97
A Subset of Retinoblastoma Lacking RB1 Gene Mutations have High-level MYCN Gene Amplification by Stephanie Yee A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Molecular Genetics University of Toronto © Copyright by Stephanie Yee 2010

A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

  • Upload
    others

  • View
    8

  • Download
    0

Embed Size (px)

Citation preview

Page 1: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

A Subset of Retinoblastoma Lacking RB1 Gene Mutations have High-level MYCN Gene Amplification

by

Stephanie Yee

A thesis submitted in conformity with the requirements for the degree of Master of Science

Department of Molecular Genetics University of Toronto

© Copyright by Stephanie Yee 2010

Page 2: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

ii

A Subset of Retinoblastoma Lacking RB1 Gene Mutations With

High-Level MYCN Gene Amplification

Stephanie Yee

Master of Science

Department of Molecular Genetics

University of Toronto

2010

Abstract

Retinoblastoma is the prototype genetic cancer caused by mutations disrupting the RB1 tumor

suppressor gene. Following loss of RB1, retinoblastoma acquires further genetic changes in a

characteristic set of oncogenes and tumor suppressors including gains of the oncogenes KIF14,

DEK, E2F3, and MYCN and loss of the tumor suppressor CDH11. The constellation of genetic

changes is the postulated genetic pathway leading to retinoblastoma. However, advances in

molecular diagnostic testing for RB1 gene mutations allows detection of at least one RB1

mutation in 98% of unilateral retinoblastomas leaving 2% of cases with undetectable RB1

mutations (RB1+/+

retinoblastoma). RB1+/+

retinoblastomas have high-level MYCN gene

amplification (>30 copies) and few other genetic changes. In addition, RB1+/+

retinoblastoma

present earlier than conventional RB1-/-

retinoblastoma and show histologic features similar to

MYCN-amplified neuroblastoma. Altogether, this study describes a distinct genetic subset of

retinoblastoma characterized by wild-type RB1 gene and high-level MYCN gene amplification.

Page 3: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

iii

Acknowledgments

I would like to express my sincerest gratitude to my supervisor Dr. Brenda L. Gallie for her

guidance, encouragement and support throughout the project. I am grateful to Dr. Sanja Pajovic

who has been a mentor to me and has provided assistance in numerous ways throughout this

thesis. I thank Clarellen Spencer for her technical assistance in the laboratory. I thank my

laboratory colleagues Dr. Ying Guo, Dr. Ghada Kurban, Dr. Brigitte Theriault, Tim To, Christine

Yurkowski and Dr. Helen Dimaras for their friendship and support and for making my time in

the lab a memorable experience.

I would like to thank Diane Rushlow, Jennifer Kennett, Dr. Paul Boutros and Anthony Mak for

their technical and intellectual contributions.

The generous support from the Vision Science Research Program Graduate Student Scholarship

is greatly appreciated.

Last but not least, I thank the people in my life who have given me years of unwavering love and

support; my parents Kim Hook Yee and Beng Cheng Yee, my sister Sylvia Yee and Klint

Ramdass.

Page 4: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

iv

Table of Contents

Acknowledgments .......................................................................................................................... iii

Table of Contents ........................................................................................................................... iv

List of Tables ................................................................................................................................ vii

List of Figures .............................................................................................................................. viii

List of Appendices ......................................................................................................................... ix

List of Abbreviations ...................................................................................................................... x

Chapter 1 ......................................................................................................................................... 1

1 Introduction ................................................................................................................................ 1

1.1 Retinoblastoma ................................................................................................................... 1

1.2 Current Retinoblastoma Treatment ..................................................................................... 2

1.3 Molecular function of pRB ................................................................................................. 2

1.4 Inactivation of pRB ............................................................................................................. 3

1.4.1 RB1 gene mutations ................................................................................................ 3

1.4.2 Inactivation of pRB or RB pathway members ........................................................ 4

1.5 Retinal Development .......................................................................................................... 5

1.6 Genomic changes in retinoblastoma ................................................................................... 5

1.6.1 1q Gain .................................................................................................................... 6

1.6.2 6p Gain .................................................................................................................... 7

1.6.3 16q Loss .................................................................................................................. 8

1.6.4 2p Gain .................................................................................................................... 8

1.7 MYCN amplification in neuronal tumors .......................................................................... 10

1.8 Genomic changes in MYCN-amplified neuroblastomas ................................................... 10

1.9 MYCN amplicon ................................................................................................................ 12

1.10 MYCN gene structure and expression ............................................................................... 13

Page 5: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

v

1.11 MYCN protein and functions ............................................................................................ 14

1.11.1 MYCN protein ...................................................................................................... 14

1.12 MYCN amplification in transgenic murine model of neuroblastoma ................................ 16

Chapter 2 ....................................................................................................................................... 18

2 Characterization of RB1+/+

retinoblastoma .............................................................................. 18

2.1 Introduction ....................................................................................................................... 18

2.2 Hypothesis ......................................................................................................................... 19

2.3 Thesis Aims and Rationale ............................................................................................... 19

2.3.1 Frequency of RB1+/+

retinoblastoma .................................................................... 19

2.3.2 Characterize genomic profile of RB1+/+

retinoblastomas ..................................... 20

2.3.3 Determine mRNA and protein levels of RB1 and MYCN genes ........................... 20

2.3.4 Analysis of clinical and pathological features of RB1+/+

retinoblastomas with

MYCN amplification ............................................................................................. 20

2.3.5 Determine the effect of MYCN silencing in MYCN-amplified retinoblastoma ..... 20

2.3.6 Designing a Mycn-overexpressing lentivirus ........................................................ 20

2.4 Materials and Methods ...................................................................................................... 21

2.4.1 Samples ................................................................................................................. 21

2.4.2 RB1 gene mutation testing .................................................................................... 21

2.4.3 Gene-specific QM-PCR ........................................................................................ 21

2.4.4 Sub-megabase resolution tiling array comparative genomic hybridization .......... 23

2.4.5 SMRT aCGH data analysis ................................................................................... 24

2.4.6 Statistics ................................................................................................................ 24

2.4.7 RT-PCR ................................................................................................................. 25

2.4.8 Immunohistochemistry ......................................................................................... 25

2.4.9 Lentivirus production ............................................................................................ 26

2.4.10 Lentivirus titration ................................................................................................ 26

Page 6: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

vi

2.4.11 Proliferation assay ................................................................................................. 27

2.4.12 Construction of Mycn-overexpression lentivirus .................................................. 27

2.4.13 Transduction of retinal explants ............................................................................ 27

2.4.14 Western blot analysis ............................................................................................ 28

2.5 Results ............................................................................................................................... 29

2.5.1 Frequency of RB1+/+

retinoblastoma across four independent sites ..................... 29

2.5.2 Genomic profile of RB1+/+

retinoblastomas ......................................................... 29

2.5.3 Expression of RB1 and MYCN mRNA transcripts and protein in RB1+/+

retinoblastomas ..................................................................................................... 41

2.5.4 Clinical features of RB1+/+

retinoblastomas ......................................................... 42

2.5.5 Functional consequence of MYCN silencing in retinoblastoma with high levels

of MYCN .............................................................................................................. 45

2.5.6 Construction of a Mycn-overexpression lentivirus ............................................... 46

Chapter 3 ....................................................................................................................................... 48

3 Discussion ................................................................................................................................ 48

3.1.1 RB1+/+

MYCNA retinoblastoma is observed in independent clinical samples ....... 48

3.1.2 RB1+/+

MYCNA: a novel genetic subset of retinoblastoma .................................... 48

3.1.3 MYCN-driven tumorigenesis ................................................................................. 49

3.1.4 Chromosome 8;13 translocation ........................................................................... 50

3.1.5 MYCN copy number as a rapid screen for RB1+/+

MYCNA

retinoblastoma ........... 51

3.1.6 Targeting MYCN ................................................................................................... 51

3.1.7 Future directions ................................................................................................... 52

References ..................................................................................................................................... 55

Appendices .................................................................................................................................... 67

Page 7: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

vii

List of Tables

Table 1 Samples used for QM-PCR .............................................................................................. 23

Table 2 List of primer sequences and expected product sizes used in RT-PCR analysis ............. 25

Table 3 Frequency of RB1+/+

retinoblastomas across four sites ................................................... 29

Table 4 Frequencies of M3-Mn changes in RB1+/+

versus RB1-/-

retinoblastomas ....................... 32

Table 5 Summary of retinoblastoma histopathological features in RB1+/+

MYCNA retinoblastomas

....................................................................................................................................................... 44

Page 8: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

viii

List of Figures

Figure 1 M3-Mn profile of M3-Mn copy number in 139 primary retinoblastomas and 6 cell lines

....................................................................................................................................................... 31

Figure 2 Summary of chromosomal changes for 47 primary retinoblastomas, 5 retinoblastoma

cell lines and 1 neuroblastoma cell line, IMR32 ........................................................................... 33

Figure 3 Number of CNAs per retinoblastoma tumors ................................................................. 34

Figure 4 Number of aberrant base pairs in the different subtypes of retinoblastoma ................... 35

Figure 5 Whole genome tiling path array CGH karyogram of RB1+/+

MYCNA retinoblastoma

FA793 ........................................................................................................................................... 36

Figure 6 Specific amplification of the MYCN locus in RB1+/+

MYCNA RB1348 ......................... 37

Figure 7 The minimal MYCN amplicon ........................................................................................ 39

Figure 8 RB381 der(8)t(8;13)(q21.2;q14.12) ins(13;8)(q14; q21.2-q23.3) translocation ............ 40

Figure 9 Expression of pRB and MYCN in primary human retinoblastoma and normal retina .. 42

Figure 10 Age of diagnosis of 11 RB1+/+

MYCNA retinoblastomas .............................................. 43

Figure 11 Large prominent nucleoli in two RB1+/+

MYCN A retinoblastomas, RB2237 and

NZ499J .......................................................................................................................................... 45

Figure 12 MYCN shRNA knockdown in Y79 retinoblastoma cells ............................................. 46

Figure 13 Lentiviral overexpression of Mycn in HEK293 cells ................................................... 47

Page 9: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

ix

List of Appendices

Table 6 Copy numbers of M3-Mn genes in retinoblastomas as measured by QM-PCR ............. 67

Table 7 SMRT aCGH alterations by sample ................................................................................ 71

Page 10: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

x

List of Abbreviations

aCGH Array comparative genomic hybridization

ACVRL-1 Activin A receptor type II-like 1

ALK Anaplastic lymphoma kinase

ANOVA Analysis of variance

ATF Activating transcription factor 1

BAC Bacterial artificial chromosome

bHLH Basic helix-loop-helix

BIM Bcl-2 interacting mediator of cell death

Bp Base pairs

BSA Bovine serum albumin

CAN NUP214, nucleoporin 214kDa

CASP9 Caspase 9, apoptosis-related cysteine peptidase

CDH8 Cadherin 8, type 2

CDH11 Cadherin 11, type 2, OB-cadherin (osteoblast)

CDH13 Cadherin 13, H-cadherin (heart)

Cdk Cyclin-dependent kinase

cDNA Complementary DNA

CGH comparative genomic hybridization

Page 11: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

xi

ChIP Chromatin immunoprecipitation

CMV Cytomegalovirus

CNA Copy number alteration

CNV Copy number variation

CpG Cytosine next to Guanine

Cy3 Cyanine 3

Cy5 Cyanine 5

DAB 3,3´-diaminodbenzidine

DAPI 4’, 6-diamidino-2-phenylindole

dCTP deoxycytidine triphosphate

DDX1 DEAD (Asp-Glu-Ala-Asp) box polypeptide 1

DEK DEK oncogene (DNA binding)

DFFA DNA fragmentation factor, 45kDa, alpha polypeptide

DFMO alpha-difluoromethylornithine

DM Double minute

DMEM Dulbecco’s modified Eagle's medium

DNA Deoxyribonucleic acid

E2F E2F transcription factor

ECL Electrochemiluminescence

EDTA Ethylenediaminetetraacetic acid

Page 12: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

xii

EGFP Enhanced green fluorescence protein

ESD Esterase D

ETAA16 Ewing tumor-associated antigen 16

Ets V-ets erythroblastosis virus E26 oncogene homolog 1

FAM49A Family with sequence similarity 49, member A

FAM84A Family with sequence similarity 84, member A

FBS Fetal bovine serum

GSK3 Glycogen synthase kinase 3

H3 Histone cluster 3, H3

HEK293 Human embryonic kidney 293

HEK293T Human embryonic kidney 293 SV40 large T-antigen

HLH Helix-loop-helix

HPV Human papilloma virus

HSR Homogeneously staining region

ID2 Inhibitor of DNA binding 2, dominant negative helix-loop-helix protein

IE1 Intermediate early 1

INL Inner nuclear layer

IRES Internal ribosomal entry site

kb Kilobase pairs

kDa Kilodalton

Page 13: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

xiii

Ki67 Antigen identified by monoclonal antibody Ki-67

KIF14 Kinesin family member 14

M1 Mutation1

M2 Mutation 2

M3-Mn Mutation 3-n

Mad MAX dimerization protein 1

MAP Small G protein signaling modulator 3

MAX MYC associated factor X

Mb Megabase pairs

mCMV Murine cytomegalovirus

MDM2 Mdm2 p53 binding protein homolog

MEIS Meis homeobox

miR microRNA

MLPA Multiplex ligation-dependent probe amplification

MRG Minimal region of gain

mRNA Messenger ribonucleic acid

Mnt MAX binding protein

Mxi MAX interactor 1

Myc v-myc myelocytomatosis viral oncogene homolog (avian)

MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma derived (avian)

Page 14: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

xiv

MYCNA High-level MYCN amplification (>10 copies)

MYCNOS N-myc opposite strand

NAG Neuroblastoma amplified sequence

NAHR Nonallelic homologous recombination

ODC1 Ornithine decarboxylase 1

ONL Outer nuclear layer

P0 Postnatal day 0

p19ARF

cyclin-dependent kinase inhibitor 2A (melanoma, p16, inhibits CDK4), alternate

reading frame

p21 cyclin-dependent kinase inhibitor 1A

p27 cyclin-dependent kinase inhibitor 1B

p53 Tumor protein p53

p107 Retinoblastoma-like 1

p130 Retinoblastoma-like 2

Pax Paired-box 1

PBS Phosphate-buffered saline

PCAN1 Gene differentially expressed in prostate

PRC1 Protein-regulating cytokinesis 1

pRB Retinoblastoma protein

PP1 Protein phosphatase 1

PCR Polymerase chain reaction

Page 15: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

xv

PNA Peptide nucleic acid

QM-PCR Quantitative multiplex-polymerase chain reaction

RB Retinoblastoma

RB1 Retinoblastoma 1

REXOIL1 REX1, RNA exonuclease 1 homolog (S. cerevisiae)-like 1

RIPA Radioimmunoprecipitation assay buffer

RNA ribonucleic acid

RPE Retinal pigment epithelium

rRNA Ribosomal RNA

RT-PCR Reverse transcriptase PCR

S phase Synthesis phase

SDS Sodium dodecyl sulfate

shRNA Short hairpin RNA

siRNA Small interfering RNA

SMRT Sub-megabase resolution tiling

SP1 SP1 transcription factor

SSC Saline sodium citrate

SSTR2 Somatostatin receptor 2

TAg SV40 large T antigen

TBP TATA binding protein

Page 16: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

xvi

TBS Tris-buffered saline

TH Tyrosine hydroxylase

tRNA Transfer RNA

UL97 Tegument serine/threonine protein kinase

Wnt Wingless-type MMTV integration site family

WPRE Woodchuck hepatitis post-transcriptional regulatory element

Page 17: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

1

Chapter 1

1 Introduction

1.1 Retinoblastoma

Retinoblastoma is a childhood cancer of the eye that affects approximately 1:18000 live birth

children (Devesa 1975). In the two-hit hypothesis, Alfred Knudson correctly postulated that

retinoblastoma is caused by at least two mutational events (Knudson 1971). The first clues to the

location of the mutated gene came from studies of chromosome 13q deletion syndrome in which

affected children presented with retinoblastoma along with developmental defects (Lele, Penrose

et al. 1963; Grace, Drennan et al. 1971; Wilson, Towner et al. 1973). Linkage between

retinoblastoma and the esterase D gene (ESD) narrowed the search to chromosomal band 13q14

(Sparkes, Sparkes et al. 1980; Connolly, Payne et al. 1983). Restriction endonuclease mapping

identified different restriction fragment length polymorphisms of DNA isolated from

chromosome 13 (Cavenee, Dryja et al. 1983; Dryja, Rapaport et al. 1986). This was followed by

cloning of a DNA fragment present in many tumor types but missing in retinoblastomas and

osteosarcomas which led to the discovery of the gene we now know as RB1 (Friend, Bernards et

al. 1986; Lee, Bookstein et al. 1987).

Retinoblastoma can present either in one eye (unilateral) or both eyes (bilateral). About 60% of

patients are unilaterally affected with sporadic disease i.e. no family history. About 40% of

patients are bilaterally affected, often with multifocal tumors in both eyes. In general, bilateral

patients are diagnosed earlier than unilaterally affected patients with median ages of 11 and 22

months respectively. In the unilateral form of the disease, two mutations to RB1 occur in a

susceptible retinal cell. In the bilateral form of the disease, one mutation is either inherited or

occurs de novo in a germ cell and the second mutation is acquired in the somatic retinal cell.

Individuals with a germline RB1 mutation have a lifetime susceptibility to second primary

tumors such as osteosarcoma (Matsunaga 1980; Draper, Sanders et al. 1986; Marees, Moll et al.

2008).

Page 18: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

2

1.2 Current Retinoblastoma Treatment

Since retinoblastoma can be a fatal disease if metastasis occurs, the first goal of treatment is to

save the patient’s life and secondly to salvage vision. In developed countries it is often detected

early and the survival rate is > 95% (Chintagumpala, Chevez-Barrios et al. 2007). Treatment

given depends on whether one or both eyes are affected. In unilateral cases, the patient is often

cured by enucleation or removal of the affected eye. In bilateral cases, a variety of treatments

are available, including enucleation, external beam therapy, cryotherapy, laser photocoagulation,

thermotherapy, brachytherapy and systemic chemotherapy (Lin and O'Brien 2009). Although

current treatments are successful in curing retinoblastoma, many patients experience serious side

effects from chemotherapy and radiation therapy. In cases where external beam radiation is

used, there is a significant increased risk of secondary tumors later in life. There remains a need

for treatments with less toxicity and this might be achieved through targeting specific molecular

targets. The oncogene MYCN is gained in 16% of primary retinoblastomas (Bowles, Corson et

al. 2007). Tonelli et al developed a peptide that specifically targeted the MYCN transcript and

found that growth in neuroblastoma cells could be inhibited (Tonelli, Purgato et al. 2005). Thus,

there is potential to direct therapies that specifically inhibit MYCN-driven tumorigenesis. Though

developing targeted therapies specific for secondary genomic changes may treat only a small

subset of retinoblastoma patients, it may be one way to reduce side effects associated with

general systemic therapies given today.

1.3 Molecular function of pRB

The RB1 gene was the first tumor suppressor to be discovered. It is a principal regulator of the

cell cycle and also has roles in differentiation, apoptosis and senescence (van den Heuvel and

Dyson 2008). pRB, along with the proteins p107 and p130, belong to a family of proteins

containing a pocket domain. Together they work at different times in the cell cycle to coordinate

the expression of S phase genes by binding to different targets, most notably the E2Fs. There are

at least 8 different E2Fs in the mouse. E2F1, E2F2 and E2F3a are activators of gene

transcription and E2F3b, E2F4 and E2F5 repress transcription by recruitment of chromatin

modifying enzymes. The E2F C-terminal domain mediates binding to the pocket domains of the

pRB family proteins. Each pRB family protein interacts with different subsets of E2Fs, and they

have overlapping but unique roles in cell cycle control. For example, pRB mainly associates

Page 19: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

3

with E2F1, E2F2, and E2F3a. When cyclin dependent kinases phosphorylate pRB, a

conformational shift in pRB results in the release of the E2Fs allowing them to bind to promoters

of S phase genes. The functional inactivation of pRB due to mutation or inactivation by binding

to cellular or viral proteins can result in uncontrolled cell cycling, failed differentiation and

apoptosis.

1.4 Inactivation of pRB

1.4.1 RB1 gene mutations

The RB1 gene is located on chromosome 13q14.2 and is composed of 27 exons distributed along

183 kb of genomic sequence. At its 5’ end, lies a promoter with a CpG island that is normally

unmethylated. The promoter has sequences recognized by transcription factors such as Sp1, ATF

but does not contain TATA or CAAT motifs. In addition, the RB1 promoter has an E-box

(Martelli, Cenciarelli et al. 1994) which can be recognized by the Myc family of transcription

factors. Patients who are heterozygous at the RB1 locus can develop tumors in one eye or both

eyes (variable expressivity) and in rare cases none at all (reduced penetrance) since it is due to

chance that a second mutation will occur in the other normal allele (Lohmann and Gallie 2004).

However, not all of the phenotypic variations can be accounted for by stochastic effects and it is

now known that the penetrance and phenotypes vary in part due to the nature of the predisposing

mutation (Lohmann and Gallie 2004). The RB1 gene does not contain any hot spots for

mutations and all classes of mutations can be detected in retinoblastomas and are distributed

throughout the gene (Richter, Vandezande et al. 2003). The majority of germline mutations are

null mutations which include whole gene and exonic gene deletions, splice mutations and

nonsense mutations (Richter, Vandezande et al. 2003). Nonsense mutations make up the

majority of the null mutations in both bilateral and unilateral tumors (Richter, Vandezande et al.

2003). In some genetic diseases, the phenotype varies depending on the location of the stop

codon, however, in retinoblastoma nonsense mutations result in no transcript being produced,

presumably by nonsense mediated decay (Frischmeyer and Dietz 1999; Wen and Brogna 2008).

In a heterozygous cell, this results in only transcripts produced by the normal allele. Aberrant

splicing is the second most important class of mutations and is caused by point mutations

affecting splice acceptor or donor sequences in intronic and exonic sequences. Splice mutations

in set splice sites can lead to premature stop codons or exon skipping resulting in complete

Page 20: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

4

penetrance. However, splice mutations in less conserved sequences are more likely to result in

incomplete penetrance (Lohmann and Gallie 2004). Missense mutations are in-frame changes to

nucleotide sequence that result in substitution of an amino acid residue. The majority of

missense mutations (81%) occur in the A/B “pocket” domain (Richter, Vandezande et al. 2003)

essential for interaction of pRB with E2F transcription factors (DiCiommo, Gallie et al. 2000).

Missense mutations often result in incomplete penetrance because some mutant alleles retain

partial activity (Otterson, Chen et al. 1997).

Richter et al developed a highly sensitive set of molecular tests to determine RB1 gene mutations

(Richter, Vandezande et al. 2003). The tests consists of sequencing of all 27 exons and

promoter, quantitative-multiplex PCR (QM-PCR) to detect gains or deletions in the promoter

and exons, promoter methylation assay and allele-specific PCR to detect 11 point mutations that

recur with significant frequency (Richter, Vandezande et al. 2003). Using this method, both

mutations can be identified in >95% of bilateral tumors and at least one mutation can be

identified in >98% (94%, both mutations identified and 4.8% one mutation identified) of

unilateral tumors leaving 1.6% of unilateral retinoblastoma with no evidence of mutations

(Rushlow, Piovesan et al. 2009).

1.4.2 Inactivation of pRB or RB pathway members

Aside from mutations to the RB1 gene sequence, there are many ways to inactivate the wild-type

protein, pRB. The holoenzyme protein phosphatase 1, PP1, binds to and dephosphorylates pRB

restoring its cell cycle negative regulatory function at mitotic exit (Vietri, Bianchi et al. 2006).

The loss of any of the catalytic subunits of PP1 could result in the deactivation of pRB.

Likewise, overexpression of proteins that phosphorylate and inactivate pRB such as cyclin

D/cdk4/6 and cyclin E/cdk2 could lead to absence of pRB function. When overexpressed the

cellular protein, inhibitor of differentiation 2, ID2, can inhibit pRB by binding to and

sequestering pRB away from its normal binding partners (Iavarone, Garg et al. 1994). Genomic

gain or overexpression of genes suppressed by pRB such as E2F transcription factors can

ultimately lead to progression through the cell cycle and unrestrained proliferation.

Viral proteins such as human papillomavirus (HPV) E7 and human cytomegalovirus UL97 can

bind and destabilize or hyperphosphorylate pRB (Kamil, Hume et al. 2009). However, a recent

screen was performed to look for the presence of several pRB-inactivating DNA tumor viruses

Page 21: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

5

which included subtypes of HPV, adenovirus and others in 40 RB1-/-

retinoblastomas but could

not find any evidence of viral sequences in the tumors (Gillison, Chen et al. 2007). Hence,

viruses may not play a role in the development of retinoblastoma.

1.5 Retinal Development

Eye development begins in the 18-day embryo with outpocketing of the forming neural tube to

form two optic grooves on either side. The optic grooves grow larger to become optic vesicles

which make contact with the surface ectoderm. Together the surface ectoderm and the optic

vesicle invaginate to form the lens placode and optic cup respectively. The lens placode fuses

with itself separating from the surface ectoderm to become the lens vesicle and later the lens.

The optic cup has two layers separated by a lumen, called the intraretinal space. The outer layer

gives rise to the retinal pigmented epithelia (RPE) and the inner layer gives rise to the neural

retina (Chow and Lang 2001). Starting from the layer bordering the intraretinal space, the neural

retina gives rise to the rod and cone photoreceptors whose cell bodies make up the outer nuclear

layer (ONL) followed by bipolar, horizontal, amacrine, and Müller cells which make up the inner

nuclear layer (INL) (Dyer and Cepko 2001). Internal to INL is the ganglion cell layer which also

includes some amacrine cells.

1.6 Genomic changes in retinoblastoma

Following loss of RB1 a specific set of genomic losses and gains drives benign non-proliferative

retinomas into malignant retinoblastomas (Dimaras, Khetan et al. 2008). Early cytogenetic

studies identified gains of chromosome 1q, 2p and 6p and loss of 16q to be the most common

abnormalities in retinoblastoma tumors (Kusnetsova, Prigogina et al. 1982; Squire, Gallie et al.

1985; Pogosianz and Kuznetsova 1986). Using the higher resolution of comparative genomic

hybridization several groups confirmed that these changes occurred frequently (Mairal, Pinglier

et al. 2000; Chen, Gallie et al. 2001; Herzog, Lohmann et al. 2001; Lillington, Kingston et al.

2003; van der Wal, Hermsen et al. 2003; Zielinski, Gratias et al. 2005). A summary of these six

studies showed that gain of 1q, 2p and 6p and loss of 16q occurred in 53%, 34%, 54% and 32%

of retinoblastomas respectively (Corson and Gallie 2007).

Each of the chromosomal regions of gain or loss contained many genes. To differentiate true

oncogenes and tumor suppressors from the “passengers,” i.e. genes that were gained or loss due

Page 22: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

6

to close proximity to the “driver” or causative gene, differential overexpression or decreased

expression of genes in minimal regions of gain or loss in tumor versus normal tissue respectively

was assessed. To determine the minimal overlapping regions of gain and loss, copy numbers of

sequence tagged sites spanning evenly across the chromosomal regions of interest were

measured using QM-PCR or real-time PCR. Eventually a peak or “hotspot” was found to be the

most common site gained or lost in the sample of primary tumors. This approach narrowed the

search to a few genes. Expression of these genes was then assessed at the mRNA and protein

levels in tumor versus normal tissue to identify the overexpressed potential oncogenes or under

expressed potential tumor suppressor genes. In the next sections, the candidate genes found on

each of the above mentioned chromosomes will be discussed.

1.6.1 1q Gain

1.6.1.1 KIF14

Using the QM-PCR approach described above, KIF14 was identified as a target of 1q gain

(Corson, Huang et al. 2005). KIF14 is a mitotic kinesin motor protein that interacts with

microtubule bundling protein PRC1 (protein-regulating cytokinesis 1) and citron kinase and has

an essential role in regulating cytokinesis (Gruneberg, Neef et al. 2006). Corson et al reported

that out of 14 genes in 1q32 minimal region of gain only KIF14 was overexpressed at 341-fold

higher compared to normal human retina (Corson, Huang et al. 2005). In addition, KIF14 was

gained frequently not only in retinoblastoma but in other cancers including breast, lung and

medulloblastoma. Higher levels of KIF14 mRNA expression in breast cancer correlated with

more aggressive tumors (Corson and Gallie 2006). siRNA-mediated knockdown of KIF14 in a

cervical and non-small cell lung cancer cells resulted in decreased proliferation and ability to

form colonies in soft-agar (Corson, Zhu et al. 2007). KIF14 knockdown in ovarian cancer cells

lead to similar results and overexpression of KIF14 in ovarian cancer cells significantly

increased proliferation and soft-agar colony formation (Brigitte Theriault, personal

communication).

Page 23: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

7

1.6.2 6p Gain

1.6.2.1 DEK and E2F3

Early karyotypic studies showed that the 6p isochromosome is one of the most common regions

of genomic gains in retinoblastoma (Squire, Gallie et al. 1985). QM-PCR analysis of 70

retinoblastoma tumors was used to narrow down the minimal region of gain on 6p to a 0.6-Mb

size region at 6p22 (Chen, Pajovic et al. 2002). Through the study of expression level of mRNA

and protein levels of 6 genes in the 6p22 MRG the oncogenes DEK and E2F3 were identified as

targets of 6p22 gain since they were the only 2 genes to show overexpression in tumor compared

to normal adjacent retina (Orlic, Spencer et al. 2006). In addition, 3 out 4 retinoblastoma cell

lines showed increased copy number of DEK and E2F3 genes due to isochromosome 6p

formation and the cell lines that showed further rearrangements on 6p shared the common

translocation breakpoint located at 6p22 (Paderova, Orlic-Milacic et al. 2007). DEK is a nuclear

protein (Kappes, Burger et al. 2001) that binds to chromatin and is involved in modifying DNA

structure through the introduction of supercoils (Kappes, Scholten et al. 2004). It is highly

expressed in proliferating cells and its phosphorylation status oscillates with the cell cycle

peaking during G1 phase (Kappes, Damoc et al. 2004). In acute myeloid leukemia it is involved

in a fusion gene called DEK-CAN resulting from a t(6;9) translocation (von Lindern, Breems et

al. 1992), however the transforming ability of this fusion gene is debated as overexpression of

DEK-CAN failed to inhibit differentiation of myeloid precursor cell line (Boer, Bonten-Surtel et

al. 1998). Nevertheless, DEK is frequently overexpressed in other types of tumor cells as well

including hepatocellular carcinoma (Kondoh, Wakatsuki et al. 1999), melanoma (Grottke,

Mantwill et al. 2000) and acute myeloid leukemia (Casas, Nagy et al. 2003).

E2F3 is an important cell cycle gene. The E2F3 locus encodes two protein products E2F3a and

E2F3b through two alternate promoters (Leone, Nuckolls et al. 2000). The expression patterns

of E2F3a and E2F3b contrast each other during the cell cycle with E2F3a expressed in

proliferating cells and peaking during G1 and E2F3b expressed at a constant level during the cell

cycle (Leone, Nuckolls et al. 2000). E2Fs can be activators or repressors of gene transcription.

E2F3a is considered an activator which controls DNA synthesis and cell cycle progression genes

(Humbert, Verona et al. 2000). E2F3b is considered a repressor which has been shown to

interact with pRB in quiescent cells (Leone, Nuckolls et al. 2000). One of the genes E2F3b

represses is the p19ARF

tumor suppressor gene which activates the p53 pathway (Aslanian,

Page 24: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

8

Iaquinta et al. 2004). E2F3 overexpression works in concert with inactivation of the RB

pathway. A study by Hurst et al showed that 6p22.3 amplification and E2F3 overexpression

were always associated with loss of pRB expression in bladder cancer (Hurst, Tomlinson et al.

2008). These data suggest that DEK and E2F3 play important roles in cell proliferation and may

be potential targets in retinoblastoma treatment.

1.6.3 16q Loss

1.6.3.1 CDH11

CDH11 (Cadherin 11) is a member of the cadherin family of molecules. They are cell-cell

adhesion molecules that have important roles in a wide variety cellular functions including cell

polarity, cell signaling, most notably through the β-catenin-Wnt pathway and regulation of

growth factor signaling. Chromosome 16q is lost in 32% of retinoblastomas (Corson and Gallie

2007). Using a combination of loss of heterozygosity and QM-PCR analyses, the minimal

region of loss was narrowed to a 2.62 Mb region at 16q22 (Marchong, Chen et al. 2004). The

16q22-24 region harbours a cluster of cadherin genes including CDH8, CDH11 and CDH13.

Marchong et al demonstrated that a sequence tagged site, WI5835, located in intron 2 of the

CDH11 gene was lost in 54% of retinoblastomas and that 91% of retinoblastomas with loss of

this marker also had reduced or no expression of the CDH11 protein (Marchong, Chen et al.

2004). The study also showed that advanced transgenic murine SV40 large T antigen–induced

(TAg) retinoblastoma tumors displayed a loss of Cdh11 mRNA transcript in contrast to smaller

earlier tumors which still expressed Cdh11 protein thus supporting the hypothesis that CDH11

loss promotes progression (Marchong, Chen et al. 2004). In a more recent study, the same

authors used TAg Cdh11 null mice to show that Cdh11 loss caused larger tumors and higher

levels of programmed cell death than in mice with normal Cdh11 alleles, suggesting that Cdh11

functions as a tumor suppressor by promoting apoptosis in tumor cells (Marchong, Yurkowski et

al. 2009, submitted).

1.6.4 2p Gain

1.6.4.1 MYCN

MYCN (v-myc avian myelocytomatosis viral-related oncogene, neuroblastoma-derived), located

on chromosome 2p24.3 is thought to be the major target of 2p gain and amplification in

Page 25: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

9

retinoblastoma, neuroblastoma and several other neuroectodermal cancers. It was first identified

in the early 1980s by Khol et al who cloned a gene with sequence homology to the oncogene c-

myc from neuroblastoma cell lines (Kohl, Kanda et al. 1983). MYCN amplification commonly

manifests as extrachromosomal DNA units called double minutes, DM (Kohl, Kanda et al.

1983), or intrachromosomal tandem repeats called homogeneously staining regions (HSR)

(Amler and Schwab 1989). Its role as an oncogene was supported by studies showing

amplification occurred in advanced metastatic stages of neuroblastoma (Brodeur, Seeger et al.

1984; Brodeur, Azar et al. 1992; Chan, Gallie et al. 1997). Regardless of whether MYCN occurs

as DMs or HSRs, there is no difference in survival outcome, and amplification of MYCN in

either form is associated with poor prognosis in neuroblastoma (Moreau, McGrady et al. 2006).

In retinoblastoma MYCN amplification was first observed in primary tumors and the

retinoblastoma cell line Y79 by Lee et al (Lee, Murphree et al. 1984). Many retinoblastomas

highly express MYCN (Squire, Goddard et al. 1986) and 3% of primary tumors and 29% of

retinoblastoma cell lines have MYCN genomic amplification (Bowles, Corson et al. 2007)

suggesting MYCN amplification gives the cell a proliferative advantage.

1.6.4.2 ID2

The gene ID2 is a potential target of 2p gain and is located at 2p25. It is a member of the HLH

family of transcription factors and is a transcriptional target of MYCN. Although ID proteins

contain the HLH domain they lack the basic domain required for DNA binding and therefore act

as dominant negative antagonists of bHLH proteins by sequestering them in non-functional

complexes. The ID proteins are also known as inhibitors of differentiation because the bHLH

proteins that ID proteins bind to, such as Ets and Pax, are transcription factors that regulate

differentiation. Interestingly, Id2 was found to bind specifically to the hypophosphorylated form

(active) of pRB and both of its related proteins p107 and p130 and could reverse the growth

suppressive activities of pRB, p107 and p130 (Iavarone, Garg et al. 1994; Lasorella, Iavarone et

al. 1996). Its role in tumorigenesis was further demonstrated when it was shown that the Id2-null

mutation could prevent the formation of pituitary tumors in Rb1+/-

mice (Lasorella, Rothschild et

al. 2005). ID2 may represent a possible means by which MYCN can exert its pRB inhibitory

action. The significance of ID2 in neuroblastoma however is controversial. On one hand,

clinical studies fail to find a correlation between ID2 overexpression and MYCN expression or

survival and thus that evidence suggests it lacks prognostic significance (Alaminos, Gerald et al.

Page 26: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

10

2005). On the other hand, functional studies using MYCN-targeting silencing RNA in

neuroblastoma cell lines show that ID2 is regulated by MYCN (Woo, Tan et al. 2008). Thus how

ID2 contributes to tumorigenesis in neuroblastoma and retinoblastoma remains to be fully

defined.

1.7 MYCN amplification in neuronal tumors

MYCN amplification occurs in tumors of neuroectodermal origin. In addition to retinoblastoma

and neuroblastoma, other cancers include glioblastoma (Hui, Lo et al. 2001), medulloblastoma

(Bayani, Zielenska et al. 2000; Fruhwald, O'Dorisio et al. 2000), rhabdomyosarcoma (Barr, Duan

et al. 2009), and small cell lung carcinoma (Nau, Brooks et al. 1986; Dietzsch, Lukeis et al.

1994; Salido, Arriola et al. 2009). In neuroblastoma, MYCN amplification occurs in 25-30% of

primary tumors (Fix, Lucchesi et al. 2008) and correlates strongly with advanced stages and

indicates poor prognosis (Brodeur, Seeger et al. 1984; Brodeur, Azar et al. 1992; Chan, Gallie et

al. 1997; Fix, Lucchesi et al. 2008). In many cases, MYCN amplifications occur in the form of

DMs or HSRs (Bown 2001; Moreau, McGrady et al. 2006). In the large nucleolar

neuroblastoma subset, MYCN amplification is associated with distinct histology characterized by

large prominent nucleoli (Tornoczky, Semjen et al. 2007). Large prominent nucleoli are

significantly associated with poor prognosis in neuroblastoma (Ambros, Hata et al. 2002). In

retinoblastoma, the prognostic significance of MYCN amplification is not as clear since

retinoblastomas with high level MYCN amplification do not seem to show adverse histology nor

do the patients show worse survival (Lillington, Goff et al. 2002). It is important to note,

however, that treatment of retinoblastoma has > 95% cure rate (Chintagumpala, Chevez-Barrios

et al. 2007) largely due to enucleation prior to extension of tumor outside the eye, precluding

outcome analysis. In addition, MYCN amplification in neuronal tumors is often accompanied by

other genomic changes, most commonly 1p36 loss and/or 17q gain. Overall, MYCN-amplified

tumors have a less complex pattern genomic copy number alterations compared to other low or

high risk neuroblastoma subtypes (Mosse, Diskin et al. 2007).

1.8 Genomic changes in MYCN-amplified neuroblastomas

Unlike retinoblastoma, neuroblastoma is a very heterogeneous disease with outcomes ranging

from spontaneously regressing to aggressive metastatic with poor prognosis. This heterogeneity

is reflected in the pattern of genetic changes in the different subtypes of neuroblastoma. In the

Page 27: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

11

past few years, array comparative genomic hybridization (aCGH) technology has been used to

characterize genomic changes and stratify stage and outcomes (Mosse, Diskin et al. 2007; Fix,

Lucchesi et al. 2008; Janoueix-Lerosey, Schleiermacher et al. 2009). The most aggressive forms

of disease are divided into two classes, with MYCN amplification and without. In

neuroblastoma, 1p36 deletion is strongly associated with MYCN amplification, however, 1p

deletion also occurs in high-risk tumors without MYCN amplification (Chen, Bilke et al. 2005;

Janoueix-Lerosey, Schleiermacher et al. 2009; Lavarino, Cheung et al. 2009). 10q loss occurs in

53% of MYCN-amplified neuroblastoma (Mosse, Diskin et al. 2007). It had been speculated that

amplification of MYCN would be associated with higher genomic instability (Schwab 1999) but

surprisingly, recent findings show that MYCN-amplified neuroblastomas tend to have fewer

genomic changes in comparison to other subtypes including high risk neuroblastomas without

MYCN amplification (Chen, Bilke et al. 2005; Mosse, Diskin et al. 2007). Gains of 17q are a

common change across all neuroblastomas (Mosse, Diskin et al. 2007; Janoueix-Lerosey,

Schleiermacher et al. 2009). There is a strong inverse relationship between 11q deletion and

neuroblastomas without MYCN amplification (Guo, White et al. 1999; Chen, Bilke et al. 2005;

Lavarino, Cheung et al. 2009). Despite identification of common regions of copy number

alterations, few candidate genes have been identified aside from MYCN. Candidate targets of 1p

loss have been suggested but most have since been rejected (White, Maris et al. 1995; Grenet,

Valentine et al. 1998). Abel et al proposed apoptotic pathway genes CASP9 and DFFA as

candidate targets of 1p36 loss since the two genes are located within the minimal region of loss

(Abel, Sjoberg et al. 2002). However, even though higher stage neuroblastoma showed a slight

decrease in expression of both genes compared to lower stage neuroblastoma, the study failed to

show functional evidence that either CASP9 or DFFA were definitive targets of 1p36 loss.

Recently, Wei et al identified a microRNA miR-34a, located on 1p36 that directly targeted

MYCN (Wei, Song et al. 2008); exogenous expression of miR-34a in neuroblastoma cell lines

with MYCN-amplification decreased proliferation by increasing apoptosis. For 17q gain, the

gene SSTR2 was proposed as a potential target but no correlation between 17q gain and SSRT2

expression was found, nor were any mutations found in the gene in neuroblastoma tumors (Abel,

Ejeskar et al. 1999). Four independent groups identified ALK, anaplastic lymphoma kinase, as

the cause of hereditary neuroblastoma in 2008 (Chen, Takita et al. 2008; George, Sanda et al.

2008; Janoueix-Lerosey, Lequin et al. 2008; Mosse, Laudenslager et al. 2008). Mosse et al

demonstrated linkage to chromosomal bands 2p23-24 in neuroblastoma pedigrees (Mosse,

Page 28: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

12

Laudenslager et al. 2008) and Chen et al showed that ALK was a target of recurrent gains and

amplification on 2p. All four groups found activating somatic mutations in high risk

neuroblastomas and provided functional evidence that ALK had transforming ability (Chen,

Takita et al. 2008; George, Sanda et al. 2008; Janoueix-Lerosey, Lequin et al. 2008; Mosse,

Laudenslager et al. 2008). However, mutations and gains of ALK only account for a small

percentage of neuroblastomas and continued analysis of genomic changes is needed to identify

more candidate oncogenes and tumor suppressors.

1.9 MYCN amplicon

The MYCN amplicon ranges in size from 100kb to >1Mb (reviewed in Schwab 2004). The

amplicon is arranged in tandem repeats of DNA segments with the intact MYCN coding region in

the central location (Amler and Schwab 1989; Pandita, Godbout et al. 1997). Sequence analysis

has not detected mutations within the MYCN gene (Stanton, Schwab et al. 1986; Ibson and

Rabbitts 1988) therefore it is likely that the increased gene dosage of the wild type gene is what

contributes to tumorigenesis. MYCN is often co-amplified with other neighboring genes. The

two most frequently co-amplified genes are DDX1 and NAG at 65% and 20-40% of MYCN-

amplified neuroblastomas, respectively (Scott, Board et al. 2003; Weber, Imisch et al. 2004).

Co-amplification of these genes has also been documented in retinoblastoma (Godbout and

Squire 1993) and other neuronal cancers (Fruhwald, O'Dorisio et al. 2000; Barr, Duan et al.

2009; Hodgson, Yeh et al. 2009). DDX1 is located 400kb telomeric to MYCN and NAG just

telomeric to DDX1 (Amler, Schurmann et al. 1996; Scott, Board et al. 2003). There have been

conflicting reports as to the prognostic significance of DDX1 co-amplification. A few groups

have reported that co-amplification of DDX1 in high risk MYCN-amplified neuroblastomas

correlate with a more favorable outcome and higher survival rate within the MYCN-amplified

group (Weber, Imisch et al. 2004; Kaneko, Ohira et al. 2007). However, De Preter et al provided

evidence that DDX1 co-amplification is only coincidental due to proximity to MYCN and that

there is no significant correlation to better event free or overall survival (De Preter, Speleman et

al. 2005). Despite these contradicting reports on DDX1, it is important to note that MYCN has

been the only consistent gene on the amplicon and that none of the co-amplified genes have been

reported to amplify independent of MYCN.

Page 29: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

13

1.10 MYCN gene structure and expression

The MYCN gene was first identified in neuroblastoma cells and was so named due to its

similarity in nucleotide and protein sequence to the well characterized oncogene MYC (Kohl,

Kanda et al. 1983). Like MYC, MYCN is made up of three exons; the first exon contributes to a

long 5’ untranslated region and the second and third exons make up the coding regions sharing

an overall 32% amino acid sequence identity with MYC (Stanton, Schwab et al. 1986). MYCN

transcription gives rise to two forms of mRNA resulting from use of two separate promoters each

with a different first exon (Stanton and Bishop 1987). Both MYCN mRNAs are unstable and

have short half lives of approximately 15 minutes (Stanton and Bishop 1987).

Despite the homology of MYCN and MYC, their patterns of gene expression differ spatially and

temporally. Both are expressed in proliferating cells; however, MYCN is expressed almost

exclusively in embryonic tissue whereas MYC is expressed in proliferating cells of both

embryonic and adult tissues. In the developing mouse embryo, Mycn and Myc have

complementary patterns of expression; Mycn is expressed mainly in neural tissues and myc is

expressed in proliferating cells that do not express Mycn (Hurlin, Queva et al. 1997; Hurlin

2005). Human developing brain normally expresses levels of MYCN that are comparable to

expression from 150 gene copies (Grady, Schwab et al. 1987) thus it has been speculated that

high levels of MYCN protein in tumors with normal copy number of MYCN may reflect the cell

of tumor origin or undifferentiated state of the tumor (Squire, Goddard et al. 1986). Expression

declines when cells become differentiated (Martins, Zindy et al. 2008). MYCN amplification

does not always translate to high expression levels (Matthay 2000; Tang, Zhao et al. 2006).

High expression of MYCN transcript in neuroblastomas without amplification does not indicate

poor prognosis (Tang, Zhao et al. 2006). However, several explanations have been proposed to

reconcile discordance between genomic copy number and levels of expression. Matthay et al

noted that discrepancies could be a result of the use of different methods of quantification such

as northern blot, reverse transcriptase PCR, real-time PCR immunohistochemistry and Western

blot or that clinical factors such as stage, age and treatment protocols were not always consistent

within and between studies (Matthay 2000). In an effort to explain the MYCN expression

paradox, a recent study proposed that high levels of the antisense transcript of MYCN, MYCNOS,

could contribute to decreased MYCN expression in neuroblastomas with high levels of MYCN

transcript (Jacobs, van Bokhoven et al. 2009). This group showed that MYCNOS expression

Page 30: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

14

correlated with advanced disease but overexpression of MYCNOS in MYCN-amplified

neuroblastoma cell line, IMR32, did not decrease levels of MYCN mRNA ruling out the

mechanism of RNA interference. Further analysis will be required to determine of the

relationship between amplification and gene expression.

1.11 MYCN protein and functions

1.11.1 MYCN protein

MYCN is a member of the basic helix-loop-helix (bHLH) family of transcription factors. The

MYCN gene encodes two protein products with apparent molecular weights of 65 and 67 kDa

that localize in the nucleus (Ramsay, Stanton et al. 1986). The C-terminal contains a basic

domain which binds DNA and an HLH domain which mediates dimerization with other HLH

domain-containing proteins such as MAX (Wenzel and Schwab 1995). MYCN can act as a

transcriptional activator when bound to MAX or a repressor when bound to Mnt, Mxi, Mad or

other cofactors. MYCN-MAX heterodimers recognize conserved sequences called E-boxes.

The N-terminus contains the four evolutionarily conserved myc boxes which together make up

the transactivation domain (Cowling and Cole 2006). The N-terminus also contains

phosphorylation sites for casein kinase II (Hamann, Wenzel et al. 1991) as well as

phosphorylation sites for MAP kinase and GSK3 (Henriksson, Bakardjiev et al. 1993).

1.11.1.1 MYCN and RB pathway

MYCN regulates genes involved in cell proliferation and is thought to control cell cycle genes,

however, the precise mechanism remained elusive until recently. Woo et al showed that MYCN

controls S phase genes (Woo, Tan et al. 2008). Through the use of MYCN silencing RNA, the

authors demonstrated that inactivation of MYCN in amplified neuroblastoma cell lines resulted in

increase of p27 and decrease of cell cycle genes E2F1, E2F2, and CDK6 as well as the

differentiation gene ID2. E2F promoters have E-boxes that MYC can bind to. ChIP analysis

showed that E2F1 recruitment to E2F elements of target genes is dependent on the binding of

MYC to E-boxes of E2F promoters (Leung, Ehmann et al. 2008). This provides evidence of a

direct link between MYC and transition from G1/G0 to S phase. MYCN can also drive

proliferation independent of E2Fs.

Page 31: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

15

In the developing murine retina, cells with triple knock out of E2f 1, 2 and 3 can retain the ability

to divide (Chen 2009, in press). Proliferation is only inhibited in a quadruple knock out of four

genes E2f1, 2, 3 and Mycn, indicating that Mycn provides a compensatory or redundant cell

division promoting mechanism (Chen 2009, in press). Mycn-mediated cell proliferation in the

absence of E2fs is accomplished through maintenance of E2f targets and down regulation of

Cdk1a and c. MYCN was recently shown to directly upregulate a cluster of miRNAs 17-5p-92

which inhibit the cyclin-dependent kinase inhibitor p21 (Fontana, Fiori et al. 2008). p21 lies

upstream of pRB and negatively regulates the cell cycle by inactivating Cdk2-cyclin E

complexes that inhibit pRB through phosphorylation. Fontana et al showed that ectopic

expression of miRNA 17-5p-92 in neuroblastoma cells increased proliferation and

downregulated p21 and that primary neuroblastomas with MYCN-amplification also had

upregulation of miRNA 17-5p-92 coupled with low p21 expression. In addition, miRNA 17-5p-

92 also downregulated expression of the pro-apoptotic protein BIM (Bcl-2 interacting mediator

of cell death) shutting down the apopotic pathway (Fontana, Fiori et al. 2008).

1.11.1.2 Other MYCN functions

While MYCN can induce proliferation and cell cycle progression, its overexpression also

strongly activates apoptosis (Hogarty 2003). This opposing function of MYCN is likely a

protective mechanism and indeed, high levels of MYCN protein in neuroblastoma cells without

MYCN amplification has been shown to inhibit proliferation and induces apoptosis (Peirce and

Findley 2009). Consequently, in order for the tumor cell to survive, a balance must be struck

between MYCN-driven proliferation and MYCN-induced cell death. The tumor suppressor p53

can induce apoptosis, cell cycle arrest and DNA repair mechanisms in response to a variety of

cell stress. In an unstressed cell, p53 is kept inactive mainly by E3 ubiquitin ligase MDM2

which targets p53 for degradation by the proteasome. MDM2 was recently identified as a

transcriptional target of MYCN (Slack, Chen et al. 2005). MYCN was shown to bind to E boxes

of the MDM2 promoter and when MYCN was inhibited in amplified neuroblastoma cells,

resultant decrease in MDM2 was accompanied by stabilization of p53 (Slack, Chen et al. 2005).

The same group confirmed their in vitro findings in vivo in a follow-up study. Mdm2+/-

MYCN+/+

mice had significantly delayed tumor development and lower overall incidence of

tumors (Chen, Lin et al. 2009). p19Arf

which suppresses Mdm2 was found to be epigenetically

Page 32: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

16

silenced in Mdm2+/-

mice suggesting that reduction of the Mdm2 inhibitor is another mechanism

by which MYCN circumvents induction of apoptosis.

The MYC family of proteins is considered to be weak activators of gene transcription yet it is

estimated that they activate 15% of all human genes (Patel, Loboda et al. 2004; Dang, O'Donnell

et al. 2006). In an effort to reconcile this apparent paradox, Cotterman et al, studied the effects

of MYCN on chromatin regulation and found that a surprising 90-95% of histone H3 acetylation

and methylation marks were dependent on MYCN expression (Cotterman, Jin et al. 2008). Using

ChIP coupled with array technology, the group found that MYCN bound extensively to the entire

genome and predicted that there were an estimated ~20 000-40000 MYCN binding sites with

40% of sites at least 10kb away from transcriptional start sites (Cotterman, Jin et al. 2008). This

indicated that MYCN can not only regulate genes as a transcription factor but it can indirectly

activate transcription of potentially thousands of genes by opening up large stretches

euchromatin to transcription. Altogether, the evidence presented above shows that MYCN can

exert its proliferative effect in a wide variety of ways ranging from the manipulation of the

multiple arms of the RB and apoptotic pathways as well acting as a general transcriptional

stimulus in the cell.

1.12 MYCN amplification in transgenic murine model of neuroblastoma

Functional evidence that MYCN overexpression can initiate neuroblastoma tumors came when

Weiss et al used the tyrosine hydroxylase promoter to target a Mycn transgene to neural crest

cells in mice (Weiss, Aldape et al. 1997). The TH-MYCN mice developed tumors with similar

histopathology and expression of neuronal markers consistent with human neuroblastoma and

CGH analysis of tumors showed genomic changes accompanying tumor progression occurred in

the regions syntenic to those often gained and lost in human neuroblastomas, such as gains of 11

and loss of 17 (Weiss, Aldape et al. 1997; Cheng, Cheng et al. 2007). Further characterization of

the TH-MYCN mice showed that the transgene specifically became amplified as the disease

progressed (Hansford, Thomas et al. 2004; Cheng, Cheng et al. 2007). Taken together, these

data provide evidence that overexpression of MYCN can initiate tumorigenesis and gives good

justification that a similar model in which MYCN overexpression in the retina will be a useful

tool to dissect the mechanism of MYCN tumorigenesis in retinoblastoma.

Page 33: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

17

1.12.1.1 Role of MYCN in retinal development

Recently, it was shown that MYCN plays an important role in the coordination of growth of the

murine retina. Transgenic mice lacking Mycn had a smaller but properly proportioned eyes

compared to their littermates with normal Mycn (Martins, Zindy et al. 2008). The authors

showed that in mice, the level of cyclin-dependent kinase inhibitor p27 expression was increased

and that the small eye phenotype could be rescued by also knocking out p27, an inhibitor of pRB

phosphorylation/inactivation thus demonstrating that Mycn acts through the RB pathway.

Page 34: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

18

Chapter 2

2 Characterization of RB1+/+

retinoblastoma

N.B.: Diane Rushlow, Jennifer Kennett, Paul Boutros and Anthony Mak contributed to part of

the work presented in this chapter. Diane Rushlow provided the M3-Mn gene-specific QM-PCR

copy numbers depicted in figure 1 and table 6 (Appendices), Jennifer Kennett at Dr. Wan Lam’s

laboratory performed the sub-megabase resolution tiling array comparative genomic

hybridization and assisted in the data analysis, Paul Boutros assisted in the statistical analysis

depicted in Tables 3 and 4 and Anthony Mak assisted in the construction of the Mycn-

overexpression lentiviral construct depicted in figure 13. I would also like to acknowledge Tim

Corson for providing the SKY analysis in figure 8.

2.1 Introduction

Genetic screening is performed to identify RB1 mutations in order to diagnose retinoblastoma

earlier and to provide genetic counseling to families. However, genetic testing is a difficult task

for 2 reasons: (1) the RB1 gene is large and made up of 27 exons distributed over 183 kb of

genomic sequence and has a promoter containing a normally unmethylated CpG island; (2)

almost all mutations are unique and scattered along the entire gene with no real hot spots. To

date a sensitive and efficient series of molecular tests have been developed. Mutation screening

consists of QM-PCR of all 27 exons and the core promoter to detect copy number changes,

sequencing of the core promoter and 27 exons as well as 25 intronic nucleotides flanking each

exon, and testing for hypermethylation of the RB1 core promoter (Richter, Vandezande et al.

2003). Currently, sensitivity for detecting both mutations in bilateral patients is 95% (443/467)

and the remaining 5% are predicted to be mosaic in blood (Rushlow, Piovesan et al. 2009). In

unilateral patients 94% (413/441) of patients have both mutations identified in retinoblastoma

tumor, 4.8% (21/441) have one mutation identified and in 1.6% (7/441) of cases, no mutations

can be detected. This 1.6% of unilateral retinoblastomas also does not show loss of

heterozygosity; hence, they will be referred to as RB1+/+

retinoblastomas and will be the focus of

this research project (See Table 3).

Retinoblastoma is caused by inactivating mutations on both alleles of the RB1 gene. However,

recent work by Dimaras et al, demonstrated that these first two mutations (M1 and M2) cause a

Page 35: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

19

benign precursor called retinoma and further mutational events termed M3-Mn are required for

progression to malignant retinoblastoma (Dimaras, Khetan et al. 2008). Using techniques such

as karyotype analysis, metaphase comparative genomic hybridization (CGH) and aCGH,

retinoblastoma was shown to display a specific constellation of genomic changes (Squire, Gallie

et al. 1985; Chen, Gallie et al. 2001; Corson and Gallie 2007; Sampieri, Amenduni et al. 2009).

Candidate M3-Mn genes have been characterized, including oncogenes KIF14 on 1q (Corson,

Huang et al. 2005), MYCN on 2p (Bowles, Corson et al. 2007), E2F3 and DEK on 6p (Orlic,

Spencer et al. 2006), and potential tumor suppressor CDH11 on 16q (Marchong, Chen et al.

2004). A QM-PCR was developed to profile retinoblastoma M3-Mn progressive genomic

changes, including the MYCN gene. QM-PCR results showed that M3-Mn changes occur

frequently in retinoblastomas. In RB1-/-

unilateral retinoblastomas, the oncogenes KIF14, MYCN,

DEK and E2F are gained at frequencies of 50%, 15%, 40% and 70% and the tumor suppressor

CDH11 is lost at a frequency of 45% (Bowles, Corson et al. 2007). When gene-specific QM-

PCR was used to profile the RB1+/+

retinoblastomas, it was discovered that they showed a

completely different genomic profile than RB1-/-

retinoblastomas (Rushlow and Gallie, personal

communication). First, a high proportion of RB1+/+

retinoblastomas (57%, 4/7) showed high-

level MYCN amplification (33-121 gene copies) whereas out of 70 RB1-/-

unilateral

retinoblastomas tested, none showed copy numbers greater than 10 copies. Second, RB1+/+

showed few M3-Mn genomic alterations characteristic of retinoblastoma. These observations

led to the hypothesis that in addition to the two known genetic forms of retinoblastoma, both of

which are caused by RB1 mutations, there may be a third form of retinoblastoma in which no

mutations to RB1 are required.

2.2 Hypothesis

RB1+/+

retinoblastomas represent a previously unrecognized subset of retinoblastoma that have a

genetic signature distinct from conventional RB1-/-

retinoblastomas.

2.3 Thesis Aims and Rationale

2.3.1 Frequency of RB1+/+ retinoblastoma

To confirm that RB1+/+

retinoblastomas were not isolated cases limited to the Toronto subset of

tumors, collaborations with three other RB1 testing centers were set up to collect a larger number

Page 36: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

20

of samples. Once collected, statistical analysis was performed to determine frequency of RB1+/+

retinoblastomas across four RB1 gene testing centers.

2.3.2 Characterize genomic profile of RB1+/+ retinoblastomas

Gene-specific QM-PCR was performed on samples from the three other RB1 testing sites to

determine whether they shared the M3-Mn retinoblastoma genomic signature of RB1-/-

or RB1+/+

retinoblastomas with the Toronto subset. Following gene-specific QM-PCR analysis, sub-

megabase resolution tiling aCGH (SMRT aCGH) was used to profile the entire genomes of each

RB1+/+

retinoblastoma compared to RB1-/-

retinoblastomas.

2.3.3 Determine mRNA and protein levels of RB1 and MYCN genes

The levels of RB1 and MYCN mRNA and protein were confirmed for two reasons: (1) to

determine whether full-length RB1 transcript and protein were expressed consistent with RB1+/+

status and (2) to determine the levels at which they were expressed, particularly whether levels of

MYCN transcript and protein correlated with the amplified genomic status of the tumor.

2.3.4 Analysis of clinical and pathological features of RB1+/+ retinoblastomas with MYCN amplification

Clinical features such as age of diagnosis and histology were assessed to determine whether or

not RB1+/+

retinoblastoma showed the characteristic features of RB1-/-

retinoblastoma.

2.3.5 Determine the effect of MYCN silencing in MYCN-amplified retinoblastoma

The MYCN-amplified retinoblastoma cell line Y79 was treated with MYCN-targeting shRNA

lentivirus to determine the effect on proliferation rate.

2.3.6 Designing a Mycn-overexpressing lentivirus

A lentivirus overexpressing Mycn was developed for the purpose of assessing the effect of high-

levels of Mycn in vivo. The lentivirus will potentially be used for injection into the murine retina

to determine whether exogenous expression of Mycn can initiate tumors.

Page 37: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

21

2.4 Materials and Methods

2.4.1 Samples

Analysis was performed on 410 DNA samples from primary retinoblastoma tumors of probands

with sporadic unilateral retinoblastoma collected at Retinoblastoma Solutions, Toronto, Ontario,

Canada. The Research Ethics Boards of the Wellesley Hospital, the Hospital for Sick Children,

the University Health Network, and the University of Toronto approved research use of tumor

material with parental consent. Additional samples were collected from 3 other sites: Essen,

Germany; Paris, France and Christchurch, New Zealand (Table 1).

2.4.2 RB1 gene mutation testing

For samples from Toronto, DNA was extracted using the Gentra PuregeneTM

kit (now Qiagen,

Mississauga, ON). Samples were primarily submitted as clinical samples for RB1 mutation

detection, and were screened for RB1 mutations or epigenetic changes using QM-PCR of all 27

exons and the core promoter to detect copy number changes, sequencing of the core promoter

and 27 exons (as well as 25 intronic nucleotides flanking each exon), and testing for

hypermethylation of the RB1 core promoter (Richter, Vandezande et al. 2003; Rushlow,

Piovesan et al. 2009) .

Tumors samples from each of the three additional sites had been tested for any changes in the

RB1 gene, including sequence analysis, testing for whole or multi-exon copy number changes

using either QM-PCR or MLPA (MRC-Holland), methylation of the RB1 promoter, and for loss

of heterozygosity at RB1 using microstaellite analysis (Raizis, Schmitt et al. 1995; Stirzaker,

Millar et al. 1997; Raizis, Clemett et al. 2002; Schouten, McElgunn et al. 2002; Houdayer,

Gauthier-Villars et al. 2004; Schüler, Weber et al. 2005; Mitter, Rushlow et al. 2009).

2.4.3 Gene-specific QM-PCR

After completion of RB1 mutation screening, gene-specific primers were used in a QM-PCR

reaction to determine genomic copy number for KIF14 (1q32.1), DEK (6p22) E2F3 (6p22), and

CDH11 (16q22). Copy number of these four genes were determined in 91 tumor DNA samples

collected from all four sites with both mutations identified (12 of these previously reported in

Bowles, Corson et al. 2007), in 27 tumor DNA samples with no RB1 tumor mutation identified

(RB1+/+

), one tumor reported to be bilateral with no RB1 tumor mutation identified (RB522) and

Page 38: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

22

20 tumors with only one RB1 tumor mutation identified (RB1+/-

) (see Table 1). Copy number for

MYCN (2p24.3) were determined using a second QM-PCR reaction and MYCN specific primers

(Bowles, Corson et al. 2007), in 70 primary tumors with both RB1 mutations identified, in 21

tumors with one tumor mutation identified (RB1+/-

), and in 27 tumors with neither RB1 mutation

identified (RB1+/+

) .

Gene-specific primers were used for KIF14, DEK, E2F3, CDH11, and MYCN, previously

described (Bowles, Corson et al. 2007). Each reaction tube contained 7.5µl of Qiagen Multiplex

PCRTM

2X Master Mix (Qiagen, Mississauga), 0.2 µl of gene-specific primer pool (12.5-25ng/µl

of each primer ), 0.5 µl of C4 control primers, 0.3 µl of ALK-1 exon 5 control primers, 3.5 µl of

water and 3 µl of DNA at 30ng/µl. Cycling required 15 minutes at 95ºC to activate the hot- start

enzyme followed by 19 cycles of 94 ºC for 30 seconds, 60 ºC for 1’30 seconds, 72 ºC for 1’30

seconds, and a final extension of 10 minutes at 72 ºC. One primer of each pair was Cy5.0

labeled; product peak sizes were quantified using Visible Genetics’TM

sequencers (Siemens) and

GeneObjectsTM 3.1

software.

Primers for two internal control fragments were included in each assay: a 329-bp fragment (C4)

from exon 4 of the retinaldehyde-binding protein (chromosome 15) and a 198-bp product from

exon 5 of the ACVRL-1 (ALK-1) gene (chromosome 12). One internal control peak was set to 2

copies and the ratios of the other peak heights to the control peak were compared to ratios

obtained for normal two-copy DNA from blood samples to establish copy number for each gene.

The second internal control peak in each assay acted as a check and was expected to give close to

two copies to verify that a DNA sample was amplifying consistently and that there was no

significant degradation of the DNA. Each run included at least four normal control samples and

two samples previously characterized as showing gain/loss for each of the genes of interest.

Tumor samples showing gain or loss were confirmed by repeat analysis.

Page 39: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

23

Table 1 Samples used for QM-PCR

Test Site # of RB1-/-

tested for KIF14, DEK,

E2F3, CDH11 copy number

# of RB1-/- tested for

MYCN copy number

# of RB1+/- tested for

KIF14, DEK, E2F3, CDH11 copy number

# of RB1+/- tested for

MYCN copy number

# of RB1+/+ tested for

KIF14, DEK, E2F3, CDH11

and MYCN copy number

Toronto, Canada

69 48 16 17 8*

Essen, Germany

12 12 4 4 12

Paris, France 10 10 0 0 5

Christchurch, New Zealand

0 0 0 0 2

Total 91 70 20 21 27 *This number includes RB522 which was originally diagnosed as bilateral.

2.4.4 Sub-megabase resolution tiling array comparative genomic hybridization

The array platform, comprised of 26,363 overlapping elements, was manufactured on site, as

previously described (Ishkanian, Malloff et al. 2004; Watson, deLeeuw et al. 2007). The

effective resolution of the array is 79 kb (Ishkanian, Malloff et al. 2004). Briefly, 200 ng of test

and reference (single male) DNA were separately labeled with Cyanine-3 and Cyanine-5 dCTPs

Using the BioPrime DNA labeling system (Invitrogen, Burlington, Ontario, Canada). DNA

probes were then pooled and unincorporated nucleotides were removed with a YM-30 Microcon

centrifugation tube (Millipore). Next, 100 μg of Cot-1 DNA (Invitrogen) was added and the

entire mixture was precipitated. This material was then re-suspended in a 45 μl cocktail

consisting of DIG Easy hybridization solution (Roche), sheared herring sperm DNA (Sigma-

Aldrich), and yeast tRNA (Calbiochem). Probe denaturing and blocking steps followed at 85°C

and 45°C for 10 minutes and for one hour respectively. Subsequently, the probe mixture was

applied to the surface of the array, coverslips were applied, and arrays were incubated at 45°C

for 36 hours. Slides next underwent five agitating washes in 0.1× SSC, 0.1% SDS at 45°C (each

wash ~5 min). Rinses with 0.1× SSC followed, then drying by centrifugation.

Page 40: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

24

2.4.5 SMRT aCGH data analysis

CGH array images were obtained with the Array-WoRxCCDscanner (Applied Precision,

Issaquah,WA) at a resolution of 10 mm with median intensity channel normalization. Image

analysis was performed with the Softworx software suite (Applied Precision). The raw data was

normalized for spatial and printing intensity bias with CGH Normalize suite (Khojasteh, Lam et

al. 2005). Data was imported into SeeGH (Chi, DeLeeuw et al. 2004) a program allowing

electronic representation visualization, multiple alignment, and copy number annotation of the

data. To minimize the potential noise due to dust or scratches on the array, all results were

screened using the variance between the duplicate spots. A clone was not included in analysis if

the variance exceeded 0.075. Additionally, the calculated signal to background ratio of 10 for

each spot was used to omit any spots.

Breakpoint boundaries were determined by the end sequence position of the BAC clone on either

side of the breakpoint. When a BAC clone exhibited a ratio that was an intermediate value of the

two flanking copy number ratio levels, the clone was considered to contain the breakpoint. When

such a clone did not exist, the two flanking clones were considered to encompass the breakpoint

event. Breakpoints at the centromeres were indiscernible due to their repetitive DNA and

subsequent incomplete mapping, which prohibits precise loci determination. Segmentation in

the samples were analyzed with the CNA Hmmer algorithm (Shah, Xuan et al. 2006).

2.4.6 Statistics

Comparisons of the frequency of mutations in RB1+/+

and RB1-/-

patients were made using a two-

tailed proportion test with Yates' continuity correction, as implemented in the R statistical

environment (v2.7.2).

The Mann-Whitney rank sum test was used to make pair-wise comparisons of number of CNAs

and aberrant base pairs between all combinations of the four retinoblastoma subsets

RB1+/+

MYCNA (MYCN > 10 copies), RB1

+/+ (2-copy MYCN), RB1

+/-, and RB1

-/- and cell lines

were made using the the GraphPad Prism software (v5.02).

Comparisons of age of diagnosis were made using a one-way analysis of variance (ANOVA)

using the GraphPad Prism software (v5.02).

Page 41: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

25

2.4.7 RT-PCR

Total RNA was extracted from fetal, adult and primary retinoblastoma samples using TRIzol

(Invitrogen) according to the manufacturer’s instructions. RNA concentration was measured

using the NanoDrop-1000 spectrophotometer (Thermo Scientific). For cDNA synthesis, 1μg of

total RNA was reverse transcribed using random primers (Invitrogen) and SuperScript II Reverse

Transcriptase (Invitrogen) at 42°C for 50 minutes. The reaction was inactivated by heating at

70°C for 15 minutes. The resulting cDNA library was used in end-point PCR gene expression

analyses in a reaction mixture consisting of 200 μM dNTPs, 2.5 mM MgSO4, 0.5 μM each of

forward and reverse primers, 0.5 U KOD hot start DNA polymerase (Novagen), 1x PCR buffer

(Novagen) and 1 μl of product from cDNA synthesis, in a final volume of 25 μl. PCR was

performed using the RoboCycler Gradient 96 thermal cycler (Stratagene). Primers are listed in

Table 2. Cycling conditions are as follows: 2 minutes at 94°C, next 30 cycles of amplification

(30 seconds 94°C, 30 seconds 65°C and 1 minute 30 seconds 72°C) and lastly 10 minutes

extension at 72°C.

Table 2 List of primer sequences and expected product sizes used in RT-PCR analysis

Gene Primer Sequence Expected Size (bp)

RB1 5’-ATGCCGCCCAAAACCCCCCGAAAA-3’ 5’-TCATTTCTCTTCCTTGTTTGAGGT-3’

2787

MYCN 5’-CACAAGGCCCTCAGTACCTC-3’ 5’-TCTTCTGTGGGGGTGCAT-3’

283

Ki67 5’-GCTAAAACATGGAGATGTAAT-3’ 5’-ATTTTGGTCTTGACTTACGC-3’

631

TBP 5’-ACAACAGGCTGCCACCTTAC-3’ 5’-GCTGGAAAACCCAACTTCTG-3’

743

2.4.8 Immunohistochemistry

Formalin-fixed, paraffin-embedded sections of human retina and retinoblastoma were studied.

Slides were re-hydrated by incubating two times 10 minutes each in xylene, two times 5 minutes

each in 100% ethanol, once for 2 minutes in 95%, 70%, and 50% ethanol, followed by 5 minute

incubation in TBS. For antigen retrieval, sections were treated with 0.1% trypsin for 5 min at

37°C or heated in PBS citrate for 17 min in a pressure cooker prior to incubation with primary

antibody. Slides were then incubated in 5% Triton-X for 10 minutes at room temperature.

Blocking was carried out for 30 minutes at room temperature in TBS with 10% DAKO Protein

Page 42: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

26

Block (DAKO-Cytomation), 1% BSA and 0.05% Tween-20. Sections were stained for pRB-N-

terminus, 1:200 (BD Pharmingen, Missisauga, ON), pRB-C-terminus, 1:200 (Santa Cruz) and

MYCN 1:100 (Santa Cruz) in TBS with 1% BSA, 0.05% Tween-20 and 10% Antibody Diluent

(DAKO-Cytomation), followed by three washes in TBS with 0.1% BSA and 0.05% Tween-20.

Human pRB-N-terminus immunoreactivity was detected using Immunopure DAB substrate kit

(Pierce). Human pRB-C-terminus and MYCN immunoreactivity was detected using fluorescent

staining by Alexa™ 488 Streptavidin conjugate from Molecular Probes. DAPI was used to

visualize nuclei of cells. Slides were mounted using the DAKO-Cytomation Fluorescent

Mounting Medium. Slides were visualized using a Zeiss LSM510 confocal microscope (Zeiss,

Toronto, Canada)

2.4.9 Lentivirus production

Bacterial stocks of MYCN-targeting lentiviral Mission® shRNA vectors were purchased from

Sigma-Aldrich. The pLKO.1-shRNA lentiviral plasmid vector DNA was isolated according to

standard phenol-chloroform isolation procedure. 8 X 105

human embryonic kidney 293T (HEK

283T) cells were plated in 10 cm plates in 10 ml of growth media with antibiotics and incubated

overnight for 24 hours at 37°C in 5% CO2. The next day, the cells were transfected with

pLKO.1-shRNA lentiviral plasmid vector along with Pax2 and MD2.G packaging vectors using

the Lipofectamine 2000®

transfection reagent (Invitrogen) for shRNA lentivirus and FuGENE® 6

transfection reagent (Roche) for the Mycn-overexpression vector according to manufacturer’s

instructions. From this point onwards, the cells were incubated in an incubator reserved only for

virus work. After 18 hours of incubation, the media was replaced with 30% serum growth

medium. At 48 and 72 hrs after transfection, virus was harvested by collecting media from cells.

Debris was spun down and the supernatant was aliquoted and frozen at -70°C. Media was

replaced on the cells after 48 and incubated again overnight for the 72 hour harvest.

2.4.10 Lentivirus titration

HEK293 cells were seeded onto 6-well dishes at a density of 2 x 104/ml. The next day, cells

were transduced with serial dilutions of virus (10-2

to 10-6

) in DMEM 10% FBS media in

triplicate and incubated overnight. The next day, virus was removed and cells were

supplemented with 2 ml of fresh media and incubated overnight. The next day, media was

changed to DMEM with 1 μg/ml of puromycin. Media was changed every 3-4 days for 14 days.

Page 43: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

27

Cells were then rinsed twice with PBS and fixed with 4% paraformaldehyde for 10 minutes.

Following rinsing with PBS two times, cells were stained with crystal violet stain (0.1% crystal

violet powder in 10% ethanol) and rinsed 6 times with double distilled water. Number of viral

colonies were counted and averaged across the triplicate wells to give transforming units per ml

which was multiplied by the volume (1 ml) to give the titer.

2.4.11 Proliferation assay

1 x 106

Y79 retinoblastoma cells were transduced with a 1/20 dilution of the undiluted virus

stock of each of the 5 MYCN Mission® shRNA lentiviral particles as well as the empty pLKO-

puro lentivirus in T75 flasks and incubated overnight. The next day, virus was removed and

fresh RB media (500 Iscove’s medium, 89.4 ml Fetal Clone III serum, 5ml 100X penicillin-

streptomycin, 2.38 μl β-mercaptoethanol, and 596 μl 10mg/ml insulin) was replaced. Forty-eight

hours after transduction, media was replaced with RB media containing 1μg/ml puromycin.

Media was changed every 3-4 days for one week. Cells were then plated at a density of 2 X 104

cells/well in 24-well plates in triplicate for each of the 6 viruses in 7 sets. The number of cells in

each well was counted everyday for 7 days and the triplicate counts were averaged.

2.4.12 Construction of Mycn-overexpression lentivirus

Mycn cDNA was PCR-amplified from mouse fetal retina cDNA library using the following

primers: forward 5’-CGAACCCATGCCCAGCTGCA-3’ and reverse 5’-

GAAACGTTAGCAAGTCCGA-3’. The amplified Mycn product was cloned into the

StrataClone™ PCR cloning vector according to manufacturer’s instructions. Mycn cDNA was

sequenced and verified and further subcloned into the pSY series of lentiviral vectors (Figure

13). Lentiviral particles were then produced as described above. HEK293 cells were transduced

with undiluted virus stock with polybrene at a concentration of 8μg/ml to test the expression of

Mycn protein. Media was changed 24 hours post-infection and fresh growth media was replaced

every 2 days for 4 days at which time, cells were scraped and lysed with RIPA buffer. The lysed

cell debris was spun down and supernatant collected for western blot analysis.

2.4.13 Transduction of retinal explants

Transduction of explants has been previously described (DiCiommo, Duckett et al. 2004). Mice

were treated in accordance with the Canadian Council on Animal Care and with approval from

Page 44: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

28

the University Health Network Animal Care Committee. Briefly, to test expression of Mycn in

murine retinal explants, P0 B-6 mice (Ontario Cancer Institute) were sacrificed and retinas were

dissected and placed on a cell culture membrane (Millipore) in explant media (DMEM/F-12

supplemented with 5% FBS, insulin (5μg/mL), pyruvate, and glutamate for 24 hours. To

transduce explants, a sterile 1ml pipette tip was cut to 0.5 cm at the base and placed around the

retina, 200 μl of virus stock was placed in the pipette tip barrier and incubated overnight at 37°C,

5% CO2. The next day, virus was removed and replaced with fresh media and media was

changed every 48 hrs for 5 days. The retinal explants were then harvested by removing media

and fixed with 4% paraformaldehyde on ice for 1 hour. The paraformaldehyde was removed and

replaced with 70% ethanol overnight at 4°C. The retinal explants were then embedded in

paraffin and slides were cut for immunohistochemistry analysis of Mycn and EGFP protein

expression.

2.4.14 Western blot analysis

Total protein was extracted from harvested cell pellets. Samples were mixed with cold Triton-X

buffer (1% Triton-X, 20mM Tris (pH 7.5), 150mM NaCl, 1mM EDTA, 1X Roche complete

protease) and incubated for 20 minutes at 4°C. After centrifugation at 12,600x g for 15 minutes,

supernatants were recovered and protein concentrations were determined using a Bradford

protein assay (Bio-Rad, Hercules, CA). Proteins (30μg) were separated by 4-20% gradient pre-

cast SDS-PAGE (Lonza, Rockland, ME) at 120V for 2 hours and transferred to a polyvinylidene

fluoride membrane (Bio-Rad). The membrane was blocked [5% Blotto (Bio-Rad)] overnight

and incubated with either rabbit polyclonal anti-MYCN (NCM II 100, Santa Cruz) (1:200) or

rabbit isotype IgG control. X-ray film (Kodak) was used to detect chemiluminescence generated

using ECL reagent (GE Healthcare).

Page 45: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

29

2.5 Results

2.5.1 Frequency of RB1+/+ retinoblastoma across four independent sites

To establish the frequency of RB1+/+

retinoblastomas over a larger set of patients, retinoblastoma

samples were collected from three other RB1 testing centers performing similar testing to

Retinoblastoma Solutions, Toronto (Table 3). Of a total of 400, 152, and 30 unilateral

retinoblastomas screened for RB1 mutations at the Institute für Humangenetik, Essen, Germany,

the Institut Curie, Paris, France and the Christchurch School of Medicine, Christchurch, New

Zealand, 12 (3%), 5 (3%) and 2 (7%) respectively were found to be RB1+/+

(Table 3). These

were similar frequencies to the Toronto data set (P = 0.168, 0.475 and 0.269 pair-wise proportion

test) and all 3 sets of RB1+/+

retinoblastomas were used in gene-specific QM-PCR analysis

(Table 3 and see Aim 2.1 below). In total 26 RB1+/+

retinoblastomas out of 992 tumors tested

were collected from 4 independent sites as part of a multi-site analysis. RB1+/+

retinoblastomas

occur at a frequency similar between all sites thus justification could be made to pool them into

one set making the frequency of RB1+/+

retinoblastomas 2.6% across 4 independent sites.

Table 3 Frequency of RB1+/+

retinoblastomas across four sites

Test Site Total Number of Unilateral

retinoblastomas

Number of RB1-/- and

RB1+/-

Number of RB1+/+ (Freq.)

P-value (pair-wise

proportion test)

Number of RB1+/+ MYCNA

Canada 441 434 7 (2%) 4

Germany 400 388 12 (3%) P=0.168 3

France 152 147 5 (3%) P=0.475 2

New Zealand 30 28 2 (7%) P=0.269 1

Total 1023 997 26 (3%) 10

2.5.2 Genomic profile of RB1+/+ retinoblastomas

2.5.2.1 Copy number changes in M3-Mn genes in RB1+/+ retinoblastomas

Several studies have shown that, following the initial two mutations to RB1, further characteristic

genomic gains and losses (M3-Mn) are common in retinoblastoma including the gain of

oncogenes KIF14, MYCN, DEK, and E2F3, and loss of the tumor suppressor CDH11 (Marchong,

Chen et al. 2004; Corson, Huang et al. 2005; Orlic, Spencer et al. 2006; Bowles, Corson et al.

Page 46: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

30

2007; Dimaras, Khetan et al. 2008). A previously described gene-specific QM-PCR method

(Bowles, Corson et al. 2007) was used to measure copy number changes of the above genes in

the RB1+/+

and RB1-/-

retinoblastoma subsets (Table 6, Appendices). Gain is defined as 2.5 to 10

copies, loss as less than 1.5 copies and amplification as more than 10 copies (Figure 1). Figure 1

shows the copy numbers of M3-Mn genes in the 26 RB1+/+

, 20 RB1+/-

, 91 RB1-/-

retinoblastomas

and 6 cell lines. With the larger sample sizes available for the current analysis, it was found that

gains of KIF14, MYCN, DEK, and E2F3, and loss of CDH11 are more frequent in RB1-/-

retinoblastomas than previously thought (Bowles, Corson et al. 2007) as they were gained and

lost at frequencies of 61%, 64%, 57%, 58% and 64% respectively. In RB1+/+

retinoblastomas,

however, it was found that gains in KIF14, DEK, and E2F3, and loss of CDH11, were much less

frequent at 23%, 34%, 25%, and 19% respectively compared to RB1-/-

retinoblastomas (Table 4).

However, four tumors MC945, RB1348 and MA94 did show gains of KIF14, DEK, and E2F3,

and RB1700 showed loss of CDH11 (see Figure 1 and Table 6 in Appendices). Most striking

was the frequent occurrence of high-level amplification of the MYCN gene (MYCNA) in the

RB1+/+

retinoblastomas (10/26; 38%) of the RB1+/+

samples showed MYCN genomic copy

numbers ranging from 38 to 121 (see Table 6 Appendices). This was in stark contrast to and

significantly different from RB1-/-

retinoblastomas where, although low level gain of 3-5 copies

of MYCN was frequently observed and occasionally moderate gain of up to 9 copies of MYCN,

MYCN amplification of over ten copies was never seen in any of the 91 RB1-/-

unilateral tumor

samples tested (P-value 9.55 x 10-9

).

Of the 21 RB1+/-

unilateral tumor DNA samples tested for MYCN copy number, one sample,

RB2285, showed high level MYCN amplification. The one non-germline RB1 mutation found in

this sample was deletion of one copy of most of the 13q arm (see Whole genomic profiling

below), including RB1. This sample showed high similarity to the RB1+/+

MYCN A

subset,

including a very early age of diagnosis (4 months). Thus it is hypothesized that the loss of 13q in

this tumor may be a secondary event and that this sample may in fact belong in the RB1+/+

MYCN A set.

RB522 was originally diagnosed to be from a bilaterally affected child, however, since the

clinical evidence of bilateral retinoblastoma is not definitive (two small white retinal areas were

ablated with cryotherapy, no images recorded), no RB1 mutations were found in either tumor nor

blood sample of the patient and the M3-Mn profile including high-level MYCN amplification are

Page 47: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

31

similar to the RB1+/+

MYCNA retinoblastomas, this sample has been subsequently included in the

analysis as an RB1+/+

retinoblastoma.

Figure 1. M3-Mn profile of M3-Mn copy number in 139 primary retinoblastomas and 6 cell lines. The unilateral tumors are grouped by RB1 mutation status, RB1+/+, RB1+/-, RB1-/-. Each row represents an individual tumor or cell line while across the top is the genes KIF14, DEK, E2F3, CDH11, and MYCN. Pink indicates gain; green amplification; blue loss; white no change; and gray copy number not determined. For copy numbers of each gene see Table 6 in Appendices.

Page 48: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

32

Table 4 Frequencies of M3-Mn changes in RB1+/+

versus RB1-/-

retinoblastomas

KIF14 DEK E2F3 CDH11 MYCN Gain

(2.5-10)

MYCN Amplificati

on (>10)

France RB1+/+

RB1-/-

0/5 6/10

1/5 6/10

1/5 6/10

0/5 6/10

2/5 9/10

2/5 0/10

Germany RB1+/+

RB1-/-

3/17 5/11

9/17 6/11

7/17 6/11

3/17 5/11

10/17 11/11

3/17 0/11

Toronto RB1+/+

RB1-/-

3/8 45/70

1/8 41/70

0/8 42/70

2/8 38/70

2/8 25/91

4/8 0/91

New Zealand RB1+/+

RB1-/-

1/2 NA

0/2 NA

0/2 NA

1/2 NA

1/2 NA

1/2 NA

Pooled RB1+/+

RB1-/-

7/32 56/91

11/32 53/91

8/32 54/91

6/32 49/91

15/32 45/112

10/32 0/112

P-Value 2.57 x 10-4 0.034 1.71 x 10-3 1.25 x 10-3 0.635 9.55 x 10-9

2.5.2.2 Whole genomic profiling of RB1+/+ retinoblastomas

DNA copy number alterations (CNA) are present in almost all tumor cells. CNAs can range in

sizes from a few kilobases to whole chromosomal arm deletions or amplifications. Thus sub-

megabase resolution tiling array CGH (SMRT aCGH) (Ishkanian, Malloff et al. 2004) was used

to profile and identify CNAs in 49 primary retinoblastomas and 22 of corresponding blood

samples and 6 cell lines. The 49 primary retinoblastomas included 11 RB1+/+

MYCNA, 13

RB1+/+

, 15 RB1+/-

, 10 RB1-/-

(Figure 2). The 6 cell lines included 5 retinoblastoma cell lines

(RB247, RB383, RB1021, WERI and Y79) and one neuroblastoma cell line, IMR32 (Figure 2).

Both Y79 and IMR32 are well characterized and long known to have MYCN amplification (Reid,

Albert et al. 1974; Schwab, Ellison et al. 1984). A complete list of alterations and sizes of each

CNA is listed in Table 7 Appendices. CNAs were divided into two categories: whole

chromosomal arm changes and segmental DNA copy changes (Figure 2).

Page 49: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

33

Figure 2. Summary of chromosomal changes for 47 primary retinoblastomas, 5 retinoblastoma cell lines and 1 neuroblastoma cell line, IMR32. Samples are grouped into RB1+/+MYCNA, RB1+/+, RB1+/-, RB1-/-, RB1-/- bilateral and Cell lines. A blue box indicates the presence of at least one segmental change on the chromosome arm and a red box represents a whole arm alteration. Case numbers are listed to the left and chromosomal regions are listed at the top.

Page 50: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

34

CNAs are a contiguous segment of aberrant DNA as defined by the algorithm CNA HMMer that

is separated by normal DNA or a CNA in the opposite direction. The size of each CNA can be

measured as the number of aberrant base pairs contained in the altered DNA segment. RB1+/+

MYCNA retinoblastomas did not show a significant difference in number of CNAs per tumor

when pair-wise comparisons were made between RB1+/+

, RB1+/-

, and RB1-/-

retinoblastomas

(Mann-Whitney test, P<0.05) except for when compared to the retinoblastoma and

neuroblastoma cell lines (Mann-Whitney test, P value = 0.012) (Figure 3).

Figure 3. Number of CNAs per retinoblastoma tumors. Horizontal lines represent the mean while vertical bars represent standard error of the mean. Breakpoints were determined by CNA Hmmer. X and Y chromosomes were excluded from the analysis.

However, when pair-wise comparisons of number of aberrant base pairs between tumor types

were performed it was found that there was a significant difference between RB1+/+

MYCNA and

RB1-/-

retinoblastomas (Mann-Whitney test, P value = 0.0433) and between RB1+/+

MYCNA and

the cell lines (P value = 0.0160). Overall, RB1+/+

MYCNA retinoblastomas had fewer aberrant

base pairs in their genomes than RB1-/-

retinoblastomas and cell lines (Figure 4) consistent with

reports that MYCN-amplified tumors are more genomically stable (Chen, Bilke et al. 2005;

Mosse, Diskin et al. 2007). Figure 5 shows a typical RB1+/+

MYCNA retinoblastoma with few

genomic alterations.

Page 51: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

35

Figure 4. Number of aberrant base pairs in the different subtypes of retinoblastoma. Breakpoints were determined by CNA Hmmer. X and Y chromosomes were excluded from the analysis.

In the 11 RB1+/+

MYCNA retinoblastomas, chromosome 1q was gained in 3 (27%) samples,

chromosome 6p was gained in 1 (9%) sample and chromosome 16q was lost in 4 (36%) samples,

however loss of 16q in RB2237 occurred at 16q24.1-qter and did not encompass CDH11. None

of the 11 RB1+/+

MYCNA retinoblastomas had rearrangements on chromosome 13. In contrast to

RB1+/-

and RB1-/-

retinoblastomas with low-level gain of the whole 2p arm, RB1+/+

retinoblastomas showed a small, highly amplified region specifically at the MYCN locus (Figure

6).

Page 52: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

36

Figure 5. Whole genome tiling path array CGH karyogram of RB1+/+MYCNA retinoblastoma FA793. RB is shown. Each dark blue dot on the karyogram represents the average signal ratio for an individual BAC clone from the array. Clones were plotted vertically against known chromosomal position. Log2 signal intensity ratios for each clone were plotted horizontally, with colored vertical lines denoting log2 signal ratios from -0.5 to 0.5. Where the signal intensity ratio equals zero (purple line), equivalent DNA copy number between the sample and the reference DNA was inferred. DNA copy number increases were indicated by log2>0 (red line) and losses indicated by log2<0 (green line). MYCN amplification is magnified in the orange box.

Page 53: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

37

Figure 6. Specific amplification of the MYCN locus in RB1+/+ MYCNA RB1348. This is contrasted by low-level whole 2p gain seen in RB1777 (RB1+/-).

In the 13 RB1+/+

with 2-copy number MYCN, chromosome 1q was gained in 5 (38%) samples,

chromosome 6p was gained in 5 (38%) samples, chromosome 16q was lost in 1 (8%) samples

and chromosome 13q was normal in 11 samples. Two RB1+/+

retinoblastomas with 2-copy

number MYCN, MC561 and MC887, showed loss of chromosome 13 that did not encompass

RB1 but involved 13q32.1-ter and 13q21.1-ter, respectively, both telomeric to RB1. In the 16

RB1+/-

retinoblastomas chromosome 1q was gained in 4 (27%) samples, chromosome 6p was

gained in 7 (44%) samples and chromosome 16q was lost in 1 (7%) samples. One RB1+/-

retinoblastoma sample RB2285 showed MYCN amplification but normal copy numbers in

chromosomes 1q, 6p and 16q and normal copy number in the rest of the 2p arm. The one non-

germline RB1 mutation found in this sample consisted of a deletion of one copy of most of the

13q arm, including RB1. This sample showed high similarity to the RB1+/+

samples with MYCN

amplification, including an early age of diagnosis (4 months). It is hypothesized that the loss of

13q in this tumor may be a secondary event and that this sample may in fact belong in the RB1+/+

group with MYCN amplification. In the 10 RB1-/-

retinoblastomas analyzed, chromosome 1q was

Page 54: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

38

gained in 5 (50%) samples, chromosome 6p was gained in 5 (50%) samples, chromosome 16q

was lost in 8 (80%) samples and rearrangements and deletions of chromosome 13 occurred in 4

(40%) consistent with their deleted RB1 gene status (data not shown). In the 5 retinoblastoma

cell lines chromosome 1q was gained in 4 (80%), chromosome 6p was gained in 3 (60%),

chromosome 16q was lost in 3 (60%) and 2p was gained in 4 (80%). Y79 and IMR32 showed

amplification of the MYCN locus but normal copy of the rest of the 2p arm. IMR32 showed

gain of 1q, gain of 6p and consistent with a previous report (Spieker, van Sluis et al. 2001) had a

second amplicon on chromosome 2p14 which included only two genes, MEIS and ETAA16.

2.5.2.3 Minimal MYCN amplicon in RB1+/+ MYCNA retinoblastomas

To determine the size of the minimal MYCN amplicon, SeeGH software was used to perform a

multiple alignment of 14 samples that showed amplification of MYCN. These included 11

primary RB1+/+

retinoblastomas (RB1348, RB1700, RB2237, RB2285, RB2532, MA72, MA94,

MC945, FA337, FA793, NZ499J, and RB522), one RB1+/-

retinoblastoma RB2285, and the Y79

retinoblastoma and IMR32 neuroblastoma cell lines (Figure 7). The minimal amplification spans

448 kb, located on chromosome 2, cytoband 2p24.3, between BAC clones RP11-451A14 and

RP11-463P22, and between base pairs 15703698 and 16152619. The minimal region of

amplification was bounded by primary sample RB2285 and Y79 and contains only the MYCN

gene (see Figure 7 dark blue box). Excluding the two cell lines and using only the primary

retinoblastomas to determine the minimal region of overlap, the region was bounded by samples

RB2285 and FA337 (see Figure 7 pink box). This region was 513 kb in size and still only

contained MYCN. Both of the minimal regions of overlap excluded neighboring genes such as

NAG, DDX1, FAM49A and FAM84A. The NAG and DDX1 genes are commonly co-amplified

with MYCN in neuroblastomas (Weber, Imisch et al. 2004), so the exclusion of NAG and DDX1

is significant since the importance of co-amplified surrounding genes has been disputed in a

number of neuroblastoma studies (Squire, Thorner et al. 1995; Weber, Imisch et al. 2004; De

Preter, Speleman et al. 2005; Weber, Starke et al. 2006; Kaneko, Ohira et al. 2007).

Page 55: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

39

Figure 7. The minimal MYCN amplicon. The minimal region of amplification contains only the MYCN oncogene within cytoband 2p24.3, based on genomic amplification in 11 RB1+/+MYCNA retinoblastomas, one RB1+/- primary retinoblastomas, and two cell lines, Y79 and IMR32. It spans 448kb. It lies between BAC clones RP11-451A14 and RP11-463P22, and between base pairs 15703698 and 16152619. The dark blue line highlights the smallest minimal amplicon including the two amplified cell lines Y79 and IMR32 and is bounded by primary retinoblastoma RB2285 and the cell line Y79. The pink band underneath indicates the common region of gain in the primary retinoblastomas that is bounded by RB2285 and FA337.

2.5.2.4 Detection of translocation breakpoints

Translocation is a chromosomal aberration caused by a recombination event. Translocations can

be balanced, resulting in no net loss or gain of DNA, or unbalanced resulting in duplication or

loss of DNA. Translocations are common in precancerous and progressing tumor cells and can

not only cause loss or gain of DNA but can cause silencing of genes or form fusion genes that

lead to tumorigenesis. Thus the precise identification of chromosomal breakpoints is important

in the characterization of events that could contribute to oncogenesis. Recently it was shown that

Page 56: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

40

translocations that appeared balanced by cytogenetic characterization methods such as spectral

karyotyping (SKY) were discovered on SMRT aCGH to be associated with focal DNA CNAs

that could pinpoint with higher resolution the location of the breakpoint (Watson, deLeeuw et al.

2007). The cell line derived from primary retinoblastoma RB381 was previously characterized

using SKY (Corson, personal communication) and it was determined that despite detecting no

mutation in RB1 by the methods described above, a portion of chromosome 13 was found to be

translocated to chromosome 8 der(8)t(8;13)(q21;q14) ins(13;8)(q14;?q?q) however the exact

location of the second translocation could not be identified. By SMRT aCGH on the original

primary tumor DNA from RB381, the breakpoints for der(8)t(8;13)(q21;q14) can now be

confirmed and pinpointed to their chromosomal sub-bands der(8)t(8;13)(q21.2;q14.12) and the

breakpoint for the chromosome 8 insertion into the chromosome 13q14.12 can be determined to

be at sub bands 8q21.2-23.3 ins(13;8)(q14; q21.2-q23.3) (Figure 8).

Figure 8. RB381 der(8)t(8;13)(q21.2;q14.12) ins(13;8)(q14; q21.2-q23.3) translocation. A. (left) SeeGH karyogram of chromosome 8. (center) SKY data (Tim Corson, unpublished data) for the translocation. (right) SeeGH karyogram of chromosome 13. Red lines indicate breakpoints corresponding to both SMRT array and SKY data (Tim Corson, unpublished data). Yellow line indicates REXO1L1 gene. Green line indicates RB1 gene. B. CNA loss of 0.5 Mb at locus 8q21.2. C. CNA loss of 0.6 Mb at locus 8q23.3. D. CNA loss of 0.9 Mb at locus 13q14.2.

Page 57: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

41

The gene REXO1L1 is located in the small region of loss at chromosome 8q21.2 breakpoint and

is a known site of copy number variation (CNV), in humans and chimpanzees (Perry, Tchinda et

al. 2006). CNVs are naturally occurring structural variations that can be found across

populations and are not usually pathologic however it is postulated that they can contribute to

phenotypic variation and inherent susceptibility to diseases. Indeed it was found that 13

retinoblastomas and RB247 cell line from all four types of RB1 mutation status groups showed

CNV of the REXO1L1 gene. Blood DNA for RB381 did not show deletion at 8q21.2. However,

array CGH showed that tumor sample RB2052 had complex rearrangements on chromosomes 13

and 8 while the corresponding blood had a normal karyogram except for deletion of REXO1L1 in

blood. In addition, 3 other samples RB2532, RB2903, RB2589 that showed REXO1L1 deletion

in tumor also had deletion in blood DNA consistent with the observation that REXO1L1 CNV is

a common in the population. However, whether it predisposes to chromosomal rearrangement in

retinoblastoma needs to be further investigated.

2.5.3 Expression of RB1 and MYCN mRNA transcripts and protein in RB1+/+ retinoblastomas

Whether RB1+/+

genomic status corresponded with expression of mRNA and full-length

retinoblastoma protein (pRB) in the tumor (Figure 9) was verified next. To do this, primers were

designed that targeted the RB1 and MYCN transcripts and reverse transcriptase PCR was used to

analyze levels of the transcript in two RB1+/+

retinoblastomas for which mRNA was available.

The expected 2.8 kb coding-containing transcript of RB1 was detected in two of the primary

RB1+/+

retinoblastomas compared to normal retina and MYCN transcript was detected in fetal

retina and two primary RB1+/+

retinoblastoma but not adult retina as expected (Figure 9A). Ki67

transcript, indicative of proliferation, was expressed abundantly in fetal retina and in the two

primary tumors but not in adult retina. To confirm the presence of full-length pRB protein,

formalin-fixed, paraffin-embedded slides from three primary RB1+/+

retinoblastomas (RB2237 is

shown) were stained with two different pRB antibodies, recognizing an N-terminus and the other

a C-terminus epitope (Figure 9B). All three tumors stained positively with both pRB antibodies

suggesting that full-length pRB was expressed. The tumor but not adjacent retina stained

strongly with MYCN antibody confirming that tumors amplified for MYCN expressed abundant

protein (Figure 9C).

Page 58: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

42

Figure 9. Expression of pRB and MYCN in primary human retinoblastoma and normal retina. A. RT-PCR was performed using primers that spanned the entire 2.8kb coding region of RB1 and primers spanning the 283 bp coding region of MYCN to determine if RB1+/+ MYCNA

retinoblastomas MA94 and FA793 expressed full-length RB1 transcript and to look for presence of MYCN transcript. Normal fetal and adult retinas were included as positive controls. Presence of Ki67 mRNA transcript in fetal and both RB1+/+ MYCNA retinoblastomas indicates proliferation while absence of Ki67 mRNA was expected in non-proliferative adult retina. TBP housekeeping gene was included as a loading control. B and C. Paraffin sections of RB1+/+ MYCNA

retinoblastoma RB2237 were stained with pRB C-terminus, pRB N-terminus and MYCN antibodies. Positively staining cells are indicated by green fluorescent or DAB (brown) staining. Scale bar in b is 50 µm in c is 100 µm.

2.5.4 Clinical features of RB1+/+ retinoblastomas

The median age at diagnosis of the 12 RB1+/+

MYCNA including RB2285 (RB1

+/-) was 6 months

(Figure 10). This is significantly younger than the median ages of diagnosis of 15 RB1+/+

with

2-copy MYCN, 10 RB1+/-

and 147 RB1-/-

retinoblastomas randomly selected from the Toronto

data set which were diagnosed at 23, 24, and 24 months respectively (P-value 0.0023, one-way

ANOVA). A previous study also reported a median age of diagnosis of 23 months in unilateral

retinoblastomas (Schüler, Weber et al. 2005).

Four enucleated RB1+/+

MYCNA retinoblastomas RB2237, RB522, NZ499, and MA94 were

stained with hematoxylin and eosin and assessed for histological features of retinoblastoma

(Table 5). In addition, two pathology reports were available for RB1348 and RB1700. None of

the tumors had Flexner-Wintersteiner rosettes in the sections analyzed, which are specific for

retinoblastoma and seen in 70% of cases (Poulaki and Mukai 2009). All of the tumors were

described as large filling most of the eye, showed undifferentiated cells, large areas of necrosis

and calcification. Apoptosis was present in RB2237, RB522, NZ499 and, MA94.

Page 59: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

43

Of note is that RB2237, RB522 and NZ499 displayed large prominent nucleoli (Figure 11), a

feature that is atypical of retinoblastoma but common in other neuroectodermal or embryonal

type tumors such as neuroblastoma (Tornoczky, Semjen et al. 2007). Large nucleolar

neuroblastomas are associated with a high incidence of MYCN amplification and are also largely

undifferentiated (Tornoczky, Semjen et al. 2007).

Figure 10. Age of diagnosis of 11 RB1+/+ MYCNA retinoblastomas. Samples include RB2973 (RB1+/-) (circle), 15 RB1+/+ retinoblastomas with 2-copy MYCN (square), 10 RB1+/- retinoblastomas (triangle) and 147 RB1-/- retinoblastomas (diamond). Lines represent medians.

Page 60: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

44

Table 5 Summary of retinoblastoma histopathological features in RB1+/+

MYCNA retinoblastomas

Pathologic features

RB1348 (report only)

RB1700 (report only)

RB522 RB2237 MA94 NZ499 RB2903 (2-copy MYCN )

Flexner-Wintersteiner

rosettes

- - - - - - +

Homer-Wright NS + - + - - -

Pseudorosettes - - - - - - -

Retinoma - - - - - - -

Mitotic figures NS NS + + + + +

Necrosis + + + + + + +

Calcification + + + + + + +

Vitreous seeding

NS NS + + - + +

Nuclear moulding

NS NS - - - - +

Apoptosis NS NS + + + + +

Optic nerve involvement

- - NS - - - -

Choroid and Sclera

involvement

+ + - - - + -

Anterior segment

NS - - - - - -

Large prominent

nuclei

NS NS + + - + -

NS, Not scored

Page 61: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

45

Figure 11. Large prominent nucleoli in two RB1+/+MYCNA retinoblastomas, RB2237 and NZ499. RB1+/- retinoblastoma RB2903 is shown for contrast.

2.5.5 Functional consequence of MYCN silencing in retinoblastoma with high levels of MYCN

The retinoblastoma cell line Y79 has genomic amplification of MYCN and expresses high levels

of MYCN protein and mRNA. Gene-specific QM-PCR indicated that Y79 has 53 copies of

MYCN. To determine whether decreased MYCN expression would affect the growth and cellular

fate of RB cell lines with MYCNA amplification, 5 different shRNA-expressing viruses (Mission

shRNA system, Sigma) targeting the human MYCN transcript were used to transduce Y79 cells.

All 5 shRNA vectors could efficiently knockdown MYCN expression in Y79 cells (Figure 12A).

Four out of 5 Y79 clones stably transduced with shRNA vectors showed decreased growth rate

compared to Y79 cells transduced with empty vector (Figure 12B).

Page 62: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

46

A B

Figure 12. MYCN shRNA knockdown in Y79 retinoblastoma cells. A) Western blot showing MYCN and β-tubulin protein expression in Y79 cells infected with virus vector carrying the empty PLKO construct or one of 5 different shRNAs targeting different regions of the MYCN transcript. B) Growth curve of Y79 cells infected with empty pLKO vector or 5 shRNAs targeting MYCN. For each day and each shRNA, three wells (24-well plate) were counted and averaged. Error bars show standard deviation of triplicate samples.

2.5.6 Construction of a Mycn-overexpression lentivirus

Similar to the TH-MYCN murine model for MYCN-amplified neuroblastoma (Weiss, Aldape et

al. 1997), it is hypothesized that overexpression of MYCN in the mouse retina during early

development would result in tumor growth. To determine if overexpression of Mycn is sufficient

to initiate tumors during murine retinal development, a Mycn-overexpression lentiviral construct

was developed for injection into eyes of P0 mice of various genetic backgrounds. Lentiviral

delivery of the oncogene driven by a strong viral promoter, human cytomegalovirus (CMV), was

chosen since it is not known which cell type in the retina is the tumor-initiating cell in RB1+/+

MYCNA retinoblastoma. Three Mycn-expression vectors; all containing a truncated human CMV

promoter to drive Mycn expression and two vectors that co-expressed EGFP were constructed

(Figure. 13A). Mycn cDNA was cloned from fetal mouse cDNA and the sequence was verified

in the lentiviral plasmids. HEK293 cells were infected with the three viruses followed by

western blot analysis to determine level of Mycn expression. Mycn protein was detected in

infected HEK293 cells by all three constructs (Fig. 13B).

Page 63: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

47

Figure 13. Lentiviral overexpression of Mycn in HEK293 cells. A) pSY-hCMV-Mycn-F (pJLM backbone, truncated human CMV, mouse Mycn, C-terminal Flag), pSY-hCMV-MycnF-IRES-WPRE (pJLM backbone, truncated human CMV, mouse Mycn, C-terminal Flag, internal ribosomal entry site, enhanced green fluorescent protein, woodchuck hepatitis post-transcriptional regulatory element), pSY- hCMV- MycnF-EGFP-WPRE (pJLM backbone, truncated human CMV, mouse Mycn, C-terminal Flag, human phosphoglycerate kinase promoter, enhanced green fluorescent protein, woodchuck hepatitis post-transcriptional regulatory element). B) Expression of Mycn protein in lentivirus infected HEK293 cells. Only cells infected with pSY-hCMV-MycnF, pSY-hCMV-MycnF-IRES-WPRE, and pSY-hCMV-MycnF-EGFP-WPRE lentivirus constructs expressed murine Mycn protein. Cells were either untreated, infected with pJLM-eGFP-Flag control and pJLM empty vectors or pSY-hCMV-MycnF lentiviral constructs. Cells were harvested lysed and western blot analysis was performed using anti-Mycn antibody and anti-β-tubulin (loading control).

To test whether these viruses could infect the murine retina, P0 murine retinal explants were

dissected and infected with lentivirus followed by immunohistochemistry to determine level and

pattern of expression of Mycn protein. Fluorescence was detected in cells infected by virus with

EGFP; however, Mycn protein was not detected in paraffin embedded sections (data not shown).

This may indicate that the human CMV promoter may not drive Mycn expression in murine

cells. Replacement of the CMV promoter with the murine CMV intermediate early promoter 1

(mCMV IE1) is underway since the mCMV IE1 promoter has been shown to drive strong

expression of exogenous proteins in a variety of cell types (Dorsch-Hasler, Keil et al. 1985).

Page 64: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

48

Chapter 3

3 Discussion

3.1.1 RB1+/+ MYCNA retinoblastoma is observed in independent clinical samples

The RB1+/+

MYCNA retinoblastoma subset was initially identified in clinical retinoblastoma

samples at Retinoblastoma Solutions when mutations could not be identified in 7/441 unilateral

retinoblastoma tumors. Four (57%) of these samples showed high-level MYCN amplification

with no other M3-Mn gene copy changes. In this study, collaborations were set up with three

other centers from around the world, each independently performing similar clinical RB1 gene

testing, to collect data for a total of 1023 unilateral retinoblastomas. It was determined that each

center had a similar frequency of RB1+/+

MYCNA retinoblastomas and overall, the frequency of

RB1+/+

MYCNA retinoblastomas was 3%. It is expected that future studies involving samples to

be collected from Africa, China and India will yield a similar frequency of RB1+/+

MYCNA

retinoblastomas.

3.1.2 RB1+/+ MYCNA: a novel genetic subset of retinoblastoma

This study showed that RB1+/+

MYCNA retinoblastomas display a very different genetic signature

to the well-characterized RB1-/-

retinoblastomas. They possess fewer copy number gains and

losses in the M3-Mn genes, KIF14, DEK, E2F3, and CDH11. Instead, 7/11 RB1+/+

MYCNA

retinoblastomas showed normal copy numbers for those genes. Mutation analysis for each of the

M3-Mn genes was not performed in this study, thus there remains a possibility that activating or

inactivating point mutations exist in the potential oncogenes and tumor suppressor genes

respectively, however there has been no evidence to date suggesting that M3-Mn genes are

mutated in retinoblastoma.

With a larger sample set combined with optimized protocol for gene-specific QM-PCR for M3-

Mn genes as previously described (Bowles, Corson et al. 2007), it was observed that the

frequencies of M3-Mn changes in RB1-/-

retinoblastomas are actually higher than previously

thought. Of note is that the frequencies of DEK and E2F3 gains were reported as 40% and 70%

respectively however in this study, in almost every sample where gain of DEK was observed the

Page 65: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

49

same level gain was observed in E2F3 which is consistent with the fact that both genes are

within close proximity to each other on chromosome 6p22.

Consistent with the observations that RB1+/+

MYCNA retinoblastomas display fewer M3-Mn

gene-specific changes, whole genomic profiling showed that overall, RB1+/+

MYCNA

retinoblastomas had fewer altered base pairs than RB1-/-

retinoblastomas further supporting that

they are a distinct genetic subset of retinoblastoma and are more genomically stable. This

observation is consistent with the array CGH study by Mosse et al showing that neuroblastoma

samples with MYCN amplification had overall less complex genome-wide pattern of CNAs

(Mosse, Diskin et al. 2007).

It was also confirmed that the minimal amplified region contained only the MYCN gene. These

results suggest that MYCN-amplification drives tumorigenesis in this unique subset of

retinoblastomas that may be similar to neuroblastomas with high-level MYCN amplification. In

further support that RB1+/+

MYCNA retinoblastomas are similar to neuroblastomas with MYCN-

amplification, it was shown that RB1+/+

MYCNA retinoblastoma histology was uncharacteristic of

conventional retinoblastoma but similar to large nucleolar neuroblastomas, most notably they

shared the feature of multiple large prominent nucleoli. Overexpression of MYCN in

neuroblastoma cells strongly upregulates genes involved in ribosome biogenesis (Boon, Caron et

al. 2001). rRNAs and genes involved in ribosome biogenesis reside in nucleoli, which provides

an explanation for the increased nucleoli size in MYCN-amplified cells.

3.1.3 MYCN-driven tumorigenesis

Several lines of evidence in this thesis and other studies support the hypothesis that MYCN-

amplification drives tumorigenesis. This study showed that RB1+/+

MYCNA retinoblastomas

expressed high levels of MYCN transcript. RB1+/+

MYCNA retinoblastomas displayed histology

that suggest an activated cell state most likely involving increased ribosome biogenesis and

proliferation. These tumors presented at a much earlier age than RB1-/-

retinoblastomas

suggesting that they may initiate earlier, grow faster and/or may be more aggressive. In vitro,

this study showed that inhibition of MYCN in the highly proliferative and MYCN-amplified

retinoblastoma cell line, Y79, significantly decreased proliferation. This study also showed that

fetal retina, but not adult retina, expressed high levels of MYCN transcript (see Figure 9A)

consistent with studies that show normal fetal retina and brain express high levels of MYCN

Page 66: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

50

(Squire, Goddard et al. 1986; Grady, Schwab et al. 1987; Martins, Zindy et al. 2008). Given that

amplification of a MYCN transgene expressed in a precursor neuronal cell occurs in the TH-

MYCN murine model, it can be hypothesized that perhaps it is a normal precursor retinal cell

expressing high levels of MYCN that gives rise to the MYCN-amplified retinoblastoma cell.

Failure to downregulate MYCN and differentiate during embryogenesis may give this cell a

proliferative advantage. Due to as yet unknown factors, this cell may begin to amplify MYCN

providing a selective advantage, giving rise to a larger and earlier presenting tumor than a tumor

initiated by loss of RB1. Future in vivo studies involving the Mycn overexpression in the murine

retina with wild-type Rb1 may validate this model of MYCN-driven tumorigenesis.

3.1.4 Chromosome 8;13 translocation

In this study, it was found that a focal deletion of the gene REXO1L1 was associated with

translocation of chromosome 8 to chromosome 13 in one sample RB381 and that many

retinoblastoma samples regardless of RB1 status exhibited CNV at this gene, mostly in the form

of deletion and in two cases it was gained. CNV at REXO1L1 is common in both chimpanzees

and humans (Perry, Tchinda et al. 2006). Though most CNVs are common and usually

considered benign variations in the human genome, they can be associated with complex genetic

diseases (Zhang, Gu et al. 2009). Due to the inherent instability at those regions, they can be

hotspots for genomic structural rearrangements. Nonallelic homologous recombination (NAHR)

is one mechanism postulated to cause CNVs. It is the process by which paralogous genes,

different genes which share highly similar DNA sequences, realign and crossover during meiosis

or mitosis resulting in deletion or fusion of genes. NAHR occurring on separate chromosomes

can result in chromosomal translocations (Lupski 1998). In this study, tumor sample RB381

showed deletion of REXO1L1 at 8q21.2 but normal in blood. However, it was observed that 3

samples in which REXO1L1 was deleted in both tumor and blood DNA. It is tempting to

speculate that this common CNV could be a potential recombination hotspot that when combined

with other factors could predispose an individual to t(8;13) translocations. To investigate this

hypothesis further, a larger sample set of tumor and corresponding blood DNA combined with

cytogenetic analyses would be required.

Page 67: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

51

3.1.5 MYCN copy number as a rapid screen for RB1+/+MYCNA

retinoblastoma

This study has shown that retinoblastomas with high-level MYCN amplification are a distinct

subset characterized by the absence of RB1 mutations and lack of the retinoblastoma genetic

signature. Based on these observations, it may be possible to quickly distinguish RB1+/+

MYCNA

retinoblastomas from RB1-/-

retinoblastomas by performing a rapid quantitative-PCR screen for

MYCN copy number as an initial test before performing standard RB1 gene testing. In

conjunction with clinical data: age of diagnosis and histology of tumors, the identification of

samples with high level MYCN amplification would suggest that an RB1 mutation might not be

found. Functional assays such as immunohistochemistry to show the presence of pRB might be

performed before the full RB1 gene testing. In addition, identified RB1+/+

MYCNA patients could

benefit from MYCN-specific therapies that may become available in the future.

3.1.6 Targeting MYCN

Several groups have developed various methods that inhibit MYCN. Ornithine decarboxylase 1

(ODC1) is a rate limiting enzyme in the polyamine synthesis pathway and bona fide target of

MYCN (Hogarty, Norris et al. 2008). Using a MYCN reporter construct consisting of a luciferase

reporter driven by the ornithine decarboxylase (ODC1) promoter, Lu et al developed a chemical

screen for small molecules that could inhibit MYCN/MAX dimerization in a neuroblastoma cell

line and successfully identified 8 compounds that could reduce luciferase activity by at least 50%

(Lu, Pearson et al. 2003). A few groups have targeted ODC1 directly using the Odc inhibitor

alpha-difluoromethylornithine (DFMO) with the reasoning that polyamine synthesis is essential

for cell growth and proliferation (Hogarty, Norris et al. 2008; Rounbehler, Li et al. 2009).

MYCN has also been suggested to be a potential target for immunotherapy. In a very different

approach, Himoudi et al proposed that MYCN would be a good “candidate antigen” to elicit a

specific and sustained immune response since it satisfies three essential criteria; it is expressed

highly in tumor tissue, it is virtually undetectable in normal tissue and is required for

tumorigenesis and will likely not be downregulated by the tumor cell (Himoudi, Yan et al. 2008).

The authors performed a proof-of-principle experiment to show that MYCN is indeed

immunogenic. T cell lines from normal blood donors could be stimulated to kill MYCN

expressing cells and importantly, cytotoxic T-cells specific for MYCN could be generated from a

neuroblastoma patient with advanced disease indicating that the generation of a MYCN peptide

Page 68: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

52

vaccine is a potential avenue for therapy (Himoudi, Yan et al. 2008). Using yet another

approach, an anti-gene peptide nucleic acid (PNA) was developed to inhibit MYCN transcription

(Tonelli, Purgato et al. 2005). The PNA consisted of an antisense sequence targeting the second

exon of MYCN conjugated to a peptide with a nuclear localization signal. The researchers

developed the PNA in an effort to overcome the limitation that antisense oligonucleotides are

rapidly degraded. They showed that the anti-gene PNA could inhibit MYCN transcription and

protein production and could inhibit growth of neuroblastoma cells (Tonelli, Purgato et al. 2005).

Altogether, these studies indicate there is great interest in developing a MYCN-specific therapy

that could one day be used to treat RB1+/+

MYCNA

retinoblastomas.

3.1.7 Future directions

3.1.7.1 RB1+/+ retinoblastomas

By the most current methods, mutations in the RB1+/+

retinoblastomas could not be detected.

Though it was shown that RB+/+

MYCNA retinoblastomas were a distinct subset, it cannot be

ruled that there may be deep intronic mutations, promoter mutations and translocations that are

not detectable by current technologies. With the advent of next generation sequencing, a future

study could resequence the entire RB1 gene in all RB1+/+

retinoblastomas to look for such

mutations. This approach has the potential to first further delineate RB+/+

MYCNA

retinoblastomas from RB1+/+

retinoblastomas with 2-copy MYCN grouping them as either RB1-/-

or an entirely different subset with no RB1 mutations or MYCN amplification and secondly find

the second mutations in RB1+/-

retinoblastomas that would categorize them as RB1-/-

retinoblastomas.

3.1.7.2 Functional status of pRB in RB1+/+ MYCNA retinoblastomas

This study showed that RB1+/+

MYCNA retinoblastoma express full-length pRB, however it has

yet to be determined whether the pRB expressed is functional. The p16 is a potent inhibitor of

CDK4, which phosphorylates and inactivates pRB. Overexpression of p16 therefore has the

effect of activating pRB and arresting cells in G1 phase. The cell line RB522 is derived from a

primary tumor in the mid-1980s and was identified to be RB1+/+

MYCNA (Godbout and Squire

1993). These results were confirmed in this current study. To test whether pRB is functions

normally in RB1+/+

MYCNA retinoblastomas, a future experiment can be set up such that p16 is

exogenously expressed in RB522 cell line and pRB phosphorylation and cell cycle status

Page 69: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

53

assayed. If pRB is hypophosphorylated and G1 cell cycle arrest is induced, this would indicate

the presence of functional pRB and would suggest that MYCN induces proliferation independent

of the RB pathway.

3.1.7.3 Determine effect of MYCN amplification in extraocular retinoblastoma

This study has provided new insight to the role of MYCN in retinoblastoma which can perhaps be

applied to other neuroectodermal tumors, especially neuroblastoma where MYCN amplification

has prognostic significance. In retinoblastoma, it is not clear whether MYCN amplification

causes more aggressive disease since in developed nations, a high cure rate (>95%) is achieved

by surgically removing the affected eye (Chintagumpala, Chevez-Barrios et al. 2007). However

in developing nations where metastasis is more prevalent, extraocular tumors can be analyzed for

MYCN amplification to determine if there is a correlation with more aggressive disease. In

conjunction with the clinical analysis of extraocular tumors in developing nations, an in vivo

experiment can be set up to test whether Mycn amplification causes more aggressive disease and

extraocular spread by injecting the eyes of nude mice with either the WERI cell line which does

not cause metastasis (Chevez-Barrios, Hurwitz et al. 2000), or the RB522 cell line, which this

study has identified as RB1+/+

MYCNA.

3.1.7.4 In vivo model of MYCNA in mice with intact pRB

It is expected that MYCN overexpression alone will initiate tumors in the murine retina similar to

the TH-MYCN murine model of neuroblastoma (Weiss, Aldape et al. 1997). A lentiviral MYCN-

overexpression vector was developed for the purpose of injection into the murine retina in this

study. When human embryonic kidney cells were transduced with virus, Mycn protein was

detected compared to cells transduced with an empty control virus. However, when murine

retinal explants were transduced no protein expression was detected indicating that the human

CMV promoter was incompatible with murine cells. This experiment highlights an important

obstacle in the development of RB+/+

MYCNA retinoblastoma model. It is important to find a

promoter that can achieve expression in the majority of cell types since it is not known which

cell in the retina is the tumor initiating cell. Future experiments must first replace the human

CMV promoter with a promoter that can drive Mycn expression to develop the model. Three

potential promoters are initially proposed: the Pax 6 α-enhancer, which has been used to drive

Page 70: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

54

expression of Cre recombinase in Rbf/f

; p107-/-

mice, the enhancer that drives expression in SV40

large Tag retinoblastoma mouse model (Windle, Albert et al. 1990) and is active in Müller Glia

cells and finally the PCAN1 promoter which is expressed in the neural retina (Cross, Reding et

al. 2004). Once a promoter is selected and the model developed, tumor growth, histology of

tumors and CGH could be performed to determine whether syntenic regions to human

retinoblastoma are altered. This model could be used to validate therapies for MYCN-amplified

retinoblastoma as well as other tumors of neuroectodermal origin.

Page 71: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

55

References

Abel, F., K. Ejeskar, et al. (1999). "Gain of chromosome arm 17q is associated with

unfavourable prognosis in neuroblastoma, but does not involve mutations in the

somatostatin receptor 2(SSTR2) gene at 17q24." Br J Cancer 81(8): 1402-9.

Abel, F., R. M. Sjoberg, et al. (2002). "Analyses of apoptotic regulators CASP9 and DFFA at

1P36.2, reveal rare allele variants in human neuroblastoma tumours." Br J Cancer 86(4):

596-604.

Alaminos, M., W. L. Gerald, et al. (2005). "Prognostic value of MYCN and ID2 overexpression

in neuroblastoma." Pediatr Blood Cancer 45(7): 909-15.

Ambros, I. M., J. Hata, et al. (2002). "Morphologic features of neuroblastoma (Schwannian

stroma-poor tumors) in clinically favorable and unfavorable groups." Cancer 94(5): 1574-

83.

Amler, L. C., J. Schurmann, et al. (1996). "The DDX1 gene maps within 400 kbp 5' to MYCN

and is frequently coamplified in human neuroblastoma." Genes Chromosomes Cancer

15(2): 134-7.

Amler, L. C. and M. Schwab (1989). "Amplified N-myc in human neuroblastoma cells is often

arranged as clustered tandem repeats of differently recombined DNA." Mol Cell Biol

9(11): 4903-13.

Aslanian, A., P. J. Iaquinta, et al. (2004). "Repression of the Arf tumor suppressor by E2F3 is

required for normal cell cycle kinetics." Genes Dev 18(12): 1413-22.

Barr, F. G., F. Duan, et al. (2009). "Genomic and clinical analyses of 2p24 and 12q13-q14

amplification in alveolar rhabdomyosarcoma: a report from the Children's Oncology

Group." Genes Chromosomes Cancer 48(8): 661-72.

Bayani, J., M. Zielenska, et al. (2000). "Molecular cytogenetic analysis of medulloblastomas and

supratentorial primitive neuroectodermal tumors by using conventional banding,

comparative genomic hybridization, and spectral karyotyping." J Neurosurg 93(3): 437-

48.

Boer, J., J. Bonten-Surtel, et al. (1998). "Overexpression of the nucleoporin CAN/NUP214

induces growth arrest, nucleocytoplasmic transport defects, and apoptosis." Mol Cell Biol

18(3): 1236-47.

Boon, K., H. N. Caron, et al. (2001). "N-myc enhances the expression of a large set of genes

functioning in ribosome biogenesis and protein synthesis." EMBO J 20(6): 1383-93.

Bowles, E., T. W. Corson, et al. (2007). "Profiling genomic copy number changes in

retinoblastoma beyond loss of RB1." Genes Chromosomes Cancer 46(2): 118-29.

Page 72: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

56

Bown, N. (2001). "Neuroblastoma tumour genetics: clinical and biological aspects." J Clin

Pathol 54(12): 897-910.

Brodeur, G. M., C. Azar, et al. (1992). "Neuroblastoma. Effect of genetic factors on prognosis

and treatment." Cancer 70(6 Suppl): 1685-94.

Brodeur, G. M., R. C. Seeger, et al. (1984). "Amplification of N-myc in untreated human

neuroblastomas correlates with advanced disease stage." Science 224(4653): 1121-4.

Casas, S., B. Nagy, et al. (2003). "Aberrant expression of HOXA9, DEK, CBL and CSF1R in

acute myeloid leukemia." Leuk Lymphoma 44(11): 1935-41.

Cavenee, W. K., T. P. Dryja, et al. (1983). "Expression of recessive alleles by chromosomal

mechanisms in retinoblastoma." Nature 305(5937): 779-84.

Chan, H. S., B. L. Gallie, et al. (1997). "MYCN protein expression as a predictor of

neuroblastoma prognosis." Clin Cancer Res 3(10): 1699-706.

Chen, D., B. L. Gallie, et al. (2001). "Minimal regions of chromosomal imbalance in

retinoblastoma detected by comparative genomic hybridization." Cancer Genet Cytogenet

129(1): 57-63.

Chen, D., S. Pajovic, et al. (2002). "Genomic amplification in retinoblastoma narrowed to 0.6

megabase on chromosome 6p containing a kinesin-like gene, RBKIN." Cancer Res 62(4):

967-71.

Chen , P. M., Wenzel P, Knoepfler PS, Leone G, Bremner R (2009). "Division and apoptosis of

E2f-deficient retinal progenitors." Nature.

Chen, Q. R., S. Bilke, et al. (2005). "High-resolution cDNA microarray-based comparative

genomic hybridization analysis in neuroblastoma." Cancer Lett 228(1-2): 71-81.

Chen, Y., J. Takita, et al. (2008). "Oncogenic mutations of ALK kinase in neuroblastoma."

Nature 455(7215): 971-4.

Chen, Z., Y. Lin, et al. (2009). "Mdm2 deficiency suppresses MYCN-Driven neuroblastoma

tumorigenesis in vivo." Neoplasia 11(8): 753-62.

Cheng, A. J., N. C. Cheng, et al. (2007). "Cell lines from MYCN transgenic murine tumours

reflect the molecular and biological characteristics of human neuroblastoma." Eur J

Cancer 43(9): 1467-75.

Chevez-Barrios, P., M. Y. Hurwitz, et al. (2000). "Metastatic and nonmetastatic models of

retinoblastoma." Am J Pathol 157(4): 1405-12.

Chi, B., R. J. DeLeeuw, et al. (2004). "SeeGH--a software tool for visualization of whole

genome array comparative genomic hybridization data." BMC Bioinformatics 5: 13.

Page 73: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

57

Chintagumpala, M., P. Chevez-Barrios, et al. (2007). "Retinoblastoma: review of current

management." Oncologist 12(10): 1237-46.

Chow, R. L. and R. A. Lang (2001). "Early eye development in vertebrates." Annu Rev Cell Dev

Biol 17: 255-96.

Connolly, M. J., R. H. Payne, et al. (1983). "Familial, EsD-linked, retinoblastoma with reduced

penetrance and variable expressivity." Hum Genet 65(2): 122-4.

Corson, T. W. and B. L. Gallie (2006). "KIF14 mRNA expression is a predictor of grade and

outcome in breast cancer." Int J Cancer 119(5): 1088-94.

Corson, T. W. and B. L. Gallie (2007). "One hit, two hits, three hits, more? Genomic changes in

the development of retinoblastoma." Genes Chromosomes Cancer 46(7): 617-34.

Corson, T. W., A. Huang, et al. (2005). "KIF14 is a candidate oncogene in the 1q minimal region

of genomic gain in multiple cancers." Oncogene 24(30): 4741-53.

Corson, T. W., C. Q. Zhu, et al. (2007). "KIF14 messenger RNA expression is independently

prognostic for outcome in lung cancer." Clin Cancer Res 13(11): 3229-34.

Cotterman, R., V. X. Jin, et al. (2008). "N-Myc regulates a widespread euchromatic program in

the human genome partially independent of its role as a classical transcription factor."

Cancer Res 68(23): 9654-62.

Cowling, V. H. and M. D. Cole (2006). "Mechanism of transcriptional activation by the Myc

oncoproteins." Semin Cancer Biol 16(4): 242-52.

Cross, D., D. J. Reding, et al. (2004). "Expression and initial promoter characterization of

PCAN1 in retinal tissue and prostate cell lines." Med Oncol 21(2): 145-53.

Dang, C. V., K. A. O'Donnell, et al. (2006). "The c-Myc target gene network." Semin Cancer

Biol 16(4): 253-64.

De Preter, K., F. Speleman, et al. (2005). "No evidence for correlation of DDX1 gene

amplification with improved survival probability in patients with MYCN-amplified

neuroblastomas." J Clin Oncol 23(13): 3167-8; author reply 3168-70.

Devesa, S. S. (1975). "The incidence of retinoblastoma." Am J Ophthalmol 80(2): 263-5.

DiCiommo, D., B. L. Gallie, et al. (2000). "Retinoblastoma: the disease, gene and protein

provide critical leads to understand cancer." Semin Cancer Biol 10(4): 255-69.

DiCiommo, D. P., A. Duckett, et al. (2004). "Retinoblastoma protein purification and

transduction of retina and retinoblastoma cells using improved alphavirus vectors." Invest

Ophthalmol Vis Sci 45(9): 3320-9.

Page 74: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

58

Dietzsch, E., R. E. Lukeis, et al. (1994). "Characterization of homogeneously staining regions in

a small cell lung cancer cell line, using in situ hybridization with an MYCN probe."

Genes Chromosomes Cancer 10(3): 213-6.

Dimaras, H., V. Khetan, et al. (2008). "Loss of RB1 induces non-proliferative retinoma:

increasing genomic instability correlates with progression to retinoblastoma." Hum Mol

Genet 17(10): 1363-72.

Dorsch-Hasler, K., G. M. Keil, et al. (1985). "A long and complex enhancer activates

transcription of the gene coding for the highly abundant immediate early mRNA in

murine cytomegalovirus." Proc Natl Acad Sci U S A 82(24): 8325-9.

Draper, G. J., B. M. Sanders, et al. (1986). "Second primary neoplasms in patients with

retinoblastoma." Br J Cancer 53(5): 661-71.

Dryja, T. P., J. M. Rapaport, et al. (1986). "Chromosome 13 homozygosity in osteosarcoma

without retinoblastoma." Am J Hum Genet 38(1): 59-66.

Dyer, M. A. and C. L. Cepko (2001). "Regulating proliferation during retinal development." Nat

Rev Neurosci 2(5): 333-42.

Fix, A., C. Lucchesi, et al. (2008). "Characterization of amplicons in neuroblastoma: high-

resolution mapping using DNA microarrays, relationship with outcome, and

identification of overexpressed genes." Genes Chromosomes Cancer 47(10): 819-34.

Fontana, L., M. E. Fiori, et al. (2008). "Antagomir-17-5p abolishes the growth of therapy-

resistant neuroblastoma through p21 and BIM." PLoS One 3(5): e2236.

Friend, S. H., R. Bernards, et al. (1986). "A human DNA segment with properties of the gene

that predisposes to retinoblastoma and osteosarcoma." Nature 323(6089): 643-6.

Frischmeyer, P. A. and H. C. Dietz (1999). "Nonsense-mediated mRNA decay in health and

disease." Hum Mol Genet 8(10): 1893-900.

Fruhwald, M. C., M. S. O'Dorisio, et al. (2000). "Gene amplification in

PNETs/medulloblastomas: mapping of a novel amplified gene within the MYCN

amplicon." J Med Genet 37(7): 501-9.

George, R. E., T. Sanda, et al. (2008). "Activating mutations in ALK provide a therapeutic target

in neuroblastoma." Nature 455(7215): 975-8.

Gillison, M. L., R. Chen, et al. (2007). "Human retinoblastoma is not caused by known pRb-

inactivating human DNA tumor viruses." Int J Cancer 120(7): 1482-90.

Godbout, R. and J. Squire (1993). "Amplification of a DEAD box protein gene in retinoblastoma

cell lines." Proc Natl Acad Sci U S A 90(16): 7578-82.

Grace, E., J. Drennan, et al. (1971). "The 13q- deletion syndrome." J Med Genet 8(3): 351-7.

Page 75: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

59

Grady, E. F., M. Schwab, et al. (1987). "Expression of N-myc and c-src during the development

of fetal human brain." Cancer Res 47(11): 2931-6.

Grenet, J., V. Valentine, et al. (1998). "Duplication of the DR3 gene on human chromosome

1p36 and its deletion in human neuroblastoma." Genomics 49(3): 385-93.

Grottke, C., K. Mantwill, et al. (2000). "Identification of differentially expressed genes in human

melanoma cells with acquired resistance to various antineoplastic drugs." Int J Cancer

88(4): 535-46.

Gruneberg, U., R. Neef, et al. (2006). "KIF14 and citron kinase act together to promote efficient

cytokinesis." J Cell Biol 172(3): 363-72.

Guo, C., P. S. White, et al. (1999). "Allelic deletion at 11q23 is common in MYCN single copy

neuroblastomas." Oncogene 18(35): 4948-57.

Hamann, U., A. Wenzel, et al. (1991). "The MYCN protein of human neuroblastoma cells is

phosphorylated by casein kinase II in the central region and at serine 367." Oncogene

6(10): 1745-51.

Hansford, L. M., W. D. Thomas, et al. (2004). "Mechanisms of embryonal tumor initiation:

distinct roles for MycN expression and MYCN amplification." Proc Natl Acad Sci U S A

101(34): 12664-9.

Henriksson, M., A. Bakardjiev, et al. (1993). "Phosphorylation sites mapping in the N-terminal

domain of c-myc modulate its transforming potential." Oncogene 8(12): 3199-209.

Herzog, S., D. R. Lohmann, et al. (2001). "Marked differences in unilateral isolated

retinoblastomas from young and older children studied by comparative genomic

hybridization." Hum Genet 108(2): 98-104.

Himoudi, N., M. Yan, et al. (2008). "MYCN as a target for cancer immunotherapy." Cancer

Immunol Immunother 57(5): 693-700.

Hodgson, J. G., R. F. Yeh, et al. (2009). "Comparative analyses of gene copy number and mRNA

expression in glioblastoma multiforme tumors and xenografts." Neuro Oncol 11(5): 477-

87.

Hogarty, M. D. (2003). "The requirement for evasion of programmed cell death in

neuroblastomas with MYCN amplification." Cancer Lett 197(1-2): 173-9.

Hogarty, M. D., M. D. Norris, et al. (2008). "ODC1 is a critical determinant of MYCN

oncogenesis and a therapeutic target in neuroblastoma." Cancer Res 68(23): 9735-45.

Houdayer, C., M. Gauthier-Villars, et al. (2004). "Comprehensive screening for constitutional

RB1 mutations by DHPLC and QMPSF." Hum Mutat 23(2): 193-202.

Page 76: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

60

Hui, A. B., K. W. Lo, et al. (2001). "Detection of multiple gene amplifications in glioblastoma

multiforme using array-based comparative genomic hybridization." Lab Invest 81(5):

717-23.

Humbert, P. O., R. Verona, et al. (2000). "E2f3 is critical for normal cellular proliferation."

Genes Dev 14(6): 690-703.

Hurlin, P. J. (2005). "N-Myc functions in transcription and development." Birth Defects Res C

Embryo Today 75(4): 340-52.

Hurlin, P. J., C. Queva, et al. (1997). "Mnt, a novel Max-interacting protein is coexpressed with

Myc in proliferating cells and mediates repression at Myc binding sites." Genes Dev

11(1): 44-58.

Hurst, C. D., D. C. Tomlinson, et al. (2008). "Inactivation of the Rb pathway and overexpression

of both isoforms of E2F3 are obligate events in bladder tumours with 6p22

amplification." Oncogene 27(19): 2716-27.

Iavarone, A., P. Garg, et al. (1994). "The helix-loop-helix protein Id-2 enhances cell proliferation

and binds to the retinoblastoma protein." Genes Dev 8(11): 1270-84.

Ibson, J. M. and P. H. Rabbitts (1988). "Sequence of a germ-line N-myc gene and amplification

as a mechanism of activation." Oncogene 2(4): 399-402.

Ishkanian, A. S., C. A. Malloff, et al. (2004). "A tiling resolution DNA microarray with complete

coverage of the human genome." Nat Genet 36(3): 299-303.

Jacobs, J. F., H. van Bokhoven, et al. (2009). "Regulation of MYCN expression in human

neuroblastoma cells." BMC Cancer 9: 239.

Janoueix-Lerosey, I., D. Lequin, et al. (2008). "Somatic and germline activating mutations of the

ALK kinase receptor in neuroblastoma." Nature 455(7215): 967-70.

Janoueix-Lerosey, I., G. Schleiermacher, et al. (2009). "Overall genomic pattern is a predictor of

outcome in neuroblastoma." J Clin Oncol 27(7): 1026-33.

Kamil, J. P., A. J. Hume, et al. (2009). "Human papillomavirus 16 E7 inactivator of

retinoblastoma family proteins complements human cytomegalovirus lacking UL97

protein kinase." Proc Natl Acad Sci U S A 106(39): 16823-8.

Kaneko, S., M. Ohira, et al. (2007). "Relationship of DDX1 and NAG gene

amplification/overexpression to the prognosis of patients with MYCN-amplified

neuroblastoma." J Cancer Res Clin Oncol 133(3): 185-92.

Kappes, F., K. Burger, et al. (2001). "Subcellular localization of the human proto-oncogene

protein DEK." J Biol Chem 276(28): 26317-23.

Page 77: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

61

Kappes, F., C. Damoc, et al. (2004). "Phosphorylation by protein kinase CK2 changes the DNA

binding properties of the human chromatin protein DEK." Mol Cell Biol 24(13): 6011-

20.

Kappes, F., I. Scholten, et al. (2004). "Functional domains of the ubiquitous chromatin protein

DEK." Mol Cell Biol 24(13): 6000-10.

Khojasteh, M., W. L. Lam, et al. (2005). "A stepwise framework for the normalization of array

CGH data." BMC Bioinformatics 6: 274.

Knudson, A. G., Jr. (1971). "Mutation and cancer: statistical study of retinoblastoma." Proc Natl

Acad Sci U S A 68(4): 820-3.

Kohl, N. E., N. Kanda, et al. (1983). "Transposition and amplification of oncogene-related

sequences in human neuroblastomas." Cell 35(2 Pt 1): 359-67.

Kondoh, N., T. Wakatsuki, et al. (1999). "Identification and characterization of genes associated

with human hepatocellular carcinogenesis." Cancer Res 59(19): 4990-6.

Kusnetsova, L. E., E. L. Prigogina, et al. (1982). "Similar chromosomal abnormalities in several

retinoblastomas." Hum Genet 61(3): 201-4.

Lasorella, A., A. Iavarone, et al. (1996). "Id2 specifically alters regulation of the cell cycle by

tumor suppressor proteins." Mol Cell Biol 16(6): 2570-8.

Lasorella, A., G. Rothschild, et al. (2005). "Id2 mediates tumor initiation, proliferation, and

angiogenesis in Rb mutant mice." Mol Cell Biol 25(9): 3563-74.

Lavarino, C., N. K. Cheung, et al. (2009). "Specific gene expression profiles and chromosomal

abnormalities are associated with infant disseminated neuroblastoma." BMC Cancer 9:

44.

Lee, W. H., R. Bookstein, et al. (1987). "Human retinoblastoma susceptibility gene: cloning,

identification, and sequence." Science 235(4794): 1394-9.

Lee, W. H., A. L. Murphree, et al. (1984). "Expression and amplification of the N-myc gene in

primary retinoblastoma." Nature 309(5967): 458-60.

Lele, K. P., L. S. Penrose, et al. (1963). "CHROMOSOME DELETION IN A CASE OF

RETINOBLASTOMA." Ann Hum Genet 27: 171-4.

Leone, G., F. Nuckolls, et al. (2000). "Identification of a novel E2F3 product suggests a

mechanism for determining specificity of repression by Rb proteins." Mol Cell Biol

20(10): 3626-32.

Leung, J. Y., G. L. Ehmann, et al. (2008). "A role for Myc in facilitating transcription activation

by E2F1." Oncogene 27(30): 4172-9.

Page 78: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

62

Lillington, D. M., L. K. Goff, et al. (2002). "High level amplification of N-MYC is not

associated with adverse histology or outcome in primary retinoblastoma tumours." Br J

Cancer 87(7): 779-82.

Lillington, D. M., J. E. Kingston, et al. (2003). "Comparative genomic hybridization of 49

primary retinoblastoma tumors identifies chromosomal regions associated with

histopathology, progression, and patient outcome." Genes Chromosomes Cancer 36(2):

121-8.

Lin, P. and J. M. O'Brien (2009). "Frontiers in the management of retinoblastoma." Am J

Ophthalmol 148(2): 192-8.

Lohmann, D. R. and B. L. Gallie (2004). "Retinoblastoma: revisiting the model prototype of

inherited cancer." Am J Med Genet C Semin Med Genet 129C(1): 23-8.

Lu, X., A. Pearson, et al. (2003). "The MYCN oncoprotein as a drug development target."

Cancer Lett 197(1-2): 125-30.

Lupski, J. R. (1998). "Genomic disorders: structural features of the genome can lead to DNA

rearrangements and human disease traits." Trends Genet 14(10): 417-22.

Mairal, A., E. Pinglier, et al. (2000). "Detection of chromosome imbalances in retinoblastoma by

parallel karyotype and CGH analyses." Genes Chromosomes Cancer 28(4): 370-9.

Marchong, M. N., D. Chen, et al. (2004). "Minimal 16q genomic loss implicates cadherin-11 in

retinoblastoma." Mol Cancer Res 2(9): 495-503.

Marchong, M. N., C. Yurkowski, et al. (2009). "Cdh11 acts as a tumor suppressor in a murine

retinoblastoma model by facilitating tumor cell death."

Marees, T., A. C. Moll, et al. (2008). "Risk of second malignancies in survivors of

retinoblastoma: more than 40 years of follow-up." J Natl Cancer Inst 100(24): 1771-9.

Martelli, F., C. Cenciarelli, et al. (1994). "MyoD induces retinoblastoma gene expression during

myogenic differentiation." Oncogene 9(12): 3579-90.

Martins, R. A., F. Zindy, et al. (2008). "N-myc coordinates retinal growth with eye size during

mouse development." Genes Dev 22(2): 179-93.

Matsunaga, E. (1980). "Hereditary retinoblastoma: host resistance and second primary tumors." J

Natl Cancer Inst 65(1): 47-51.

Matthay, K. K. (2000). "MYCN expression in neuroblastoma: A mixed message?" J Clin Oncol

18(21): 3591-4.

Mitter, D., D. Rushlow, et al. (2009). "Identification of a mutation in exon 27 of the RB1 gene

associated with incomplete penetrance retinoblastoma." Fam Cancer 8(1): 55-8.

Page 79: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

63

Moreau, L. A., P. McGrady, et al. (2006). "Does MYCN amplification manifested as

homogeneously staining regions at diagnosis predict a worse outcome in children with

neuroblastoma? A Children's Oncology Group study." Clin Cancer Res 12(19): 5693-7.

Mosse, Y. P., S. J. Diskin, et al. (2007). "Neuroblastomas have distinct genomic DNA profiles

that predict clinical phenotype and regional gene expression." Genes Chromosomes

Cancer 46(10): 936-49.

Mosse, Y. P., M. Laudenslager, et al. (2008). "Identification of ALK as a major familial

neuroblastoma predisposition gene." Nature 455(7215): 930-5.

Nau, M. M., B. J. Brooks, Jr., et al. (1986). "Human small-cell lung cancers show amplification

and expression of the N-myc gene." Proc Natl Acad Sci U S A 83(4): 1092-6.

Orlic, M., C. E. Spencer, et al. (2006). "Expression analysis of 6p22 genomic gain in

retinoblastoma." Genes Chromosomes Cancer 45(1): 72-82.

Otterson, G. A., W. Chen, et al. (1997). "Incomplete penetrance of familial retinoblastoma linked

to germ-line mutations that result in partial loss of RB function." Proc Natl Acad Sci U S

A 94(22): 12036-40.

Paderova, J., M. Orlic-Milacic, et al. (2007). "Novel 6p rearrangements and recurrent

translocation breakpoints in retinoblastoma cell lines identified by spectral karyotyping

and mBAND analyses." Cancer Genet Cytogenet 179(2): 102-11.

Pandita, A., R. Godbout, et al. (1997). "Relational mapping of MYCN and DDXI in band 2p24

and analysis of amplicon arrays in double minute chromosomes and homogeneously

staining regions by use of free chromatin FISH." Genes Chromosomes Cancer 20(3):

243-52.

Patel, J. H., A. P. Loboda, et al. (2004). "Analysis of genomic targets reveals complex functions

of MYC." Nat Rev Cancer 4(7): 562-8.

Peirce, S. K. and H. W. Findley (2009). "High level MycN expression in non-MYCN amplified

neuroblastoma is induced by the combination treatment nutlin-3 and doxorubicin and

enhances chemosensitivity." Oncol Rep 22(6): 1443-9.

Perry, G. H., J. Tchinda, et al. (2006). "Hotspots for copy number variation in chimpanzees and

humans." Proc Natl Acad Sci U S A 103(21): 8006-11.

Pogosianz, H. E. and L. E. Kuznetsova (1986). "Nonrandom chromosomal changes in

retinoblastomas." Arch Geschwulstforsch 56(2): 135-43.

Poulaki, V. and S. Mukai (2009). "Retinoblastoma: genetics and pathology." Int Ophthalmol

Clin 49(1): 155-64.

Raizis, A., R. Clemett, et al. (2002). "Improved clinical management of retinoblastoma through

gene testing." N Z Med J 115(1154): 231-4.

Page 80: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

64

Raizis, A. M., F. Schmitt, et al. (1995). "A bisulfite method of 5-methylcytosine mapping that

minimizes template degradation." Anal Biochem 226(1): 161-6.

Ramsay, G., L. Stanton, et al. (1986). "Human proto-oncogene N-myc encodes nuclear proteins

that bind DNA." Mol Cell Biol 6(12): 4450-7.

Reid, T. W., D. M. Albert, et al. (1974). "Characteristics of an established cell line of

retinoblastoma." J Natl Cancer Inst 53(2): 347-60.

Richter, S., K. Vandezande, et al. (2003). "Sensitive and efficient detection of RB1 gene

mutations enhances care for families with retinoblastoma." Am J Hum Genet 72(2): 253-

69.

Rounbehler, R. J., W. Li, et al. (2009). "Targeting ornithine decarboxylase impairs development

of MYCN-amplified neuroblastoma." Cancer Res 69(2): 547-53.

Rushlow, D., B. Piovesan, et al. (2009). "Detection of mosaic RB1 mutations in families with

retinoblastoma." Hum Mutat 30(5): 842-51.

Salido, M., E. Arriola, et al. (2009). "Cytogenetic characterization of NCI-H69 and NCI-H69AR

small cell lung cancer cell lines by spectral karyotyping." Cancer Genet Cytogenet

191(2): 97-101.

Sampieri, K., M. Amenduni, et al. (2009). "Array comparative genomic hybridization in

retinoma and retinoblastoma tissues." Cancer Sci 100(3): 465-71.

Schouten, J. P., C. J. McElgunn, et al. (2002). "Relative quantification of 40 nucleic acid

sequences by multiplex ligation-dependent probe amplification." Nucleic Acids Res

30(12): e57.

Schüler, A., S. Weber, et al. (2005). "Age at diagnosis of isolated unilateral retinoblastoma does

not distinguish patients with and without a constitutional RB1 gene mutation but is

influenced by a parent-of-origin effect." Eur J Cancer 41(5): 735-40.

Schwab, M. (1999). "Human neuroblastoma: from basic science to clinical debut of cellular

oncogenes." Naturwissenschaften 86(2): 71-8.

Schwab, M. (2004). "MYCN in neuronal tumours." Cancer Lett 204(2): 179-87.

Schwab, M., J. Ellison, et al. (1984). "Enhanced expression of the human gene N-myc

consequent to amplification of DNA may contribute to malignant progression of

neuroblastoma." Proc Natl Acad Sci U S A 81(15): 4940-4.

Scott, D. K., J. R. Board, et al. (2003). "The neuroblastoma amplified gene, NAG: genomic

structure and characterisation of the 7.3 kb transcript predominantly expressed in

neuroblastoma." Gene 307: 1-11.

Shah, S. P., X. Xuan, et al. (2006). "Integrating copy number polymorphisms into array CGH

analysis using a robust HMM." Bioinformatics 22(14): e431-9.

Page 81: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

65

Slack, A., Z. Chen, et al. (2005). "The p53 regulatory gene MDM2 is a direct transcriptional

target of MYCN in neuroblastoma." Proc Natl Acad Sci U S A 102(3): 731-6.

Sparkes, R. S., M. C. Sparkes, et al. (1980). "Regional assignment of genes for human esterase D

and retinoblastoma to chromosome band 13q14." Science 208(4447): 1042-4.

Spieker, N., P. van Sluis, et al. (2001). "The MEIS1 oncogene is highly expressed in

neuroblastoma and amplified in cell line IMR32." Genomics 71(2): 214-21.

Squire, J., B. L. Gallie, et al. (1985). "A detailed analysis of chromosomal changes in heritable

and non-heritable retinoblastoma." Hum Genet 70(4): 291-301.

Squire, J., A. D. Goddard, et al. (1986). "Tumour induction by the retinoblastoma mutation is

independent of N-myc expression." Nature 322(6079): 555-557.

Squire, J. A., P. S. Thorner, et al. (1995). "Co-amplification of MYCN and a DEAD box gene

(DDX1) in primary neuroblastoma." Oncogene 10(7): 1417-22.

Stanton, L. W. and J. M. Bishop (1987). "Alternative processing of RNA transcribed from

NMYC." Mol Cell Biol 7(12): 4266-72.

Stanton, L. W., M. Schwab, et al. (1986). "Nucleotide sequence of the human N-myc gene." Proc

Natl Acad Sci U S A 83(6): 1772-6.

Stirzaker, C., D. S. Millar, et al. (1997). "Extensive DNA methylation spanning the Rb promoter

in retinoblastoma tumors." Cancer Res 57(11): 2229-37.

Tang, X. X., H. Zhao, et al. (2006). "The MYCN enigma: significance of MYCN expression in

neuroblastoma." Cancer Res 66(5): 2826-33.

Tonelli, R., S. Purgato, et al. (2005). "Anti-gene peptide nucleic acid specifically inhibits MYCN

expression in human neuroblastoma cells leading to cell growth inhibition and

apoptosis." Mol Cancer Ther 4(5): 779-86.

Tornoczky, T., D. Semjen, et al. (2007). "Pathology of peripheral neuroblastic tumors:

significance of prominent nucleoli in undifferentiated/poorly differentiated

neuroblastoma." Pathol Oncol Res 13(4): 269-75.

van den Heuvel, S. and N. J. Dyson (2008). "Conserved functions of the pRB and E2F families."

Nat Rev Mol Cell Biol 9(9): 713-24.

van der Wal, J. E., M. A. Hermsen, et al. (2003). "Comparative genomic hybridisation divides

retinoblastomas into a high and a low level chromosomal instability group." J Clin Pathol

56(1): 26-30.

Vietri, M., M. Bianchi, et al. (2006). "Direct interaction between the catalytic subunit of Protein

Phosphatase 1 and pRb." Cancer Cell Int 6: 3.

Page 82: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

66

von Lindern, M., D. Breems, et al. (1992). "Characterization of the translocation breakpoint

sequences of two DEK-CAN fusion genes present in t(6;9) acute myeloid leukemia and a

SET-CAN fusion gene found in a case of acute undifferentiated leukemia." Genes

Chromosomes Cancer 5(3): 227-34.

Watson, S. K., R. J. deLeeuw, et al. (2007). "Cytogenetically balanced translocations are

associated with focal copy number alterations." Hum Genet 120(6): 795-805.

Weber, A., P. Imisch, et al. (2004). "Coamplification of DDX1 correlates with an improved

survival probability in children with MYCN-amplified human neuroblastoma." J Clin

Oncol 22(13): 2681-90.

Weber, A., S. Starke, et al. (2006). "The coamplification pattern of the MYCN amplicon is an

invariable attribute of most MYCN-amplified human neuroblastomas." Clin Cancer Res

12(24): 7316-21.

Wei, J. S., Y. K. Song, et al. (2008). "The MYCN oncogene is a direct target of miR-34a."

Oncogene 27(39): 5204-13.

Weiss, W. A., K. Aldape, et al. (1997). "Targeted expression of MYCN causes neuroblastoma in

transgenic mice." EMBO J 16(11): 2985-95.

Wen, J. and S. Brogna (2008). "Nonsense-mediated mRNA decay." Biochem Soc Trans 36(Pt 3):

514-6.

Wenzel, A. and M. Schwab (1995). "The mycN/max protein complex in neuroblastoma. Short

review." Eur J Cancer 31A(4): 516-9.

White, P. S., J. M. Maris, et al. (1995). "A region of consistent deletion in neuroblastoma maps

within human chromosome 1p36.2-36.3." Proc Natl Acad Sci U S A 92(12): 5520-4.

Wilson, M. G., J. W. Towner, et al. (1973). "Retinoblastoma and D-chromosome deletions." Am

J Hum Genet 25(1): 57-61.

Windle, J. J., D. M. Albert, et al. (1990). "Retinoblastoma in transgenic mice." Nature

343(6259): 665-9.

Woo, C. W., F. Tan, et al. (2008). "Use of RNA interference to elucidate the effect of MYCN on

cell cycle in neuroblastoma." Pediatr Blood Cancer 50(2): 208-12.

Zhang, F., W. Gu, et al. (2009). "Copy number variation in human health, disease, and

evolution." Annu Rev Genomics Hum Genet 10: 451-81.

Zielinski, B., S. Gratias, et al. (2005). "Detection of chromosomal imbalances in retinoblastoma

by matrix-based comparative genomic hybridization." Genes Chromosomes Cancer

43(3): 294-301.

Page 83: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

67

Appendices

Table 6 Copy numbers of M3-Mn genes in retinoblastomas as measured by QM-PCR

Sample AOD (months)

RB1 KIF14 DEK E2F3 CDH11 MYCN

RB1348 9 +/+ 3.92 2.23 2.30 1.77 38

RB1700 7 +/+ 2.18 2.33 2.18 1.13 49

RB2237 1 +/+ 2.14 2.24 2.11 2.13 76

RB2532 16 +/+ 2.07 2.40 1.99 1.83 73

MA72 4.5 +/+ 1.86 1.66 2.13 1.90 48

MA94 4.3 +/+ 2.14 2.85 2.98 2.47 43

MC945 12.5 +/+ 3.13 2.74 2.34 1.61 57

FA337 12 +/+ 2.16 2.29 2.11 2.38 121

FA793 3 +/+ 2.05 2.15 2.01 2.09 54

NZ499J 10 +/+ 2.37 2.04 1.73 1.93 93

RB522 2 +/+ 2.62 2.01 2.12 2.09 33

RB2285 4 +/- 2.10 1.75 1.71 2.17 73

Total 2/10 (20%) 2/10 (20%) 1/10 (10%) 1/10 (10%) 10/10 (100%)

RB818 38.5 +/+ 1.96 2.31 2.39 2.20 2.02

RB2583 56.5 +/+ 2.25 1.98 2.05 1.88 3.17

MA43 83 +/+ 2.55 2.66 2.33 1.20 2.59

MA89 47 +/+ 2.06 1.76 2.25 2.54 2.12

MC140 24.5 +/+ 3.20 7.82 5.98 1.55 5.65

MC336 23 +/+ 2.04 3.95 3.42 1.68 4.48

MC385 17 +/+ 2.20 3.00 2.90 1.85 4.06

MC431 8.5 +/+ 2.38 2.60 2.46 2.08 4.25

MC561 18 +/+ 2.29 2.17 2.03 1.84 3.00

MC887 45.5 +/+ 1.97 2.32 2.04 2.80 4.21

MC972 10.5 +/+ 2.12 4.36 3.66 1.81 3.34

FA319 20 +/+ 2.15 3.50 3.27 1.82 2.99

FA448 8 +/+ 2.02 1.81 1.91 1.93 1.67

FA502 63 +/+ 1.81 1.63 1.66 1.87 3.35

NZ945 15 +/+ 3.44 2.25 2.06 1.16 3.72

Total 2/16 (15%) 7/16 (44%) 5/16 (31%) 3/16 (19%) 11/16 (69%)

Total RB1

+/+MYCN

A

and RB1+/+

4/26 (15%) 9/26 (35%) 6/26 (23%) 4/26 (15%) 10/26 (38%)

Toronto, Canada and Essen, Germany

RB2903 9 +/- 2.19 2.30 2.02 1.91 1.77

RB 3132 NI +/- 3.29 2.74 2.29 1.58 3.90

RB 2285 4 +/- 2.10 1.75 1.71 2.17 73

RB 374 NI +/- 1.90 2.18 2.00 1.68 2.10

RB 1451 NI +/- 2.05 2.46 2.30 1.99 1.60

RB 1466 NI +/- 1.92 2.85 2.33 1.81 3.25

RB 1777 27 +/- 2.73 3.80 3.49 1.83 4.39

Page 84: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

68

Sample AOD (months)

RB1 KIF14 DEK E2F3 CDH11 MYCN

RB 1790 20 +/- 2.39 1.80 1.94 2.01 3.34

RB 1962 NI +/- 3.09 2.50 2.20 1.57 4.27

RB 2625 49 +/- 2.27 4.60 4.20 2.05 3.05

RB 2733 30 +/- 2.90 3.00 3.12 1.06 2.42

RB 1530 54 +/- 3.65 3.21 3.12 1.32 4.21

RB 1979 96 +/- 2.40 2.79 5.11 2.48 6.85

RB 2780 20 +/- 2.36 2.31 2.08 2.10 2.75

RB 2854 12 +/- 1.84 2.17 2.02 1.74 3.45

RB 3100 15 +/- 3.03 3.50 3.55 2.16 4.78

MA41 NI +/- 2.23 1.70 2.36 2.36 2.42

MA49 NI +/- 3.32 3.28 3.87 1.95 3.07

MA80 NI +/- 1.73 1.99 2.15 2.03 2.41

MC951 NI +/- 2.48 2.25 2.15 1.96 4.30

Toronto, CANADA

RB1436 NI -/- NA NA NA NA 3.47

RB613 NI -/- NA NA NA NA 1.60

RB1545 NI -/- 3.03 2.24 2.26 1.01 6.80

RB381 NI -/- 2.57 5.31 4.67 1.20 7.70

RB2589 NI -/- 2.18 2.20 2.21 1.10 2.02

RB2631 53.7 -/- 3.10 4.11 3.82 1.45 1.69

RB2641 NI -/- 2.95 2.87 2.92 1.03 1.66

RB2647 15.7 -/- 2.87 2.35 2.17 1.51 2.03

RB2683 27 -/- 3.40 5.29 5.54 1.45 1.70

RB2687 NI -/- 2.35 3.66 3.52 1.82 2.18

RB2306 59.9 -/- 3.89 1.62 1.95 1.01 2.30

RB2699 NI -/- 2.16 2.06 2.13 1.78 2.90

RB2686 NI -/- 2.78 2.60 2.76 2.13 2.74

RB2639 NI -/- 2.35 2.35 2.24 2.44 3.40

RB2637 45.1 -/- 2.61 4.62 3.77 1.17 2.70

RB2708 NI -/- 3.15 2.90 3.50 1.41 1.48

RB2598 9.4 -/- 2.20 3.92 3.33 1.77 2.60

RB2838 18.4 -/- 2.70 2.21 2.47 1.69 3.57

RB2582 NI -/- 2.28 1.93 1.70 2.16 3.45

RB2274 NI -/- 2.64 4.86 5.27 1.61 9.58

RB2934 NI -/- 2.16 2.94 3.24 1.98 3.98

RB2820 22.9 -/- 2.84 3.68 4.08 1.25 2.14

RB2960 50.6 -/- 4.78 7.25 5.72 1.50 3.59

RB2280 NI -/- 2.07 2.56 1.98 2.20 2.21

RB2391 1.2 -/- 2.73 1.66 2.58 1.17 2.08

RB1796 NI -/- 2.26 2.08 1.89 0.92 1.86

RB1707 NI -/- 3.07 2.95 3.41 0.81 1.99

RB1738 NI -/- 2.27 2.40 2.11 2.25 3.47

RB1760 NI -/- 3.26 3.13 3.53 1.00 2.05

RB2527 32.5 -/- 1.96 1.61 2.20 2.35 1.80

RB1519 NI -/- 1.70 2.88 3.09 1.06 6.07

RB2327 NI -/- 1.83 1.14 1.72 1.07 4.48

MO13 NI -/- 4.09 3.27 2.69 1.03 4.99

MO15 NI -/- 2.98 2.14 2.10 1.85 3.21

MO21 NI -/- 2.69 2.27 2.27 1.49 2.17

Page 85: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

69

Sample AOD (months)

RB1 KIF14 DEK E2F3 CDH11 MYCN

MO33 NI -/- 2.67 2.36 2.06 1.31 3.41

MO37 NI -/- 2.66 3.89 3.38 2.20 2.76

MO38 NI -/- 3.48 2.81 2.91 1.86 3.38

MO39 NI -/- 2.81 4.47 3.66 1.44 3.58

MO41 NI -/- 3.00 4.81 5.26 1.17 2.32

MO45 NI -/- 2.83 3.42 3.91 1.48 3.57

MO46 NI -/- 2.46 2.39 2.50 1.72 4.67

MO49 NI -/- 3.82 3.37 3.31 1.25 3.42

MO50 NI -/- 2.10 1.94 1.87 1.04 2.34

MO637 NI -/- 2.19 3.79 3.74 1.92 1.96

RB1575 NI -/- 2.97 2.64 2.92 0.96 NA

RB2437 14.8 -/- 3.18 2.84 3.22 1.00 NA

RB2621 31 -/- 2.86 4.24 3.77 1.00 NA

RB2651 NI -/- 3.03 7.06 6.14 2.61 NA

RB2669 NI -/- 3.02 2.17 4.14 2.10 NA

RB2670 33.3 -/- 3.29 3.76 4.17 1.10 NA

RB2667 NI -/- 2.76 2.00 1.96 1.84 NA

RB2674 25.4 -/- 4.68 2.51 2.61 1.16 NA

RB2671 NI -/- 5.20 5.40 3.23 1.79 NA

RB2675 8.5 -/- 2.83 NA 2.10 1.06 NA

RB2646 NI -/- 3.43 3.08 3.35 1.03 NA

RB2676 10.4 -/- 2.15 2.79 2.80 2.08 NA

RB2680 NI -/- 2.87 4.19 3.65 1.93 NA

RB2661 22.6 -/- 3.54 1.85 1.73 1.13 NA

RB2630 6.7 -/- 2.45 3.63 2.84 1.69 NA

RB2599 NI -/- 4.46 5.91 5.48 1.01 NA

RB2672 50.2 -/- 3.46 NA 2.19 0.68 NA

RB2591 7.9 -/- 1.91 2.33 2.16 1.72 NA

RB2253 NI -/- 2.57 2.65 2.67 2.30 NA

RB2284 NI -/- 2.76 2.25 2.47 1.59 NA

RB2389 9.8 -/- 2.41 2.16 2.36 2.09 NA

RB2396 13.2 -/- 2.38 2.56 2.55 2.01 NA

RB2409 NI -/- 3.16 3.42 3.20 1.16 NA

RB3110 16.5 -/- 2.17 3.22 2.86 1.59 NA

Essen, GERMANY

MB109 NI -/- 3.00 2.22 1.82 1.10 2.70

MB190 NI -/- 2.10 4.45 3.66 0.98 2.72

MB209 NI -/- 3.30 5.00 3.87 1.71 3.17

MB213 NI -/- 2.03 2.25 1.97 1.78 5.97

MB429 NI -/- 2.17 1.98 1.75 1.00 3.59

MB449 NI -/- 2.11 2.25 1.77 1.94 3.91

MB456 NI -/- 2.06 1.94 1.67 1.89 3.53

MB486 NI -/- 1.83 2.93 2.66 1.29 3.50

MB607 NI -/- 2.63 3.13 2.80 1.31 3.35

MB703 NI -/- 3.96 6.44 5.89 1.68 3.52

MC480 NI -/- 2.04 4.47 4.60 2.03 2.21

Paris, FRANCE

FB014 NI -/- 2.98 4.54 4.00 0.99 2.70

Page 86: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

70

Sample AOD (months)

RB1 KIF14 DEK E2F3 CDH11 MYCN

FB103 NI -/- 2.18 2.26 2.02 1.63 2.60

FB162 NI -/- 2.11 2.43 2.04 1.78 2.30

FB204 NI -/- 2.54 4.38 3.54 1.16 3.18

FB307 NI -/- 3.49 5.95 4.96 1.25 3.18

FB327 NI -/- 4.09 1.77 2.03 1.01 2.86

FB343 NI -/- 2.60 3.63 3.06 0.82 2.94

FB539 NI -/- 2.15 3.51 3.51 1.75 2.73

FB809 NI -/- 3.17 5.04 3.49 0.85 7.36

FB987 NI -/- 2.94 2.57 2.50 2.32 3.74

Total 54/89 (61%)

50/87 (57%)

52/89 (58%)

49/89 (55%)

43/67 (64%)

NI, No information. NA, Not assessed.

Page 87: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

71

Table 7 SMRT aCGH alterations by sample

Sample Name

Locus Start clone End clone Base pair start Base pair end Size (kb)

RB1348

Gains 1q arm 2p24.3

N0510I18 N0451A14

N0068F13 N0065N17

142647117 15703698

246833917 16825910

104187 1122

Deletions

2p24.3 2p24.2 10q25.2-ter 16q arm

N0091E09 N0701F16 N0466I19 N0180E11

N0788G01 N0631D03 N0091E02 N0163C18

14051860 16693859 114682303 45081598

15454023 17748109 135262317 86413580

1402 1054 20580 41332

RB1700 Gains

2p24.3-p24.2 4q33-q35.2 18q21.1 19q13.31

N0451A14 N0132L10 N0776B03 N0653D16

N0723P04 N0555D07 N0774B22 N0160A19

15703698 170864319 42660453 47715841

18180341 191239174 42846788 48626174

2477 20375 186 910

Deletions

11p and q 16q

N0182E22 N0708O13

M2013A02 F0600M14

79527 44997310

134436514 88699594

134357 43702

RB2237 Gains 2p24.3 N0231J10 N0701F16 14410329 16915017 2505

Deletions 10q26.2-ter 16q24.1-q24.3

N0317I13 N0150H19

N0106C07 F0600M14

129057100 83749375

135271097 88699594

6214 4950

RB2532

Gains

2p25.1-25.3 2p24.3-p24.2 2p24.21-2q35.1 14q21.3-q32.33 18q21.1

N0158D10 N0571E19 N0597D07 N0016G17 N0776B03

N0641J22 N0102G08 N0267H19 M2011A05 N0093N16

87030 14565626 21369662 45462518 42660453

11754223 17282900 223774942 106302057 42968870

11667 2717 202405 60840 308

Deletions

1p36.11 2p24.3 2p24.1-p24.2 2q36.1-ter 8q21.2

N0335G20 N0005H04 N0149C19 N0060D20 N0509F16

N0157K08 N0316B08 N0452B12 N0321A15 M2067O20

25454993 11750125 17417308 224111722 86620140

25639555 14632932 21011770 242359512 87055825

185 2882 3594 18247 436

RB522

Gains

1q arm 2p24.2-24.3 6p24.1-25.3 7q31.33-ter 11q14.1-q24.1 17q21.31-ter

N0510I18 N0091E09 N0812K10 N0618G22 N0444N24 M2245G16

N0068F13 N0554B24 N0805G18 N0083D03 N0381C13 N0196O11

142647117 14051860 71610 123744612 77769745 39583819

246833917 18350885 13047861 158783389 122631670 78615238

104187 4299 13041 35039 44862 39031

Deletions 11p13-ter 17p-13.3

N0182E22 N0411G07

M2270H09 N0189D22

79527 415552

32378741 18114697

32299 17699

MA72 Gains 2p24.2-24.3 N0723F23 N0422A06 14978618 17691585 2713

MA94 Gains

2p24.3 6p21.1-ter 14q22.1-ter

N0619O15 N0812K10 M2075E15

N0631D03 N0323A09 M2011A05

15374591 71610 48962539

17177195 44642253 106302057

1803 44571 574340

Deletions 11q14.1-ter N0671D11 M2013A02 83754905 134436514 50682

MC945

Gains

1q21.1-q41 2p25.1-ter 2p24.2-24.3 2p12-2p24.1

N0510I18 N0463H16 N0220H05 N0452B12

N0503C11 N0005H04 N0631D03 N0543B23

142647117 79317 13269910 20842473

212366365 11931175 17177195 80228525

69719 11852 3907 59386

Deletions

8p22-ter 16q12.1-ter 7p11.2-13.3 7q12

N0418D21 N0242N20 N0411G07 N0342F22

N0533K07 F0600M14 N0064J19 N0722D15

30472 50079561 415552 31583727

18942661 88699594 21191548 33809137

18912 38620 20776 2225

Page 88: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

72

Sample Name

Locus Start clone End clone Base pair start Base pair end Size (kb)

FA337

Gains

2p24.3-25.1 14q31.3-ter 17q21.31-ter 19p13.11-ter

N0686G09 N0557O19 N0419E16 N0009F15

N0062M03 N0012F16 N0196O11 N0715L15

10097116 90694710 40291911 189657

16432161 106218118 78615238 19436075

6335 15523 38323 19246

Deletions 8p21.1-ter 8q21.2

N0521M14 N0509F16

N0418D21 N0639P04

30472 86620140

26980569 87082985

26950 463

FA793 Gains 2p24.2-24.3 13q32.1-ter

N0541K19 N0517L15

N0554B24 N0226B11

15467410 96024440

18350885 114103214

2883 18079

NZ499J

Gains 2p24.2-24.3 7q34-ter 18q22.1-22.3

N0723F23 N0119F21 N0607G19

M2305P22 N0083D03 N0399L12

14978618 142897566 60553398

18851300 158783389 67415521

3873 15886 6862

Deletions

1p35.3-ter 2p24.3 2p24.2 4p14-15.1 8q21.2 16q22.1-23.1 17p-17q12

N0045C18 N0571E19 N0424F04 N0325H01 N0509F16 N0598D24 N0411G07

N0026P17 N0592G02 N0118G07 N0260B15 N0639P04 N0594G15 N0592L16

38264 14565626 18945057 30463631 86620140 65200179 415552

29279256 14892009 19624269 37236400 87082985 78178406 34833159

29241 326 679 6773 463 12798 34418

RB818 Deletions 8q21.2 N0509F16 N0639P04 86620140 87082985 463

MA43

Gains

1p34.2-ter 1q21.1-23.2 1q32.1-32.2 1q42.12-44 2p23.2-ter 4p16.1-16.3 6p21.1-ter 6q24.3-ter 9q33.2-ter 17p13.3-q21.3 17q22-24.2 17q24.3-ter 19p13.11-ter 19q arm 20p12.3-ter 20p11.21-12.1 21q22.2-ter 22q arm

N0045C18 N0026E04 N0017B07 N0014D01 N0651P03 N0071F05 N0328C17 N0117P04 N0147E03 N0411G07 N0695B13 N0353I13 N0110A24 N0719O04 N0640A09 N0176D18 N0017J10 N0423L23

N0350G05

N0646D10 N0345I23 N0332D17 N0371D08 N0640N05 N0323A09 N0159J07 N0668B20 N0607H13 N0120M18 N0196O11 N0715L15 N0493D23 M2130I11 N0269F15 N1000I21 N0040G15

38264 143217450 198807950 223882340 148491 358140 177604 160357898 125161993 415552 52856819 68705146 134914 33507965 60370 13794775 40973387 15935029

40504667 160958046 208088220 245744383 28477624 8437036 44642253 170880179 140237228 46208463 64137803 78615238 19436075 63696484 5961085 26013217 46940213 49569190

40466 17741 9280 21862 28329 8079 44465 10522 15075 45793 11281 9911 19301 30189 5901 12218 5967 33643

Deletions

3p26.1-ter 3p22.1-24.3 4p16.1-q34.3 8q arm 9q21.1-ter 14q11.1-23.2 16q12.1-21

N0385A18 N0015O03 N1338A24 N0691F16 N0632I19 N0404K10 N0708O13

N0810H01 N0092J20 N0231C10 N0639O03 N0143M01 M2285E05 F0600M14

38685 16938767 8655779 46999570 22027 18071243 44997310

8163492 43774860 181250916 146236298 32152961 61866126 88699594

8125 26836 172595 99237 32131 43795 43702

Page 89: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

73

Sample Name

Locus Start clone End clone Base pair start Base pair end Size (kb)

MA89 Gains

1p34.1-ter 6p21.1 9p13.2-13.3 9q33.3-ter 11p15.5 17p13.1-ter 17q12-21.33 17q22-ter 19p and q arms 20q13.31 21q22.3 22q12.3

N0379K15 N0380E17 N0284F01 N0205K06 N0182E22 N0634P19 N0342F22 N0468D03 N0519F09 N0267N05 N0891L10 N0452N11

N0802K22 N0375K06 N0644E22 N0668B20 N0474E20 N0657A11 N0021I09 N0196O11 N0493D23 N0476I15 N1000I21 N0040G15

95421 41356867 33879013 128256342 79527 415552 31583727 52252199 27679 54593535 42282192 33919042

44425691 44392522 37940151 140237228 1930170 10130238 46560288 78615238 63696484 62434320 46940213 49569190

44330 3036 4061 11981 1851 9715 14977 26363 63669 7841 4658 15650

Deletions 8q21.2 N0038K03 M2067O20 86757546 87055825 299

MC140 Gains

1p36.13 1q arm 2p arm 6p arm 10q26.2-ter 13q34-ter 21q22.3

N0777P08 N0510I18 N0463H16 N0812K10 N0223P11 N0412K14 N0447A17

N0148H11 N0068F13 N0785H17 N0325M17 N0106C07 N0226B11 N0457P07

16665329 142647117 79317 71610 128657820 110600643 44437000

16891342 246833917 91633812 58872610 135271097 114103214 46926492

266 104187 91554 58801 6613 3503 2489

MC336 Gains

2p13.3-ter 6p arm 9q33.3-ter 19p13.11-ter 20q11.21-13.2 20q13.31-ter

N0463H16 N0812K10 N0661B09 N0744L24 N0620H13 N0231B02

N0482J04 N0325M17 N0350O14 N0657O13 N0694L10 N0134L13

79317 71610 127160749 189657 29986634 55955329

70893187 58872610 139220879 18874065 51672430 62416964

70814 58802 12060 18684 21686 6461

Deletions 20q13.2 N0474C21 N0262B23 53355489 53684580 329

MC385 Gains

1q21.1-25.3 1q32.1-ter 2p24.3-ter

N0510I18 N0119D06 N0463H16

N0804A08 N0059M10 N0733B22

142647117 199215670 79317

186845681 246789440 74792843

44199 47574 74714

Deletions 2p12-13.1 N0123I06 N0755O06 74988529 83668734 8680

MC431 Gains 8q21.2 N0038K03 F0574H12 86757546 86973008 215

MC561 Gains

1q arm 13q32.1-ter

N0510I18 N0504C17

N0068F13 N0226B11

142647117 96334426

246833917 114103214

104187 17769

Deletions 22q12.3-ter N0564B15 N0040G15 31062997 49569190 18506

MC887

Gains

10p13-15.1 10q11.21-23.1 13q21.1-ter 17q23.1-ter 18p and q arms 19p arm

N0453H02 N0770F09 N0114F16 M2001K22 N0059I11 N0009F15

N0606G04 N0470J18 N0226B11 N0196O11 N0565D23 N0717E18

6485574 42409119 53055272 55060077 37518 189657

17251721 86380414 114103214 78615238 76103181 24391802

10766 43971 61048 23555 76066 24202

Deletions

3p arm 4q21.1-22.1 8q21.2 10p15.1-ter 13q14.3

N0385A18 N0077J09 N0038K03 N0797F08 N0435C23

M2185K04 N0737D22 F0574H12 N0284M10 N0715B19

38685 76868319 86757546 65726 52101811

90584932 90108053 86973008 4908726 52938369

90546 13240 215 4843 837

MC972 Gains

6p arm 9q33.3-ter 19p13.11-ter 19q12-ter

N0812K10 N0121C13 N0519F09 N0109B11

N0325M17 N0668B20 N0715L15 N0493D23

71610 126474147 27679 36871853

58872610 140237228 19436075 63696484

58801 13763 19408 26825

Deletions 8q21.2 N0038K03 F0574H12 86757546 86973008 215

Page 90: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

74

Sample Name

Locus Start clone End clone Base pair start Base pair end Size (kb)

FA448 No alterations

FA502 Gains 5q13.2 15q13.3 20q13.2

N0155O16 N0336F16 N0006L15

N0313J05 N0732H03 N0790B05

69070334 30112611 52483586

70698370 30621426 54304290

1628 509 1821

NZ945 Gains

1q arm 15q13.3

N0510I18 N0336F16

N0068F13 N0732H03

142647117 30112611

246833917 30621426

104187 509

Deletions 16q arm N0708O13 F0600M14 44997310 88699594 43702

RB2903

Gains

13q14.3-21.1 13q21.32 13q32.1 13q32.1 15q13.3

N0470H04 N0583P19 N0432C10 N0638C04 N0336F16

N0435C23 N0731A24 N0080B03 N0297E16 N0732H03

50664473 66046052 94004743 96178610 30112611

52278216 66805219 94578199 96782355 30621426

1614 759 573 604 509

Deletions

5q23.2 8q21.2 13q13.1 13q13.3-14.13 13q14.13 13q21.2 13q21.32 13q21.33 13q22.3 13q31.3 17p12-ter

N0619L15 N0509F16 N0379M14 N0624H01 N0651I05 N0164E20 M2026N21 N0298G13 N0133E12 N0411G07

N0009E08 M2067O20 N0584M10 N0164I01 N0307O11 N0640A08 N0440F07 N0026J21 N0459D15 N0687M21

124791998 86620140 32574544 35802418 59901013 66882861 70875970 76201583 93005803 415552

125696404 87055825 33186613 46132877 60620577 67681892 71173005 77651205 93348854 13940898

904 436 612 10330 720 799 297 1450 343 13525

RB2285 Gains

2p24.2-24.3 16p12.3-ter

N0674F13 N0766H16

N0631D03 N0164A06

15919161 73492

17177195 18186964

1258 18113

Deletions 13q13.3-ter N0336L17 M2323L19 35268823 113989403 78721

RB374 Deletions 13q14.13 N0071H01 N0454H21 46787216 53074718 6288

RB1466 Deletions 5q11.2-21.3 13q13.3-14.11

N0619H18 N0051K07

N0099I23 N0316D04

53825498 38740354

109768296 43148508

55943 4408

RB1777

Gains

1q arm 2p arm 6p arm 14q arm 19p13.2

N0510I18 N0463H16 N0812K10 N0643D12 N0282G19

N0068F13 N0447F08 N0325M17 M2011A05 N0203K06

142647117 79317 71610 21377600 8665783

246833917 89958830 58872610 106302057 8832847

104187 89880 58801 84924 167

Deletions

13q14.11 13q13.11 13q14.2 13q21.1-14.3 13q21.2 13q21.31 13q21.33 13q22.3 13q31.1

N0756F10 N0632G17 N0192F23 N0435C23 N0750I15 N0109J06 N0626I10 N0598D17 N0398A22

N0131B13 N0467G16 N0602C22 N0458G10 N0675B18 N0586C17 M2026N21 N0203P02 N0467P19

39917249 42205257 47196559 52101811 58083246 60883541 69505102 76679454 85215055

40286991 42694359 48889915 52701353 58915551 61395239 71053759 77764214 86720366

370 489 1693 600 832 512 1549 1085 1505

RB1790 Gains

1q arm 2p23.1-ter 10p arm 17q21.22-ter

N0510I18 N0463H16 N0797F08 N0472H05

N0068F13 N0450L18 N0787P11 N0196O11

142647117 79317 65726 44810989

246833917 31455726 39116217 78615238

104187 31376 39050 33804

Page 91: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

75

Sample Name

Locus Start clone End clone Base pair start Base pair end Size (kb)

Deletions

10q arm 13q12.11 13q12.3 13q14.11 13q14.2 13q21.2-32.1 13q33.1 17p arm 22q13.1-ter

N0496O18 N0717M17 N0058M19 N0110C17 N0685I15 N0616E14 N0624M01 N0411G07 N0806D02

N0106C07 N0064F04 N0706H02 N0267N09 N0795F23 N0621G18 N0681O22 N0399C02 N0040G15

41753627 19491702 28785074 41349120 46601076 53146615 102099748 415552 36789448

135271097 20304995 29739221 43792169 47823050 96075230 107271825 22128721 49569190

93517 813 954 2443 1222 42929 5172 21713 12780

RB2625

Gains

2p and q 5q14.1-pter 6p25.3-p22.3 13q31.3-q34 15q11.2 19q12-13.11 20p and q

N0463H16 N0348B13 N0812K10 N0487A02 N0607H20 N0738N05 N0766B22

N0321A15 N0043N06 N0159C08 N0226B11 N0034K18 N0306G07 N0476I15

79317 102972 71610 90908618 18273500 32864434 67103

242359512 79524609 20622576 114103214 19941698 40136267 62434320

242280 79422 20551 23195 1668 7172 62367

Deletions

1p34.3-p11.2 6p22.3-qter 13q14.11-q31.3 14q13.1-q21.3

N0799L22 N0648P12 N0350A18 N0345M21

N0115N23 N0113J06 N0035H02 N0634H08

43367715 20726090 39817963 34291680

121064448 170851849 90730242 46587232

77697 150126 50912 12296

RB1530

Gains

1q21.1-25.3 1q32.1-ter 2p and q 6p12.3-ter 7p21.1-ter 13q 19p and q 20p and q

N0510I18 N0051H18 N0463H16 N0812K10 N0669C22 N0563G05 N0519F09 N0766B22

N0703I24 N0068F13 N0321A15 N0734G07 M2245C05 N0226B11 N0493D23 N0476I15

142647117 199695815 79317 71610 40844 18014607 27679 67103

183125425 246833917 242359512 48046066 18783666 114103214 63696484 62434320

40478 47138 242280 47974 18743 96089 63669 62367

Deletions

3p and q 4q 9p and q 12p and q 14q 16p and q 22q

N0038B22 N0365H22 N0143M01 M2094C14 N0404K10 N0568F01 M2177M20

N0192L23 N0555D07 N0668B20 M2140B24 M2011A05 F0600M14 N0040G15

16865 52388942 22027 16595 18071243 74714 14440103

199240276 191239174 140237228 132289487 106302057 88699594 49569190

199223 138850 140215 132273 88231 88625 35129

RB1979

Gains

6p23 6p22.2-23 6p22.1 6p21.31 12q24.21-24.22 12q24.31-ter 18p and q

N0144A19 N0597G24 N0600F15 N0043G08 N0749J02 N0387F15 N0683L23

N0810G03 N0006N23 N0313H11 N0528P20 N0612H13 N0503G07 N0565D23

15608023 19267498 26943704 33689655 112884293 122580551 17653

16239152 24574853 27628148 35027691 116443044 131701422 76103181

631 5307 684 1338 3559 9121 76086

Deletions

8p and q 9p and q 12p-q24.21 12q24.22-q24.31 13q14.12 15q arm

N0091J19 N0143M01 N0574G08 N0791N06 N0012E03 N0095I09

N0639O03 N0668B20 N0666G06 N0197N18 M2012N23 N0558B22

304176 22027 38722 116308201 44251042 18459439

146236298 140237228 112890869 122155363 73915995 100221959

145932 140215 112852 5847 29665 81763

RB2780 Gains 13q31.3-ter 19p13.2

N0618L13 N0282G19

N0226B11 N0203K06

93475025 8665783

114103214 8832847

20628 167

Page 92: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

76

Sample Name

Locus Start clone End clone Base pair start Base pair end Size (kb)

RB3100

Gains 1q arm 2p arm 4q28.3-ter

N0510I18 N0463H16 N0090M18

N0068F13 N0785H17 N0555D07

142647117 79317 140693364

246833917 91633812 191239174

104187 91554 50546

Deletions

4q28.3 5q21.2-ter 6p arm 8q21.2 13q14.13-14.2

N0685M01 N0358E06 N0812K10 N0509F16 N0417P21

N0745L13 N0324K20 N0325M17 N0639P04 N0685I15

131288820 103000264 71610 86620140 46257295

133157751 180730036 58872610 87082985 46771856

1869 77730 58801 463 515

MA41 Gains

1p34.1-ter 6p21.1-ter 9q33.2-ter 17q12-ter 19p and q 22q

N0045C18 N0812K10 N0498E02 N1330O06 N0657O13 N0437O02

N0767N06 N0375K06 N0035I18 N0196O11 N0493D23 N0620A14

38264 71610 123966918 31801769 189657 15930262

45939116 44392522 140185674 78615238 63696484 45478948

45901 44321 16219 46813 63507 29549

Deletions 13q14.3 N0790J06 N0746C24 49769922 77566504 27797

MA49 Gains

1q32.1-ter 6p21.1-ter 9q33.3 17q12-21.33 17q22 17q25.1-ter 19p and q 20q 20q13.12 21q22.2-ter 22q

N0133P06 N0812K10 N0258M22 N0208D07 N0016A07 N0647F02 N0657O13 N0702M08 N0014E17 N0035C04 N0437O02

N0068F13 N0323A09 N0668B20 N0167K20 N0013C05 N0196O11 N0493D23 N0151C05 N0260O01 N1000I21 N0040G15

198475140 71610 126850503 30340614 52448493 69684458 189657 29644114 43271693 42127232 15930262

246833917 44642253 140237228 46634199 63066270 78615238 63696484 35444952 50137940 46940213 49569190

48359 44571 13387 16294 10618 8931 63507 5801 6866 4813 33639

Deletions 13q14.3-21.1 N0572P15 N0196C09 49941858 55373857 5432

MA80

Gains

2p21-ter 13q22.2-ter 18p11.32-q11.2 19p12-ter

N0371D08 N0639I16 N1035E02 N0657O13

N0749C14 N0226B11 N0009E17 N0468G14

148491 75781463 18338 189657

46586091 114103214 22399113 20746786

46438 38322 22381 20557

Deletions

1p32.1-31.1 4p16.2-qter 8p and q 9q33.2-pter 13q13.1-13.2 13q21.31-21.32 16p13.3-q23.3

N0010A17 N1235J08 N0418D21 N0143M01 N0045L14 N0108P18 N0063H12

N0794G09 N0555D07 N0639O03 N0804N08 N0090F05 N0816F07 N0813D14

59319690 3576555 30472 22027 32165473 60620617 4772802

77102399 191239174 146236298 122729099 34512960 65283696 82400842

17783 187663 146206 122707 2347 4663 77628

RB381

Gains 2p25.1-25.3 2p24.2-24.3 7q21.1-36.1

N0129I01 N0541K19 N0634B10

N0517E08 M2123N14 F0620M21

5022521 15467410 77767314

9267587 18028443 153591479

4245 2561 75824

Deletions

2p 8q21.2 13q14.12 13q32.3 19p13.2

N0119K02 N0509F16 N0564M19 N0044I07 N0282G19

N0788G01 N0639P04 N0164H01 N0418I10 N0203K06

9249270 86620140 45178511 100614576 8665783

15454023 87082985 46121109 101707600 8832847

6205 463 943 1093 167

RB1336 Gains

6p arm 16q 19p-q13.42 20p and q 21q

N0328C17 N0708O13 N0009F15 N0640A09 N0073I15

N0325M17 F0600M14 N0066H23 N0476I15 N0457P07

177604 44997310 189657 60370 14327625

58872610 88699594 59985032 62434320 46926492

58695 43702 59795 62374 32599

Page 93: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

77

Sample Name

Locus Start clone End clone Base pair start Base pair end Size (kb)

RB1740 Deletions 17p12-ter N0411G07 N0687M21 415552 13940898 13525

RB2052 Gains

1q32.1 4p16.1-ter 4p15.31-15.33 4p15.1 4p13-14 4q13.3-21.1 4q31.3-32.1 6p and q 8p21.2-21.3 8p11.23-8p12 13q11-12.11 13q12.11-12.12 13q13.3 13q14.3 13q22.1-22.2 13q33.3-ter 18p and q 19p and q 20 p and q 21q

N0165E10 N0335H03 M2185J18 N0092M20 N0142P03 N0632A23 N0509I22 N0812K10 N0116M17 N0745K06 N0563G05 N0273F15 N0162F21 N0630I06 N0441J10 N0014G15 N0683L23 N0519F09 N0766B22 N0675D22

N0595K11 N0238O15 N0013H04 N0103P18 N0384C20 N0641E03 N0071A06 N0113J06 N0795G08 N0156L03 N0506L23 N0499O19 N0718A20 N0655C11 N0010A23 N0226B11 N0565D23 N0493D23 N0476I15 N0457P07

200361154 39105 15392919 29923977 38295574 73533568 154805443 71610 23526552 36910075 18014607 21528172 35606696 51289896 73593525 109310260 17653 27679 67103 21358972

205125953 9220751 19998505 31661464 43297704 78634666 156270117 170851849 26272272 38058325 20335446 24215466 38322375 52555670 74860115 114103214 76103181 63696484 62434320 46926492

4765 9182 4606 1737 5002 5101 1465 170780 2746 1148 2321 2687 2716 1266 1267 4793 76086 63669 62367 25568

Page 94: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

78

Sample Name

Locus Start clone End clone Base pair start Base pair end Size (kb)

Deletions

4p16.1 4q13.1 4q21.1-21.3 4q26 4q28.1 4q32.1 4q34.1-34.2 4q34.3-35.1 4q35.1 4q35.1-35.2 8p23.3-ter 8p21.3-22 8p12-8p21.2 8p12 8p11.21 8q11.22-11.23 8q12.1 8q12.1 8q21.11 8q21.2-21.3 8q22.3-23.1 8q23.3-24.13 8q24.22 8q24.3 10p and q 13q12.13-12.3 13q13.3-14.11 13q14.13-14.3 13q21.1-21.2 13q21.33-22.1 13q31.1-32.1 13q33.1-33.3 15q arm 16p and q

N0063P01 N0371B02 N0638G02 N0111L14 N0728C08 N0423E20 M2220L14 N0442N05 N0099L17 N0571N01 N0111E15 N0711D03 N0521M14 N0608P11 N0745M10 N0818C13 N0022E14 N0661A03 N0503K13 N0509F16 N0109C19 N0096J05 N0238O02 N0265N12 N0797F08 N0662C02 N0050D16 N0417P21 N0685E08 N0521B14 N0605B09 N0790J08 M2200G17 N0568F01

N0065O09 N0484B23 N0272G21 N0122K18 N0427N08 N0295F08 N0520H06 N0692E14 N0472G22 N0016L12 N0521J16 N0652H24 N0051J09 N0654O09 N0117K13 N0669G08 N0314N12 N0051L11 N0064K14 N0023B03 N0659A24 N0047A23 N0213I02 N0792G19 N0106C07 N0629E24 N0025N03 N0686G10 M2012K04 N0709B02 N0747M20 N0359M07 N0558B22 F0600M14

10208020 60325977 79651029 115365486 125060157 159055244 174532621 181621943 185082952 186855084 156973 23535579 26980569 32968313 41575316 52218489 57031842 59655134 75247524 86620140 103542026 116012122 132286383 140919525 65726 24792776 38419312 46257295 53909558 68182005 78322396 104048543 19970520 74714

10941759 65150139 87642586 119579104 127718213 161426872 177417393 184692674 186877885 189851441 3647702 16420058 30264501 34431087 42689304 53575828 58582905 60743597 78302251 93815239 109058233 122584169 133463867 142491552 135271097 29602194 39705077 50401135 58001888 73283703 95902058 109061691 100221959 88699594

734 4824 7992 4214 2658 2372 2885 3071 1795 2996 3491 7116 3284 1463 1114 1357 1551 1088 3055 7195 5516 6572 1177 1572 135205 4809 1286 4144 4092 5102 17580 5013 80251 88625

RB2589

Gains 6p 17q24.3-ter 20q arm

N0157M05 N0075H13 N0559K10

N0159J07 N0196O11 N0476I15

65779919 66875850 29294627

170880179 78615238 62434320

105100 11739 33140

Deletions

1p 8q21.2 8q24.13-ter 10q24.31-ter 16p13.2-ter 16q12.2-ter 17p arm

N0045C18 N0509F16 N0636H23 F0628D17 N0766H16 N0533J12 N0411G07

N0385C11 M2067O20 N0639O03 N0091E02 N0107G06 F0600M14 N0399C02

38264 86620140 123692681 102443708 73492 52309608 415552

120345972 87055825 146236298 135262317 9954815 88699594 22128721

120308 436 22544 32819 9881 36390 21713

RB2631 Gains

1p arm 6p arm 13q14.2-ter 13q14.3-21.1 13q31.1-32.3 13q33.2-ter 14q24.3

N0510I18 N0059N17 N0563G05 N0157B12 N0514P01 N0111G22 N0306K22

N0059M10 N0325M17 N0685I15 N0715B19 N0813H05 N0226B11 M2011A05

142647117 351266 18014607 51626286 89114601 105217625 74286435

246789440 58872610 46771856 52938369 98902788 114103214 106302057

104142 58521 28757 1312 9788 8886 32016

Page 95: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

79

Sample Name

Locus Start clone End clone Base pair start Base pair end Size (kb)

Deletions

8p23.1-ter 13q14.2-14.3 16p and q 17p 20p and q

N0418D21 N0438K10 N0773G07 N0411G07 N0649H22

N0802F15 N0509N17 N0665E03 N0728E14 N0134L13

30472 47560571 235519 415552 6122936

11564386 51364049 88686715 21322111 62416964

11534 3803 88451 20907 56294

RB2641

Gains

1q 6p 9q 13q 16q

N0510I18 N0537J16 N0088D03 N0563G05 N0046B20

N0059M10 N0325M17 N0668B20 N0226B11 N0665E03

142647117 367602 66405147 18014607 84705552

246789440 58872610 140237228 114103214 88686715

104142 58505 73832 96089 3981

Deletions 1p35.3 14q 19p and q

N0414L23 N0521F15 N0081I08

N0467D18 N0046B20 N0265J21

608751 62108060 902662

29444707 84867117 58496669

28836 22759 57594

RB2647 Gains

1q 5p

N0510I18 N0597A21

N0059M10 M2220G19

142647117 350757

246789440 45985067

104142 45634

Deletions 3q26.1-ter 16q

N0593A09 N0584H05

M2110L16 N0665E03

167465632 45532097

199131621 88686715

31666 43155

RB2683

Gains

1q 6p 7q32.1-ter 14q21.3

N0114B18 N0812K10 N0475K08 N0279J20

N0068F13 N0325M17 N0083D03 N0012F16

149888055 71610 127479042 47251765

246833917 58872610 158783389 106218118

96946 58801 31304 58966

Deletions

1p35.1pter 13q13.3-21.31 16p and q 17p13.2

N0045C18 N0336L17 N0773G07 N0411G07

N0114D07 N0109J06 N0665E03 N0220M19

38264 35268823 235519 415552

34244419 61051693 88686715 5373014

34206 25783 88451 4957

RB2306

Gains 1q23.2-ter 9q 16q11.2-12.1

N0297K08 N0211E19 N0471D09

N0059M10 N0644H13 N0545E02

159374906 70091012 45229845

246789440 139839547 47619108

87415 69749 2389

Deletions

1p35.3-ter 5q31.3-ter 8p and q 16p13.11-ter 16p11.2-12.3 16q

N0776O18 N0035N12 N0091J19 N0568F01 N0813D06 N0001F10

N0333N08 N0324K20 N0639O03 N0103G05 N0590H03 N0665E03

307737 140231805 304176 74714 18938492 47811144

30171718 180730036 146236298 15025243 33871612 88686715

29864 40498 145932 14951 14933 40876

RB247

Gains

1q 2p 5p and q 6p 9p31.2-ter 11p15.1-ter 17q21.31-ter 18p11.31

N0510I18 N0420M07 N0811I15 N0059N17 N0069E16 N0182E22 N0135M15 N0683L23

N0059M10 N0785H17 N0324K20 N0325M17 N0668B20 N0583F24 N0196O11 N0297J24

142647117 264456 72312 351266 109899366 79527 40812466 17653

246789440 91633812 180730036 58872610 140237228 19004570 78615238 6128065

104142 91369 180658 58521 30338 18925 37803 6110

Deletions 8q21.2 15q26.1

N0509F16 N0267B15

F0574H12 N0558B22

86620140 88814205

86973008 100221959

353 11408

RB383 Gains

1p21.3-22 2p 5q31.1-33.2 6p 13q 22q

N0212D05 N0371D08 N0254M05 N0812K10 N0563G05 N0619K17

N0263K19 N0495B16 N0096J04 N0325M17 N0226B11 N0040G15

150227817 148491 132519214 71610 18014607 22778020

153541655 82715436 152951910 58872610 114103214 49569190

3314 82567 20433 58801 96089 26791

Page 96: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

80

Sample Name

Locus Start clone End clone Base pair start Base pair end Size (kb)

Deletions 5q33.2-ter 18p

N0565D08 N0059I11

N0324K20 N0390I06

153948314 37518

180730036 15399258

26782 15362

RB1021

Gains

1q 7q 14q22.3 21q21.2-ter

N0510I18 N0665G16 N0118M18 N0705P12

N0059M10 N0083D03 M2011A05 N0457P07

142647117 80719268 55340193 25638123

246789440 158783389 106302057 46926492

104142 78064 50962 21288

Deletions

5q11.2-13.2 5q33.2-ter 10p 11p 11q14.3-23.3 16q 19q13.2-ter

N0143O12 N0096J04 N0592K16 N0063A07 N0798B05 N0471D09 F0497A07

N0808I04 N0324K20 F0547M11 N0699N10 N0152O02 F0600M14 N0493D23

55718971 152764659 286972 621027 91188732 45229845 46383400

68938439 180730036 39012541 50692538 118910577 88699594 63696484

13219 27965 38726 50072 27722 43470 17313

WERI

Gains

1q21.1-23.3 1q24.3-24.3 1q31.3-32.2 1q41 1q41-42.3 2p12-ter 3p22.3-ter 5p14.3 7p15.3-ter 13q12.12-12.2 13q31.1-ter 17q12-ter 21q22.13-ter

N0510I18 N0448B23 N0124P19 N0101K19 N0381O10 N0463H16 N0038B22 N0811I15 M2245C05 N0294L03 N0089A14 N0032H06 N0777J19

N0578G06 N0118D10 N0002P02 N0514C19 N0739C15 N0543B23 N0384L08 N0638C22 N1151M13 N0624L24 N0226B11 N0196O11 N0457P07

142647117 170841028 192123480 213850669 219492439 79317 16865 209225 40844 23076054 82412127 35262985 37641258

162556579 178664532 206526445 219396428 233391573 80228525 32142422 18922411 25288541 27970959 114103214 78615238 46926492

19909 7824 14403 5546 13899 80149 32126 18713 25248 4895 31691 43352 9285

Deletions

1q24.1-24.3 1q25.2-25.3 1q31.1-31.2 1q41-42.13 3q28-ter 5q11.2-12.1 10p 10q11.23 13q12.3-13.1 13q13.3-14.11 13q14.2-21.33 13q22.2-31.1 16q23.2-ter 18q22.1-22.3 21p 21q11.2-21.2

N0713G21 N0796G10 M2245J12 N0638A13 N0456E14 N0008N21 N0109N22 N0423G02 N0057H24 N0289J04 N0795F23 N0715H21 N0474K20 N0607G19 N0376P20 N0429H22

N0341B16 N0450F15 N0272B08 N0638A13 N0192L23 N0593B07 N0797F08 N0640E12 N0645A10 N0039L22 N0465K12 N0103P23 F0600M14 N0798A24 N0259G22 N0709D12

164981867 178395125 185734306 219624020 190215218 55340491 65726 50166065 27927907 35991523 47624175 75329929 78991561 60553398 9721646 13270806

169464909 181243272 191129912 228145219 199240276 62741971 35812347 54430559 31278920 43637987 70603880 82201310 88699594 68679304 10197104 24687098

4483 2848 5396 8521 9025 7401 35747 4264 3351 7646 22980 6871 9708 8126 475 11416

Y79

Gains

2p24.3 7q35-ter 11q24.1-ter 13q21.32-ter 18q12.3-21.1 18q23 21q22.11-ter

N0619O15 N0564O04 N0381C13 N0001L24 M2032O09 N0531M16 N0004F08

N0463P22 N0083D03 M2013A02 N0226B11 N0002E13 N0703M10 N0457P07

15175256 147151220 122460473 64741928 41713136 71676600 32359411

16152619 158783389 134436514 114103214 46266399 72686216 46926492

977 11632 11976 49361 4553 1010 14567

Deletions 5p13.3-15.33 16q11.2-12.2

N0128A03 N0180E11

M2335O24 N0070N16

4054083 45081598

32497485 54541026

28443 9459

Page 97: A Subset of Retinoblastoma Lacking RB1 Gene Mutations have ... · Myc v-myc myelocytomatosis viral oncogene homolog (avian) MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma

81

Sample Name

Locus Start clone End clone Base pair start Base pair end Size (kb)

IMR32 Gains

1p31.3-33 1q21.1-25.3 1q32.1-ter 2p24.3-24.3 2q14 6p21.1-21.22 15q 17q 20q-ter

N0682P18 N0510I18 N0133P06 N0571E19 N0678O18 N0065L23 N0137P24 N0606M07 N0004O09

N0185E18 N0522B03 N0332D17 N0427M01 N0812M06 N0061H05 N0584I15 N0196O11 N0476I15

51504350 142647117 198475140 14565626 66473520 30666043 59763464 37680398 29737772

62616624 182897083 245744383 17376543 67573409 44747012 91451479 78615238 62434320

11112 40250 47269 2811 1100 14081 31688 40935 32697

Deletions 1p33-ter 16q22.2-ter

N0379K15 N0113E03

N0670L22 N0655C18

95421 69787193

50599578 85754878

50504 15968