromosome mutations involving 13q14 in retinoblastoma patients. Hum Genet 79:118-123. 56. Keith CG, Webb GC (1985): Retinoblastoma and retinoma occurring in a child with a translocation and deletion of the long arm of chromosome 13. Arch Ophthalmo1103:941-944. 57. Lalande M, Dryja TP, Schreck RR, Shipley J, Flint A, Latt SA (1984): Isolation of human chromosome 13-specific DNA sequences cloned from flow sorted chromosomes and potentially linked to the retinoblastoma locus. Cancer Genet Cytogenet 13:283-295. 58. Dryja TP, Rapaport JM, Joyce JM, Petersen RA (1986): Molecular detection of deletions involving band q14 of chromosome 13 in retinoblastomas. Proc Natl Acad Sci USA 83: 7391-7394. 59. Friend SH, Bernards R, Rogelj S, Weinberg RA, Rapaport JM, Albert DM, Dryja TP (1986): A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma. Nature 323:643-646. 60. Lee W-H, Bookstein R, Hong F, Young L-J, Shew J-Y, Lee EYHP (1987): Human retinoblastoma susceptibility gene: cloning, identification, and sequence. Science 235:1394-1399. 61. Fung YT, Murphree AL, T'Ang A, Qian J, Hinrichs SH, Benedict WF (1987): Structural evidence for the authenticity of the human retinoblastoma gene. Science 236:1657-1661. 62. Goddard AD, Balakier H, Canton M, Dunn J, Squire J, Reyes E, Becker A, Phillips RA, Gallie BL (1988): Infrequent genomic rearrangement and normal expression of the putative Rbl gene in retinoblastoma tumours. Mo] Cell Biol 8:2082-2088. 63. Friend SH, Horowitz JM, Gerber MR, Wang X-F (1987): Deletions of a DNA sequence in retinoblastoma and mesenchymal tumors: organization of the sequence and its encoded protein. Proc Natl Acad Sci USA 84:9059-9063. 64. T'Ang A, Wu K-J, Hashimoto T, Liu W-Y, Takahashi R, Shi X-H, Mihara K, Zhang F-H, Chen YY, Du C, Qian J, Lin Y-G, Murphree AL, Qiu W-R, Thompson T, Benedict WF, Fung Y-KT (1989): Genomic organization of the human retinoblastoma gene. Oncogene 4:401-407. 65. McGee TL, Yandell DW,Dryja TP (1989): Structure and partial genomic sequence of the human retinoblastoma susceptibility gene. Gene 80:119-128. 66. Hong FD, Huang H-JS, To H, Young L-JS, Oro A, Bookstein R, Lee EY-HP, Lee W-H (1989): Structure of the human retinoblastoma gene. Proc Natl Acad Sci USA 86:5502-5506. 67. Ford GM, Gallie BL, Phillips RA, Becker AJ (1990): A physical map around the retinoblastoma gene. Genomics 6:284-292. 68. Sakai T, Ohtani N, McGee TL, Robbins PD, Dryja TP (1991): Oncogenic germ-line mutations in Spl and ATF sites in the human retinoblastoma gene. Nature 353:83-86. 69. Wiggs J, Nordenskjeld M, Yandell D, Rapaport J, Grondin V, Janson M, Werelius B, Petersen R, Craft A, Riedel K, Lieberfarb R, Walton D, Wilson W, Dryja TP (1988): Prediction of the risk of hereditary retinoblastoma using DNA polymorphisms within the retinoblastoma gene. New Eng J Med 318:151-157. 70. Cowell JK (1985): Tracking the cancer genes in paediatric

9

predisposition syndromes; opportunities for prenatal diagnosis. Cancer Surv 3:573-601. 71. Scheffer H, Meerman GJ, Kruise YCM, van den Berg AHM, Penninga DP, Tan KEWP, der Kinderen DJ, Buys CHCM (1989): Linkage analysis of families with hereditary retinoblastoma: Nonpenetrance of mutation, revealed by combined use of markers within and flanking the RB1 gene. Am J Hum Genet 45:252-260. 72. Onadim Z, Mitchell CD, Rutland PC, Buckle BG, Jay M, Hungerford JL, Harper K, Cowell JK (1990): Application of intragenic DNA probes in prenatal screening for retinoblastoma gene carriers in the United Kingdom. Arch Dis Child 65:651-656. 73. Mitchell CD, Nicolaides K, Kingston J, Hungerford J, Jay M, Cowell JK (1988): Prenatal exclusion of hereditary retinoblastoma. Lancet i:826. 74. Onadim Z, Hungerford J, Cowell JK (1992): Follow-up of retinoblastoma patients having prenatal and perinatal predictions for mutant gene carrier status using intragenic polymorphic probes from the RB1 gene. Br J Cancer 65:711-716. 75. McGee TL, Cowley GS, Yandell DW, Dryja TP (1990): Detection of the Xbal RFLP within the retinoblastoma locus by PCR. Nucleic Acids Res 18:207. 76. Bookstein R, Lai C-C, To H, Lee W-H (1990): PCR-based detection of a polymorphic BamHI site in intron 1 of the human retinoblastoma (RB) gene. Nucleic Acids Res 18:1666. 77. Yandell DW, Dryja TP (1989): Detection of DNA sequence polymorphisms by enzymatic amplification and direct genomic sequencing. Am J Hum Genet 1989:547-555. 78. Onadim Z, Cowell JK (1991): Application of PCR-amplification to DNA from paraffin-embedded tissue sections to linkage analysis in familial retinoblastoma. J Med Genet 28:312-316. 79. Grand RJA, Byrd PJ, Grabham PW, Gregory CD, Huen DS, Merrick RM, Young LS, Gallimore PH (1989): The expression of the retinoblastoma gene product Rbl in primary and adenovirus-transformed human cells. Oncogene 4:1291-1298. 80. Bernards R, Schackleford GM, Gerber MR, Horowitz JM, Friend SH, Schartl M, Bogenmann E, Rapaport JM, McGee T, Dryja TP, Weinberg RA (1989): Structure and expression of the murine retinoblastoma gene and characterisation of its encoded protein. Proc Natl Acad Sci USA 86:6474-6478. 81. Horowitz JM, Park S-H, Bogenmann E, Cheng J-C, Yandell DW, Kaye FJ, Minna JD, Dryja TP, Weinberg RA (1990): Frequent inactivation of the retinoblastoma anti-oncogene is restricted to a subset of human tumour cells. Proc Natl Acad Sci 87:101-107. 82. Huang HS, Yee J, Shew Y, Chen P, Bookstein R, Friedmann T, Lee EYP (1990): Suppression of the neoplastic phenotype by replacement of the RB gene in human cancer cells. Science 242:1563-1566. 83. Bookstein R, Rio P, Madreperla SA, Hong F, Allred C, Grizzle WE, Lee W-H (1990): Promotor deletion and loss of retinoblastoma gene expression in human prostate carcinoma. Proc Natl Acad Sci USA 87:7762-7766. 84. Sumegi J, Uzolgyi E, Klein G (1990): Expression of the RB gene under the control of MuLC-LTR suppresses tumorigenicity of WERI-Rb-27 retinoblastoma cells in immunodefective mice. Cell Growth Differentiation 1:247-249. 85. Madreperla SA, Whittum-Hudson JA, Predergast RA, Chen P-L, Lee W-H (1991): Intraocular tumor suppression of retinoblastoma gene-reconstituted retinoblastoma cells. Cancer Res 51:6381-6384. 86. Xu H-J, Sumegi J, Hu S-X, Banerjee A, Uzvolgyi E, Klein G, Benedict WF (1991): Intraocular tumor formation of RB reconstituted retinoblastoma cells. Cancer Res 51:4481-4485.

10

87. Muncaster MM, Cohen BL, Phillips RA, Gallie BL (1992): Failure of RB1 to reverse the malignant phenotype of human tumor cell lines. Cancer Res 5:654-661. 88. Lee WH, Shew JY, Hong FD, Sery TW, Donoso LA, Young LJ, Bookstein R, Lee YP (1987): The retinoblastoma susceptibility gene encodes a nuclear phosphoprotein associated with DNA binding activity. Nature 329:642-645. 89. Ludlow JW, DeCaprio JA, Huang CM, Lee WH, Paucha E, Livingston DM (1989): SV40 large T antigen binds preferentially to an underphosphorylated member of the retinoblastoma susceptibility gene product family. Cell 56:57-67. 90. Whyte P, Buchkovich K, Horowitz JM, Friend SH, Raybuck M, Weinberg RA, Harlow E (1988): Association between an oncogene and an anti-oncogene: The adenovirus EIA proteins bind to the retinoblastoma gene product. Nature 334:124-129. 91. Mihara K, Cao X-R, Yen A, Chandler S, Driscoll B, Murphree AL, T'Ang A, Fung Y-KT (1989): Cell cycle-dependent regulation of phosphorylation of the human retinoblastoma gene product. Science 246:1300-1303. 92. Chen P-L, Scully P, Shew Y-T, Wang JYT, Lee W-H (1989): Phosphorylation of the retinoblastoma gene product is modulated during the cell cycle and cellular differentiation. Cell 58:1193-1198. 93. DeCaprio JA, Ludlow JW, Lynch D, Furukawa Y, Griffin J, Piwnica-Worms H, Huang C-M, Livingston DM (1989): The product of the retinoblastoma susceptibility gene has properties of a cell cycle regulatory element. Cell 58:1085-1095. 94. Buchkovich K, Duffy LA, Harlow E (1989): The retinoblastoma protein is phosphorylated during specific phases of the cell cycle. Cell 58:1097-1105. 95. DeCaprio JA, Ludlow JW, Figge J, Shew J-Y, Huang C-M, Lee W-H, Marsilio E, Paucha E, Livingston DM (1988): SV40 large tumour antigen forms a specific complex with the product of the retinoblastoma susceptibility gene. Cell 54:275-283. 96. Dyson N, Howley PM, Munger K, Harlow E (1989): The human papilloma virus-16 E7 oncoprotein is able to bind to the retinoblastoma gene product. Science 243:934-936. 97. Whyte P, Williamson NM, Harlow E (1989): Cellular targets for transformation by the adenovirus EIA proteins. Cell 56:67-75. 98. Goodrich DW, Wang NP, Qian Y-W, Lee EY-HP, Lee W-H (1991): The retinoblastoma gene product regulates progression through the G1 phase of the cell cycle. Cell 67:293-302. 99. Hu Q, Dyson N, Harlow E (1990): The regions of the retinoblastoma protein needed for binding to adenovirus EIA or SV40 large T antigen are common sites for mutation. EMBO J 9:1147-1566. 100. Kaelin WGJ, Ewen ME, Livingston DM (1990): Definition of the minimal simian virus large T antigen- and adenovirus EIA-binding domain in the retinoblastoma gene product. Mol Cell Biol 10:3761-3769. 101. Huang S, Wang N-P, Tseng BY, Lee W-H, Lee EH-HP (1990): Two distinct and frequently mutated regions of the retinoblastoma protein are required for binding to SV40 T antigen. EMBO J 9:1815-1822. 102. Nevins R (1989): Mechanisms of viral-mediated trans-activation of transcription. Adv Virus Res 37:35-83. 103. Bagchi S, Weinmann R, Raychaudhuri P (1991): The retinoblastoma protein copurifies with E2F-1, and EIA-regulated inhibitor of the transcription factor E2F. Cell 65:1063-1072. 104. Kaelin WG, Pallas DC, DeCaprio JA, Kaye FJ, Livingston DM (1991): Identification of cellular proteins that can interact specifically with the T/EiA-binding region of the retinoblastoma gene product. Cell 64:521-532. 105. Bandara LR, La Thangue NB (1991): Adenovirus Ela prevents

J . K . C o w e l l a n d A. Hogg

106.

107.

108.

109.

110.

111.

112.

113.

114.

115. 116.

117.

118.

119.

120.

121.

122. 123.

124.

125.

the retinoblastoma gene product from complexing with a cellular transcription factor. Nature 351:494-497. Chellappan SP, Hiebert S, Mudryj M, Horowitz JM, Nevins JR (1991): The E2F transcription factor is a cellular target for the RB protein. Cell 65:1053-1061. Bandara LR, Adamczewski JP, Hunt T, La Thangue NB (1991): Cyclin A and the retinoblastoma gene product complex with a common transcription factor. Nature 352:249-251. Partridge JF, La Thangue NB (1991): A developmentally regulated and tissue-dependent transcription factor complexes with the retinoblastoma gene product. EMBO J 10: 3819-3827. Defoe-Jones D, Huang PS, Jones RE, Haskell KM, Vuocolo GA, Hanobik MG, Huber HE, Oliff A (1991): Cloning of cDNAs for cellular proteins that bind to the retinoblastoma gene product. Nature 352:251-254. Rustgi AK, Dyson N, Bernards R (1991): Amino-terminal domains of c-myc and N-myc proteins mediate binding to the retinoblastoma gene product. Nature 353:541-544. Robbins PD, Horowitz JM, Mulligan RC (1990): Negative regulation of human c-fos expression by the retinoblastoma gene product. Nature 346:668-671. Kim S-J, Lee H-D, Robbins PD, Busam K, Sporn MB, Roberts AB (1991): Regulation of transforming growth factor fll gene expression by the product of the retinoblastoma-susceptibility gene. Proc Natl Acad Sci USA 88:3052-3056. Laiho M, DeCaprio JA, Ludlow JW, Livingston DM, Massague J (1990): Growth inhibition by TGF-fl linked to suppression of retinoblastoma protein phosphorylation. Cell 62:175-185. Ong G, Sikora K, Gullick WJ (1991): Inactivation of the retinoblastoma gene does not lead to loss of TGF-/3 in breast cancer cell lines. Oncogene 6:761-763. Hunter T, Pines J (1991): Cyclins and cancer. Cell 66: 1071-1074. Lin BY-T, Gruenwald S, Morla AO, Lee W-H, Wang JYJ (1991): Retinoblastoma cancer suppressor gene product is a substrate of the cell cycle regulator cdc2 kinase. EMBO J 10:857-864. Cao L, Faha B, Dembski M, Tsai L-H, Harlow E, Dyson N (1992): Independent binding of the retinoblastoma protein and p107 to the transcription factor E2F. Nature 355:176-179. Mudryj M, Devoto SH, Hiebert SW, Hunter T, Pines J, Nevins JR (1991): Cell cycle regulation of the E2F transcription factor involves an interaction with cyclin A. Cell 65:1243-1253. Faha B, Ewen ME, Tsai L-H, Livingston DM, Harlow E (1992): Interaction between human cyclin A and adenovirus EIAassociated p107 protein. Science 255:87-90. Shirodkar S, Ewen M, DeCaprio JA, Morgan J, Livingston DM, Chittenden T (1992): The transcription factor E2F interacts with the retinoblastoma product and a plO7-cyclin A complex in a cell cycle-regulated manner. Cell 68:157-166. Ewen ME, Xing Y, Lawrence JB, Livingston DM (1991): Molecular cloning, chromosomal mapping and expression of the cDNA for p107, a retinoblastoma gene product-related protein. Cell 66:1155-1164. Kitchin FD, Ellsworth RM (1974): Pleiotropic effects of the gene for retinoblastoma. J Med Genet 11:244. Abramson DH, Ellsworth RM, Kitchin DF, Tung G (1984): Second nonocular tumors in retinoblastoma survivors, are they radiation-induced? Ophthalmology 91:1351-1355. Draper GJ, Sanders BM, Kingston JE (1986): Second primary neoplasms in patients with retinoblastoma. Br J Cancer 53:661-671. Sanders BM, Jay M, Draper GJ, Roberts EM (1989): Non-ocular

G e n e t i c s a n d C y t o g e n e t i c s of R e t i n o b l a s t o m a

126.

127.

128.

129.

130.

131.

132.

133.

134.

135.

136.

137.

138.

139.

140.

cancer in relatives of retinoblastoma patients. Br J Cancer 60:358-365. Toguchida J, Ishizaki K, Sasaki MS, Ikenaga M, Sugimoto M, Kotoura Y, Yamamuro T (1988): Chromosomal reorganization for the expression of recessive mutation of retinoblastoma susceptibility gene in the development of osteosarcoma. Cancer Res 48:3939-3943. Shew J-Y, Ling N, Yang Z, Fodstad O, Lee W-H (1989): Antibodies detecting abnormalities of the retinoblastoma susceptibility gene product (pp110rb) in osteosarcomas. Oncogene 1:205-214. Reissmann PT, Simon MA, Lee W-H Slamon DJ (1989): Studies of the retinoblastoma gene in human sarcomas. Oncogene 4:839-843. Lee EYP, To H, Shew J-H, Bookstein R, Scully P, Lee W-H (1988): Inactivation of the retinoblastoma susceptibility gene in human breast cancers. Science 241:218-221. Varley JM, Armour J, Swallow JE, Jeffreys AJ, Ponder BAJ, T 'Ang AT, Fung Y-K, Brammer WJ, Walker RA (1989): The retinoblastoma gene is frequently altered leading to loss of expression in primary breast tumours. Oncogene 4:725729. Harbour JW, Lai S-L, Whang-Peng J, Gazdar AF, Minna J, Kaye FJ (1988): Abnormalities in structure and expression of the human retinoblastoma gene in SCLC. Science 241: 353-357. Yokota J, Aiyama T, Fung Y-KT, Benedict WF, Namba Y, Hanaoka M, Wada M, Terasaki T, Shimosato Y, Sugimura T, Terada M (1988): Altered expression of the retinoblastoma (RB) gene in small-cell carcinoma of the lung. Oncogene 3:471-475. Hensel CH, Hsieh C-L, Gazdar AF, Johnson BE, Sakaguchi AY, Naylor SL, Lee W-H, Lee EY-HL (1990): Altered structure and expression of the human retinoblastoma susceptibility gene in small cell lung cancer. Cancer Res 50:30673067. Horowitz JM, Yandell DW, Park S-H, Canning S, Whyte P, Buchkovich K, Harlow E, Weinberg RA, Dryja TP (1989): Point mutational inactivation of the retinoblastoma antioncogene. Science 243:937-940. Ishikawa J, Xu H-j, Hu S-X, Yandell DW, Maeda S, Kamidono S, Benedict WF, Takahashi R (1991): Inactivation of the retinoblastoma gene in human bladder and renal cell carcinomas. Cancer Res 51:5736-5743. Cairns P, Proctor AJ, Knowles MA (1991): Loss of heterozygosity at the RB locus is frequent and correlates with muscle invasion in bladder carcinoma. Oncogene 6:2305-2309. Boynton RF, Huang, Y, Blount PL, Reid BJ, Raskind WH, Haggitt RC, Newkirk C, Resau JH, Yin J, McDaniel T, Meltzer SJ (1991): Frequent loss of heterozygosity at the retinoblastoma locus in human esophageal cancers. Cancer Res 51: 5766-5769. Murakami Y, Hayashi K, Hirohashi S, Sekiya T (1991): Aberrations of the tumor suppressor p53 and retinoblastoma genes in human hepatocellular carcinomas. Cancer Res 51:5520-5525. Furukawa Y, DeCaprio JA, Belvin M, Griffin JD (1991): Heterogeneous expression of the product of the retinoblastoma susceptibility gene in primary human leukaemia cells. Oncogene 6:1343-1346. Bader JL, Meadows AT, Zimmerman LE, Rorke LB, Voute PA,

11

141.

142.

143.

144.

145.

146.

147.

148.

149.

150.

151.

152.

153.

154.

155.

Champion LA, Miller RW (1982): Bilateral retinoblastoma with ectopic intracranial retinoblastoma: trilateral retinoblastoma. Cancer Genet Cytogenet 5:203-213. Holladay DA, Montebello JF, Redmond KP (1991): Clinical presentation, treatment, and outcome of trilateral retinoblastoma. Cancer 67:710-715. Canning S, Dryja TP (1989): Short direct repeats at the breakpoints of deletions of the retinoblastoma gene. Proc Natl Acad Sci USA 86:5044-5048. Kloss K, Wahrisch P, Greger V, Messmer E, Fritze H, Hopping W, Passarge E, Hortshemke B (1991): Characterization of deletions at the retinoblastoma locus in patients with bilateral retinoblastoma. Am J Med Genet 39:196-200. Hashimoto T, Takahashi R, Yandell DW, Xu H-J, Hu S-X, Gunnell S, Benedict WF (1991): Characterization of intragenic deletions in two sporadic germinal mutation cases of retinoblastoma resulting in abnormal gene expression. Oncogene 6:463-469. Dunn JM, Phillips RA, Becker A, Gallie BL (1988): Identification of germline and somatic mutations affecting the retinoblastoma gene. Science 241:1797-1800. Dunn JM, Phillips RA, Zhu X, Becker A, Gallie BL (1989): Mutations in the RB1 gene and their effects on transcription. Mol Cell Biol 9:4596-4604. Yandell DW, Campbell TA, Dayton SH, Petersen R, Walton D, Little JB, McConkie-Rosell A, Buckley EG, Dryja TP (1989): Oncogenic point mutations in the human retinoblastoma gene: their application to genetic counselling. New Engl J Med 321:1689-1695. Hogg A, Onadim Z, Baird PN, Cowell JK (1992): Detection of heterozygous mutations in the RB1 gene in retinoblastoma patients using single strand conformation polymorphism (SSCP) analysis and PCR sequencing. Oncogene 7:14451451. Mori N, Yokata J, Akiyama T, Sameshima Y, Okamoto A, Mizoguchi H, Toyoshima K, Sugimura T, Terada M (1990): Variable mutations of the RB gene in small-cell lung carcinoma. Oncogene 5:1713-1717. Orita M, Suzuki Y, Sekiya T, Hayashi K (1989): Rapid and sensitive detection of point mutations and DNA polymorphisms using the polymerase chain reaction. Genomics 5:874-879. Weir-Thompson E, Condie A, Leonard RCF, Prosser J (1991): A familial RB1 mutation detected by the HOT technique is homozygous in a second primary neoplasm. Oncogene 6:2353-2356. Bookstein R, Shew J-Y, Chen P-L, Scully P, Lee W-H (1990): Suppression of tumorigencity of human prostate carcinoma cells by replacing a mutated RB gene. Science 247:712-715. Onadim Z, Hogg A, Baird PN, Cowell JK. (1992). Oncogenic point mutations in exon 20 of the RB1 gene in families showing incomplete penetrance and mild expression of the retinoblastoma phenotype. Proc. Nat. Acad. Sci. USA. 89: 6177-6181. Chittenden T, Livingston DM, Kaelin WG (1991): The T/EIAbinding domain of the retinoblastoma product can interact selectively with a sequence specific DNA-binding protein. Cell 65:1073-1082. Mittnacht S, Weinberg RA (1991): G1/S phosphorylation of the retinoblastoma protein is associated with an altered affinity for the nuclear compartment. Cell 65:381-383.

Genetics and cytogenetics of retinoblastoma.

LEAD ARTICLE Genetics and Cytogenetics of Retinoblastoma John K. Cowell and Annette Hogg INTRODUCTION Cancer cells are referred to as " i m m o r t a...
1MB Sizes 0 Downloads 0 Views