102
UNDIFFERENTIATED SARCOMAS: UNDERSTANDING EARLY SARCOMA DEVELOPMENT A DETAILED GENOMIC AND TRANSCRIPTOMIC ANALYSIS OF PAEDIATRIC UNDIFFERENTIATED SARCOMAS BY Cassandra Graham A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Laboratory Medicine and Pathobiology University of Toronto Date of Defense: November 3rd, 2010 © Copyright by Cassandra Graham, 2011

UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

UNDIFFERENTIATED SARCOMAS: UNDERSTANDING

EARLY SARCOMA DEVELOPMENT

A DETAILED GENOMIC AND TRANSCRIPTOMIC ANALYSIS OF

PAEDIATRIC UNDIFFERENTIATED SARCOMAS

BY

Cassandra Graham

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

Department of Laboratory Medicine and Pathobiology University of Toronto

Date of Defense: November 3rd, 2010

© Copyright by Cassandra Graham, 2011

Page 2: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

ii

ABSTRACT

A Detailed Genomic and Transcriptomic Analysis of Paediatric Undifferentiated Sarcomas

Cassandra Graham

Master of Science, 2010

Department of Laboratory Medicine and Pathobiology

University of Toronto

November 3rd, 2010

Paediatric undifferentiated soft tissue sarcomas (USTSs) are a diagnostically challenging

group of neoplasms. We hypothesized that USTSs contain distinct subgroups that can be

identified based on their morphology, genomic aberrations and expression profiles. We sought

to characterize genomic aberrations within primitive round cell (PRC) sarcomas which may

underlie aberrant expression patterns. Using molecular and cytogenetic analyses, we identified 5

of 18 CIC-DUX4-positive PRC sarcomas. The consistent involvement of the CIC-DUX4 fusion

in a subset of PRC sarcomas suggests a central role for the fusion transcript in such tumours.

These analyses also identified a cohort of CIC-DUX4-negative USTSs with no established

genetic markers. We performed integrative copy number and expression profiling, and identified

significant genomic and transcriptomic changes. We propose that these genes are involved in

biological pathways that are important to the initiation and progression of undifferentiated

sarcoma, and may provide novel insights into the biological events responsible for

sarcomagenesis.

Page 3: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

iii

ACKNOWLEDGEMENTS

I would like to express my heartfelt appreciation to those people that have made the completion

of this thesis a reality.

This Master’s thesis is dedicated to my parents, whose never-ending patience and unconditional

support have allowed me the latitude to explore all of my personal and academic pursuits. I

thank my brother and sister for their endless teasing and loving support, which always allow me

to keep things in perspective. I would also like to thank my supportive extended family, and my

Toronto family at 470 Markham Street, who have been there for me every step of the way.

I would like to express my sincerest gratitude to my supervisors, Dr. Gino Somers and Dr. Maria

Zielenska. I am so grateful for the confidence with which you have always supported my

research. The encouragement and guidance that I have received from you were invaluable to the

successful completion of this thesis, and have helped me grow significantly both academically

and personally.

I would also like to acknowledge Dr. Jeremy Squire, who has provided me with so many

opportunities, and without whom I may not have chosen this path. I would like to thank Dr.

Maisa Yoshimoto, Dr. Georges Maire, Jane Bayani and Paula Marrano who have mentored me

for the past 6 years and have served as knowledgeable resources throughout the course of this

project. Without our lunch dates, I might have gone crazy. To Devina Ramsaroop, I couldn’t

have asked for a better labmate or partner in crime at our lab meetings and conferences.

I wish to acknowledge Dr. Cynthia Hawkins and Dr. Mary Shago, for providing me with

direction, advice and genuine enthusiasm over the course of this project. I would also like to

Page 4: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

iv

thank the following friends at Sick Kids for all of their help with this thesis: Susan Chilton-

MacNeill, Dr. Bekim Sadikovic, Michael Ho, Pawel Buczkowicz and Dr. Paul Thorner.

Lastly I would like to thank Graeme Mask, who has been so encouraging and supportive over the

past 2 years – I couldn’t have done it without you.

My deepest appreciation to each and every one of you.

Page 5: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

v

TABLE OF CONTENTS

ABSTRACT ii

ACKNOWLEDGEMENTS iii

TABLE OF CONTENTS v

LIST OF ABBREVIATIONS vii

LIST OF TABLES AND FIGURES ix

CHAPTER ONE: INTRODUCTION 1

1.1 Overview of Paediatric Soft Tissue Sarcomas 2

1.2 Classification of Paediatric Soft Tissue Sarcomas 2

1.3 Diagnostic Importance of Chromosomal Translocations in Sarcomas 3

1.4 Overview of Paediatric Undifferentiated Soft Tissue Sarcomas 5

1.4.1 Morphological Subtypes of USTS 6

1.4.2 Models of Sarcomagenesis: Cell of Origin & Acquisition of Malignant

Properties 7

1.4.3 Molecular Studies of USTS 15

1.5 Hypothesis, Objectives & Expected Outcomes 18

CHAPTER TWO: MATERIALS & METHODS 19

2.1 Tumour Specimens 20

2.2 DNA and RNA Extraction 20

2.3 Cytogenetic and Spectral Karyotyping (SKY) analyses 21

2.4 Array Comparative Genomic Hybridization (CGH) 22

2.5 Interphase Fluorescence in situ Hybridization (FISH) 23

2.5.1 Isolation of DNA from Bacterial Artificial Chromosomes (BACs) 23

2.5.2 Pilot Project Four-Colour Interphase FISH 24

2.5.3 FISH Validation of Copy Number Gain of EPHA3 25

2.6 Reverse Transcriptase Polymerase Chain Reaction (RT-PCR) for

Detection of CIC-DUX4 26

2.7 Copy Number Arrays 27

2.8 Expression Arrays 28

2.9 Analysis and Integration of Microarray Data 28

2.9.1 Partek Genomics Suite: Data Analysis and Integration 28

2.9.2 Ingenuity Pathway Analysis: Network Identification and

Pathway Analysis 30

2.10 Quantitative Polymerase Chain Reaction (qPCR) 30

2.10.1 Comparative CT Analysis of qPCR Data 31

2.11 Immunohistochemistry (IHC) 31

Page 6: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

vi

2.11.1 IHC for CD99 32

2.11.2 IHC for EPHA3 32

CHAPTER THREE: RESULTS & DISCUSSION 33

3.1 Clinicopathological Features of Tumour Samples 34

3.2 t(4;19) Pilot Project 36

3.2.1 Genomic Imbalances in Undifferentiated Sarcomas 36

3.2.2 Karyotypes of Undifferentiated Sarcomas 39

3.2.3 Detailed Comparison of Cytogenetic Karyotyping and Array CGH 40

3.2.4 Four-Colour FISH Validation of 19q Rearrangement and Detection

of Translocation Binding Partners 42

3.2.5 Detection of CIC-DUX4 Fusion Transcripts by RT-PCR 46

3.2.6 Discussion of t(4;19) Pilot Project 48

3.3 Screening of USTS Cohort for the CIC-DUX4 Fusion Transcript 49

3.3.1 Detection of the CIC-DUX4 Fusion by RT-PCR 50

3.3.2 DNA Sequence Analysis of the CIC-DUX4 Positive PCR Products 52

3.3.3 Discussion of Overall Screening of USTS cohort of CIC-DUX4

Fusion Transcript 55

3.4 Discovery of Novel Markers for CIC-DUX4-Negative Tumours 58

3.4.1 DNA Copy Number Analysis 59

3.4.2 Gene Expression Analysis 60

3.4.3 Integration of Copy Number and Gene Expression Analyses 62

3.4.4 Gene Network Analysis 64

3.4.5 Identification of Candidate Genes 65

3.4.6 Gene Expression Validation of Candidate Genes 66

3.4.7 Copy Number Validation of EPHA3 68

3.4.8 Protein Validation of EPHA3 68

3.4.9 Discussion of Discovery of Novel Markers for

CIC-DUX4-Negative Tumours 71

CHAPTER FOUR: SUMMARY, GENERAL CONCLUSIONS & FUTURE DIRECTIONS 77

4.1 Summary & General Conclusions 78

4.2 Future Directions 79

REFERENCES 81

Page 7: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

vii

LIST OF ABBREVIATIONS

ARMS Alveolar Rhabdomyosarcoma

BAC Bacterial Artificial Chromosome

CGH Comparative Genomic Hybridization

CSC Cancer Stem Cell

DAPI 4’,6’-diamidino-2-phenylindole

DEPC Diethylpyrocarbonate

DNA Deoxyribonucleic Acid

EDTA Ethylenediaminetraacetic acid

ERMS Embryonal Rhabdomyosarcoma

ES/PNET Ewing Sarcoma/Primitive Neuroectodermal Tumour

FFPE Formalin-Fixed Paraffin-Embedded

FISH Fluorescence in situ Hybridization

IHC Immunohistochemistry

IPA Ingenuity Pathway Analysis

MPNST Malignant Peripheral Nerve Sheath Tumour

MSC Mesenchymal Stem Cell

NSC Neural Stem Cell

PGS Partek Genomic Suite

PRC Primitive Round Cell

qPCR Quantitative Polymerase Chain Reaction

RMS Rhabdomyosarcoma

RNA Ribonucleic Acid

RT-PCR Reverse Transcriptase-Polymerase Chain Reaction

Page 8: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

viii

SNP Single Nucleotide Polymorphism

SSC Saline Sodium Citrate

SDS Sodium Dodecyl Sulfate

SKY Spectral Karyotyping

SS Synovial Sarcoma

t() Translocation

TCAG The Centre for Applied Genomics

USTS Undifferentiated Soft Tissue Sarcoma

Page 9: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

ix

LIST OF TABLE AND FIGURES

Chapter 1

Figure 1.1 Subclassification of paediatric soft tissue sarcomas 4

Table 1.1 Sarcomas characterized by recurrent chromosomal translocations 5

Figure 1.2 Euclidean clustering of sarcoma tumours based on eight stem cell markers 11

Figure 1.3 Immunohistochemical staining of CD56 12

Figure 1.4 Decoding the early events of sarcoma development 14

Chapter 2

Table 2.1 Primer sequences used for RT-PCR screening of USTS cohort 26

Chapter 3

Figure 3.1 Histological features and CD99 staining of USTSs used in

t(4;19) pilot study 34

Table 3.1 Clinicopathological features, CD99 staining patterns, and CIC-DUX4

fusion transcript status of the cohort of tumours used in study. 35

Figure 3.2 Composite analysis of genomic imbalances in USTSs used in

t(4;19) pilot study 38

Table 3.2 Summary of USTS karyotypes by G-banding and SKY analyses 39

Figure 3.3 SKY of USTSs with 19q13 rearrangement 40

Figure 3.4 Comparative analysis of array CGH and SKY results 42

Figure 3.5 Four-colour FISH validation of 19q rearrangement and detection

of translocation binding partners in USTS1 44

Figure 3.6 Four-colour FISH validation of 19q rearrangement and detection

of translocation binding partners in USTS2 46

Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47

Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening 51

Page 10: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

x

Figure 3.9 Nucleotide sequences of CIC and DUX4 sequences denoting

primer sequences and translocation breakpoints 53

Figure 3.10 Nucleotide and predicted amino acid sequences of the different

CIC-DUX4 fusions 54

Figure 3.11 Predicted protein structure of CIC-DUX4 fusion protein variants 55

Figure 3.12 Overall genomic profiling views from PGS of all USTS samples

interrogated for copy-number changes 60

Figure 3.13 Cluster analysis from PGS of all USTS samples interrogated for

changes in gene expression 61

Figure 3.14 Tumour-specific integration of copy number and gene expression

analyses 62

Figure 3.15 Integration of cumulative copy number and gene expression analyses 63

Figure 3.16 Gene network analysis of copy number and gene expression changes 64

Table 3.3 Candidate genes with USTS-specific genomic and transcriptomic changes 65

identified by integrative and gene network analyses.

Figure 3.17 Changes in Gene expression of candidate genes 67

Figure 3.18 FISH Validation of Copy Number Gain of EPHA3 69

Figure 3.19 IHC for EPHA3 70

Page 11: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

1

CHAPTER ONE

INTRODUCTION

Page 12: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

2

1. INTRODUCTION

1.1 Overview of Paediatric Soft Tissue Sarcomas

Paediatric soft tissues sarcomas (STSs) are a diverse group of mesenchymal tumours arising in

the connective tissues of the body. These tumours are generally found to be quite aggressive,

particularly if the tumour cells are poorly differentiated, and often present with early hematogenous

metastasis [1-6]. Collectively, STSs account for up to 10% of all paediatric cancers, making them

proportionately more common among children than adults (1% of all adult cancers) [5, 7].

Sarcomas have long been a challenging group of tumours to diagnose and classify due to the

morphological similarities between established subgroups, and the relative rarity of these

neoplasms [8]. However, advances in immunohistochemical analysis and the development of

molecular and cytogenetic techniques have allowed for the discovery of mechanisms by which we

can differentiate between different sarcoma subtypes [9]. Treatment of paediatric STSs consists of

surgery and preoperative chemotherapy. Chemotherapy has been shown to effectively manage

localized, chemosensitive tumours, and when the tumour is caught early enough, surgery can be

curative [6]. Though advances in therapeutics have improved long-term survival, the prognosis of

chemoresistant and metastasized sarcomas remains very poor, with a 5-year survival rate of less

than 60% [6, 10].

1.2 Classification of Paediatric Soft Tissue Sarcomas

Morphologically, sarcomas are divided into rhabdomyosarcomas (RMS), the most common

subtype of STS accounting for up to 50% of paediatric sarcomas, and non-rhabdomyomatous

sarcomas (NRSTSs) (Figure 1.1) [11]. NRSTSs include a very large number of pathologically

diverse tumours including Ewing sarcoma/primitive neuroectodermal tumour (ES/PNET),

Page 13: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

3

malignant fibrous histiocytoma (MFH), synovial sarcoma (SS), and many others [11]. At the

genomic level, sarcomas are divided into two major categories (Figure 1.1). The first category is

composed of sarcomas with simple, near-diploid karyotypes which consistently present with

recurrent chromosomal rearrangements [12]. Members of this group include ES/PNET which are

associated with rearrangements of the EWS gene including t(11;22) and t(21;22); alveolar

rhabdomyosarcoma (ARMS), which is associated with t(1;13) and t(2;13); and SS which is

associated with t(X;18) resulting in an SYT-SSX gene fusion product [13-15]. To date,

approximately 41 gene fusions have been associated with 17 different subtypes of sarcomas [16].

The second category is composed of tumours that have very complex karyotypes, but in which no

reproducible chromosomal aberrations have been identified. This group includes malignant

peripheral nerve sheath tumour (MPNST), embryonal rhabdomyosarcoma (ERMS) and

osteosarcoma (OS) [12].

1.3 Diagnostic Importance of Chromosomal Translocations in Sarcomas

The accurate subclassification of paediatric sarcomas has important therapeutic and prognostic

implications [17, 18]. Subclassification is achieved using a variety of diagnostic techniques

including morphological, immunohistochemistal, molecular and cytogenetic analyses [19-21]. In

some cases, the combination of specific morphological features (e.g. biphasic histology for SS, or

spindled and myxoid histology for RMS), together with positive staining for specific antibodies

(e.g. CD99 for ES [19], or myogenin for RMS [22]) allows for the identification of specific

sarcoma subtypes [23]. However certain tumours are particularly difficult for the pathologist to

diagnose due to significant overlap in the histologic and immunohistochemical features of the

tumours [24]. Because the treatment protocols and prognoses of these different sarcoma subtypes

vary immensely, a correct diagnosis is crucial.

Page 14: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

4

The discovery of specific chromosomal translocations associated with sarcoma subtypes has

markedly improved the diagnostic accuracy of paediatric sarcomas (Table 1.1) [23-27]. Tumours

with these translocations generally show low genomic complexity, with well-defined recurrent

chromosomal translocations that result in fusion genes with oncogenic fusion protein products. Not

only has the identification of these fusion genes improved the precision of sarcoma

subclassification, it has provided an increased understanding of sarcoma biology by providing a

logical starting point for more thorough functional studies of tumour development [27, 28]. The

identification of specific translocations harboured by different tumours has played a pivotal role in

differentiating between different neoplasms. As such, it has become imperative to use molecular

Figure 1.1. Subclassification of Pediatric Soft Tissue Sarcomas. Morphologically,

pediatric sarcomas are divided into rhabdomyosarcomas (RMS) and non-rhabdomyomatous

sarcomas (NRSTS) (Top). At the genomic level sarcomas are divided into two categories

(Bottom); those with simple, near-diploid karyotypes which consistently present with

recurrent chromosomal rearrangements and those with very complex karyotypes, but in

which no recurrent chromosomal aberrations have been identified.

Page 15: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

5

and cytogenetic techniques, in conjunction with traditional histopathological techniques, to identify

these chromosomal translocations so that a correct diagnosis can be made.

TUMOUR TRANSLOCATION FUSION PRODUCT

Alveolar Rhabdomyosarcoma t(2;13)(q35;q14) PAX3-FOXO1A

t(1;13)(p36;q14) PAX7-FOXO1A

Alveolar Soft- Part Sarcoma t(X;17)(p11.2;q25) TFE3-ASPL

Clear-cell Sarcoma t(12;22)(q13;q12) EWS-ATF1

Congenital Fibrosarcoma t(12;15)(p13;q25) ETV6-NTRK3

Desmoplastic Small Round

Cell Tumour t(11;22)(p13;q12) EWS-WT1

Ewing Sarcoma/PNET

t(11;22)(q24;q12) EWS-FLI1

t(21;22)(q22;q12) EWS-ERG

t(7;22)(p22;q12) EWS-ETV1

t(17;22)(q21;q12) EWS-ETV4

t(2;22)(q33;q12) EWS-FEV

Myxoid Chondrosarcoma t(9;22)(q22;q12) EWS-NR4A3

Myxoid Liposarcoma t(12;16)(q13;p11) FUS-CHOP

t(12;22)(q13;q12) EWS-CHOP

Synovial Sarcoma t(X;18)(p11;q11) SYT-SSX1

Table 1.1. Several sarcoma subtypes are characterized by specific chromosomal translocations that

produce fusion oncogenes with protein products. (Adapted from [11]).

1.4 Overview of Paediatric Undifferentiated Soft Tissue Sarcomas

Despite the advancement of diagnostic applications which allow for the identification of

sarcoma subtypes, approximately 5% of sarcomas remain unclassifiable [29-32]. These tumours,

termed undifferentiated soft tissue sarcomas (USTSs), show no specific lineage differentiation,

exhibit no well established or consistent histologic or immunohistochemical profile, and harbour no

compelling recurrent molecular aberrations associated with the traditional sarcoma subtypes [32,

33]. Furthermore, the karyotypes of these sarcomas are largely variable, with some tumours having

a very complex genomic make-up, and other tumours harbouring a near-diploid genome [34]. As

Page 16: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

6

such, a diagnosis of undifferentiated sarcoma is largely considered a diagnosis of exclusion,

evoking much debate as to whether this group is composed of tumours with common histological

and biological features, or if these tumours represent a heterogeneous group of primitive

mesenchymal tumours that can differentiate along various soft tissue lineages [9]. USTSs have

been found to be most commonly located in the extremities (46%), the trunk (38%), and the head

and neck (16%) [8, 29]. Treatment protocols included surgery, VAC (vincristine, adriamycin,

cyclophosphamide) chemotherapy and radiation therapy [30]. At the present time, patients

diagnosed with USTS have a 40-50% risk of developing distant metastases within 5 years of

diagnosis [4]. USTSs generally have a poorer prognosis compared to RMSs [8, 30, 35, 36], with a

44% overall 5-year survival rate [8, 30], though these results may be skewed due to small sample

size and inaccurate diagnoses.

1.4.1 Morphological Subtypes of USTS

Due to the rarity of these tumours, only a few studies have been published which seek to

more fully elucidate the pathology behind a USTS diagnosis. Somers et al initiated a study which

sought to examine the clinical and pathological features of the largest cohort of USTSs studied at

the time, in an attempt to identify specific characteristics which could be useful for the diagnosis,

prognosis and treatment of USTS tumours [32]. This study was unable to identify any specific

features shared by the majority of these tumours, but noted that many of the tumours showed a

sheet-like proliferation of densely packed round to plump spindled tumour cells [32]. In a similar

study, Alaggio et al. [9] examined the histology of a smaller cohort of USTSs, and furthered this

notion, determining that USTSs can be broken down into two morphological groups. The first

group contains tumours with a spindled/myxoid morphology, that have elongated cells arranged in

poorly formed bundles or fascicles [9, 32] (see Figure 3.1c taken from Somers et al., 2006 [32]).

Page 17: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

7

The second group, which our lab has entitled paediatric primitive round cell (PRC) sarcomas,

contains tumours composed of primitive mesenchymal cells showing no evidence of differentiation,

that are arranged in sheets or nests [9, 32] (see Figure 3.1b taken from Somers et al., 2006 [32]).

These PRC sarcomas represent the most primitive paediatric sarcomas described to date, and thus

form an attractive model in which to study the early events involved in sarcomagenesis.

1.4.2 Models of Sarcomagenesis: Cell of Origin and Acquisition of Malignant Properties

Much research has gone into studying the molecular mechanisms of sarcomagenesis, or the

initiation and development of sarcoma. While significant progress has been made, this complex

process remains highly debated. Two generally accepted models of tumourigenesis are the ‘cancer

stem cell’ (CSC) hypothesis and the ‘dedifferentiation’ hypothesis. The dedifferentiation

hypothesis suggests that cells with more primitive features, such as ability to self-renew and

differentiate down different lineages, may arise in an adult through the process of dedifferentiation

[37]. Overlapping with this model is the stochastic model of tumour formation which proposes that

any cell from a tumour is capable of limitless growth and the ability to spread throughout the body

[38]. These theories are less accepted by modern cancer biologists due to the heterogeneous nature

of most tumours, as well as evidence to the contrary that all tumour cells can re-initiate tumour

formation. The CSC hypothesis suggests that tumours are initiated and sustained by a fraction of

cancer cells derived from tissue stem cells which maintain pluripotency and the capacity to self-

renew [39-41]. In general, these cells are thought to be resistant to chemotherapy, and thus the cells

that are responsible for relapse and metastasis. This theory appears to hold true for numerous

cancer types including leukemia [42, 43], pancreatic cancer [44], and brain tumours [45], in which

specific tumour- initiating cells have been identified. The CSC hypothesis is not without criticisms

Page 18: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

8

however, as certain cancers, such as melanoma, have been identified which seem to harbour

numerous cells that are able to initiate and maintain a tumour [40, 46].

Though controversial, a number of publications have suggested that some sarcoma subtypes

may also follow this CSC hypothesis [41, 47-49]. Other studies however, have found that the

ability of some sarcoma-initiating cells to actually initiate tumour formation in vitro and in

xenograft models is inconsistent, and varies from tumour to tumour [50]. Thus, the initiation and

progression of sarcomas remains a complex process that is poorly understood. Crucial to our

understanding of sarcomagenesis are two central and related themes: the cell of origin and the

mechanisms by which cancer-initiating cells acquire malignant properties. Due to the complete

lack of lineage differentiation in the majority of the tumour population, PRC sarcomas are

hypothesized to be the most primitive paediatric STSs that have been identified, and understanding

the molecular processes responsible for the initiation and progression of these tumours will provide

novel insights into the critical biological events responsible for the development of sarcomas in

general.

Cell of Origin

The discovery of the cell of origin is extremely important in understanding the molecular

mechanisms involved in the formation of different sarcoma subtypes. Due to the fact that sarcomas

are mesenchymal-derived tumours, recent studies have focused on mesenchymal stem cells (MSCs)

as the cells of origin for sarcomas [37]. MSCs are capable of differentiating along numerous

differentiation pathways including adipocytic, chondrocytic, osteogeneic and myogenic lineages.

Recent studies have made progress in identifying the supposed cells of origin of specific paediatric

sarcomas, particularly mxyoid liposarcoma, ES/PNET and RMS. Data from several sources has

supported the notion that these tumours arise from mesenchymal stem cells (MSCs), with

Page 19: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

9

expression of the tumour-specific fusion proteins in mesenchymal stem cells (TLS/CHOP in

myxoid liposarcoma [51]; PAX3/FKHR in ARMS [52]; EWS/FLI1 in ES [53-55]) inducing

phenotypic changes similar to those seen in the parent tumour. Furthermore, inhibition of the

EWS/FLI1 fusion gene in ES cell lines has been shown to shift the expression profiles of these cells

lines towards the MSC expression profile [56]. These cell lines were then shown to be able to

differentiate along both adipogenic and osteogenic lineages [56]. While increasing amounts of

evidence are being published to support the notion of MSCs as the cell of origin for many sarcoma

subtypes, this concept has been challenged by theories suggesting ES may emanate from the

neuroectodermal lineage. ES has been shown by immunohistochemistry (IHC) to express neural

and neuroectodermal markers, and one study has shown that the expression of the EWS/FLI1 fusion

transcript in neural crest-derived stem cells (NSCs) induces phenotypic and migratory changes [56,

57].

Traditionally, paediatric USTSs have been classified with ERMSs for the purposes of

treatment and therapeutic trials because these tumours often share a similar response to therapy [8,

58, 59]. More recent reports have suggested that at least some paediatric USTSs have more Ewing-

like characteristics, including sheet-like, round cell morphology, focal CD99 positivity (albeit

cytoplasmic), and occasional ultrastructural features of primitive neural differentiation [9, 33, 34,

60]. As previously mentioned, there is much debate as to whether MSCs or NSCs serve as the cell

of origin of many sarcomas. A recent study from our laboratory applied antibodies against several

MSC and NSC-associated proteins to a variety of sarcoma subtypes (ES, USTS, MPNST, ARMS,

ERMS, SS) in an attempt to determine whether differential expression of stem cell-associated

proteins could be used to aid in the subclassification of paediatric sarcomas [61]. It was found that

based on the expression of eight stem cell-associated proteins, paediatric sarcomas cluster into two

major subgroups (Figure 1.3). The first cluster included ES and USTS, and the second cluster

Page 20: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

10

contained the majority of the remaining tumours. Lack of expression of CD56 was significantly

associated with the ES/USTS cluster (Figure 1.4). Interestingly, CD56 is a neural stem cell marker,

and the lack of expression in ESs and USTSs is suggestive of a mesenchymal stem cell origin for

these tumours [61]. However, the results of this study were not conclusive enough to definitively

establish a cell of origin for either ES or USTS. On the other hand, the results from this study do

indicate that USTS is much more closely related to ES than to RMS. Thus, the cell of origin of

most primitive sarcomas remains elusive. However based on the primitive nature of these tumours,

their pathological similarity to ES, and the increasing amounts of evidence supporting MSCs as the

cell of origin for ES, it is likely that MSCs serve as the cell of origin for USTSs.

Page 21: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

11

Fig

ure 1

.2.

Eu

cli

dean

Clu

sterin

g o

f S

arcom

a T

um

ou

rs

Base

d o

n E

igh

t S

tem

Cell

Mark

ers.

B

ased

on t

he

expre

ssio

n o

f ei

ght

stem

cel

l-as

soci

ated

pro

tein

s, a

s m

easu

red b

y I

HC

, ped

iatr

ic s

arco

mas

clu

ster

into

tw

o m

ajor

sub

gro

ups.

T

he

firs

t cl

ust

er i

ncl

udes

ES

and U

ST

S,

and t

he

seco

nd c

lust

er c

onta

ins

the

rem

ainin

g t

um

ours

. B

lue

repre

sents

rel

ativ

e under

expre

ssio

n;

red r

epre

sents

rel

ativ

e over

expre

ssio

n;

gre

y r

epre

sents

mid

-lev

el e

xpre

ssio

n;

and

whit

e re

pre

sents

no d

ata.

(A

dap

ted

fro

m [

61])

.

Page 22: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

12

Acquisition of Malignant Properties

The striking specificity of individual translocations for different sarcoma subtypes leads one

to question the functional basis of a specific gene rearrangement in a given tumour. More

specifically, does the acquired gene fusion directly influence the histology of the tumour, or does

the histology of the tumour influence which cell types are able to support the oncogenic activity of

the gene fusion product [28]? These concepts speak to the timing of translocation acquisition;

whether fusion transcripts are acquired at later stages of cellular differentiation and tumour

Figure 1.3. Immunohistochemical Staining of CD56. Immunohistochemical

staining of CD56 in USTS (A), ES (B), MPNST (C) and SS (D). Lack of

expression of CD56, a neural stem cell marker, was significantly associated with

the ES/USTS cluster, where positive staining was found in all other tumour types.

A-D, CD56 immunostain with hematoxylin counterstain, original magnification x

400. (Adapted from [61]).

Page 23: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

13

formation, and simply facilitate tumour development, or if the acquisition of a translocation occurs

in more primitive cells, and is the critical event in the formation of a specific histologic subtype of

sarcoma. Conflicting evidence has been shown in ARMS. Some studies have suggested that the

critical event for RMS sarcomagenesis occurs after the tumour cells have acquired myoblastic

properties [37, 62]; whereas others suggest that the malignant phenotype occurs in the more

primitive MSCs [28, 37]. The latter theory is being more widely supported, as numerous studies

have now shown that PAX3 is an important myogenic regulator [63, 64], and that dysregulated

PAX3 (in the form of the ARMS fusion transcript PAX3-FOXO1) represses myogenic

differentiation [64-66]. The ability of PAX3 to commit cells to the myogenic lineage is however

only seen in specific cell types such as those of mesenchymal origin, and cannot induce muscle

commitment in endothelial cells [67]. Thus it is conceivable that dysregulation of PAX3 through a

genomic rearrangement in a very primitive cell may generate a skeletal muscle-derived tumour

[37]. Overall much evidence is supporting the notion that the critical events responsible for tumour

formation occur in more primitive cells as opposed to fully differentiated cells. Furthermore, the

degree of tumour differentiation is being linked to the point of cellular differentiation at which a

specific translocation is acquired [37]. For example, if the translocation is acquired early in lineage

commitment, the resulting tumour is much more primitive, however if the translocation is acquired

later in lineage commitment, the tumour will be well-differentiated. This has led our lab to develop

a hypothetical model of USTS molecular aberration acquisition (Figure 1.5), in which PRC

sarcomas acquire molecular aberrations at a much earlier stage of differentiation relative to other

sarcoma subtypes. The critical events leading to the development of most sarcoma subtypes are

believed to occur in mesenchymal progenitor cells. USTSs, on the other hand, are believed to be

the proliferation of uncommitted mesenchymal cells. It is this difference in the timing of

acquisition of molecular aberrations that results in the more primitive morphology of USTSs.

Page 24: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

14

Gaining a better understanding of this process in more primitive tumour types may offer insight

into the specific mechanisms of sarcomagenesis.

Figure 1.4. Decoding the early events of sarcoma development. Many sarcomas, such as

rhabdomyosarcoma, show evidence of differentiation down a specific connective tissue

lineage. Undifferentiated sarcoma shows no specific lineage differentiation, leading to the

hypothesis that the critical molecular events leading to undifferentiated sarcoma development

are likely happening at an earlier stage of differentiation compared to other sarcoma

subtypes.

Page 25: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

15

1.4.3 Molecular Studies of USTS

Identifying diagnostic and prognostic markers which help to more accurately identify

sarcoma subtypes is invaluable. Unfortunately, as with the morphological studies of USTS, the

molecular studies of these tumours have been rather uncommon and sporadic. However, in the past

5 years progress has been made in terms of characterizing some of the molecular abnormalities

arising in these tumours, although small sample sizes and minimal clinical correlation have been

limiting factors in these studies.

Immunohistochemical Screens

In general, immunohistochemical screening of USTS have been unsuccessful because of the

uniform negativity seen for most immunohistochemical markers, and inconsistent positivity for

others. However, Somers et al. [32] recently performed a thorough molecular analysis of 13

USTSs and identified certain molecular features of this specific USTS cohort.

Immunohistochemistry identified positivity for vimentin (92%), CD117 (92%), VEGF (69%),

HER-2 (54%), WT1 (46%), and Cox-2 (31%), among others. Alaggio et al. [9] performed a similar

immunohistologial study on 7 USTS tumours and found variable positivity for smooth muscle

actin, muscle-specific actin, CD34, cytokeratin, S-100, CD117, nestin, FLI1, CD105, survivin and

INI1. Discrepancies between these studies are likely a result of the small sample sizes used in each

study. While some of these immunohistochemical results are promising, more confirmatory studies

are necessary in order to determine the importance of these findings.

INI1

A specific type of sarcoma called malignant rhabdoid tumour has been found to harbour a

characteristic loss or mutation of the INI1 gene on chromosome 22q11.2. The INI1 protein is part

Page 26: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

16

of the SWI/SNF chromatin-remodelling complex. This complex can act as either a transcriptional

activator or repressor, and has been found to be constitutively expressed in all cells [68, 69]. In a

recent study Kreiger et al [33] sought to investigate whether USTSs show a loss of INI1 protein

expression due to underlying genomic abnormalities in the INI1 gene. By immunohistochemisty,

the study found 5/17 (29%) cases of USTS showing loss of nuclear expression of INI1. They

further identified by fluorescence in situ hybridization (FISH), reverse transcription-polymerase

chain reaction (RT-PCR) and/or mutational analysis that 4 of these 5 tumours harboured genetic

abnormalities of the INI1 gene. The report goes on to suggest that this category represents a unique

subgroup of tumours. Unfortunately others have found data inconsistent with these results,

suggesting that a category of USTSs showing INI1 loss may not be significant [9, 32].

IGF2

A recent publication by Somers et al. applied gene expression array analysis and post-array

validation to a series of USTSs and identified consistent upregulation of the excitatory components

of the insulin-like growth factor pathway. Particularly high expression of the insulin-like growth

factor 2 (IGF2) ligand was identified and confirmed using RT-PCR. IHC identified overexpression

of the IGF2 protein in 19 of 21 tumours (90%) with 2 distinct staining patterns (diffuse cytoplasmic

(16/19) and punctuate perinuclear (3/19)). Convincing and consistent upregulation of this pathway

in USTS suggests that the IGF signalling pathway may be a critical early event in sarcomagenesis,

however functional studies are necessary to confirm these studies.

CIC-DUX4 translocation

Over the past decade there have been sporadic case reports of tumours with PRC

morphology that harbour a rearrangement involving chromosome 4 and chromosome 19. In 1996,

Page 27: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

17

Richkind et al. [70] reported a balanced t(4;19)(q35;q13.1) translocation in a tumour diagnosed as a

malignant extraskeletal sarcoma. This translocation did not resurface in the literature again until

2006, when Kawamura-Saito et al. [60] reported two primitive Ewing-like sarcomas harbouring

t(4;19)(q35;q13) translocations. This group further deduced that in both cases, this translocation

resulted in the fusion of the CIC gene (19q13) to the C-terminal portion of the DUX4 gene (4q35).

Finally they found that overexpression of the CIC-DUX4 transcript in vitro increased anchorage-

dependent colony formation in murine NIH3T3 fibroblasts and directly induced over-expression of

downstream ETS-family transcription factors [60]. A more detailed description of the CIC and

DUX4 genes, as well as the CIC-DUX4 translocation, can be found in section 3.3.3. Most recently,

Rakheja et al. [71] published the 4th

case of a PRC sarcoma harbouring a t(4;19) translocation.

Unfortunately no group has attempted to characterize the prevalence of this translocation in a larger

cohort of USTS.

Page 28: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

18

1.5 Hypothesis, Objectives & Expected Outcomes

The accurate sub-classification of sarcomas is of utmost importance in order to determine an

accurate prognosis and to plan optimal therapy for the patient. Based on the limited literature that

has been published on USTSs, I hypothesize that paediatric undifferentiated sarcomas contain

distinct subgroups that can be identified based on their morphology, genomic aberrations and

expression profiles. To test this hypothesis, three objectives are proposed. Firstly, we seek to

develop an assay with which to screen our cohort of USTSs for the t(4;19)(q35;q13.1), and

ultimately determine the prevalence of this translocation in our cohort of USTSs. Secondly we seek

to characterize genomic aberrations within the t(4;19)(q35;q13.1)-negative USTSs, looking

specifically for any novel recurrent regions of gain, loss or rearrangement. Lastly, we seek to

characterize the expression profiles of the t(4;19)(q35;q13.1)-negative USTSs with the aim of

identifying a unique expression signature. With these objectives, we hope to identify specific

genomic changes which will aid in the characterization and diagnosis of USTSs, establish unique

genomic and protein expression profiles for USTSs for diagnostic and prognostic use, and to

identify potential genes or proteins for targeted treatment.

Page 29: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

19

CHAPTER TWO

MATERIALS & METHODS

Page 30: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

20

2. MATERIALS & METHODS

2.1 Tumour Specimens

An electronic search of the pathology database at the Hospital for Sick Children was

performed to identify children diagnosed with USTS between 1987 and 2007. Primary tumours

involving viscera and bone, and intradural tumours, were excluded. Pretreatment biopsy specimens

were studied if available, and all tumours underwent extensive immunohistochemical and

molecular genetic screening before being included in the series of USTS (see [32] for methodology

used). Thirteen of the 22 USTSs (USTS1-4, USTS6-11, USTS16-18) were part of previous studies

looking at the clinical, pathologic and cytogenetic features of paediatric USTSs [31, 32, 34, 72].

Three of the USTSs were originally diagnosed as atypical Ewing sarcomas (USTS20-22), due to

the fact that none of these tumours harboured a demonstrable rearrangement of the EWS gene by

RT-PCR, but had PRC morphology. Primary tumour samples were used when available; for one

patient, the primary tumour specimen was not available and therefore one post-therapy specimen

was studied. Frozen tissue from five additional paediatric USTSs (USTS12-15, USTS19) were

obtained from the files of the Co-Operative Human Tissue Network (Columbus, OH). Clinical

details were not available for one such tumour. All studies were performed in accordance with the

guidelines of the Hospital for Sick Children’s Research Ethics Board.

2.2 DNA and RNA Extraction

High molecular weight genomic DNA was extracted from snap-frozen tissue digested in a

proteinase K buffer and purified by standard phenol/chloroform methods [73]. All samples were

treated with RNase A (Roche Diagnostics, Laval, QC, Canada) for 30 minutes at 37oC. Total RNA

was extracted from snap-frozen tumour tissue samples using the TRIzol reagent according to the

Page 31: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

21

manufacturer’s instructions (Invitrogen, Carlsbad, CA), and treated with RNase-free DNase I

(Message Clean; GeneHunter Corporation, Nashville, TN) for 30 minutes at 37oC to remove DNA

contaminants. RNA extraction from formalin-fixed paraffin-embedded (FFPE) tissue samples was

performed as previously described [31]. In brief, tissue sections were deparaffinized in xylene and

subsequently washed in absolute ethanol. Following centrifugation, the tissue pellet was air-dried

and digested overnight at 55oC in 350 μl of RNA lysis buffer (20mM Tris pH 7.5, 10mM EDTA,

1% SDS, 500mg of Proteinase K). RNA extraction was then carried out using the TRIzol reagent

according to the manufacturer’s instructions (Invitrogen, Carlsbad, CA), and treated with RNase-

free DNase I (Message Clean; GeneHunter Corporation, Nashville, TN) for 30 minutes at 37oC to

remove DNA contaminants. Quality and concentration of extracted RNA and DNA samples were

quantified by the NanoDrop ND 3300 fluorospectrometer (NanoDrop products, Wilmington, DE).

2.3 Cytogenetic and Spectral Karyotyping (SKY) Analyses

Tumour specimens were processed for short-term culture in high-glucose Dulbecco’s

modified Eagle’s medium (4.5g/L) supplement with 15% fetal calf serum, 1% penicillin

streptomycin, and 1% L-glutamine [34]. The cultures were harvested within 5 days using 0.1

mg/mL colcemid (Gibco/BRL) for 2-4 hours, hypotonically treated with 0.075M KCl, and fixed in

3:1 methanol:acetic acid [34, 74]. The slides were dropped and aged for 2 to 3 days, after which

they were stained with Giemsa [34]. By convention, a minimum of 10 metaphases were analyzed

using conventional G-banding methods according to standard procedures, and were immediately

destained and dehydrated for subsequent fluorescence SKY analysis. The SKY analysis was

performed on the tumour metaphase cells according to the manufacturer’s instructions (Applied

Spectral Imaging, Carlsbad, CA) and as previously published [34, 74, 75]. Briefly, slides were

destained, formalin fixed, and denatured in 70% formamide/2XSSC at 75oC for 40 seconds. The

Page 32: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

22

SKYPaints were denatured, preannealed and hybridized to the denatured slides for 24 hours at

37oC. Posthybridization washes and detections were conducted according to the manufacturer’s

instructions. Spectral images were acquired with an SD 200 spectral bioimaging system (ASI,

Migdal Ha’Emek, Israel), and were analyzed using ASI SkyView software (version 1.2). When

sufficient metaphase cells of adequate quality were present, 10 metaphase cells were analyzed.

2.4 Array Comparative Genomic Hybridization (CGH)

All tumour specimens were selected to include only those tissue samples containing >80%

of identifiable neoplastic tissue by routine histological characterization. The Agilent human

genome CGH microarray 44 k (USTS1, USTS16) and 244 k (USTS2) (Agilent Technologies, Santa

Clara, CA) were used for the array CGH experiments. Three micrograms of human genomic DNA

from multiple anonymous male donors (Promega, Madison, WI) and 3 μg of test genomic DNA

sample were digested with AluI (5 units) and RsaI (5 units) (Promega, Madison, WI) for a

minimum of 2 hours at 37oC. Digestion quality was assessed by the DNA 1000 LabChip Kit

(Agilent 2100 Bioanalyzer). Labeling reactions were performed using the Agilent genomic DNA

labeling kit PLUS according to the manufacturer’s instructions. Briefly, the reference and sample

DNA were labeled with 1.5-3 mmol/L Cy5-dUTP or Cy3-dUTP (Agilent), and purified using a

Centricon YM-30 filter (Millipore, Billerica, MA). Probe mixture of Cy3-labeled sample DNA,

Cy5-labeled reference DNA, 50 μL of 1.0 mg/mL of human Cot-1 DNA (Invitrogen Canada,

Burlington, ON, Canada), 52 μL of Agilent 10x blocking agent and 260 μL of Agilent 2X

hybridization buffer was denatured at 100oC for 1 minute 30 seconds and incubated at 37

oC for 30

minutes. The probe was applied to the array using an Agilent microarray hybridization chamber,

and hybridized for 40 hours at 65oC in a rotating oven (Robbins Scientific, Sunnyvale, CA) at 20

rpm. Dye swaps were performed for both USTS1 and USTS2 samples. Arrays were washed

Page 33: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

23

according to the manufacturer’s recommendations, and then air dried and scanned using an Agilent

2565AA DNA microarray scanner. For each tumour, the data analysis of array CGH was

performed by applying rank segmentation, with a significance threshold of 1.0x10-6

, a maximum

contiguous probe spacing of 1,000 kb, and a minimum of 5 probes per segment (Nexus Copy

Number v.4; BioDiscovery, El Segundo, CA). Genomic imbalances were assigned as either gain

[log(3/2) or threshold of 0.2] or loss [log(1/2) or threshold of -0.3], considering only genomic

intervals >100 kb. The aberrations on X and Y chromosomes were excluded from the analysis to

eliminate the sex mismatching bias.

2.5 Interphase Fluorescence in situ Hybridization (FISH)

Interphase FISH was used in the pilot project portion of this thesis for the purposes of

identifying the translocation partners for chromosome 19 in samples USTS1 and USTS2.

Interphase FISH was also used in the exploratory array portion of this thesis in an attempt to

validate copy number changes of the EPHA3 gene in USTS12-15 and USTS19.

2.5.1 Isolation of DNA from Bacterial Artificial Chromosomes (BACs)

DNA was isolated from individual BAC clones grown overnight in a 20ml culture of Luria-

Bertani (LB) medium containing 12.5 μg/ml chloramphenicol (Sigma-Aldrich Canada Limited,

Oakville, ON, Canada). BAC DNA was isolated using the Qiagen Plasmid Midi Kit as per the

manufacturer’s instructions. Briefly, cell pellets were resuspended in Tris-EDTA (50mM Tris-

Chloride, pH 8.0, 10mM EDTA) and lysed in sodium hydroxide (200mM NaOH, 1% SDS).

Following neutralization with potassium acetate (3M KOAc, pH 5.5), BAC DNA was precipitated

in isopropanol for 1 hour at -80oC, and resuspended in DEPC water. Samples were then treated

with RNase A (Roche Diagnostics, Laval, QC, Canada) for 30 minutes at 37oC to remove RNA

Page 34: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

24

contaminants and DNA was isolated by standard phenol-chloroform methods, precipitated with

isopropanol and resuspended in sterile water. Quality and concentration of extracted DNA was

quantified by the NanoDrop ND 3300 fluorospectrometer (NanoDrop products, Wilmington, DE).

2.5.2 Pilot Project Four-Colour Interphase FISH

For the pilot project portion of this study, a four-colour interphase FISH method was

applied to USTS1 and USTS2 paraffin sections to validate the genomic rearrangement involving

chromosomes 4 and 19. The following BAC clones were used for BACs located at chromosome 19

(a-e), chromosome 4 (f,g), and chromosome 20 (h): (a) 19p13.12~p13.11 (16.00-16.42 Mb, control

probe): RP11-121I1 and RP11- 451E20; (b) 19q13.2 (47.02-47.33 Mb, probe A): RP11-688M4 and

RP11-108I20; (c) 19q13.2 (47.26-47.44 Mb, probe B): RP11-317E13; (d) 19q13.2~q13.31 (47.52-

47.91 Mb, probe C): RP11-374A11 and RP11- 1029C16; (e) 19q13.2 (47.44-47.61 Mb, probe E):

CTC- 790D18; (f) 4q33 (17.05-17.07 Mb): RP11-157C21 and RP11-242A14; (g) 4q35.2 (19.10-

19.11 Mb, 111 kb upstream of DUX4, probe D): RP11-521G19; and (h) 20p12.3 (7.39-7.55 Mb,

probe F): RP11-19D2. The linear order and approximate distances are based on the March 2006

assembly (Hg18) of the University of California, Santa Cruz, genome browser

(http://www.genome.ucsc.edu). DNA from all BAC clones was extracted by standard methods and

labeled with Vysis Spectrum Green-dUTP, Spectrum Red-dUTP, Spectrum Orange-dUTP (Abbott

Molecular, Des Plaines, IL), or Cy5-dUTP (PerkinElmer Life and Analytical Sciences, Waltham,

MA), using a Vysis nick-translation kit according to manufacturer’s instructions. The presence of

CIC, DEDD2, and ERF sequences and correct chromosome location of all BAC clones were

verified by PCR and by hybridization to metaphase spreads from normal peripheral lymphocytes,

respectively. Samples were scanned using the Zeiss Axio Imager.M1 microscope (Carl Zeiss

Page 35: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

25

Canada, Toronto, ON) equipped with appropriate filter sets and analyzed with ISIS imaging

software (MetaSystems, Altlussheim, Germany).

2.5.3 FISH Validation of Copy Number Gain of EPHA3

A BAC clone was selected that covers the EPHA3 gene region on 3p11.2 (89.13-89.30 Mb,

RP11-23D18). A commercially available CEP3 DNA probe labeled with Spectrum Orange-dUTP

and specific for the centromeric region 3p11.1-q11.1 (Vysis

Inc., Downers Grove, IL, USA) was

used as a control for both locus-specificity and ploidy analyses. DNA from all BAC clones was

extracted by standard methods and labeled with Spectrum Green–dUTP using the Vysis nick-

translation kit according to manufacturer’s instructions. Locus-specificity of the selected BAC

clones was verified by hybridization to metaphase spreads

from normal peripheral lymphocytes.

Five micrometer histological formalin-fixed and paraffin-embedded tissue sections were pre-treated

as follows: deparaffinization with a series of xylene followed by immersion in 100% ethanol,

incubation in 10mM NaCitrate (pH 6.4) at 80oC for 90 minutes followed by 2XSSC rinse, pepsin

digestion (4mg/ml pepsin in 100ml 0.01N HCl) at 45oC for 20 minutes followed by 2XSSC rinse

and a final ethanol dehydration series (70%, 90% and 100%, 2 minutes each) [76]. Slides and

probes were co-denatured on a hot plate at 75oC for 10 minutes and incubated overnight at 37

oC.

Following a post-hybridization rapid-wash technique consisting of one wash in 0.4xSSC and 0.3%

NP-40 at 72 °C for 3 min, followed by a 5-min wash at room temperature in 2XSSC and 0.1% NP-

40, slides were mounted in DAPI/Antifade (Vector Laboratories, Burlington, ON, Canada) and

visualized with a Zeiss Axioscope fluorescence microscope (Carl Zeiss Canada). The establishment

of EPHA3 gene copy number status was defined relative to the CEP 3 DNA probe gene copy

number.

Page 36: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

26

2.6 Reverse Transcriptase Polymerase Chain Reaction (RT-PCR) for Detection of CIC-DUX4

Fusion Transcripts

To test for the presence of the CIC-DUX4 translocation, RT-PCR amplification was

performed using the GeneAmp RNA PCR core kit (Applied Biosystems, Foster City, CA).

Complementary DNA was synthesized by reverse transcription (RT) using 1-2μg of total RNA and

primed using random hexamers. The RT reaction consisted of 15 minute incubation at 42oC

followed by inactivation at 99oC for 5 minutes. Specific amplification of the CIC-DUX4 fusion

transcript was performed using combinations of CIC forward primers and DUX4 reverse primers at

a concentration of 0.2μM for each primer (Table 2.1).

Primer Name Primer Sequence (5'-->3')

DUX4RTr2 [60] TGAGGGGTGCTTCCAGCG

DUX4-R2 ATGCCTTGCATCTGCCC

CIC4120 [60] TGAGTTGCCTGAGTTTCG

CIC2F ATCATGCAGGCTGCCACT

CIC3F CCCTGGAGCTGAGGCT

CIC4F ACTGGCACCGCTGCT

Table 2.1. Primer sequences for the RT-PCR reactions described in the

materials and methods. (Adapted from [30]).

Page 37: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

27

Primer sets were designed such that the PCR products were small enough so that the CIC-

DUX4 fusion transcript could be detected from RNA extracted from paraffin, and that primer sets

would detect all known breakpoints. The cycling parameters for each PCR reaction consisted of 40

cycles of 94oC for 30 seconds, 62

oC for 45 seconds, 72

oC for 45 seconds, followed by a final

elongation step at 72oC for 7 minutes. RT-PCR products were size-fractionated by gel

electrophoresis on a 1.5% agarose gel. To determine RNA integrity, cDNA from the same RT

reaction was also amplified with primers from the endogenous housekeeping gene

phosphoglycerate kinase 1 (PGK1). A single product of 247bp was visualized on a 1.5% agarose

gel with ethidium bromide staining (data not shown). A full-length CIC-DUX4 cDNA subcloned

into pGEM (Promega, Madison, WI; a kind gift from Dr. Takuro Nakamura, Department of

Carcinogenesis, Japanese Foundation for Cancer Research, Japan) was used as a positive control for

the CIC-DUX4 fusion transcript.

2.7 Copy Number Arrays

Copy number analysis was performed using the Affymetrix Genome-Wide Human SNP 6.0

array platform (Affymetrix, Santa Clara CA, USA). High molecular weight genomic DNA was

extracted from five snap-frozen t(4;19)(q35;q13.2)-negative USTS tumours (USTS12-15, USTS19)

as described in section 2.2. 500ng of genomic DNA was labeled and hybridized to the array as per

the manufacturer’s instructions at the Centre for Applied Genomics (The Hospital for Sick

Children, Toronto ON, Canada). Quality control was performed by Contrast QC calculation. Data

collected by the International HapMap project [77] was used as the normal control reference

(Affymetrix SNP 6.0 data on 270 normal samples).

Page 38: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

28

2.8 Expression Arrays

Genomic RNA expression analysis was performed using the Affymetrix Gene 1.0 ST array

platform (Affymetrix, Santa Clara CA, USA). Total RNA was extracted from five snap-frozen

t(4;19)(q35;q13.2)-negative USTS tumours as described in section 2.2. To test the purity of the

RNA samples, total RNA was run on the Agilent 2100 Bioanalyzer (Agilent Technologies, Santa

Clara, CA). The software for this program provided an RNA Integrity Number (RIN) for each

sample that allowed for estimation of the integrity of the total RNA extracted from each sample

[78]. Normal Adult Human Dermal Fibroblasts were obtained from Cedarlane Canada and

ScienCell Research Laboratories (Cedarlane Canada, Burlington On, Canada; ScienCell Research

Laboratories, Carlsbad CA, USA) and used as normal reference controls. Total RNA (200ng) from

normal human fibroblasts and tumour samples was analyzed as per manufacturer’s instructions at

the Centre for Applied Genomics (The Hospital for Sick Children, Toronto ON, Canada).

2.9 Analysis and Integration of Microarray Data

For the purposes of array analysis and integration two programs were used: Partek

Genomics Suite (PGS) and Ingenuity Pathway Analysis (IPA).

2.9.1 Partek Genomics Suite: Data Analysis and Integration

Gene copy number and RNA expression analyses were performed using the PGS Software.

Data from copy number array experiments and RNA expression array experiments were received in

the form of .cel files (GCOS 1.3 software), and were imported into, analyzed, and integrated using

the PGS software.

The analysis of copy number changes followed the PGS Copy Number Workflow, which

compares signal log2 ratios to a reference of 270 normal HapMap samples [79]. The PGS Genomic

Page 39: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

29

Segmentation tool was used to detect amplified and deleted segments with the following

parameters: minimum continuous probes set at 10, p-value threshold of 0.01, and signal to noise

ratio of 0.1. Regions of significant gain or loss were annotated to the corresponding genes on the

Affymetrix Gene 1.0 Array using the publically available HuGene-1_0-st-v1.na24.hg18.-

transcript.csv file. A gene list was subsequently created which contained all genes that were

significantly gained or lost in at least 3/5 (60%) of tumours.

The analysis of the expression array data used the PGS one-way analysis of variance

(ANOVA) tool at P < 0.05 and a cut-off of ±2-fold enrichment. Changes in gene expression were

cumulatively analyzed across all tumours versus normal fibroblast controls. Significantly over- and

under-expressed genes were annotated using the Affymetrix Gene 1.0 Array, which in turn uses the

publically available HuGene-1_0-st-v1.na24.hg18.-transcript.csv file. A gene list was subsequently

created which contained all genes that were significantly over- and under-expressed cumulatively

across all tumours analyzed.

The integration of genes lists of significantly gained and lost, and significantly over- and

under-expressed genes was performed using the PGS Venn analysis tool. The genes in the cross-

section area of the Venn diagram represent genes found to have significant changes in both copy

number and gene expression. From the list of genes found in the Venn diagram cross-section,

genes that showed copy number gain and under-expression or copy number loss and over-

expression were excluded from further analysis (5 of 51 genes). Copy number and gene expression

analysis excluded sex chromosomes to avoid bias in the identification of significant regions owing

to sex differences between some tumour samples and controls.

Page 40: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

30

2.9.2 Ingenuity Pathway Analysis: Network Identification and Pathway Analysis

Functional identification of gene networks was performed using the IPA program as

previously described [80, 81]. Excel (.xls) tables representing genomic changes (as per analysis of

the Affymetrix SNP 6.0 microarray data) and differentially expressed genes (as per analysis of the

Affymetrix Gene 1.0 ST microarray data relative to normal fibroblasts) in the tumour samples were

imported as individual experiments using the IPA Core Analysis tool. The analysis was performed

using the Ingenuity Knowledge Database for Humans only, and was limited to known direct

molecular interactions.

2.10 Quantitative Polymerase Chain Reaction (qPCR)

A quality control RT-PCR using primers for the PGK1 housekeeping gene demonstrated

that RNA from all five samples used for the microarray studies were of suitable quality for RT-PCR

experiments. Thus, RNA from the five tumour samples and two normal adult human fibroblasts

were reverse transcribed into complementary DNA using 1μg of total RNA and primed using

random hexamers. The RT reaction consisted of a 15 minute incubation at 42oC followed by

inactivation at 99oC for 5 minutes. Real-time PCR was performed as previously published [82, 83].

0.5μl of cDNA was used for real-time PCR using the SYBR green master mix as per

manufacturer’s instructions and primers at a concentration of 50μM each on the Roche

Lightcycler480 (Hoffmann-La Roche Limited, Mississauga, ON, Canada). Each combination of

tumour sample and gene primer sets was run in triplicate. Reactions were incubated for 5 minutes

at 95oC, followed by 50 cycles of 95

oC for 15 seconds, 60

oC for 40 seconds, and 72

oC for 15

seconds. Product specificity was confirmed by melt-point analysis. Primer sets for the four genes

of interest (EPHA3, SNX10, ADAM9, CDC73) as well as the housekeeping control were obtained

from SABiosciences (SABiosciences: A Qiagen Company, Frederick MD, USA). Data was

Page 41: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

31

analyzed by the comparative CT method (see below) using the TATA box-binding protein (TBP) as

the housekeeping control.

2.10.1 Comparative CT Analysis of qPCR Data

Comparative CT analysis, or 2-ΔΔC

T, is a method of presenting quantitative gene expression

[83]. This method calculates the relative gene expression of a gene of interest compared to an

internal control gene using the CT, or threshold cycle, of each test sample [83]. The CT is defined

as the PCR cycle at which the fluorescent signal of the reporter dye crosses an arbitrarily placed

threshold [83]. Plots of cDNA dilutions versus delta CT were performed, and the resultant plots

were consistently close to zero, indicating that the amplification efficiencies of the target and

housekeeping genes were similar for all gene primers. The following formula [83] was used to

determine relative fold-change for genes of interest in each tumour sample:

2-ΔΔC

T = [(CT gene of interest in tumour – CT internal control gene in tumour) – (CT gene of interest

in normal – CT internal control gene in normal)]

Where TBP was used as the internal control reference gene, and pooled adult human dermal

fibroblasts were used as the normal reference sample.

2.11 Immunohistochemistry (IHC)

IHC for CD99 was performed in all tumours in order to determine CD99 expression status.

IHC for EPHA3 was performed on the t(4;19)(q35;q13.1)-negative tumours used in the array

portion of the study in order to attempt to confirm the gain of EPHA3 in some of these tumours.

Page 42: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

32

2.11.1 IHC for CD99

Immunohistochemistry was performed on dewaxed slides cut at 4-µm using the Ventana

Benchmark XTTM autostainer (Ventana Medical Systems, Tucson, Arizona, USA) and the

Ventana i-VIEW LSAB DAB kit. The CD99 antibody (Leica-Novocastra, Richmond IL; clone

HO36-1.1) was used at 1:50 following heat-induced epitope retrieval. CD99 staining was

performed on all tumours.

2.11.2 IHC EPHA3

Immunohistochemistry with an EPHA3 antibody (1:300; Abnova, Walnut CA, USA) was

performed on 4-µm thick sections of each tumour. Tumours used for IHC were the same as those

used for array analysis (USTS12-15, USTS19). Optimal conditions of heat-induced epitope

retrieval using the Ventana 3,3’diaminobenzidine tetrachloride kit according to the manufacturer’s

instructions (Ventana Medical Systems Inc., Tucson, AZ). Sections were scored using a previously

established scoring system [32]. Intensity of staining was scored as 1 for low, 2 for moderate, and

3 for high, where the positive control was used as the standard for high intensity. Distribution of

staining was as 1 if less than 10% of cells were positive, 2 if 11-50% of cells were positive, and 3 if

greater than 50% of cells were positive. A combined score of 4 or more was considered positive,

and a combined score of less than 4 was considered negative. Optimization was performed on

tissues identified as highly expressing EPHA3 by the Human Protein Atlas version 6.0

(www.proteinatlas.org) [84-87]. Tissues identified as most favorable for optimization were testis,

kidney, placenta, and breast carcinoma. Ultimately breast carcinoma was identified as the optimal

positive control reference tissue.

Page 43: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

33

CHAPTER THREE

RESULTS & DISCUSSION

Page 44: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

34

3. RESULTS & DISCUSSION

3.1 Clinicopathological Features of Tumour Samples

In total 22 USTSs were used over the course of this thesis work. Table 3.1 summarizes the

clinicopathological features of the tumours; for one patient, the age and sex was not available. The

clinicopathological features of 13 tumours have been previously described [31, 32, 34, 72]. Briefly,

17 cases were composed of sheets of primitive round to plump spindle cells; one case was

composed of nests of PRCs; and four cases were composed of pure spindle cells within a myxoid,

collagenous or cellular fascicular background (Figure 3.1). There was a slight male predominance

(12:9) and the mean and median age was 9.3 and 11 years, respectively. The most common sites of

involvement were the trunk (50%), the extremities (27%) and the head & neck (23%). CD99

showed cytoplasmic positivity in 3 cases (USTS13-15) and membranous positivity in 5 cases

(USTS1-3, USTS20-21) (Figure 3.1).

Figure 3.1. Histological features and CD99 staining pattern of USTSs used in t(4;19) pilot study.

(A,B) USTS1 and USTS2 exhibited a primitive round cell phenotype. (C) USTS16 exhibited a more

spindled and myxoid phenotype. (D) CD99 staining of USTS2 exhibited weak and inconsistent membranous

staining. (A-C) Hematoxylin-eosin stain, x 200, hematoxylin and eosin. (D) CD99 antibody (Dako,

Carpinteria, CA, 1:50) with hematoxylin countertain. Magnification x 400. (Adapted from [32]).

Page 45: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

35

Sample Sex Age Site of Primary Morphology CD99 CIC-DUX4

status

USTS1 F 16 yr Trunk (abdominal wall) PRC, sheets Pos (m) POS

USTS2 M 14 yr H&N PRC, sheets Pos (m) POS

USTS3 F 11 yr Trunk (flank) PRC, sheets Pos (m) POS

USTS4 F 9 yr Trunk (paraspinal) PRC, nested Neg POS

USTS5 F 11 yr Trunk (inguinal) PRC, sheets Neg POS

USTS6 F 12 yr Trunk (chest wall) PRC, sheets Neg NEG

USTS7 F 14 yr Trunk (back) PRC, sheets Neg NEG

USTS8 M 13 yr H&N (neck) PRC, sheets Neg NEG

USTS9 M 12 yr Extremity (knee) PRC, sheets Neg NEG

USTS10 M 3 yr Extremity (toe) PRC, sheets Neg NEG

USTS11 M 4 yr Trunk (shoulder) PRC, sheets Neg NEG

USTS12 M 9 m Trunk (retroperitoneal) PRC, sheets Neg NEG

USTS13 M 9 m Extremity (arm) PRC, sheets Pos (c) NEG

USTS14 F 11 yr H&N (neck) PRC, sheets Pos (c) NEG

USTS15 n/a n/a Extremity (thigh) PRC, sheets Pos (c) NEG

USTS16 M 12 yr Trunk (paraspinal) SC, myxoid Neg NEG

USTS17 M 11 yr H&N (orbit) SC, myxoid Neg NEG

USTS18 F 7 yr Trunk (chest wall) SC, fascicular Neg NEG

USTS19 M 10 yr Trunk (flank) SC, collagenous Neg NEG

USTS20 M 13 yr Extremity (leg) PRC, sheets Pos (m) NEG

USTS21 M 11 yr Extremity (toe) PRC, sheets Pos (m) NEG

USTS22 F 9 m H&N (orbit) PRC, sheets Neg NEG

Table 3.1. Clinicopathological features, CD99 staining pattern, and CIC-DUX4 fusion

transcript status of the cohort of tumours used in the study. USTS, undifferentiated soft tissue

sarcoma; N/A, not available; H&N, head and neck; PRC, primitive round cell; SC, spindled cell; pos,

positive; neg, negative; m, membranous; c, cytoplasmic. (Table modified from [31]).

Page 46: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

36

3.2 t(4;19) Pilot Project

The following data has been published as follows: Yoshimoto M., Graham C., Chilton-MacNeill

S. et al., Cancer Genet Cytogenet. 2009;195:1-11 [31].

Prior to the commencement of this thesis project, our laboratory initiated a pilot project to

investigate the presence of a 19q13 rearrangement in our cohort of USTSs. This study applied

high-throughput array comparative genomic hybridization (CGH) together with spectral

karyotyping (SKY), four-colour fluorescence in situ hybridization (FISH), and reverse

transcriptase-polymerase chain reaction (RT-PCR) to a series of three paediatric USTS samples

(USTS1, USTS2, USTS16). Two of these samples have PRC morphology with CD99 positivity,

and the third sample has a spindled and myxoid appearance and is negative for CD99. G-banding

and SKY analyses were initiated and completed prior to my addition to the study group. Array

CGH studies had been initiated prior to my addition to the study, however analysis of the array

CGH results were completed by both myself and Dr. Maisa Yoshimoto. I subsequently played a

major role in the development and application of the four-colour FISH and RT-PCR assays.

3.2.1 Genomic Imbalances in Undifferentiated Sarcomas

High-resolution array CGH analysis was performed to identify: (a) copy number changes of

a whole chromosome; (b) copy number changes affecting a chromosomal arm; (c) small genomic

imbalances (>750kb or ≤ 10Mb); and (d) cryptic microaberrations (>100kb or ≤ 750kb) in the

paediatric USTS samples [34]. From comparison analysis of genomic gains and losses among the

three tumour samples, USTS16 exhibited the least genomic copy number imbalance relative to the

other samples (Figure 3.2). USTS1 and USTS2 exhibited the highest frequency of genomic

complexity (15 and 26 respectively), whereas USTS16 exhibited only 1 imbalance by array CGH.

Page 47: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

37

USTS2 harboured the most genomic losses (17/26), compared with USTS1 (3/15) and USTS16

(0/1). Although small genomic imbalances were widely detected in USTS1 (7/15), the high-

resolution genome-wide analysis also identified chromosome-arm (2/15) and whole chromosomal

copy number changes (1/15). USTS2 showed chromosome-arm copy number changes, small

genomic imbalances and cryptic microaberrations (1/26, 10/26, and 15/26, respectively). These

cryptic microaberrations were randomly distributed across the genome. Each genomic imbalance

interval in USTS1 and USTS2 was converted to the corresponding cytoband at 850-band resolution.

Notably, the 19q13 region was consistently rearranged in both USTS1 and USTS2, but not

USTS16. This common 19q13 region rearranged in USTS1 and USTS2 was further interrogated

using SKY and four-colour FISH.

Page 48: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

38

Fig

ure 3

.2.

Com

posi

te a

naly

sis

of

gen

om

ic i

mb

ala

nces

in t

hree U

ST

S s

am

ple

s, w

ith

wh

ole

gen

om

ic p

rofi

les

of

(A)

US

TS

16,

(B)

US

TS

1,

an

d (

C)

US

TS

2.

Reg

ions

of

gai

n a

re s

how

n o

n t

he

right

side

of

each

ver

tica

l pro

file

and a

re h

ighli

ghte

d a

s gre

en

sect

ions

on t

he

chro

moso

mal

rep

rese

nta

tions.

Reg

ions

of

loss

are

show

n o

n t

he

right

side,

and a

re h

ighli

ghte

d a

s re

d.

Aber

rati

ons

wer

e as

sign

ed a

s ei

ther

gai

n [

log(3

/2)

or

thre

shold

of

0.2

] or

loss

[lo

g(1

/2)

or

thre

shold

of

0.3

]. G

enom

ic i

mbal

ance

s w

ere

ass

igned

as e

ither

gai

n [

log(3

/2)

or

thre

shold

of

0.2

) o

r lo

ss [

log

(1/2

) o

r th

resh

old

of

0.3

], t

akin

g i

nto

acc

ou

nt

on

ly g

eno

mic

inte

rval

s >

10

0 k

b.

(Adap

ted f

rom

[30])

.

Page 49: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

39

3.2.2 Karyotypes of Undifferentiated Sarcomas

Metaphase cells from the 3 tumours were characterized by G-banding (data not shown) and

SKY analysis. All of the tumours demonstrated both balanced and unbalanced structural

rearrangements. The karyotypes are given in Table 3.2.

Sample Karyotype

USTS16

a 46,XY,t(1;15)(q?31;q?21),t(2;6)(q?33;q12~q14),der(8)t(8;20)(p23;q11.2),t(11;20)(q12

~q13;p11.2),del(20)(q11.2)

USTS1b 47,XX,i(1)(q10),der(4)t(4;19)(q33~q35;q13.1),+8,t(15,17)(q24;p11.2~p12),der(19)t(19

;20)(q13.1;p11.2),der(22)t(20;22)(q13;q13)

USTS2 46,XY,del(4)(q33),der(6)t(6;19)(p21.1;p13.3),der(7)t(7;19)(q36;q13.2),der(13)t(6;13)(

p21.1;p11.2),del(19)(p13.3),der(19)t(6;19)(p21.3;q13.2)

SKY analysis confirmed that the 19q13 region was consistently rearranged in the PRC sarcoma

samples. In sample USTS1, chromosome 19 was involved in two unbalanced translocations, one

with chromosome 4 and one with chromosome 20 (Figure 3.3A). In sample USTS2, chromosome

19 was involved in unbalanced translocations with chromosome 6 and chromosome 7 (Figure

3.3B). In both cases it was subsequently necessary to determine which chromosome was the

binding partner for the region of interest on chromosome 19 (19q13.1).

Table 3.2. Summary of the undifferentiated sarcoma karyotypes according to G-banding and

spectral karyotyping analyses. a Cytogenetic analysis previously described by Selvarajah et al

[33]. b Cytogenetic analysis previously described by Somers et al [36].

Page 50: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

40

3.2.3 Detailed Comparison of Cytogenetic Karyotyping and Array CGH

Comparison of the array CGH profiles to the SKY results showed that apparent balanced

rearrangements affecting chromosomes 1, 2, 6, 8, 11, 15, and 20 were associated with no

acquisition of genomic imbalances in USTS16 (data not shown). In contrast, both USTS1 and

USTS2 showed unbalanced translocations associated with genomic imbalances. Array CGH

profiling of USTS1 revealed loss and gain of the short and long arm of chromosome 1, respectively.

These genomic imbalances were consistent with the presence of i(1q), as previously described from

Figure 3.3. Representative spectral karyotyping (SKY) image of two USTS samples

showing multiple structural alterations in a diploid karyotype. (A) SKY and G-band

(data not shown) analysis of USTS1 showing a complex karyotype including

isochromosome of 1q, trisomy of chromosome 8 and rearrangements of chromosomes 4,

15, 17, 19, 20 and 22. (Adapted from [33]). (B) SKY analysis of USTS2 showing

chromosomal rearrangements on chromosomes 4, 6, 7, and 19. (Adapted from [30, 36]).

Page 51: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

41

SKY analysis [34]. In addition, the extra copy of chromosome 8 was detected by both array CGH

and SKY analyses [88]. The derivative chromosomes, such as der(4)t(4;19)(q33~q35;q13.1),

der(19)t(19;20)(q13.1;p11.2), and der(22)t(20;22)(q13;q13) were also associated with gain and loss

of genomic regions (Figure 3.4A). For example, genomic gains of chromosome 20 and genomic

loss of chromosome 22q were likely generated through unbalanced translocation mechanisms

involving chromosomes 20 and 22, respectively. The der(19)t(19;20)(q13.1;p11.2) unbalanced

translocation was accompanied by a genomic loss on chromosome 19. USTS2 also showed

genomic imbalances consistent with unbalanced chromosomal rearrangements. Small genomic

losses on chromosome 4 were associated with a deletion at the 4q33 region (Figure 3.4B).

Genomic gain of chromosomal arm 6p was likely generated through mechanisms involving

unbalanced translocations. USTS2 exhibited the recurrent 19q13 region rearrangement, and the

der(19)t(6;19)(p21.3;q13.2) and der(7)t(7;19)(q36;q13.2) unbalanced translocation were

accompanied by a genomic loss and gain on chromosome 19q13 (Figure 3.4B). These recurrent

19q abnormalities may lead to haploinsufficiency or gene fusion events affecting one or more genes

mapping to either the deleted or gained interval, the translocation breakpoints, or flanking genomic

regions. Notably, array CGH analysis indicated at least two candidate genes, CIC and ERF,

potentially affected by aberrations on chromosome 19.

Page 52: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

42

3.2.4 Four-colour FISH Validation of 19q Rearrangement and Detection of Translocation Binding

Partners

In order to further investigate the binding partners of the region of interest on chromosome

19, four-colour interphase FISH analysis was performed on corresponding formalin-fixed, paraffin-

embedded sections. This approach was also used to validate the genomic imbalances and to test the

sensitivity of the platform in detecting a small gain of interest in proximity to the microdeletion.

Copy number changes for the chromosomal 19q13 region frequently showing alterations were

recorded in 50 tumour cells for USTS1 and USTS2.

Page 53: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

43

USTS1

As predicted by cytogenetic analysis, the USTS1 tumour cells did not exhibit co-

localization of the control probe (pink) with probe C (yellow). Instead, an extreme nuclear

separation of these two probes was apparent and concurrent with the detection of losses of probe A

and B (green and red signals, respectively), confirming the presence of the 19q abnormalities

described by array CGH and SKY (Figure 3.5A). The BAC probes hybridizing to the unaffected

chromosome 19 showed the normal pink, green, red and yellow co-localization pattern.

Subsequently, we sought to investigate the binding partners of the chromosomal 19q

rearrangement in USTS1. Based on hybridization in the control sample (data not shown) and

tumour samples, the detection of chromosomal 19q rearrangement was defined when the distance

between two signals was more than three times the estimated signal diameter [89]. Fusion FISH

strategy was used in the analysis of 19q rearrangement, with BAC DNA probes, producing two-

colour fusions on the derivative chromosome. To determine the translocation partner for USTS1,

the four-colour FISH approach consisted of BAC DNA probe sets proximal to the breakpoint

region involving the CIC gene on chromosome 19 (probe E, red signal), the region upstream of the

DUX4 gene on chromosome 4 (probe D, green signal), a control region on chromosome 19 (probe

A, pink signal), and region of interest on chromosome 20 (probe F, yellow signal). Co-localization

of probes for the CIC gene and the DUX4 gene confirmed the presence of a t(4;19) translocation

(Figure 3.5B). Lack of co-localization of the red and yellow signals indicates that CIC is not

involved in a rearrangement with chromosome 20p.

Page 54: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

44

Figure 3.5. Four-colour FISH validation of 19q abnormalities and detection of

translocation binding partners in USTS1. (A) FISH probes used to validate genomic

imbalances in USTS1 (arbitrarily designated as probes A-C; see Materials and Methods for

details). Gene and BAC locations are taken from the UCSC Genome Browser (May 2004

assembly, Hg17; http://www.genome.ucsc.edu). (B) Representative FISH images

confirming deletions on chromosome 19 (green and red signals). Extreme nuclear

separation of the yellow and pink signals is indicative of a rearrangement involving 19q,

confirming array CGH and SKY results. BAC probes hybridizing to the unaffected

chromosome 19 showed the normal pink, green, red, and yellow colocalization pattern. (C)

FISH probes used to determine binding partner of 19q13 in USTS1 (arbitrarily designated

as probes D-F; see Materials and Methods for details). Gene and BAC locations are taken

from the UCSC Genome Browser (May 2004 assembly, Hg17;

http://www.genome.ucsc.edu). (D) FISH analysis identifies a t(4;19)(q35;q13.1)

translocation in USTS1. Probes for the CIC gene (probe E, red signal) showed

colocalization with a probe just upstream of DUX4 (probe D, green signal), confirming the

presence of the CIC-DUX4 fusion rearrangements. Lack of colocalization of the red and

yellow (probe F) signal indicates that CIC is not involved in a rearrangement with 20p. All

captured signals were converted to default false colours: Spectrum Orange to red, Spectrum

Red to pink, and Cy5 to yellow. (Adapted from [30]).

Page 55: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

45

USTS2

As predicted by array CGH, the USTS2 tumour showed cell populations with extra copies

of probes A and B (green and red signals, respectively), ranging from three to four copies for the

chromosomal loci analyzed, and hemizygous loss of probe C (yellow signal) (Figure 3.6A). The

BAC probes hybridizing to the unaffected chromosome 19 showed the normal pink, green, red and

yellow co-localization pattern.

To determine the translocation partner for USTS2, four-colour FISH was performed using

probes for the CIC gene, a control region on chromosome 19, the region of interest on chromosome

6 and the region of interest on chromosome 7. Surprisingly, a lack of co-localization was seen for

chromosome 19 with either chromosome 6 or 7 (data not shown). To be thorough, the FISH assay

used to detect the t(4;19) translocation in USTS1 was applied to both USTS2 and USTS16. No co-

localization of the CIC and flanking DUX4 probes was seen in USTS16 (data not shown).

However, in USTS2 the CIC probe signal showed co-localization with the probe immediately

upstream of DUX4 (probe D, green signal), indicating that this sample harboured a similar t(4;19)

translocation (Figure 3.6B). The probe for DUX4 falls within a region of hemizygous deletion on

chromosome 4 (as detected by array CGH), thus co-localization is indicative of a fusion of the CIC

gene to the unaffected chromosome 4. Therefore, both USTS1 and USTS2 showed evidence of a

t(4;19) CIC-DUX4 fusion event by FISH analysis.

Page 56: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

46

3.2.5 Detection of CIC-DUX4 Fusion Transcripts by RT-PCR

A t(4;19)(q35;q13.1) translocation had been previously identified in 4 samples with a

similar PRC morphology over the past 15 years [60, 70, 71]. One group determined that in 2 cases,

Figure 3.6. Four-colour FISH validation of 19q abnormalities and detection of

translocation binding partners in USTS2. (A) FISH probes used to validate genomic

imbalances in USTS2 (arbitrarily designated as probes A-C; see Materials and Methods for

details). Gene and BAC locations are taken from the UCSC Genome Browser (May 2004

assembly, Hg17; http://www.genome.ucsc.edu). (B) Representative FISH images showing

hemizygous loss of the yellow signal and gain of the red and green signals, confirming array

CGH results. BAC probes hybridizing to the unaffected chromosome 19 showed the normal

pink, green, red, and yellow colocalization pattern. (C) FISH probes used to validate binding

partner for 19q13 in USTS1, applied to USTS2 (arbitrarily designated as probes D-F; see

Materials and Methods for details). Gene and BAC locations are taken from the UCSC

Genome Browser (May 2004 assembly, Hg17; http://www.genome.ucsc.edu). (D) FISH

analysis identifies the presence of a t(4;19)(q35;q13.1) rearrangement in USTS2. The red

signal (probe E, CIC) showed extreme nuclear separation from the chromosome 19 control

probe signal (purple); however, this signal showed colocalization with a probe immediately

upstream of DUX4 (probe D, green signal). The probe for DUX4 falls within a region of

hemizygous deletion on chromosome 4; thus, colocalization is indicative of a fusion of the

CIC gene to the unaffected chromosome 4. All captured signals were converted to default

false colours: Spectrum Orange to red, Spectrum Red to pink, and Cy5 to yellow. (Adapted

from [30]).

Page 57: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

47

this translocation resulted in the fusion of the CIC gene (19q13) to the C-terminal portion of the

DUX4 gene (4q35). The primer sequences and positive control were obtained from [60], and RT-

PCR was run with the CIC4120 and DUX4RTr2 primers. Of the three tumours analyzed by RT-

PCR, the two tumours with PRC morphology (USTS1 and USTS2) were confirmed to harbour a

CIC-DUX4 fusion transcript (Figure 3.7). One of the two positive CIC-DUX4 fusion products was

consistent in size with published findings; however the other tumour sample (USTS2) contained a

novel variant CIC-DUX4 fusion transcript. Automated DNA sequencing of gel-purified transcripts

from the positive control and USTS2 confirmed fusions of the CIC gene with the DUX4 gene.

Unfortunately USTS1 had insufficient material for sequence analysis. The positive control was

890bp, with exon 20 of the CIC gene fused to the end of exon 1 of the DUX4 gene. The novel

variant CIC-DUX4 transcript was 710bp, with a 50bp deletion in CIC and a 130bp deletion in

DUX4 relative to the positive control. Interestingly, the fusion gene event involving CIC and

DUX4 was detected by RT-PCR and FISH analysis in both PRC sarcoma cases, but by SKY

analysis in only one case.

Figure 3.7. Confirmation of the CIC-DUX4 fusion transcript in two of

the three USTS samples. RT-PCR products from the three cases (USTS16,

USTS1, and USTS2) were sized against a 100-bp ladder marker using

electrophoresis on a 1.5% agarose gel. Both USTS1 (lane 4) and USTS2

(lane 5) showed the presence of the CIC-DUX4 fusion transcript. The full-

length CIC-DUX4 cDNA was subcloned into a pGEM vector (Promega,

Madison, WI) and used as a positive control (lane 2). (Adapted from [30]).

Page 58: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

48

3.2.6 Discussion of t(4;19) Pilot Project

This pilot project describes the results of a detailed analysis of the genomic aberrations in

three paediatric USTSs. Two of the tumours were of PRC morphology, and one tumour had

spindled, less cellular architecture with a focal hemangiopericytomatous-like pattern. All three

tumours had in common a lack of lineage-specific differentiation and no common translocations

associated with paediatric sarcomas. Both tumours with PRC morphology also exhibited CD99

positivity to varying degrees. Both tumours exhibiting the PRC phenotype with CD99

immunoreactivity harboured a recurrent rearrangement between 19q13 and 4q35, as detected by a

combination of SKY, array CGH, FISH and RT-PCR. As previously discussed, four cases of PRC

sarcomas have been previously reported that harbour similar rearrangements involving a

translocation between 19q13 and 4q35 [60, 70, 71]. One of these studies identified two tumours

with this rearrangement, and found that the fusion event between the CIC gene and the DUX4 gene

resulted in a protein with transforming properties. The human CIC gene codes for a protein that is

a member of the HMG-box superfamily of transcription factors [90]. The DUX4 gene is a double-

homeobox gene belonging to the family of double homeodomain transcriptional activators [91].

The biological roles of the CIC and DUX4 genes will be discussed in more detail in section 3.3.3.

As confirmed by RT-PCR, both t(4;19)-positive cases in this pilot project harboured the CIC-DUX4

fusion transcript. Notably, one tumour had a PCR product of a different size than those previously

published, suggesting a different site of rearrangement within one or both genes. Thus, both

tumours are members of the novel category of paediatric primitive sarcomas with a t(4;19)

rearrangement resulting in a CIC-DUX4 fusion transcript.

One particularly intriguing aspect of these analyses was that the original SKY analysis of

USTS2 did not reveal the presence of a t(4;19) rearrangement. By RT-PCR, and subsequent

Page 59: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

49

sequence confirmation, a CIC-DUX4 fusion transcript was shown to be present in this sample.

Finer mapping of the genome of USTS2 did indeed confirm the presence of a t(4;19)(q35;q13)

rearrangement, together with a gain, followed by a loss of chromosome 19q at the CIC region. As

chromosomal breakage generally occurs as part of chromosomal rearrangements [92], it is probable

that a small region of CIC was fused to DUX4, and that the size of the chromosome 19 locus that

was translocated was below the threshold of detection for SKY analysis. Thus, the absence of

specific translocations by SKY analysis does not exclude the presence of cryptic or complex

rearrangements below the threshold of detection, and RT-PCR remains the method of choice for

confirming the presence of this fusion gene.

Ultimately, molecular and cytogenetic analyses identified two tumours with PRC

morphology that harbour a t(4;19)(q35;q13) translocation involving the CIC gene on chromosome

19 and the DUX4 gene on chromosome 4. One tumour of spindled and myxoid morphology was

not found to harbour this translocation. These initial findings suggest that the CIC-DUX4 fusion

events are limited to tumours with an undifferentiated round cell phenotype. More importantly, this

pilot study allowed us to optimize RT-PCR and FISH conditions that can be used to screen the

remaining USTSs in our cohort.

3.3 Screening of USTSs Cohort for CIC-DUX4 Translocation

The following data has been submitted for publication as follows: Graham et al., Pediatr Dev

Pathol, Submitted October 2010 [93].

In this portion of the study, an additional 19 tumours were screened for the CIC-DUX4

fusion transcript with an RT-PCR assay developed for both frozen and paraffin-based tissues. A

combination of primer sets were used which cover all known breakpoints of the CIC-DUX4 fusion

Page 60: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

50

transcript. Some of the clinicopathological features of these tumours have been previously

described [32, 34, 72, 88]. Fifteen cases were composed of sheets of primitive round to plump

spindle cells, one case was composed of nests of PRCs; and three cases were composed of pure

spindle cells within a myxoid, collagenous or cellular fascicular background. CD99 showed

cytoplasmic positivity in three cases (USTS9-11) and membranous positivity in 3 cases (USTS3,

USTS20, USTS21). The ultrastructural features of USTS3, USTS4, USTS6-11 and USTS17 have

been previously described [34, 72]. For seven USTSs, tissue was not available for ultrastructural

analysis. The remaining 2 USTSs with PRC morphology showed sparse intracytoplasmic

neurosecretory granules, variable amounts of intracytoplasmic glycogen and cytoplasmic processes.

3.3.1 Detection of the CIC-DUX4 fusion by RT-PCR

Total RNA from ten fresh-frozen USTS samples and nine FFPE USTS samples were

subjected to RT-PCR analysis for the CIC-DUX4 fusion transcript. All samples were subjected to a

series of PCR reactions using a combination of CIC forward primers and DUX4 reverse primers

(Table 2.1). CIC4120 and DUX4RTr2 were selected as the optimal primer set for fresh-frozen

samples as they result in the largest PCR product, however all primer sets successfully amplified

specific PCR products (data not shown). CIC4F and DUX4-R2 were selected as the optimal primer

set for RNA extracted from FFPE tissue samples. Of the nineteen tumour samples analyzed by RT-

PCR, three were positive for the CIC-DUX4 fusion transcript (USTS3, USTS4 and USTS5) (Figure

3.8). All three CIC-DUX4 positive tumours had PRC morphology, and one tumour showed

membranous positivity for CD99. Two of the three positive CIC-DUX4 fusion products (USTS3,

USTS4) were consistent in size and sequence with previously published findings [60], whereas one

product (USTS5) contained a novel variant of the CIC-DUX4 fusion transcript.

Page 61: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

51

Figure 3.8. Identification of the CIC-DUX4 fusion by reverse transcriptase-polymerase chain

reaction (RT-PCR) screening. (A) Total RNA samples from 10 fresh-frozen USTS tumours were

subjected to RT-PCR analysis for the CIC-DUX4 fusion transcript using the CIC4120 forward and

DUX4RTr2 reverse primers. These products were sized against a 100-bp ladder marker using

electrophoresis on a 1.5% agarose gel. USTS5 (lane 3) showed the presence of the CIC-DUX4 fusion

transcript. The full-length CIC-DUX4 cDNA was subcloned into a pGEM vector (Promega, Madison,

WI) and used as a positive control (lane 2). (B) Total RNA from 9 formalin fixed paraffin-embedded

USTS tumours was subjected to RT-PCR analysis for the CIC-DUX4 fusion transcript using the CIC4

forward and DUX4-R2 reverse primers. These products were sized against a 100-bp ladder marker

using electrophoresis on a 1.5% agarose gel. USTS3 (lane 3) and USTS4 (lane 4) showed the presence

of the CIC-DUX4 fusion transcript. The full-length CIC-DUX4 cDNA was subcloned into a pGEM

vector (Promega, Madison, WI) and used as a positive control (lane 2). (Adapted from [93]).

Page 62: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

52

3.3.2 DNA Sequence Analysis of the CIC-DUX4 Positive PCR Products

Gel-purified transcripts from the positive control, USTS3, USTS4 and USTS5 were directly

sequenced using automated DNA sequencing. Sequencing confirmed fusions of the CIC gene with

the DUX4 gene in all three cases, as well as the positive control. Using the CIC4120 forward

primer and DUX4rtR2 reverse primer, the positive control was 890bp. Using the same primer set

USTS5 was 821bp, with an additional 10bp in CIC and a 75bp deletion in DUX4 relative to the

positive control.

Using the CIC4F forward primer and the DUX4-R2 reverse primer, the positive control was

170bp. USTS3 and USTS4 shared the same breakpoint as the positive control. In all cases, exon

20 of the CIC gene was fused to exon 1 of the DUX4 gene. Combining the sequencing analysis of

these three additional cases with the sequencing analysis from the pilot project resulted in the

identification of three breakpoints associated with the CIC-DUX4 fusion transcript in our cohort

(Figure 3.9). Based on the sequence analysis results, the amino acid structure of the fusion

proteins was predicted (Figure 3.10). This results in an in-frame fusion between CIC and DUX4

with the CIC open reading frame. This fusion leaves intact virtually all functional regions of the

CIC gene, including the DNA-binding high-mobility group (HMG)-box and fifteen of sixteen

putative MAPK phosphorylation sites [94-98]. On the other hand, this translocation results in the

loss of the majority of the DUX4 functional regions, including both DNA-binding homeodomains

(Figure 3.11).

Page 63: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

53

CIC mRNA Exons 17-20

...GGTCCTGTCAGAAGTGGACTTCGAAGAGCGCTTTGCTGAGTTGCCTGAGTTTCGGCCTGAGGAGGTGCTGCCCTCCCCCACCCTGCAG

CIC4120

TCTCTGGCCACCTCACCCCGGGCCATCCTGGGCTCTTACCGCAAGAAGAGGAAGAACTCCACGGACCTGGATTCAGCACCCGAGGACCCCA

CCTCGCCCAAGCGCAAGATGAGAAGACGCTCCAGCTGCAGCTCGGAGCCCAACACCCCCAAGAGTGCCAAGTGCGAGGGGGACATCTTCAC

CTTTGACCGTACAGGTACAGAAGCCGAGGACGTGCTTGGGGAGCTAGAGTATGACAAGGTGCCATACTCCTCCCTGCGGCGCACCCTGGAC

CAGCGCCGGGCCCTGGTCATGCAGCTCTTTCAGGACCATGGCTTCTTCCCGTCAGCCCAGGCCACAGCCGCCTTCCAGGCCCGCTATGCAG

ACATCTTTCCCTCCAAGGTTTGTCTGCAGTTGAAGATCCGTGAGGTGCGCCAGAAGATCATGCAGGCTGCCACTCCCACGGAGCAGCCCCC

CIC2F

TGGAGCTGAGGCTCCTCTCCCTGTACCGCCCCCCACTGGCACCGCTGCTGCCCCTGCCCCCACTCCCAGCCCCGCAGGGGGCCCTGACCCC

CIC3F CIC4F

ACCTCACCCAGCTCGGACTCTGGCACGGCCCAGGCTGCCCCGCCACTGCCTCCACCCCCAGAGTCGGGGCCTGGACAGCCTGGCTGGGAGG

GGGCTCCCCAGCCCTCCCCCCCACCCCCAGGTCCCTCCACAGCTGCCACAGGCAGGTGA

- Positive Control, USTS3 & USTS4 breakpoint

- USTS1 breakpoint

- USTS5 breakpoint

DUX4 mRNA

...CCGCCCCCGCGCTGCAGCCCAGCCAGGCCGCGCCGGCAGAGGGGGTCTCCCAACCTGCCCCGGCGCGCGGGGATTTCGCCTACGCCGC

CCCGGCTCCTCCGGACGGGGCGCTCTCCCACCCTCAGGCTCCTCGGTGGCCTCCGCACCCGGGCAAAAGCCGGGAGGACCGGGACCCGCAG

CGCGACGGCCTGCCGGGCCCCTGCGCGGTGGCACAGCCTGGGCCCGCTCAAGCGGGGCCGCAGGGCCAAGGGGTGCTTGCGCCACCCACGT

CCCAGGGGAGTCCGTGGTGGGGCTGGGGCCGGGGTCCCCAGGTCGCCGGGGCGGCGTGGGAACCCCAAGCCGGGGCAGCTCCACCTCCCCA

GCCCGCGCCCCCGGACGCCTCCGCCTCCGCGCGGCAGGGGCAGATGCAAGGCATCCCGGCGCCCTCCCAGGCGCTCCAGGAGCCGGCGCCC

DUX4R2

TGGTCTGCACTCCCCTGCGGCCTGCTGCTGGATGAGCTCCTGGCGAGCCCGGAGTTTCTGCAGCAGGCGCAACCTCTCCTAGAAACGGAGG

CCCCGGGGGAGCTGGAGGCCTCGGAAGAGGCCGCCTCGCTGGAAGCACCCCTCAGCGAGGAAGAATACCGGGCTCTGCTGGAGGAGCTTTA

DUX4RTr2

G

- Positive Control, USTS3 & USTS4 breakpoint

- USTS1 breakpoint

- USTS5 breakpoint

Figure 3.9 Nucleotide sequences of CIC and DUX4 sequences denoting primer sequences and translocation

breakpoints. (Top) Partial nucleotide sequence of the CIC gene, with CIC forward primer sequences underlined,

and different translocation breakpoints denoted by coloured arrows. (Bottom) Partial nucleotide sequence of the

DUX4 gene, with DUX4 reverse primer sequences underline, and different translocation breakpoints denoted by

coloured triangles. Arrows represent direction in which the genes are fused to one another. (Adapted from [93]).

Page 64: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

54

USTS1 Fusion

... GCC ACT CCC ACG GAG CAG CCC CCT GGA GCT GAG GCT CCT CTC CCT GTA CCG CCC CCC

A A T P T E Q P P G A E A P L P V P P P

ACT GGC ACC GCT GCT GCC CCT GCC CCC CTC CCC TGC GGC CTG CTG CTG GAT GAG CTC CTG

T G T A A A P A P L P C G L L L D E L L

GCG AGC CCG GAG TTT CTG CAG CAG GCG CAA CCT CTC CTA GAA ACG GAG GCC CCG GGG GAG

A S P E F L Q Q A Q P L L E T E A P G E

CTG GAG GCC TCG GAA GAG GCC GCC TCG CTG GAA GCA CCC CTC AGC GAG GAA GAA TAC CGG

L E A S E E A A S L E A P L S E E E Y R

GCT CTG CTG GAG GAG CTT TAG

A L L E E L *

Positive Control, USTS3 and USTS4 Fusion

... GCC ACT CCC ACG GAG CAG CCC CCT GGA GCT GAG GCT CCT CTC CCT GTA CCG CCC CCC

A A T P T E Q P P G A E A P L P V P P P

ACT GGC ACC GCT GCT GCC CCT GCC CCC ACT CCC AGC CCC GCA GGG GGC CCT GAC CCC ACC

T G T A A A P A P T P S P A G G P D P T

TCA CCC AGC TCG GAC TCT GGG GGT GGA CCC CAA GCC GGG GCA GCT CCA CCT CCC CAG CCC

S P S S D S G G G P Q A G A A P P P Q P

GCG CCC CCG GAC GCC TCC GCC TCC GCG CGG CAG GGG CAG ATG CAA GGC ATC CCG GCG CCC

A P P D A S A S A R Q G Q M Q G I P A P

TCC CAG GCG CTC CAG GAG CCG GCG CCC TGG TCT GCA CTC CCC TGC GGC CTG CTG CTG GAT

S Q A L Q E P A P W S A L P C G L L L D

GAG CTC CTG GCG AGC CCG GAG TTT CTG CAG CAG GCG CAA CCT CTC CTA GAA ACG GAG GCC

E L L A S P E F L Q Q A Q P L L E T E A

CCG GGG GAG CTG GAG GCC TCG GAA GAG GCC GCC TCG CTG GAA GCA CCC CTC AGC GAG GAA

P G E L E A S E E A A S L E A P L S E E

GAA TAC CGG GCT CTG CTG GAG GAG CTT TAG

E Y R A L L E E L *

USTS5 Fusion

... GCC ACT CCC ACG GAG CAG CCC CCT GGA GCT GAG GCT CCT CTC CCT GTA CCG CCC CCC

A A T P T E Q P P G A E A P L P V P P P

ACT GGC ACC GCT GCT GCC CCT GCC CCC ACT CCC AGC CCC GCA GGG GGC CCT GAC CCC ACC

T G T A A A P A P T P S P A G G P D P T

TCA CCC AGC TCG GAC TCT GGC ACG GCC CAG CAA GGC ATC CCG GCG CCC TCC CAG GCG CTC

S P S S D S G T A Q Q G I P A P S Q A L

CAG GAG CCG GCG CCC TGG TCT GCA CTC CCC TGC GGC CTG CTG CTG GAT GAG CTC CTG GCG

Q E P A P W S A L P C G L L L D E L L A

AGC CCG GAG TTT CTG CAG CAG GCG CAA CCT CTC CTA GAA ACG GAG GCC CCG GGG GAG CTG

S P E F L Q Q A Q P L L E T E A P G E L

GAG GCC TCG GAA GAG GCC GCC TCG CTG GAA GCA CCC CTC AGC GAG GAA GAA TAC CGG GCT

E A S E E A A S L E A P L S E E E Y R A

CTG CTG GAG GAG CTT TAG

L L E E L *

Figure 3.10. Nucleotide and predicted amino acid sequences of the different CIC-DUX4 fusions. (Top) In-frame

fusion between CIC and DUX4 in USTS1. (Middle) In-frame fusion between CIC and DUX4 in the positive control,

USTS3 and USTS4. 2 additional glycine residues are present at the fusion point which do not natively belong to

either CIC or DUX4. (Bottom) In-frame fusion between CIC and DUX4 in USTS5. Legend: CIC nucleotide sequence,

DUX4 nucleotide sequence, nucleotide sequence not belonging to CIC or DUX4, CIC amino acid sequence, DUX4

amino acid sequence, amino acid sequence not belonging to CIC or DUX4.

Page 65: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

55

3.3.3 Discussion of Overall Screening of USTS cohort of CIC-DUX4 Fusion Transcript

Paediatric USTSs are a very poorly understood group of tumours on which very little data

had been published. Recently however, several groups have identified a particular variant of USTS

having a PRC phenotype, and harbouring recurrent translocations involving chromosomes 4q35 and

19q13 [31, 60, 70, 71]. One such study determined that in two cases, this rearrangement resulted in

the fusion of the CIC gene on chromosome 19q13.1 and the DUX4 gene on chromosome 4q35 [60].

Characterization of the genes involved in the t(4;19)(q35;q13.1) translocation has provided the

necessary information to develop an RT-PCR approach for the detection of the fusion transcript

resulting from this translocation. As such, the aims of this study were two-fold: (i) to identify

Figure 3.11. Predicted protein structure of CIC-DUX4 fusion protein variants.

Predicted structure of the CIC–DUX4 protein is based on the results of sequence analysis and

protein prediction. This fusion leaves intact most of the functional regions of the CIC gene,

including the DNA-binding high-mobility group-box and the majority of the putative MAPK

phosphorylation sites. However, this translocation results in the loss of the majority of the

DUX4 functional regions, including both DNA-binding homeodomains. (Adapted from [93]).

Page 66: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

56

primer sets and RT-PCR conditions that could be used to effectively detect the CIC-DUX4 fusion

transcript on RNA extracted from both fresh-frozen and FFPE tissue and (ii) to screen a large

cohort of paediatric USTSs for the presence of this rearrangement.

Overall our institution has screened 22 USTS tumours and identified 5 of 18 (28%) tumours

with PRC morphology that harbour the CIC-DUX4 fusion transcript. Three of these 5 tumours

show CD99 positivity. Given that CIC-DUX4-positive tumours have been previously described as

‘atypical Ewing sarcomas’ [31, 88] or ‘Ewing-like sarcomas’ [60], the distinction of ESs and CIC-

DUX4-positive tumours on morphological and immunohistochemical grounds appears to be

challenging. Both exhibit some degree of CD99 positivity and ultrastructural evidence of neural-

type differentiation. However, previous reports have highlighted that the PRC sarcomas appear less

differentiated, with some lacking evidence of neural differentiation [31, 71] and an overall

inconsistent CD99 staining pattern [31, 60, 71]. Nevertheless, definitive diagnosis rests upon

cytogenetic and molecular diagnostic assays for identification of this specific rearrangement.

Interestingly, 4 distinct breakpoints were identified in the 5 positive cases (Figure 3.9),

though we were unable to determine the specific breakpoint in USTS1 due to insufficient material

for sequencing. The breakpoint is variable in both CIC and DUX4, suggesting that the structure of

both of these genes may facilitate multiple rearrangements with different genomic sequences [99].

In all cases, however, exon 20 of the CIC gene was fused to exon 1 of the DUX4 gene, leaving

intact the functional regions of CIC, but resulting in a loss of all DNA-binding portions of DUX4

(Figure 3.10). This suggests that while a specific nucleotide breakpoint may not be necessary, the

fusion between specific higher-level functional domains may be of particular importance.

Page 67: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

57

The human CIC gene is an ortholog of the Drosophila capicua gene, and is a member of the

HMG-box superfamily of transcription factors [90]. This gene has 20 exons encoding a protein of

1608 amino acids [100], and contains an N-terminal DNA-binding HMG-box and sixteen possible

MAPK phosphorylation sites [94-98, 101]. Expression of CIC is predominantly limited to

immature granule cells in the central nervous system, and is important for their development [90,

101]. CIC has been shown to be involved in mediating two oncogenic signaling pathways, EGFR

and Wnt, by transcriptional repression. In cancer, CIC has been shown to be differentially

expressed in medulloblastomas [101], and has been shown to play a role in the pathogenesis of

PRC sarcomas [31, 60]. Furthermore, CIC has been shown to play a role in spinocerebellar ataxia

type 1 (SCA1) through association of the CIC protein with ATAXIN-1 (ATXN-1) [102, 103].

Binding of ATXN-1 to the N-terminal region of CIC alters the repressional activity of the CIC

protein [103].

The DUX4 gene is a double-homeobox gene belonging to the family of double

homeodomain transcriptional activators [91]. DUX4 is located within the tandem repeat locus

D4Z4 on chromosome 4 [104], and contains two DNA-binding homeoboxes at its N-terminus [91].

DUX4 has been implicated in the pathogenesis of facioscapulohumeral muscular dystrophy, in

which the D4Z4 copy number is significantly reduced [104, 105], and in ERMS where DUX4 is

involved in a chromosomal rearrangement with the EWSR1 gene [106]. Under normal conditions

DUX4 is believed to be involved in myogenic differentiation and cell-cycle control [106], and over-

expression of this gene in cultured primary myoblasts has been shown to induce caspase activity

and promote cell death [107].

Page 68: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

58

The CIC-DUX4 fusion preserves the majority of the CIC gene, including the DNA-binding

HMG-box and the majority of the MAPK phosphorylation sites, but both DUX4 homeobox

domains were lost. Preliminary evidence suggests that the chimeric protein has transforming

properties, and acts as a strong transcriptional activator of downstream CIC targets, with CIC-

DUX4 showing 130-fold enhancement of transcriptional activity against wild-type CIC [60, 108].

Nonetheless, full characterization of the CIC-DUX4 fusion product will be required to fully

understand the functional role that this rearrangement plays in oncogenesis.

The CIC-DUX4-positive cohort forms a distinct subcategory of tumours with PRC

morphology and variable membranous CD99 positivity. Of note are another 17 cases of USTS

lacking the CIC-DUX4 transcript by RT-PCR. Such tumours may harbour variants of the t(4;19)

rearrangement not detected in the current analysis, or may represent a different category of

primitive sarcomas altogether.

3.4 Discovery of Novel Markers for CIC-DUX4-Negative Tumours

The t(4;19) pilot project and screening portions of this project allowed for the identification

of 5 novel cases of paediatric PRC sarcomas that harbour the CIC-DUX4 fusion transcript.

Furthermore, this screening allowed for the identification of a cohort of CIC-DUX4-negative

USTSs with no established genetic markers. As such we sought to characterize novel genomic

aberrations that may play a role in the deregulation of gene expression in these CIC-DUX4-negative

tumours. Recent developments in microarray technologies have revolutionized the way in which

we study the relationships between genomic changes and gene expression in cancer cells, allowing

us to study this relationship on a whole genome scale [81]. Identifying changes in gene expression,

as well as the mechanisms responsible for these changes, serves to significantly enhance our

Page 69: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

59

understanding of oncogenesis [109]. Moreover, the integration of platforms that identify genomic

and transcriptomic aberrations will aid in determining genetic variations that control the

development and progression of cancer. As such, in order to study the effects of genomic

mechanisms on sarcoma-related gene networks, we used an integrative approach for genome-wide

profiling of genomic and gene expression changes. To do so we have tested DNA and RNA from

five CIC-DUX4-negative tumours on the Affymetrix Human SNP 6.0 microarray and the

Affymetrix Gene 1.0 ST expression microarray, respectively.

3.4.1 DNA Copy Number Analysis

In order to identify genome-wide copy number changes in each tumour sample, high

molecular weight genomic DNA was sent to The Centre for Applied Genomics (TCAG) microarray

facility to be run on the Affymetrix Genome-Wide Human SNP Array 6.0 microarray platform

(Affymetrix Inc., Santa Clara, CA, USA). Copy number analysis was conducted in 5 primary

tumour samples – USTS12, USTS13, USTS14, USTS15 and USTS19. Copy number analysis was

performed using the PGS software, using the publicly available normal control SNP data from the

HapMap consortium [79] as the reference for determining copy number alterations. While copy

number analysis revealed gross heterogeneity between the individual tumour samples (Figure 3.12),

numerous smaller regions could be identified which showed similar patterns of aberration across

the majority of the tumour samples. Tumour specific copy number changes ranged between 250

(USTS13) and 10019 (USTS12). In all tumours there was more copy number gain relative to loss.

A cumulative gene list was created that contained the genes showing statistically significant copy

number changes (P ≤ 0.05) in at least 3 of the 5 tumour samples.

Page 70: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

60

3.4.2 Gene Expression Analysis

To characterize the expression profiles of these tumours, total RNA from each tumour as

well as two adult human dermal fibroblast samples was sent to the TCAG microarray facility to be

run on the Affymetrix Gene 1.0 ST microarray platform (Affymetrix Inc, Santa Clara, CA, USA).

The RNA extracted from USTS15 was not of high enough quality to be used for the expression

microarray analysis. As such the tumour-specific integration analysis, cumulative expression

microarray analysis and cumulative integration analysis did not include USTS15. Expression

Figure 3.12. Overall genomic profiling view from Partek Genomic Suite (PGS). High

molecular weight DNA from five t(4;19)(q35;q13.2)-negative USTS tumour samples were

tested on the genome-wide human SNP 6.0 microarray platform , and results were analyzed

using PGS to identify tumour-specific and cumulative copy number alterations. Copy number

analysis was normalized against the publicly available data collected by the International

HapMap consortium. A cumulative gene list was created that contained genes showing

statistically significant copy number changes (P ≤ 0.05) in at least 3 of the 5 tumour samples.

The x-axis contains chromosomes 1-22, X and Y in order.

Page 71: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

61

analysis was performed using the PGS software, using human fibroblasts as reference RNA

samples for determining differential expression. Clustering analysis showed distinct clustering of

tumours relative to the normal human fibroblast samples (Figure 3.13). Tumour specific changes

ranged between 726 genes in USTS14 and 1370 genes in USTS13. An overall cumulative analysis

was performed, which compared the cumulative expression profiles of all tumours to the combined

profiles of the normal fibroblasts. This analysis produced a gene list with genes showing at least

two-fold, statistically significant (P ≤ 0.05) differential gene expression, cumulatively across the

tumours.

Figure 3.13. Clustering analysis shows distinct clustering of tumours relative to normal

human fibroblast samples. (A) Hierarchical clustering using average Euclidean cluster analysis

on 4 tumours tested on the Gene 1.0 ST expression microarray platform. Tumour samples (blue)

clustering separately from the normal fibroblast control expression profiles. (B) Principal

component analysis (PCA) of individual tumour samples and normal human fibroblast control

samples from the expression analysis. The PCA tool in PGS subjects the raw expression data to

3-dimensional clustering analysis. The PCA-generated ellipsoids denote separate clustering

between normal and tumour samples in the gene expression analysis.

Page 72: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

62

3.4.3 Integration of Copy Number and Gene Expression Analyses

Tumour-Specific Integration

Microarray and statistical analyses using PGS software allowed for the identification of

genomic and transcriptional alterations in individual tumour samples. Genes with significant

changes (P ≤ 0.05) in copy number and gene expression in individual tumour samples were

analyzed using the PGS-Venn analysis tool. Integration of these data revealed gene-specific

overlap in each tumour (Figure 3.14A). The number of overlapping genes in each tumour sample

varied from 9 genes in USTS13 to 225 genes in USTS12. In order to characterize the correlation

between gene-specific changes in the Venn analysis intersects of each tumour, the genes were

plotted based on copy number and gene expression status (Figure 3.14B). This analysis clearly

showed the strongest correlation between copy number gain and over-expression.

Figure 3.14. Tumour-specific integration of copy number and gene expression analyses. (A)

Integration of expression and copy number data in individual USTS tumours. Genes with significant

(P≤0.05) changes in copy number relative to the HapMap and gene expression relative to normal

human fibroblasts in individual tumours were analyzed using the Partek Genomics Suite Venn

Analysis tool. (B) Distribution of significant tumour-specific changes. The line graph represents the

percentage of genes (y-axis) identified with specific genomic and transcriptomic changes (x-axis).

Page 73: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

63

Cumulative Integration

Microarray and statistical analyses using the PGS software also allowed for the

identification of genomic and transcriptional alterations across all tumours. This analysis allowed

us to identify the most significant recurrent changes in USTSs as a group. Parameters for this

analysis were as follows: (1) Copy number changes present in at least 3 samples with P ≤ 0.05; and

(2) Transcriptional changes at a magnitude of at least 2-fold over- or under-expression with P ≤

0.05 cumulatively across all tumour samples relative to normal fibroblasts. Based on these

parameters, 438 genes were identified in the cumulative copy number gene analysis, and 2296

genes were identified in the gene expression analysis. Integration of these analyses was performed

using the PGS-Venn analysis tool, and resulted in the identification of 51 genes with overlapping

copy number and expression level changes (Figure 3.15A). These genes were plotted based on

copy number and gene expression status to characterize the correlation between gene-specific

changes in the Venn analysis intersect. This analysis displayed a strong correlation between copy

number gain and over-expression (66.7%), and copy number loss and under-expression (23.8%)

(Figure 3.15B).

Figure 3.15. Cumulative analysis of gene-specific changes in copy number and gene expression in

undifferentiated sarcoma. (A) Integration of cumulative copy number and expression analyses in USTS

tumours. Genes with significant (P≤0.05) changes in copy number relative to the HapMap, and gene expression

relative to normal human fibroblasts across all tumours were analyzed using the PGS Venn analysis tool. (B)

Distribution of significant overall genomic changes in undifferentiated sarcoma. The pie chart represents the

percentage of genes identified with specific genomic and transcriptomic changes.

Page 74: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

64

3.4.4 Gene Network Analysis

In order to identify USTS-related gene networks, Ingenuity Pathway Analysis (IPA) of the

genes identified in the cumulative copy number and cumulative expression analyses was performed.

This analysis allowed us to identify significant biological pathways that have been disrupted at the

genomic and transcriptomic levels, and to identify genes of interest within those pathways. We

compared the most significantly affected biological functions from copy number analysis to the

most significantly affected biological functions from gene expression analysis (Figure 3.16). All

tumours showed disruption in copy number and gene expression in similar biological functions.

Overall the biological functions that were most significantly affected by the changes in copy

number and gene expression were cell death, cell movement, cellular growth and proliferation, cell

development, cell cycle, and cellular assembly and organization.

Figure 3.16. Biological functions significantly affected by changes in copy

number and gene expression. The top six most significantly affected biological

functions in individual USTS tumours detected using the Ingenuity Pathway

Analysis (IPA) Comparative Analysis tool. IPA analysis of copy number (dark

blue bars) and gene expression (light blue bars) . Yellow line is indicative of P-

value threshold of 0.05.

Page 75: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

65

3.4.5 Identification of Candidate Genes

By combining the results from the PGS-based integrative analysis with the IPA network

analysis, a list of candidate genes were selected. This selection process focused on genes which

were either gained and over-expressed or deleted and under-expressed, and are directly involved in

the biological pathways identified in the network analysis. The candidate genes identified are

presented in Table 3.3.

Gene Genomic

Region

Copy

Number

Cumulative

Expression

Brief Description

EPHA3 3p11.2 Gain 12.11

· Transmembrane protein belonging to the Eph

receptor tyrosine kinase family.

· Eph receptors and ephrin ligands known to play

an important role in many biological functions

including axon guidance, cell migration,

angiogenesis and cytoskeletal regulation.

SNX10 7p15.2 Gain 2.3

· Member of sorting nexin family which play a role

in cellular functions including intracellular protein

trafficking, endocytosis and cell-to-cell signaling.

· May control endosome homeostasis.

ADAM9 8p11.22 Deletion -6.83

· Member of the ADAM (a disintegrin and

metalloprotease domain) family which have a role

in ectodomain shedding of membrane-bound

molecules.

· Known to cleave and release many molecules

associated with tumourigenesis and angiogenesis.

CDC73 1q25 Deletion -2.08

· Tumour suppressor involved in transcriptional

control pathways.

· Part of the PAF1 transcriptional regulatory

complex.

· Loss of function results in increased cell

proliferation and over-expression of proto-

oncogenes.

Table 3.3. Candidate genes with USTS-specific changes identified by integrative copy number

array, expression array, and gene network analysis.

Page 76: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

66

3.4.6 Gene Expression Validation of Candidate Genes

Integrative microarray analysis identified significant genomic and transcriptomic changes in

the following genes: (1) a gain in chromosome 3p11.2 and over-expression of EPHA3, (2) a gain in

chromosome 7p15.2 and over-expression of SNX10, (3) a loss in chromosome 8p11.22 and under-

expression of ADAM9, and (4) a loss in chromosome 1q25 and under-expression of CDC73. To

validate the results of the expression analysis (Figure 3.17A), quantitative PCR was performed in

all 5 tumours (Figure 3.17B). Over-expression of the EPHA3 gene was found in 4/5 tumours, with

an average of 11.3-fold over-expression relative to fibroblasts. Cumulative microarray expression

analysis found a 12.1-fold over-expression of the EPHA3 gene relative to fibroblasts. Over-

expression of the SNX10 gene was found in 3/5 tumours, with an average of 3.7-fold over-

expression relative to fibroblasts. Cumulative microarray analysis found 2.3-fold over-expression

of the SNX10 gene relative to fibroblasts. Under-expression of the ADAM9 gene was found in 5/5

tumours, with an average of 27.7-fold under-expression relative to fibroblasts. Cumulative

microarray analysis found 6.8-fold under-expression of the ADAM9 gene relative to fibroblasts.

Under-expression of the CDC73 gene was found in 5/5 tumours, with an average of 2.1-fold under-

expression relative to fibroblasts. Cumulative microarray analysis found 2.1-fold under-expression

of the CDC73 gene relative to fibroblasts.

Page 77: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

67

Figure 3.17. Expression analysis of candidate genes. (A) PGS dot plot visualization of

candidate genes from expression microarray analysis. Significant over-expression of EPHA3 and

SNX10 (top), and under-expression of ADAM9 and CDC73 (bottom) were seen cumulatively

across the four samples tested. The y-axis represents the raw lo2 signal intensity for each probe

set. (B) Quantitative RT-PCR validation of candidate gene expression. The y-axis represents fold

enrichment values generated using the comparative CT method. TATA-box binding protein

(TBP) was used as the endogenous housekeeping reference gene and human adult fibroblast

samples were used as the control samples. Each qPCR reaction was performed in triplicate and

average values were used for enrichment calculations.

Page 78: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

68

3.4.7 Copy Number Validation of EPHA3

Quantitative PCR confirmed over-expression of the EPHA3 gene in 4/5 USTS cases.

Ephrins and ephrin receptors have been found to play a role in the development of numerous types

of cancer [110-112]. Due to the high correlation between copy-number gain (3 copies; Figure

3.18A) and over-expression, subsequent qPCR validation, and the prominent role that EPHA3 has

been found to play in cellular development, we decided to further validate this potential biomarker

in our samples. We performed validation of the copy number status of EPHA3 by performing dual-

colour FISH on all five tumour samples using a commercial Spectrum-Orange labeled centromere 3

probe as a control. In order to confirm locus specificity and optimize the EPHA3-specific probe,

FISH was first performed on normal metaphase spreads and on FFPE normal tonsil tissue (Figure

3.18B and C). The expected low-level gain of EPHA3 (3 copies) was not identified in any of the 5

tumours samples. Instead, 2 copies of both the control probe (orange probe) and test probe (green

probe) were seen in the majority of the tumour cells (Figure 3.18D and E).

3.4.8 Protein Validation of EPHA3

Immunohistochemistry was performed on FFPE sections from all 5 tumours in order to

determine the protein expression patterns of the EPHA3 gene in these tumours. The EPHA3

antibody (Abnova) was applied to USTS12-15 and USTS19 using methodology described

previously. USTS12 and USTS13 showed weak, consistent cytoplasmic staining in <10% of cells;

the remaining 3 tumours showed no staining (Figure 3.19). All 5 tumours were thus scored as

negative.

Page 79: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

69

Fig

ure

3.1

8.

FIS

H v

ali

da

tion

of

cop

y n

um

ber

ga

in o

f E

PH

A3

. (

A)

PG

S v

isu

aliz

atio

n o

f th

e h

eat

map

an

d p

rofi

le v

iew

of

5 t

um

ou

r

sam

ple

s sh

ow

ing

a s

ign

ific

ant

gai

n o

f a

29

-kb

reg

ion

in

all

5 t

um

ou

r sa

mp

les.

T

his

reg

ion i

s h

igh

lig

hte

d i

nsi

de

the

red

box

. (

B-D

)

Val

idat

ion

of

the

cop

y n

um

ber

sta

tus

of

EP

HA

3 w

as p

erfo

rmed

usi

ng

du

al-c

olo

ur

inte

rph

ase

FIS

H.

(B

) L

ocu

s sp

ecif

icit

y o

f th

e E

PH

A3

-

spec

ific

BA

C p

rob

e (R

P1

1-2

3D

18

) w

as p

erfo

rmed

on

no

rmal

met

aph

ase

spre

ads.

(C

) O

pti

miz

atio

n o

f th

e E

PH

A3

-sp

ecif

ic p

rob

e w

as

per

form

ed o

n F

FP

E n

orm

al t

on

sil

tiss

ue.

(D

) R

epre

sen

tati

ve

imag

e o

f d

ual

-co

lou

r in

terp

has

e F

ISH

an

aly

sis

of

EP

HA

3 i

n U

ST

S14

. F

ISH

was

per

form

ed u

sin

g t

he

Sp

ectr

um

-Gre

en l

abel

ed E

PH

A3

-sp

ecif

ic p

rob

e an

d a

co

mm

erci

al S

pec

tru

m-O

ran

ge

lab

eled

cen

tro

mer

e 3

pro

be

as

a co

ntr

ol.

L

ow

-lev

el g

ain

of

EP

HA

3 w

as n

ot

con

firm

ed i

n a

ny

of

the

tum

ou

r sa

mp

les,

as

2 c

op

ies

of

bo

th t

he

con

tro

l p

rob

e an

d t

he

test

pro

be

wer

e se

en i

n t

he

maj

ori

ty o

f th

e tu

mo

ur

cell

s.

Page 80: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

70

Figure 3.19. Immunohistochemical staining for EPHA3. Positive control tissue

included breast, both normal (A) and cancer (B). The photomicrographs show strong

cytoplasmic positivity for EPHA3 in the normal glandular epithelium (A), as well as

moderate to weak cytoplasmic positivity in ductal carcinoma and infiltrating histiocytes

(B). The negative controls show negativity in the same tissues, as well as focally positive

inflammatory cells that stain for endogenous peroxidase (C, D). Representative sections

of two tumours (E, F; USTS12 & USTS13) show very focal, weak and inconsistent

cytoplasmic staining, scored as negative. Scale bar = 100 µm.

Page 81: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

71

3.4.9 Discussion of Discovery of Novel Markers for CIC-DUX4-Negative Tumours

As previously described, paediatric USTS remains a diagnosis of exclusion, despite the

advancement of highly specific and sensitive diagnostic applications. The similarities that these

tumours share with other sarcoma subtypes (e.g. Ewing Sarcoma) has evoked much debate as to

whether this group comprises tumours with common histological and biological features

representing a distinct subgroup of sarcomas or if these tumours have simply not been accurately

sub-classified into known sarcoma subgroups [9]. Recent studies on the CIC-DUX4 fusion

transcript have suggested the former – that there are distinct morphological and molecular

subgroups which can be identified from within the USTS family [31, 60]. However, identification

of the subgroup of USTSs harbouring the CIC-DUX4 fusion transcript left a group of USTSs

devoid of any known molecular identifiers. As such, the final studies in this thesis project sought to

identify novel genomic markers in a series of CIC-DUX4-negative USTS tumours using an

integrative genomic and transcriptomic analysis. Using bioinformatic whole-genome approaches,

the genomic and transcriptomic profiles of 5 USTS tumours were analyzed, and a series of

biological networks were identified that may play a role in sarcomagenesis. While we did find that

there was gross heterogeneity between the tumours at both the DNA and RNA levels, significant

changes were identified that were present in the majority of the tumours studied. Cumulative

analysis of the tumours found that there was a correlation between copy number gain and

overexpression, as well as copy number loss and underexpression, with more than 90% of the genes

identified following these patterns. However, these molecular correlations are present in a

relatively small proportion of the genes identified in any given tumour sample. Many of the genes

found to have changes at the copy number level, did not exhibit changes at the expression level and

vice-versa. This can be partially explained as a result of other biological phenomena present in the

cellular environment such as the actions of regulatory elements and epigenetic mechanisms such as

Page 82: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

72

DNA methylation [113, 114]. However, by identifying genes which do in fact show a

genomic/transcriptomic correlation, and investigating the biological pathways in which these genes

are involved, it may be possible to identify specific genes that are responsible for driving USTS

tumourigenesis.

Integrative analysis of changes in copy-number, gene expression, and USTS-related gene

networks, identified a set of candidate genes which may play a role in the early event involved in

USTS development. The genes identified from this analysis were EPHA3, SNX10, ADAM9 and

CDC73. EPHA3 is a transmembrane protein that belongs to the Eph receptor tyrosine kinase

(RTK) family, which can be subdivided into 2 groups (EphA and EphB receptors) based on

sequence homology of the extracellular domain (ECD) of the receptor [115]. The Eph-receptor

ECD is composed of a ligand-binding domain, a cysteine-rich region, and two fibronectin type III

repeats. The cytoplasmic domain is composed of a juxtamembrane domain, a classical tyrosine

kinase domain, a sterile α-motif and a PDZ-domain. The Eph-ephrin receptor-ligand interaction

serves as a guidance system which regulates cell positioning and the modulation of cell morphology

[116]. Eph receptors and ephrin ligands are known to play an important role in many biological

functions including axon guidance, cell migration, angiogenesis and cytoskeletal regulation [117-

119]. Over-expression of the Eph receptors may promote cellular growth by increasing

vascularization, disrupting cell-cell adhesion, and providing potential adherence in a new tumour

microenvironment [120]. As such, it is not surprising that over-expression of EPHA3 has been

implicated in the pathogenesis of numerous cancers including lung cancer, prostate cancer,

melanoma, rhabdomyosarcoma and lymphocytic leukemia [110, 112, 120, 121].

Page 83: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

73

SNX10 is a member of the sorting nexin family which are characterized by the presence of

an extracellular PX domain [122]. There are 29 known or predicted members of the sorting nexin

family, and the function of the majority of these proteins remains unknown [123]. Sorting nexins

have been shown to play a role in numerous cellular functions including intracellular protein

trafficking, endocytosis and cell-to-cell signaling [124, 125]. SNX10 specifically has been shown

to be capable of generating giant vacuoles in mammalian cells, suggesting that this protein may

serve to control endosome homeostasis [126]. EPHA3 and SNX10 both showed significant copy

number gains in 4/5 USTS samples. Interestingly, preliminary analysis also found EPHA3 to be

gained in the fifth USTS sample, however this gain was not found to be statistically significant.

These genes were also found to be significantly over-expressed in the cumulative expression

analysis.

ADAM9 is a member of the ADAM (a disintegrin and metalloprotease domain) family,

which have a predominant role in ectodomain shedding of membrane-bound molecules [127].

ADAMs are critical regulators of cell-cell signaling during development, and have been shown to

specifically play a role in muscle development, neurogenesis, cell adhesion and cell migration

[128]. ADAM9 specifically is known to cleave and release many molecules associated with

tumourigenesis and angiogenesis [129], and as such dysregulation of this protein has been

implicated in the pathogenesis of breast, prostate and lung cancer, melanoma, adenocarcinoma, and

metastasis to the brain [127, 130-133].

CDC73, or parafibromin, is a tumour suppressor that has been found to be involved in

transcriptional and post-transcriptional control pathways. Human parafibromin binds to RNA

polymerase II as part of the PAF1 transcriptional regulatory complex, and facilitates the association

Page 84: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

74

of 3’ mRNA processing factors with chromatin that is being actively transcribed [134]. It has been

documented that loss of function of CDC73 can lead to oncogenesis, though the mechanisms

underlying this phenomenon are poorly understood. Furthermore, it has been shown that down-

regulation of CDC73 expression leads to increased cell proliferation and increased expression of

known proto-oncogenes [135]. Not surprisingly, deletions and loss-of-function mutations in the

CDC73 gene have been associated with various cancers including pancreatic cancer and

parathyroid carcinoma [136, 137]. ADAM9 and CDC73 both showed copy number loss in 3/5

USTS samples, and significant under-expression in the cumulative expression analysis.

Quantitative PCR analysis confirmed that USTS12, USTS13, USTS15 and USTS18 showed over-

expression of the EPHA3 gene, USTS12, USTS14, USTS15 and USTS18 showed over-expression

of SNX10, and that all 5 tumours showed under-expression of ADAM9 and CDC73. Each of these

genes plays an important role in the USTS-related biological pathways identified by gene network

analysis.

However, EPHA3 was identified as the most biologically relevant marker in this study as

this gene showed the most significant correlation between genomic gain and over-expression. As

such, we sought to validate the copy-number gain identified in EPHA3 by dual-colour FISH.

Microarray analysis identified a low-level gain in a 29-kb region in EPHA3 in all samples, but this

gain was not found by FISH in any of the tumour samples. This does not however, negate the

results of the microarray analysis. This 29-kb region of EPHA3 encompasses the entire exon III

coding region, as well as portions of introns II and III. It is possible that this copy number gain is a

result of a segmental duplication of exon III of the EPHA3 gene. If this were the case, it is possible

that separate EPHA3 probe signals could not be seen due to the size of the duplication, and the

proximity of the duplicate signals to one another. It would have been possible to visualize this

Page 85: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

75

signal if the low-level copy-number gain were a result of rearrangement of this region, though this

does not seem to be the case. Interestingly, the ephrin ligand-binding domain has been found to be

specifically restricted to the N-terminal extracellular region of EPHA3 encoded by exon III.

Duplication of exons coding for the ligand-binding domain have been similarly documented in

another RTK, epidermal growth factor receptor (EGFR), in human gliomas [138-140]. Duplication

of the ligand-binding domain of EGFR has been shown to induce constitutive receptor

phosphorylation, while retaining the ability to bind the EGF ligand. Furthermore, it has been

shown that this segmental duplication may prevent efficient ligand-mediated receptor

internalization, resulting in prolonged RTK signaling [138-140]. Hypothetically, the same may be

true for the EPHA3 receptor. Duplication of the EPHA3 ligand-binding domain may promote

oncogenesis by increasing receptor activation, and impairing receptor attenuation. In order to

elucidate the role of this duplication in EPHA3, detailed functional studies are essential.

Furthermore, the IHC for EPHA3 was negative in all 5 samples tested, suggesting that

EPHA3 is not overexpressed at the protein level in these samples. However, negative staining for

EPHA3 may be a result of a poor quality antibody, as optimization on the positive control tissues

proved challenging. These results may also be a result of poor quality tissue received from the

Cooperative Human Tissue Network, as both FISH and IHC were difficult to perform on these

FFPE slides. Alternatively, it is possible that the EPHA3 protein is simply not expressed, or is

expressed at very low levels in these tissues. More studies using a larger cohort of samples, as well

as a different EPHA3-specific antibody are necessary to the EPHA3 expression status in USTSs.

This microarray study was limited in terms of the number of samples that were studied, as

well as the number of genes that were validated. Further studies which include more samples and

more comprehensive validation will be imperative to determine the role of these genes in

Page 86: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

76

sarcomagenesis. Importantly, the general pathways identified in this study may provide novel

insights into the critical biological events responsible for the development of USTS as well as

sarcomas in general.

Page 87: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

77

CHAPTER FOUR

SUMMARY, GENERAL CONCLUSIONS

&

FUTURE DIRECTIONS

Page 88: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

78

CHAPTER 4. SUMMARY, GENERAL CONCLUSIONS & FUTURE DIRECTIONS

4.1 Summary and General Conclusions

Paediatric sarcomas have long been a challenging group of tumours to accurately diagnose

due to the similarity in histology and clinical presentation between different subgroups [141].

Though different sarcoma subtypes may appear very similar at the histological level, the prognosis

and treatment of these subtypes varies immensely, making correct sub-categorization paramount for

optimal therapy. The development of ancillary techniques such as immunohistochemistry,

cytogenetics and RT-PCR has significantly enhanced the pathologists’ ability to make an accurate

diagnosis [142]. Until very recently no markers had been identified in USTS, and consequently

undifferentiated sarcoma has remained a diagnosis of exclusion.

In this study, extensive and detailed genomic analysis of paediatric undifferentiated soft

tissue sarcomas has identified a specific variant of USTS with PRC morphology harbouring the

CIC-DUX4 rearrangement. Using RT-PCR and FISH, we have established a reliable and specific

screening platform with which any tumour can be screened for this rearrangement. In our cohort of

22 USTS samples, we identified 18 tumours with PRC morphology, 5 of which harbour the CIC-

DUX4 rearrangement (Objective 1).

Following this screening we were left with 17 tumours in which no specific molecular

markers had been identified. In order to identify potentially novel molecular markers for these

tumours, we performed an integrative copy-number and expression microarray analysis to identify

genes with changes at both the DNA and RNA levels. This study identified specific genes

(EPHA3, SNX10, ADAM9 and CDC73) which may play an important role in sarcomagenesis.

More importantly this study identified key USTS-related genomic pathways that may play a pivotal

Page 89: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

79

role in the acquisition of the USTS phenotype (Objectives 2 and 3). While it remains unclear if it

will be possible to sub-classify the 17 tumours without the CIC-DUX4 translocation, we have

certainly made progress into identifying pathways responsible for the development of USTS.

Although USTS currently remains a diagnosis of exclusion, the cumulative data surrounding

the CIC-DUX4 translocation suggests that a portion of these tumours may in fact be identifiable by

a distinct chromosomal rearrangement. In CIC-DUX4-negative tumours, we have made progress in

identifying specific genes and biological pathways that may play a role in the development and

progression of these tumours. These findings help to further define this novel genetic subset of

paediatric sarcomas and provide an additional diagnostic tool for their classification and diagnosis.

4.2 Future Directions

The results of this project have given rise to several issues that are of utmost importance to

address in future studies. This study identified a subset of USTSs with PRC morphology that

harbour the CIC-DUX4 fusion gene. These tumours show minimal evidence of differentiation,

suggesting that they are able to maintain a stem cell-like phenotype. Thus, it is believed that the

presence of the CIC-DUX4 fusion transcript is associated with transforming properties [60], as well

as the maintenance of the undifferentiated state. However, significantly more evidence is needed in

order to prove both of these concepts. Future work in our laboratory will seek to determine the

specific biological and pathological consequences of the CIC-DUX4 fusion transcript. To do so, it

will be necessary to identify specific target molecules of the CIC-DUX4 fusion protein, as well as

to identify overall biological pathways that affected by this fusion. Furthermore, the diagnosis of

CIC-DUX4-positive tumours would be greatly aided by the identification of novel diagnostic

markers for these tumours. For example, the development of an immunohistochemcial-based

Page 90: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

80

diagnostic assay for these tumours would allow for a timely and cost-efficient diagnosis to be made

where RT-PCR is unavailable. Additionally, the testing of other sarcoma subtypes for the presence

of this fusion transcript with help further elucidate the diagnostic utility of the CIC-DUX4 fusion

transcript. Future studies on CIC-DUX4-positive tumours will help to further elucidate the

biological features of these tumours, the prognostic implications of carrying this translocation, and

potential therapeutic options for this challenging subgroup of sarcomas.

A more in-depth study of the CIC-DUX4-negative tumours will provide a greater

understanding of the specific mechanisms of sarcomageneis in these tumours. Analysis of a greater

number of CIC-DUX4- negative tumours will allow for the accurate determination of both genomic

and transcriptomic features of these tumours, and will certainly enhance our understanding of

paediatric primitive sarcomas. Furthermore, it will be necessary to perform functional studies on

the candidate genes in order to gain a better understanding of the roles that these genes play in

sarcomagenesis. Additional work will hopefully give rise to diagnostic markers that can be used to

recognize this challenging group of tumours.

Future work in this field will provide a greater understanding of the early events involved in

the acquisition of the sarcoma phenotype. Furthermore, the analysis of both the CIC-DUX4 fusion

transcript and dysregulated pathways identified in CIC-DUX4-negative tumours will help to

improve diagnosis and treatment of such aggressive and poorly-understood sarcomas. These

findings will have relevance not only for paediatric sarcomas, but for sarcomas in general.

Page 91: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

81

REFERENCES

Page 92: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

82

1. Rosenthal, T.C. and W. Kraybill, Soft tissue sarcomas: integrating primary care recognition

with tertiary care center treatment. Am Fam Physician, 1999. 60(2): p. 567-72.

2. Davicioni, E., D.H. Wai, and M.J. Anderson, Diagnostic and prognostic sarcoma

signatures. Mol Diagn Ther, 2008. 12(6): p. 359-74.

3. van de Rijn, M. and J.A. Fletcher, Genetics of soft tissue tumors. Annu Rev Pathol, 2006. 1:

p. 435-66.

4. Chibon, F., et al., Validated prediction of clinical outcome in sarcomas and multiple types

of cancer on the basis of a gene expression signature related to genome complexity. Nat

Med. 16(7): p. 781-7.

5. Mackall, C.L., P.S. Meltzer, and L.J. Helman, Focus on sarcomas. Cancer Cell, 2002. 2(3):

p. 175-8.

6. Riggi, N., M.L. Suva, and I. Stamenkovic, Ewing's sarcoma origin: from duel to duality.

Expert Rev Anticancer Ther, 2009. 9(8): p. 1025-30.

7. Wolden, S.L. and K.M. Alektiar, Sarcomas across the age spectrum. Semin Radiat Oncol.

20(1): p. 45-51.

8. Pawel, B.R., et al., Undifferentiated sarcomas of children: pathology and clinical behavior-

-an Intergroup Rhabdomyosarcoma study. Med Pediatr Oncol, 1997. 29(3): p. 170-80.

9. Alaggio, R., et al., Undifferentiated sarcoma: does it exist? A clinicopathologic study of 7

pediatric cases and review of literature. Hum Pathol, 2009. 40(11): p. 1600-10.

10. Ries LAG, S.M., Gurney JG, Linet M, Tamra T, Young JL, Bunin GR (eds). , Cancer

Incidence and Survival among Children and Adolescents: United States SEER Program

1975-1995. National Cancer Institute, SEER Program. NIH Pub. No. 99-4649. Bethesda,

MD, 1999.

11. Meyer, W.H. and S.L. Spunt, Soft tissue sarcomas of childhood. Cancer Treat Rev, 2004.

30(3): p. 269-80.

12. Helman, L.J. and P. Meltzer, Mechanisms of sarcoma development. Nat Rev Cancer, 2003.

3(9): p. 685-94.

13. Eilber, F.C. and S.M. Dry, Diagnosis and management of synovial sarcoma. J Surg Oncol,

2008. 97(4): p. 314-20.

14. Burchill, S.A., Molecular abnormalities in Ewing's sarcoma. Expert Rev Anticancer Ther,

2008. 8(10): p. 1675-87.

15. Barr, F.G., Gene fusions involving PAX and FOX family members in alveolar

rhabdomyosarcoma. Oncogene, 2001. 20(40): p. 5736-46.

Page 93: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

83

16. Mitelman, F., B. Johansson, and F. Mertens, The impact of translocations and gene fusions

on cancer causation. Nat Rev Cancer, 2007. 7(4): p. 233-45.

17. Coffin, C.M., L.P. Dehner, and P.A. O'Shea, Pediatric Soft Tissue Sarcomas: A Clinical,

Pathological and Therapeutic Approach. 1997, Baltimore: Williams and Wilkins.

18. Rubin, B.P., Recent progress in the classification of soft tissue tumors: role of genetics and

clinical implications. Curr Opin Oncol, 2001. 13(4): p. 256-60.

19. Coindre, J.M., Immunohistochemistry in the diagnosis of soft tissue tumours.

Histopathology, 2003. 43(1): p. 1-16.

20. Fletcher, C.D.M., K.K. Unni, and F. Mertens, World Health Organisation Classification of

Tumors: Tumors of Soft Tissue and Bone. 2002, Lyon: IARC Press.

21. Sorensen, P.H., et al., A second Ewing's sarcoma translocation, t(21;22), fuses the EWS

gene to another ETS-family transcription factor, ERG. Nat Genet, 1994. 6(2): p. 146-51.

22. Cessna, M.H., et al., Are myogenin and myoD1 expression specific for rhabdomyosarcoma?

A study of 150 cases, with emphasis on spindle cell mimics. Am J Surg Pathol, 2001. 25(9):

p. 1150-7.

23. Kast, R., et al., Differentiation of small round blue cell tumors using Raman spectroscopy. J

Pediatr Surg. 45(6): p. 1110-4.

24. Tanas, M.R. and J.R. Goldblum, Fluorescence in situ hybridization in the diagnosis of soft

tissue neoplasms: a review. Adv Anat Pathol, 2009. 16(6): p. 383-91.

25. Ladanyi, M., C.R. Antonescu, and P. Dal Cin, Cytogenetic and molecular genetic pathology

of soft tissue tumors in Enzinger and Weiss's Soft Tissue Tumors, S.W. Weiss and J.R.

Goldblum, Editors. 2008, Mosby Elsevier: Philadelphia. p. 73-102.

26. Knuutila, S., Cytogenetics and molecular pathology in cancer diagnostics. Ann Med, 2004.

36(3): p. 162-71.

27. Ordonez, J.L., et al., The clinical relevance of molecular genetics in soft tissue sarcomas.

Adv Anat Pathol. 17(3): p. 162-81.

28. Skapek, S.X. and C.H. Chui, Cytogenetics and the biologic basis of sarcomas. Curr Opin

Oncol, 2000. 12(4): p. 315-22.

29. Sebire, N.J., et al., Aberrant immunohistochemical expression in nonrhabdomyosarcoma

soft tissue sarcomas of infancy: retrospective review of clinical material. Pediatr Dev

Pathol, 2002. 5(6): p. 579-86.

Page 94: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

84

30. Qualman, S.J., et al., Intergroup Rhabdomyosarcoma Study: update for pathologists. Pediatr

Dev Pathol, 1998. 1(6): p. 550-61.

31. Yoshimoto, M., et al., Detailed cytogenetic and array analysis of pediatric primitive

sarcomas reveals a recurrent CIC-DUX4 fusion gene event. Cancer Genet Cytogenet, 2009.

195(1): p. 1-11.

32. Somers, G.R., et al., Pediatric undifferentiated sarcoma of the soft tissues: a

clinicopathologic study. Pediatr Dev Pathol, 2006. 9(2): p. 132-42.

33. Kreiger, P.A., et al., Loss of INI1 expression defines a unique subset of pediatric

undifferentiated soft tissue sarcomas. Mod Pathol, 2009. 22(1): p. 142-50.

34. Selvarajah, S., et al., Characterization of trisomy 8 in pediatric undifferentiated sarcomas

using advanced molecular cytogenetic techniques. Cancer Genet Cytogenet, 2007. 174(1):

p. 35-41.

35. Pappo, A.S., et al., Survival after relapse in children and adolescents with

rhabdomyosarcoma: A report from the Intergroup Rhabdomyosarcoma Study Group. J Clin

Oncol, 1999. 17(11): p. 3487-93.

36. Raney, R.B., et al., Rhabdomyosarcoma and undifferentiated sarcoma in the first two

decades of life: a selective review of intergroup rhabdomyosarcoma study group experience

and rationale for Intergroup Rhabdomyosarcoma Study V. J Pediatr Hematol Oncol, 2001.

23(4): p. 215-20.

37. Charytonowicz, E., et al., Alveolar rhabdomyosarcoma: is the cell of origin a mesenchymal

stem cell? Cancer Lett, 2009. 279(2): p. 126-36.

38. Mani, S.A., et al., The epithelial-mesenchymal transition generates cells with properties of

stem cells. Cell, 2008. 133(4): p. 704-15.

39. Subramaniam, D., et al., Cancer stem cells: a novel paradigm for cancer prevention and

treatment. Mini Rev Med Chem. 10(5): p. 359-71.

40. Tan, B.T., et al., The cancer stem cell hypothesis: a work in progress. Lab Invest, 2006.

86(12): p. 1203-7.

41. Suva, M.L., et al., Identification of cancer stem cells in Ewing's sarcoma. Cancer Res, 2009.

69(5): p. 1776-81.

42. Bonnet, D. and J.E. Dick, Human acute myeloid leukemia is organized as a hierarchy that

originates from a primitive hematopoietic cell. Nat Med, 1997. 3(7): p. 730-7.

43. Passegue, E., et al., Normal and leukemic hematopoiesis: are leukemias a stem cell disorder

or a reacquisition of stem cell characteristics? Proc Natl Acad Sci U S A, 2003. 100 Suppl

1: p. 11842-9.

Page 95: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

85

44. Li, C., et al., Identification of pancreatic cancer stem cells. Cancer Res, 2007. 67(3): p.

1030-7.

45. Singh, S.K., et al., Identification of human brain tumour initiating cells. Nature, 2004.

432(7015): p. 396-401.

46. Quintana, E., et al., Efficient tumour formation by single human melanoma cells. Nature,

2008. 456(7222): p. 593-8.

47. Levings, P.P., et al., Expression of an exogenous human Oct-4 promoter identifies tumor-

initiating cells in osteosarcoma. Cancer Res, 2009. 69(14): p. 5648-55.

48. Naka, N., et al., Synovial sarcoma is a stem cell malignancy. Stem Cells. 28(7): p. 1119-31.

49. Murase, M., et al., Side population cells have the characteristics of cancer stem-like

cells/cancer-initiating cells in bone sarcomas. Br J Cancer, 2009. 101(8): p. 1425-32.

50. Jiang, X., et al., CD133 expression in chemo-resistant Ewing sarcoma cells. BMC Cancer.

10: p. 116.

51. Riggi, N., et al., Expression of the FUS-CHOP fusion protein in primary mesenchymal

progenitor cells gives rise to a model of myxoid liposarcoma. Cancer Res, 2006. 66(14): p.

7016-23.

52. Ren, Y.X., et al., Mouse mesenchymal stem cells expressing PAX-FKHR form alveolar

rhabdomyosarcomas by cooperating with secondary mutations. Cancer Res, 2008. 68(16):

p. 6587-97.

53. Miyagawa, Y., et al., Inducible expression of chimeric EWS/ETS proteins confers Ewing's

family tumor-like phenotypes to human mesenchymal progenitor cells. Mol Cell Biol, 2008.

28(7): p. 2125-37.

54. Riggi, N., et al., EWS-FLI-1 expression triggers a Ewing's sarcoma initiation program in

primary human mesenchymal stem cells. Cancer Res, 2008. 68(7): p. 2176-85.

55. Riggi, N., et al., Development of Ewing's sarcoma from primary bone marrow-derived

mesenchymal progenitor cells. Cancer Res, 2005. 65(24): p. 11459-68.

56. Tirode, F., et al., Mesenchymal stem cell features of Ewing tumors. Cancer Cell, 2007.

11(5): p. 421-9.

57. Hu-Lieskovan, S., et al., EWS-FLI1 fusion protein up-regulates critical genes in neural

crest development and is responsible for the observed phenotype of Ewing's family of

tumors. Cancer Res, 2005. 65(11): p. 4633-44.

Page 96: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

86

58. Newton, W.A., Jr., et al., Classification of rhabdomyosarcomas and related sarcomas.

Pathologic aspects and proposal for a new classification--an Intergroup

Rhabdomyosarcoma Study. Cancer, 1995. 76(6): p. 1073-85.

59. Davis IJ, P.-A.A., Fisher DE. , Nonrhabdomyosarcomas and other soft tissue tumors.

Oncology of Infancy and Childhood. , ed. F.D. Orkin SH, Look AT, et al., editors. 2009:

Philadelphia: Saunders Elsevier.

60. Kawamura-Saito, M., et al., Fusion between CIC and DUX4 up-regulates PEA3 family

genes in Ewing-like sarcomas with t(4;19)(q35;q13) translocation. Hum Mol Genet, 2006.

15(13): p. 2125-37.

61. Sadikovic, B., Graham, C., Ho, M., Zielenska, M., Somers, G., Immunohistochemical

expression and cluster analysis of mesenchymal and neural stem cell-associated proteins in

pediatric soft tissue sarcomas. submitted to Pediatr Dev Pathol, 2010.

62. Keller, C., et al., Pax3:Fkhr interferes with embryonic Pax3 and Pax7 function:

implications for alveolar rhabdomyosarcoma cell of origin. Genes Dev, 2004. 18(21): p.

2608-13.

63. Tajbakhsh, S., et al., Redefining the genetic hierarchies controlling skeletal myogenesis:

Pax-3 and Myf-5 act upstream of MyoD. Cell, 1997. 89(1): p. 127-38.

64. Maroto, M., et al., Ectopic Pax-3 activates MyoD and Myf-5 expression in embryonic

mesoderm and neural tissue. Cell, 1997. 89(1): p. 139-48.

65. Epstein, J.A., et al., Pax3 inhibits myogenic differentiation of cultured myoblast cells. J Biol

Chem, 1995. 270(20): p. 11719-22.

66. Lang, D., et al., Pax3 functions at a nodal point in melanocyte stem cell differentiation.

Nature, 2005. 433(7028): p. 884-7.

67. Gang, E.J., et al., Pax3 activation promotes the differentiation of mesenchymal stem cells

toward the myogenic lineage. Exp Cell Res, 2008. 314(8): p. 1721-33.

68. Hoot, A.C., et al., Immunohistochemical analysis of hSNF5/INI1 distinguishes renal and

extra-renal malignant rhabdoid tumors from other pediatric soft tissue tumors. Am J Surg

Pathol, 2004. 28(11): p. 1485-91.

69. Judkins, A.R., et al., INI1 protein expression distinguishes atypical teratoid/rhabdoid tumor

from choroid plexus carcinoma. J Neuropathol Exp Neurol, 2005. 64(5): p. 391-7.

70. Richkind, K.E., S.G. Romansky, and J.Z. Finklestein, t(4;19)(q35;q13.1): a recurrent

change in primitive mesenchymal tumors? Cancer Genet Cytogenet, 1996. 87(1): p. 71-4.

Page 97: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

87

71. Rakheja, D., et al., Translocation (4;19)(q35;q13.1)-associated primitive round cell

sarcoma: report of a case and review of the literature. Pediatr Dev Pathol, 2008. 11(3): p.

239-44.

72. Somers, G.R., et al., IGF2 is highly expressed in pediatric undifferentiated sarcomas and

reveals two distinct cytoplasmic trafficking patterns. Pediatr Dev Pathol, 2010. 13(3): p.

169-77.

73. Ausubel FM, B.R., Kingston RE, Moore DD, Seidman JG, Smith JA, Sturhl K. , Short

protocols in molecular biology. . 5th ed, ed. N.Y.J. Wiley. 2002.

74. Bayani, J., et al., Spectral karyotyping identifies recurrent complex rearrangements of

chromosomes 8, 17, and 20 in osteosarcomas. Genes Chromosomes Cancer, 2003. 36(1): p.

7-16.

75. Bayani, J., et al., Molecular cytogenetic analysis of medulloblastomas and supratentorial

primitive neuroectodermal tumors by using conventional banding, comparative genomic

hybridization, and spectral karyotyping. J Neurosurg, 2000. 93(3): p. 437-48.

76. Chin, S.F., et al., A simple and reliable pretreatment protocol facilitates fluorescent in situ

hybridisation on tissue microarrays of paraffin wax embedded tumour samples. Mol Pathol,

2003. 56(5): p. 275-9.

77. Sabeti, P.C., et al., Genome-wide detection and characterization of positive selection in

human populations. Nature, 2007. 449(7164): p. 913-8.

78. Schroeder, A., et al., The RIN: an RNA integrity number for assigning integrity values to

RNA measurements. BMC Mol Biol, 2006. 7: p. 3.

79. The International HapMap Project. Nature, 2003. 426(6968): p. 789-96.

80. Sadikovic, B., et al., Genome-wide H3K9 histone acetylation profiles are altered in

benzopyrene-treated MCF7 breast cancer cells. J Biol Chem, 2008. 283(7): p. 4051-60.

81. Sadikovic, B., et al., Identification of interactive networks of gene expression associated

with osteosarcoma oncogenesis by integrated molecular profiling. Hum Mol Genet, 2009.

18(11): p. 1962-75.

82. Durbin, A.D., et al., JNK1 determines the oncogenic or tumor-suppressive activity of the

integrin-linked kinase in human rhabdomyosarcoma. J Clin Invest, 2009. 119(6): p. 1558-

70.

83. Schmittgen, T.D. and K.J. Livak, Analyzing real-time PCR data by the comparative C(T)

method. Nat Protoc, 2008. 3(6): p. 1101-8.

84. Berglund, L., et al., A genecentric Human Protein Atlas for expression profiles based on

antibodies. Mol Cell Proteomics, 2008. 7(10): p. 2019-27.

Page 98: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

88

85. Uhlen, M., et al., A human protein atlas for normal and cancer tissues based on antibody

proteomics. Mol Cell Proteomics, 2005. 4(12): p. 1920-32.

86. Nilsson, P., et al., Towards a human proteome atlas: high-throughput generation of mono-

specific antibodies for tissue profiling. Proteomics, 2005. 5(17): p. 4327-37.

87. Ponten, F., K. Jirstrom, and M. Uhlen, The Human Protein Atlas--a tool for pathology. J

Pathol, 2008. 216(4): p. 387-93.

88. Somers, G.R., et al., Primary subcutaneous primitive neuroectodermal tumor with

aggressive behavior and an unusual karyotype: case report. Pediatr Dev Pathol, 2004. 7(5):

p. 538-45.

89. Yoshimoto, M., et al., Absence of TMPRSS2:ERG fusions and PTEN losses in prostate

cancer is associated with a favorable outcome. Mod Pathol, 2008. 21(12): p. 1451-60.

90. Lee, C.J., et al., CIC, a member of a novel subfamily of the HMG-box superfamily, is

transiently expressed in developing granule neurons. Brain Res Mol Brain Res, 2002.

106(1-2): p. 151-6.

91. Gabriels, J., et al., Nucleotide sequence of the partially deleted D4Z4 locus in a patient with

FSHD identifies a putative gene within each 3.3 kb element. Gene, 1999. 236(1): p. 25-32.

92. Mathas, S., et al., Gene deregulation and spatial genome reorganization near breakpoints

prior to formation of translocations in anaplastic large cell lymphoma. Proc Natl Acad Sci

U S A, 2009. 106(14): p. 5831-6.

93. Graham, C., Chilton-MacNeill, S., Zielenska, M., Somers, G.R., The CIC-DUX4 fusion

transcript is present in a distinct subgroup of pediatric primitive round cell sarcomas.

Submitted to Pediatr Dev Pathol., 2010.

94. Beausoleil, S.A., et al., Large-scale characterization of HeLa cell nuclear phosphoproteins.

Proc Natl Acad Sci U S A, 2004. 101(33): p. 12130-5.

95. Olsen, J.V., et al., Global, in vivo, and site-specific phosphorylation dynamics in signaling

networks. Cell, 2006. 127(3): p. 635-48.

96. Cantin, G.T., et al., Combining protein-based IMAC, peptide-based IMAC, and MudPIT for

efficient phosphoproteomic analysis. J Proteome Res, 2008. 7(3): p. 1346-51.

97. Dephoure, N., et al., A quantitative atlas of mitotic phosphorylation. Proc Natl Acad Sci U S

A, 2008. 105(31): p. 10762-7.

98. Mayya, V., et al., Quantitative phosphoproteomic analysis of T cell receptor signaling

reveals system-wide modulation of protein-protein interactions. Sci Signal, 2009. 2(84): p.

ra46.

Page 99: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

89

99. Hernandez, L., et al., Diversity of genomic breakpoints in TFG-ALK translocations in

anaplastic large cell lymphomas: identification of a new TFG-ALK(XL) chimeric gene with

transforming activity. Am J Pathol, 2002. 160(4): p. 1487-94.

100. Grimwood, J., et al., The DNA sequence and biology of human chromosome 19. Nature,

2004. 428(6982): p. 529-35.

101. Lee, C.J., W.I. Chan, and P.J. Scotting, CIC, a gene involved in cerebellar development and

ErbB signaling, is significantly expressed in medulloblastomas. J Neurooncol, 2005. 73(2):

p. 101-8.

102. Lim, J., et al., Opposing effects of polyglutamine expansion on native protein complexes

contribute to SCA1. Nature, 2008. 452(7188): p. 713-8.

103. Lam, Y.C., et al., ATAXIN-1 interacts with the repressor Capicua in its native complex to

cause SCA1 neuropathology. Cell, 2006. 127(7): p. 1335-47.

104. Wijmenga, C., et al., Chromosome 4q DNA rearrangements associated with

facioscapulohumeral muscular dystrophy. Nat Genet, 1992. 2(1): p. 26-30.

105. Dixit, M., et al., DUX4, a candidate gene of facioscapulohumeral muscular dystrophy,

encodes a transcriptional activator of PITX1. Proc Natl Acad Sci U S A, 2007. 104(46): p.

18157-62.

106. Sirvent, N., et al., Fusion of EWSR1 with the DUX4 facioscapulohumeral muscular

dystrophy region resulting from t(4;22)(q35;q12) in a case of embryonal

rhabdomyosarcoma. Cancer Genet Cytogenet, 2009. 195(1): p. 12-8.

107. Kowaljow, V., et al., The DUX4 gene at the FSHD1A locus encodes a pro-apoptotic

protein. Neuromuscul Disord, 2007. 17(8): p. 611-23.

108. de Launoit, Y., et al., The PEA3 group of ETS-related transcription factors. Role in breast

cancer metastasis. Adv Exp Med Biol, 2000. 480: p. 107-16.

109. Farber, C.R. and A.J. Lusis, Integrating global gene expression analysis and genetics. Adv

Genet, 2008. 60: p. 571-601.

110. Clifford, N., et al., The EphA3 receptor is expressed in a subset of rhabdomyosarcoma cell

lines and suppresses cell adhesion and migration. J Cell Biochem, 2008. 105(5): p. 1250-9.

111. Wykosky, J. and W. Debinski, The EphA2 receptor and ephrinA1 ligand in solid tumors:

function and therapeutic targeting. Mol Cancer Res, 2008. 6(12): p. 1795-806.

112. Timar, J., et al., Melanoma genomics reveals signatures of sensitivity to bio- and targeted

therapies. Cell Immunol, 2006. 244(2): p. 154-7.

Page 100: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

90

113. Sakabe, N.J. and M.A. Nobrega, Genome-wide maps of transcription regulatory elements.

Wiley Interdiscip Rev Syst Biol Med. 2(4): p. 422-37.

114. Sadikovic, B., et al., Cause and consequences of genetic and epigenetic alterations in

human cancer. Curr Genomics, 2008. 9(6): p. 394-408.

115. Himanen, J.P., N. Saha, and D.B. Nikolov, Cell-cell signaling via Eph receptors and

ephrins. Curr Opin Cell Biol, 2007. 19(5): p. 534-42.

116. Pasquale, E.B., Eph receptor signalling casts a wide net on cell behaviour. Nat Rev Mol

Cell Biol, 2005. 6(6): p. 462-75.

117. Klein, R., Excitatory Eph receptors and adhesive ephrin ligands. Curr Opin Cell Biol, 2001.

13(2): p. 196-203.

118. Egea, J. and R. Klein, Bidirectional Eph-ephrin signaling during axon guidance. Trends

Cell Biol, 2007. 17(5): p. 230-8.

119. Kullander, K. and R. Klein, Mechanisms and functions of Eph and ephrin signalling. Nat

Rev Mol Cell Biol, 2002. 3(7): p. 475-86.

120. Dodelet, V.C. and E.B. Pasquale, Eph receptors and ephrin ligands: embryogenesis to

tumorigenesis. Oncogene, 2000. 19(49): p. 5614-9.

121. Surawska, H., P.C. Ma, and R. Salgia, The role of ephrins and Eph receptors in cancer.

Cytokine Growth Factor Rev, 2004. 15(6): p. 419-33.

122. Worby, C.A. and J.E. Dixon, Sorting out the cellular functions of sorting nexins. Nat Rev

Mol Cell Biol, 2002. 3(12): p. 919-31.

123. Carlton, J., et al., Sorting nexins--unifying trends and new perspectives. Traffic, 2005. 6(2):

p. 75-82.

124. Kurten, R.C., D.L. Cadena, and G.N. Gill, Enhanced degradation of EGF receptors by a

sorting nexin, SNX1. Science, 1996. 272(5264): p. 1008-10.

125. Horazdovsky, B.F., et al., A sorting nexin-1 homologue, Vps5p, forms a complex with

Vps17p and is required for recycling the vacuolar protein-sorting receptor. Mol Biol Cell,

1997. 8(8): p. 1529-41.

126. Qin, B., et al., Sorting nexin 10 induces giant vacuoles in mammalian cells. J Biol Chem,

2006. 281(48): p. 36891-6.

127. Duffy, M.J., et al., Role of ADAMs in cancer formation and progression. Clin Cancer Res,

2009. 15(4): p. 1140-4.

Page 101: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

91

128. Edwards, D.R., M.M. Handsley, and C.J. Pennington, The ADAM metalloproteinases. Mol

Aspects Med, 2008. 29(5): p. 258-89.

129. Mancia, F. and L. Shapiro, ADAM and Eph: how Ephrin-signaling cells become detached.

Cell, 2005. 123(2): p. 185-7.

130. Shintani, Y., et al., Overexpression of ADAM9 in non-small cell lung cancer correlates with

brain metastasis. Cancer Res, 2004. 64(12): p. 4190-6.

131. O'Shea, C., et al., Expression of ADAM-9 mRNA and protein in human breast cancer. Int J

Cancer, 2003. 105(6): p. 754-61.

132. Peduto, L., et al., Critical function for ADAM9 in mouse prostate cancer. Cancer Res, 2005.

65(20): p. 9312-9.

133. Grutzmann, R., et al., ADAM9 expression in pancreatic cancer is associated with tumour

type and is a prognostic factor in ductal adenocarcinoma. Br J Cancer, 2004. 90(5): p.

1053-8.

134. Rozenblatt-Rosen, O., et al., The parafibromin tumor suppressor protein is part of a human

Paf1 complex. Mol Cell Biol, 2005. 25(2): p. 612-20.

135. Lin, L., et al., Nuclear localization of the parafibromin tumor suppressor protein implicated

in the hyperparathyroidism-jaw tumor syndrome enhances its proapoptotic function. Mol

Cancer Res, 2007. 5(2): p. 183-93.

136. Hahn, M.A., et al., CDC73/HRPT2 CpG island hypermethylation and mutation of 5'-

untranslated sequence are uncommon mechanisms of silencing parafibromin in parathyroid

tumors. Endocr Relat Cancer. 17(1): p. 273-82.

137. Chaudhary, K., et al., Human RNA polymerase II-associated factor complex: dysregulation

in cancer. Oncogene, 2007. 26(54): p. 7499-507.

138. Ciesielski, M.J. and R.A. Fenstermaker, Oncogenic epidermal growth factor receptor

mutants with tandem duplication: gene structure and effects on receptor function.

Oncogene, 2000. 19(6): p. 810-20.

139. Fenstermaker, R.A. and M.J. Ciesielski, Deletion and tandem duplication of exons 2 - 7 in

the epidermal growth factor receptor gene of a human malignant glioma. Oncogene, 2000.

19(39): p. 4542-8.

140. Fenstermaker, R.A., M.J. Ciesielski, and G.J. Castiglia, Tandem duplication of the

epidermal growth factor receptor tyrosine kinase and calcium internalization domains in A-

172 glioma cells. Oncogene, 1998. 16(26): p. 3435-43.

Page 102: UNDIFFERENTIATED SARCOMAS: …...Figure 3.7 RT-PCR confirmation of the CIC-DUX4 fusion for t(4;19) pilot study 47 Figure 3.8 Identification of the CIC-DUX4 fusion by RT-PCR screening

92

141. Lewis, T.B., C.M. Coffin, and P.S. Bernard, Differentiating Ewing's sarcoma from other

round blue cell tumors using a RT-PCR translocation panel on formalin-fixed paraffin-

embedded tissues. Mod Pathol, 2007. 20(3): p. 397-404.

142. Downing, J.R., et al., Detection of the (11;22)(q24;q12) translocation of Ewing's sarcoma

and peripheral neuroectodermal tumor by reverse transcription polymerase chain reaction.

Am J Pathol, 1993. 143(5): p. 1294-300.