+ All Categories
Home > Documents > MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory...

MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory...

Date post: 24-Jun-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
81
ID1 mediates glioblastoma chemoresistance to temozolomide by Angela Maria Louisa Celebre A thesis submitted in conformity with the requirements for the degree of Master of Science Laboratory Medicine and Pathobiology University of Toronto © Copyright by Angela Celebre 2016
Transcript
Page 1: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

ID1  mediates  glioblastoma  chemoresistance  to  temozolomide  

  by    

Angela  Maria  Louisa  Celebre  

A  thesis  submitted  in  conformity  with  the  requirements  for  the  degree  of  Master  of  Science  Laboratory  Medicine  and  Pathobiology  

University  of  Toronto  

©  Copyright  by  Angela  Celebre  2016  

Page 2: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

ii

ID1  mediates  glioblastoma  chemoresistance  to  temozolomide  

Angela Celebre

Master of Science

Laboratory Medicine and Pathobiology University of Toronto

2016

Abstract  

Glioblastoma (GBM) is the most lethal primary brain tumour in adults, and represents a

therapeutic challenge. We investigated the role of inhibitor-of-DNA-binding-1 (ID1), a

transcriptional regulator implicated in cancer cell survival and cancer progression in breast and

colon cancer, as a mediator of chemoresistance in glioblastoma. Using immunohistochemical

and Western blot analysis, we determined that increased ID1 expression correlates with

decreased progression-free survival in GBM patient samples following chemotherapy, and TMZ

resistancy in GBM cell lines. Inhibition of ID1 using pimozide (chemical ID1 inhibitor), siRNA-

mediated knockdown, and CRISPR/Cas9-mediated knockout enhanced the effect of TMZ in

GBM cell lines and intracranial xenografts. In vitro, ID1 inhibition sensitized cells to TMZ-

treatment and ID1-knockout reduced colony formation post-TMZ. In vivo, ID1-knockout delayed

GBM tumour initiation and progression, and increased survival; pimozide combined with TMZ

delayed tumour recurrence. Our studies demonstrate that ID1 may serve as a potential

therapeutic target in glioblastoma.

Page 3: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

iii

Acknowledgments  My time in the Das Laboratory has been an incredible and exciting experience. Over the past two

years I have grown tremendously, both on an academic and personal level. I have met so many

inspiring, hard-working individuals who I have had the privilege of interacting with on a daily

basis and now have the honor of calling my friends. These individuals offered insight, guidance,

and support when I encountered challenges during my project, and celebrated in my successes

when experiments finally worked and my project moved forward.

I would like to sincerely thank my supervisor, Dr. Sunit Das, for his guidance and support

throughout my graduate studies, for his mentorship, and for allowing me to pursue numerous

opportunities both academic and extra-curricular. A very special thank-you goes to my

committee members, Dr. Jane McGlade and Dr. Cindi Morshead, for their instrumental guidance

and critique of my work. I would also like to thank the Das Lab Manager, Megan Wu, for her

mentorship, patience, and contagious optimism. Megan took me under her wing and showed me

the ropes of working in a wet-lab. She taught me how to prioritize my time in the lab, design

meaningful experiments, and critically analyze my work. Another huge thank-you goes to Dr.

Rohit Sachdeva for constantly challenging me, and making me think for myself. Many thanks to

the following members of the Das Lab for all their wonderful help; Uswa Shahzad, Chris Li,

Sandra Smiljanic, Jennifer Guan, Jeffrey Chan, and Karan Dhand. I am also grateful for the help

provided by Brian Golbourn, Patricia Rakopoulos, and Dr. Vijay Ramaswamy. Finally, I would

like to thank my family and friends for all their emotional support and patience.

Page 4: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

iv

Table  of  Contents  

 

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

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

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

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

Abbreviations  ......................................................................................................................................  x  

Chapter  1  Introduction  .......................................................................................................................  1  

  Glioblastoma  ...............................................................................................................................  1  

1.1   Clinical  Relevance  ............................................................................................................................  1  

1.2   Pathology  and  Molecular  Biology  ....................................................................................................  1  

1.3   Current  Standard  of  Care  ................................................................................................................  3  

  Treatment  Resistance  and  Tumour  Recurrence  ........................................................................  3  

2.1   TMZ:  Structure  and  Mechanism  of  Action  ......................................................................................  3  

2.2   Mechanisms  of  Chemoresistance  in  GBM  .......................................................................................  4  

2.2.1   Direct  Repair  via  MGMT  ..............................................................................................................  4  

2.2.2   Mismatch  Repair  .........................................................................................................................  5  

2.2.3   Base  Excision  Repair  ....................................................................................................................  6  

2.2.4   Acquired  Drug  Resistance  ...........................................................................................................  6  

2.3   Cancer  Stem  Cells  ............................................................................................................................  6  

2.4   Recurrent  GBM  ................................................................................................................................  7  

2.4.1   Molecular  Biology  .......................................................................................................................  7  

2.4.2   Treatment  of  Recurrent  GBM  ......................................................................................................  8  

  ID1  in  Normal  Biology  .................................................................................................................  8  

3.1   The  ID  Family:  Structure  and  Function  ............................................................................................  8  

3.2   ID1  in  Cellular  Differentiation  ..........................................................................................................  8  

3.3   ID1  in  the  Developmental  Process  ..................................................................................................  9  

3.4   ID1  in  Cell  Cycle  Regulation  and  Apoptosis  .....................................................................................  9  

Page 5: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

v

3.5   ID1  in  Invasion,  Migration,  and  Angiogenesis  ...............................................................................  10  

  ID1  in  Cancer  Biology  ................................................................................................................  11  

4.1   ID1  expression  in  Primary  Tumour  Cells  ........................................................................................  11  

4.2   Multimodal  Activation  of  ID1  in  Cancer  ........................................................................................  11  

4.3   ID1  in  Tumorigenesis  .....................................................................................................................  13  

4.4   ID1  in  Tumour  Invasion  and  Metastasis  ........................................................................................  14  

4.5   ID1  in  Tumour  Angiogenesis  ..........................................................................................................  14  

4.6   The  Role  of  ID1  in  Cancer  Stem  Cells  .............................................................................................  15  

4.7   ID1  in  Contributing  to  Treatment  Failure  ......................................................................................  15  

4.8   ID1  in  Glioblastoma  .......................................................................................................................  16  

  ID1  as  a  Therapeutic  Target  .....................................................................................................  17  

5.1   IDs  in  Cancer  Therapy  ....................................................................................................................  17  

5.2   ID1:  Potential  Therapeutic  Target  in  Combination  with  Chemotherapy  .......................................  18  

  Hypothesis  ................................................................................................................................  18  

Chapter  2  Methods  ...........................................................................................................................  20  

  Methods  ....................................................................................................................................  20  

7.1   Cell  Culture  ....................................................................................................................................  20  

7.2   Clinical  Chemotherapeutic  Agents  ................................................................................................  20  

7.3   Immunocytochemistry  ..................................................................................................................  20  

7.4   Alamar  Blue  ...................................................................................................................................  21  

7.5   Western  Blotting  ...........................................................................................................................  21  

7.6   Cell  Viability  Counter  .....................................................................................................................  21  

7.7   ID1  Silencing  ..................................................................................................................................  22  

7.8   Pimozide  (Clinical  ID1  Inhibitor)  ....................................................................................................  22  

7.9   Immunohistochemical  Staining  and  Scoring  .................................................................................  22  

7.10   CRISPR/Cas9  System  ......................................................................................................................  23  

7.11   Colony  Forming  Assay  ...................................................................................................................  23  

7.12   Evaluation  of  ID1  Knockout  in  an  In  Vivo  Model  of  Glioblastoma  .................................................  23  

7.13   Evaluation  of  ID1  Inhibition  in  Combination  With  Temozolomide  Chemotherapy  in  an  In  Vivo  

Model  of  Glioblastoma  ...............................................................................................................................  24  

7.14   Bioluminescence  Imaging  ..............................................................................................................  24  

Page 6: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

vi

7.15   Quantitative  Real-­‐time  Polymerase  Chain  Reaction  .....................................................................  24  

7.16   Polymerase  Chain  Reaction  Amplification  ....................................................................................  24  

7.17   Statistical  Analysis  .........................................................................................................................  25  

Chapter  3  Results  ..............................................................................................................................  26  

  Results  .......................................................................................................................................  26  

8.1   ID1  Expression  Correlates  with  Response  to  Temozolomide  Treatment  in  Glioblastoma  ............  26  

8.2   ID1  Expression  Increases  Following  Temozolomide  Treatment  ....................................................  29  

8.3   ID1  Inhibition  Sensitizes  GBM  Cells  to  TMZ-­‐Chemotherapy  Treatment  .......................................  32  

8.4   ID1  Knockout  Reduces  Colony  Formation  Capacity  following  TMZ-­‐Treatment  ............................  34  

8.4.1   Development  of  an  ID1-­‐knockout  cell  line  .................................................................................  34  

8.4.2   ID1-­‐knockout  decreases  colony  formation  following  treatment  with  TMZ  ...............................  37  

8.5   ID1  Knockout  Delays  GBM  Tumour  Initiation/Progression  and  Increases  Overall  Survival  ..........  39  

8.6   Pimozide  Enhances  the  Effect  of  TMZ-­‐Treatment  and  Provides  a  Therapeutic  Advantage  in  

Glioblastoma  ..............................................................................................................................................  42  

Chapter  4  Discussion  and  Future  Directions  ....................................................................................  45  

  Discussion  and  Future  Directions  .............................................................................................  45  

9.1   Glioblastoma:  Discovering  an  Ideal  Therapeutic  Target  ................................................................  45  

9.2   Treating  Glioblastoma  in  vitro:  ID1  Inhibition  Enhances  the  Effect  of  Temozolomide  .................  47  

9.3   Treating  Glioblastoma  in  vivo:  Targeting  ID1  Provides  a  Therapeutic  Advantage  ........................  48  

9.4   Conclusions  ...................................................................................................................................  49  

9.5   Future  Directions  ...........................................................................................................................  49  

References  ........................................................................................................................................  51  

Page 7: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

vii

List  of  Tables    

Table 1: Top off-target hits in CRISPR/Cas9 ID1-knockout system. 36

Table 2: Survival outcomes in ID1-knockout in vivo study. 41

Page 8: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

viii

List  of  Figures    

Figure 1: Key DNA repair mechanisms that mediate TMZ cytotoxicity. 5

Figure 2: The role of ID1 in cell cycle regulation. 10

Figure 3: ID1 protein stabilization and degradation. 13

Figure 4: Increased ID1 expression following chemotherapy correlates with

decreased progression-free survival in glioblastoma human patient tumour

samples.

27

Figure 5: ID1 expression correlates with resistance to TMZ in vitro. 28

Figure 6: ID1 protein expression increases following TMZ-treatment in GBM

cells.

30

Figure 7: Increased ID1 expression is post-treatment is TMZ-specific. 31

Figure 8: ID1 inhibition (via siRNA) increases GBM cell sensitivity to TMZ-

chemotherapy treatment.

33

Figure 9: Development of an ID1-knockout line using CRISPR/Cas9. 35

Figure 10: Three top off-target hits in the CRISPR/Cas9 ID1-knockout model

display no mutation in the target region.

36

Figure 11: ID1-knockout reduces colony formation capacity following

treatment with temozolomide.

38

Figure 12: ID1-knockout delays glioblastoma tumour initiation and progression. 40

Figure 13: ID1-knockout increases overall survival time in glioblastoma. 41

Figure 14: Pimozide (small molecule inhibitor that targets ID1 degradation)

increases GBM cell sensitivity to TMZ-treatment.

43

Page 9: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

ix

Figure 15: Pimozide enhances the effect of TMZ-treatment and provides a

therapeutic advantage in glioblastoma.

44

Page 10: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

x

Abbreviations  

ACC Animal care committee

AKT Protein kinase B

ATCC American type culture collection

AUP Animal use protocol

BCA Bicinechoninic acid

BER Base excision repair

bHLH Basic helix-loop-helix

BMPs Bone morphogenetic proteins

BSA Bovine serum albumin

Cas9 CRISPR associated protein 9

CDK Cyclin-dependent kinase

CDKN2A Cyclin-dependent kinase inhibitor 2A

CDKN2B Cyclin-dependent kinase inhibitor 2B

CNS Central nervous system

CO2 Carbon dioxide

CRISPR Clustered regularly interspaced short palindromic repeats

CSCs Cancer tem cells

DAPI 4’,6-diamidino-2-phenylindole

DMEM Dulbecco’s modified eagle media

Page 11: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

xi

DMSO Dimethyl sulfoxide

DNA Deoxyribonucleic acid

DUBs Deubiquitylases

EGFR Epidermal growth factor receptor

EGR1 Early growth response 1

EMT Epithelial-to-mesenchymal transition

ETS E26 transformation-specific

FBS Fetal bovine serum

FDA Food and drug administration

FFPE Formalin-fixed paraffin-embedded

G1 Gap 1

G2 Gap 2

GAPDH Glyceraldehyde 3-phosphate dehydrogenase

GBM Glioblastoma

GIC Glioma-initiating cell

hEGF Human epidermal growth factor

hFGF Human basic fibroblast growth factor

HLH Helix-loop-helix

IC50 Half maximal inhibitory concentration

ICC Immunocytochemistry

Page 12: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

xii

ID1 Inhibitor of DNA binding 1

ID1-KO ID1-knockout

IDH1 Isocitrate dehydrogenase 1

IHC Immunohistochemistry

IP Intraperitoneal injection

M Mitosis

MDM2 Mouse double minute 2 homolog

MeA Methyl adenine

MeG Methyl guanine

MET Mesenchymal-to-epithelial-transition

MGMT Methylguanine-DNA methyltransferase

Min Minutes

MMPs Matrix metalloproteinases

MMR Mismatch repair

mRNA Messenger RNA

MTIC Monomethyl triazone imidazole carboxamide

MYOD1 Myoblast determination protein 1

NF-kB NF-kappaB

NF1 Neurofibromatosis type I

NOD Non-obese diabetic

Page 13: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

xiii

NPC Nasopharyngeal carcinoma

NSC Neocarzinostatin

NSG NOD/SCID/Gamma immunodeficient mice

NT No treatment

OS Overall survival

PAM Protospacer adjacent motif

PARP-1 Poly(ADP-ribose) polymerase-1

PBS Phosphate buffered saline

PCAO Peptide-conjugated antisense oligonucleotide

PCR Polymerase chain reaction

PDGFRA Platelet-derived growth factor receptor, alpha polypeptide

PFS Progression-free survival

PI3K Phosphatidylinositol-4,5-bisphosphate 3-kinase

PID Post-implantation day

PTEN Phosphatase and tensin homolog

RB Retinoblastoma

rMA Relative mask area

RNA Ribonucleic acid

S Synthesis

SCID Severe combined immunodeficiency

Page 14: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

xiv

SDV Standard devation

Sec Seconds

SEM Standard error of the mean

SEMA3F Semaphorin 3F

sgRNA Single guide RNA

si-RNA Small (or short) interfering RNA

SMURF2 SMAD ubiquitylation regulatory factor 2

TALENs Transcription activator-like effector nucleases

TCGA The cancer genome atlas

TCP Toronto Centre for Phenogenomics

TGF-β Transforming growth factor-β

TMA Tissue microarray

TMZ Temozolomide

TP53 Tumor protein p53

USP1 Ubiquitin-specific protease 1

UV Ultraviolet

V Volts

VEGF Vascular endothelial growth factor

VEGFR1 Vascular endothelial growth factor receptor 1

VEGFR2 Vascular endothelial growth factor receptor 2

Page 15: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

xv

WHO World Health Organization

ug Microgram

um Micrometer

uM Micromolar

nM Nanomolar

Page 16: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

1

Chapter  1      

Introduction  

  Glioblastoma  

1.1   Clinical  Relevance      Glioblastoma (GBM) is the most common malignant primary brain tumour in adults, accounting

for over 50% of primary central nervous system (CNS) gliomas1. Approximately 0.59 to 3.69

GBM cases per 100,000 are diagnosed annually worldwide2. Moreover, GBM affects over

20,000 patients in North America every year3, and is responsible for the highest number of years

of life lost of any cancer, exceeding even breast and lung cancer4. This malignancy is associated

with an aggressive clinical course and poor prognosis, with a median overall survival (OS) of

less than 15 months despite maximal treatment5. OS of patients with newly diagnosed GBM is

17-30% at 1 year, and only 3-5% at 2 years6. The etiology of GBM remains largely unknown.

Only ionizing radiation7, and certain genetic syndromes are well-defined risk factors, including

Cowden, Turcot, Li-Fraumeni, neurofibromatosis type 1 and type 2, tuberous sclerosis, and

familial schwannomatosis8,9.

Treatment of GBM involves maximal site surgical resection followed by a combination of

radiotherapy and chemotherapy with temozolomide (TMZ), an alkylating agent that has been

shown to potentiate radiation and possess cytotoxic properties in GBM5. Tumour recurrence

typically occurs 6 months after the patient receives first-line therapy, and this is mainly attributed

to the development of treatment resistance10. Therefore, meaningful treatment of glioblastoma

requires the identification of new therapies that target chemoresistance, in order to effectively

prevent tumour recurrence.

1.2   Pathology  and  Molecular  Biology    The World Health Organization (WHO) classification system groups gliomas into 4 histological

grades defined by increasing degrees of undifferentiation, anaplasia, and aggressiveness11. These

grades carry prognostic and survival correlates. Glioblastoma is a WHO grade IV astrocytoma,

the most malignant grade11.

Page 17: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

2

GBMs are morphologically heterogeneous tumours that are characterized by considerable

variability in biological behavior12. The histopathology of GBM includes considerable cellularity

and mitotic activity, poorly differentiated neoplastic astrocytes, vascular proliferation, and

necrosis13. Glioblastomas are highly invasive tumours, and considered pleomorphic because their

tumour cells vary in size and shape13. However, despite being highly invasive, they are typically

confined to the CNS and do not metastasize.

Recent comprehensive genetic screens of GBM have confirmed that genetic alterations are

scattered across the entire genome, and affect a number of chromosomes. These studies have

revealed that loss of heterozygosity on chromosome 10 is the most frequent genetic alteration in

GBM (60-80% cases)14. However, there are numerous regions of loss in GBM, including areas

on 1p, 6q, 9p, 10p, 10q, 13q, 14q, 15q, 17p, 18q, 19q, 22q, and Y15,16,17,18,19,20; many of these

regions of loss represent loss of specific tumour suppressor genes. For example, regions

frequently lost at chromosome 10 include those containing PTEN and MGMT21,6. Loss of

chromosome 9p is also frequently seen, which contains a variety of tumor-suppressor genes,

including CDKN2A and CDKN2B22,6. This impacts both the RB and P53 pathways because

CDKN2A and CDKN2B encode the cell cycle proteins, p14, p15, and p16, implicated in these

pathways23. Gains of gene expression due to genetic alteration have also been demonstrated in

GBM, however, oncogenic expression is much less frequent than loss of gene expression. The

most common oncogenic event is the amplification of EGFR gene on chromosome 724,25. Focal

amplification of EGFR correlates with mutations and deletions in the EGFR gene, and EGFR

overexpression26. EGFR amplification also correlates with subsequent activation of the

PI3K/AKT pathway27, which is associated with poor prognosis28. Oncogenic amplification of

CDK4 and MDM2 results in the disruption of the RB AND TP53 pathways29.

From a molecular standpoint, GBM is classified into 4 distinct transcriptional subgroups, as

based on genome-wide expression studies: classical, mesenchymal, proneural, and neural30,28.

Although many molecular abnormalities and mutations overlap across the transcriptional

subclasses, each subgroup displays its own unique mutational landscape. Verhaak and colleagues

described how each subtype was characterized by a distinct pattern of somatic mutations and

DNA copy number30. A key finding was that aberrations and gene expression of EGFR, NF1,

and PDGFRA/IDH1 each defined classical, mesenchymal, and proneural, respectively. Also,

Page 18: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

3

different subtypes varied in their response to aggressive therapy, with the classical subtype

showing the greatest benefit, and the proneural subtype showing no benefit.

1.3   Current  Standard  of  Care  First-line treatment for GBM consists of maximal site surgical resection followed by adjuvant

radiotherapy and TMZ-chemotherapy. TMZ is a DNA alkylating agent that is administered

orally, concomitantly with radiotherapy, followed by an adjuvant course. The addition of TMZ to

first-line treatment is supported by a randomized phase 3 trial lead by Stupp5, which found an

increase in median survival from 12.1 to 14.6 months with the combination of TMZ and

radiotherapy, compared to radiotherapy alone; this represents a median survival benefit of 2.5

months. Furthermore, the two-year survival rate was 26.5% and 10.4% in the radiotherapy-plus-

temozolomide compared to radiotherapy alone group, respectively5. A study conducted by Hegi

and colleagues found that patients harbouring tumours with promoter methylation of the DNA

repair enzyme O6-methylguanine-DNA methyltransferase (MGMT), which results in gene

silencing, were more likely to benefit from the addition of TMZ to first-line treatment31.

Unfortunately, despite optimal treatment, virtually all patients will experience treatment

resistance and subsequent tumour recurrence.

  Treatment  Resistance  and  Tumour  Recurrence  

2.1   TMZ:  Structure  and  Mechanism  of  Action  TMZ is an imidazotetrazinone derivative of dacarbazine and functions as an alkylating agent

prodrug32. It is a small molecule with a molecular weight of 194 daltons, allowing it to be readily

absorbed in the digestive track33. TMZ is also lipophilic, which means that it can cross the blood-

brain barrier. It serves as a monofunctional agent with good tissue distribution, and is schedule

dependent in terms of anti-tumour activity34.

TMZ is stable at the acidic pH of the stomach, which allows it to be absorbed intact after oral

administration. However, once in contact with the slightly basic physiologic pH of the blood and

tissues, TMZ undergoes spontaneous hydrolysis to the active compound monomethyl triazone

imidazole carboxamide (MTIC)32. MTIC then rapidly breaks down to form the reactive

methyldiazonium ion, which primarily methylates guanine residues in the DNA molecule. More

specifically, the methyldiazonium ion preferentially methylates DNA at N7 positions of guanine

Page 19: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

4

(N7-MeG; 70%), but also methylates N3 adenine (N3-MeA; 9%) and O6 guanine residues (O6-

MeG; 6%)35,36. TMZ has a half-life of 1.81h37 and studies have shown that the level of drug in

the brain and cerebrospinal fluid are 30-40% plasma concentration38. TMZ does not result in

chemical cross-linking of DNA strands, therefore is considered to be less toxic in comparison to

other chemotherapeutic agents.

TMZ cytotoxicity is primarily mediated through O6-MeG, a toxic lesion39,40. During DNA

replication, O6-MeG mispairs with thymine (not cytosine), which alerts DNA mismatch repair

(MMR)41. MMR recognises and excises the mispaired base on the daughter strand, but is unable

to recognise O6-MeG on the template strand. This mispairing at O6-MeG repeats itself over

numerous replication cycles, which leads to persistent DNA strand breaks, replication fork

collapse, G2/M cell cycle arrest, and ultimately apoptosis42,43,44. The cytotoxic effect of TMZ is

hindered at N7-MeG and N3-MeA because these sites are rapidly repaired by DNA base excision

repair (BER)45.

2.2   Mechanisms  of  Chemoresistance  in  GBM  Patients with newly diagnosed GBM are typically given TMZ as first-line chemotherapy. Yet, all

patients succumb to the disease due to treatment failure and subsequent tumour recurrence. Four

of the main mechanisms of chemoresistance include direct repair via MGMT, mismatch repair

(MMR), base excision repair (BER), and acquired drug resistance.

2.2.1   Direct  Repair  via  MGMT  

The major DNA repair mechanism contributing to TMZ resistance in GBM is direct repair

(Figure 1). MGMT repairs O6-alkylguanine adducts in a single step by protecting cancer cells

from chemotherapeutic alkylating agents like TMZ39. Although MGMT activity can vary up to

300-fold in gliomas46, studies have found a strong positive correlation between MGMT activity

and TMZ resistance47,48. Zhang et al. showed MGMT is upregulated when U373 GBM cells are

exposed to incremental concentrations of TMZ, increasing resistancy by >4-fold49. In GBM

patient tumour samples, MGMT activity has been shown to be increased in recurrent tumours

post-chemoradiotherapy in comparison to the primary, untreated sample50. This finding can be

attributed to either TMZ selecting for MGMT-expressing cells or TMZ inducing MGMT

expression.

Page 20: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

5

TMZ O6#G

+Me

O6#Me#G

G

MGMT)absent

Mutation)tolerated

#Me

MGMT)present

Cell)survival

CytotoxicityMMR+

MMR#

Cell)survival

2.2.2   Mismatch  Repair  

DNA mismatch repair is another mechanism of chemoresistance in GBM. MMR is the

recognition and correction of mispaired bases, which plays a very important role in correcting

replicative mismatches that have escaped polymerase proofreading. Loss of MMR results in a

drastic increase in the number of mutations. Cellular response to TMZ is influenced by MMR

status: TMZ requires functional MMR to produce its desired cytotoxic effect. Hence, MMR

deficiency in glioma can be caused by somatic mutations harboured within MMR proteins (i.e.

MSH2, MLH1 and MSH6), and is associated with TMZ tolerance49,51,52. A study conducted by

Yip et al. found MSH6 inactivation in vitro lead to an increase in TMZ resistance; conversely,

reconstitution of MSH6 expression restored TMZ sensitivity52. Furthermore, GBM MSH

mutations selected during TMZ treatment correlate with TMZ resistance, when studied in vivo. A

study investigating clinical GBM patient tumour samples found that MSH6 alterations in tumour

cell genome are associated with TMZ resistance. For example, MSH6 mutations are found in

26% of recurrent GBM cases following TMZ chemotherapy52.

Figure 1. Key DNA repair mechanisms that mediate TMZ cytotoxicity. TMZ, a DNA alkylating agent, exerts its cytotoxic effect by adding a methyl group to O6-G (O6-Me-G). In the presence of MGMT, MGMT will repair O6-methylguanine adducts in a single step by removing the methyl group, resulting in cell survival. If MGMT is absent and MMR is intact, MMR will repeatedly mispair O6-MeG, resulting in a cytotoxic response; if MMR is not intact, the mutation will be tolerated, resulting in cell survival. (Figure adapted from Zhang J, Stevens MFG, Bradshaw TD. Temozolomide: Mechanisms of Action, Repair and Resistance. Current Molecular Pharmacology 2012. 5: 102-114.)

Page 21: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

6

2.2.3   Base  Excision  Repair  

The third DNA repair mechanism contributing to TMZ resistance in GBM is base excision

repair. The BER pathway is responsible for the removal and repair of non-bulky damaged

nucleotides, abasic sites, and DNA single strand breaks generated by alkylating agents53. In

gliomas, the BER pathway repairs N-Me purine lesions, conferring resistance to TMZ54. A key

protein in DNA damage signaling and the BER pathway is poly(ADP-ribose) polymerase-1

(PARP-1). PARP-1 is activated in response to DNA damage, and facilitates efficient DNA repair

and survival of cells undergoing genotoxic stress55. Inhibition of PARP-1 increases the frequency

of DNA strand breaks, and may provide a means to overcome resistance to TMZ56.

2.2.4   Acquired  Drug  Resistance  

Acquired drug resistance in GBM occurs when tumours that are initially sensitive to TMZ-

chemotherapy develop resistance, as a consequence of selective pressures in the presence of the

chemotherapeutic agent. Acquired resistance to TMZ can be attributed to a variety factors,

including selection of pre-existing resistant cell clones, genetic/epigenetic changes in neoplastic

cells induced by the drug, and selection of genes with a survival advantage57. Specific

mechanisms that can confer acquired resistance to TMZ include intracellular drug inactivation,

enhanced repair of drug-induced DNA damage, and alteration of drug target by mutation58,59. In

GBM, a highly heterogeneous tumour, chemotherapy eliminates TMZ-sensitive malignant cells;

however, it is hypothesized that chemoresistant cancer stem cells (CSCs) are able to survive, and

are later capable of driving tumour recurrence.

2.3   Cancer  Stem  Cells  Cancer stems cells were first identified in acute myeloid leukemia (AML) by John Dick and

colleagues60. They determined that the tumour-initiating cell in AML possesses similar traits to a

normal stem cell61. Furthermore, they found that AML was organized in a cellular hierarchy, and

that tumorigenicity was coupled to the ability to self renew62. This seminal work gave rise to the

cancer stem cell hypothesis, which posits that tumour cells are heterogeneous and hierarchically

arranged; tumour growth is driven by a subpopulation of cells that possess behavioral and

phenotypic characteristics of normal stem cells, and are thus called cancer stem cells. CSCs are

characterized by their ability to proliferate and self-renew. Furthermore, they are multipotent and

Page 22: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

7

can give rise to the diverse set of cells that make up a given tumour. Current therapies are

extremely cytotoxic to the bulk of the proliferative tumour cells, but appear to have a decreased

effect on CSCs, which are able to survive chemotherapy and later drive tumour recurrence. Also,

several studies have shown that prolonged treatment with TMZ enriches for tumour cells with

stem cell properties, and induces stemness, consequently increasing tumor aggressiveness63,64,65.

The role of CSCs has now been described in numerous solid tumours66,67,68,69,70, including

glioblastoma71,72.

CSCs in glioblastoma were first identified by Peter Dirks, who determined that glioblastoma is

arranged in a cellular hierarchy and tumorigenicity is limited to glioma cells that possess the

CSC identity71. Glioma stem cells (GSCs) have been shown to be enriched by chemoradiation,

suggesting that they may serve as a repository for tumour recurrence73. Moreover, Auffinger et

al. found that treatment of differentiated glioma cells with TMZ induced stem cell properties in

these cells63. The role of GSCs in glioblastoma tumour recurrence has been characterized in a

mouse model, where GSCs were shown to drive tumour recurrence following chemotherapy with

temozolomide (TMZ)74. It has therefore been postulated that meaningful treatment of

glioblastoma will require the development of new therapies that target glioma stem cells.

2.4   Recurrent  GBM  

2.4.1   Molecular  Biology  

The biology of recurrent GBM remains largely unknown, for two reasons. First and foremost,

only 20-30% of recurrent GBMs are accessible to surgery, therefore there are very few recurrent

samples available in the tissue bank for experimental use75. Secondly, recurrent tumours usually

contain much more necrotic tissue76. Thus, recurrence typically develops from tumour cells

located in the tumour periphery. Studies have shown that approximately two-thirds of GBM

tumours recur locally (i.e. within 2 cm of original tumour margin), and one-third recur far away

from the initial tumour bulk77,78,79. Furthermore, local recurrrences share ~70% mutations with

primary tumour; whereas distant recurrences only share ~25% mutations, despite the recurrence

stemming from the original tumour80. A study conducted by Andor et al. using the cancer

genome atlas (TCGA) genome sequencing data found that there are approximately ~7 genetically

different subclones per primary GBM sample (per 100 mg of vital tumour tissue)81. This number

did not vary in recurrent GBMs.

Page 23: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

8

2.4.2   Treatment  of  Recurrent  GBM  

Nearly all patients experience disease progression after a median progression-free survival (PFS)

of 7-10 months, following first-line therapy82. At present, there is no standard treatment to

improve survival in recurrent GBM, therefore clinical trials should be strongly considered and

treatment individualized for each patient. The current available options include surgical

resection, salvage chemotherapy, and radiotherapy. Salvage chemotherapy includes

temozolomide rechallenge, bevacizumab, and other alkylating agents (i.e. carboplatin and

nitrosoureas)83. However, after TMZ and bevacizumab fail, prognosis is approximately 3-4

months. With no curable treatment being available for GBM, the field requires the development

of new therapies aimed at preventing chemoresistance and tumour recurrence.

  ID1  in  Normal  Biology    

3.1   The  ID  Family:  Structure  and  Function  Inhibitor of DNA binding 1 (ID1) is a dominant negative helix-loop-helix (HLH) protein that is

part of a family of highly conserved transcriptional regulators, composed of four known

members in vertebrates (ID1-4)84. The basic HLH (bHLH) transcription factors are a family of

proteins that control cell fate determination, differentiation, and cell proliferation85. Because ID

proteins lack a basic DNA binding domain, ID- bHLH heterodimers are unable to bind to the

DNA, blocking bHLH-directed transcription86. Therefore, ID proteins are also termed inhibitors

of differentiation. Moreover, early studies found ID proteins control differentiation of muscle86,

neurons87, mammary88, and B89 and T-cells90. Since then, further investigation into the function

of these proteins have demonstrated their important role in coordinating various cellular

processes including proliferation, cell-cycle regulation, angiogenesis, invasion and

migration91,92,93.

3.2   ID1  in  Cellular  Differentiation  Mutational studies of the Drosophilia emc locus provided the first direct evidence that ID

proteins may play a role in regulating cellular differentiation. Drosophilia emc is a Drosophilia

HLH protein ortholog of ID. Mutation of emc in Drosophilia via loss- and gain-of-function

studies revealed that emc inhibits the function of bHLH proteins known to be involved in

processes of cellular differentiation, such as neurogenesis and sex determination94.

Page 24: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

9

The role of ID proteins in cellular differentiation was then further studied in mammalian cell

culture systems84. These studies demonstrated that there was a correlation between

downregulation of ID expression and differentiation of various cell lineages. In vivo work using

targeted expression of ID genes to specific tissues and cell types in mice has demonstrated

inhibition of cellular differentiation in these systems95,96. For example, transduction of

retroviruses that express ID1 into embryonic mouse brains lead to significant impairment in

neuronal differentiation97. Furthermore, transgenic mice overexpressing ID1 in lymphoid

progenitors experienced cell-cycle arrest at an early developmental stage, the pro-B cell stage89.

3.3   ID1  in  the  Developmental  Process    ID proteins play a critical role throughout the developmental process. In situ analyses of ID gene

expression during mouse development from early gestation to birth, showed widespread

expression of ID proteins throughout the developing organism98. This study also highlighted that

ID1 and ID3 expression patterns were highly similar. Furthermore, several animal knockout

models have been generated for the purpose of identifying the role of ID genes in development.

A knockout animal model possessing a deletion of the ID1 gene did not show any major

abnormalities, therefore failing to produce an obvious phenotype99,90. Conversely, ID1/ ID3

double knock-out mice are embryonic lethal, and were found to possess aberrant angiogenesis

and neuronal differentiation87. Therefore, these two animal models taken together would suggest

redundant functions between ID1 and ID3 proteins.

3.4   ID1  in  Cell  Cycle  Regulation  and  Apoptosis    Several studies investigating the function of ID proteins have shown that they play a significant

role in regulating the cell cycle, more specifically, in promoting G1-S cell cycle transitions. A

study conducted by Hara and colleagues demonstrated that ID1 and ID2 levels are induced in

response to mitogenic signaling in fibroblasts100 (Figure 2). Conversely, when this response is

inhibited by antisense oligonucleotides directed against these IDs, the cells are no longer able to

enter the S phase101. ID1 has also been shown to inhibit E-protein and Ets-protein-mediated-

activation of p16, also known as cyclin-dependent kinase (CDK) inhibitor 2A102,103. An in vivo

study found that ID1 null mouse embryo fibroblasts have increased expression of p16, and these

mice experienced premature senescence103. Elevation of ID1 is associated with the

Page 25: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

10

Mitogenic)signal

bHLH

CANNTG

bHLHbHLH

bHLH

ID1

ID1

G1)arrestDifferentiation

p15.genep16.genep21.gene

bHLH

downregulation of the CDK2 inhibitor p27, which correlated to increased RB phosphorylation

and inactivation104.

ID genes have been implicated in promoting apoptosis. ID1 has been shown to induce apoptosis

in neonatal and adult cardiac myocytes via a redox-dependent mechanism105. In a transgenic

model, mice with targeted ID1 expression in T cells showed a 96% decrease in total number of

thymocytes, due to significant apoptosis106. Finally, ID1 expression has also been shown to

induce apoptosis in dense mammary epithelial cell cultures107.

Figure 2. The role of ID1 in cell cycle regulation. Mitogenic signaling induces ID1 gene expression. ID1 binds to bLH proteins, and because ID proteins lack a basic DNA binding domain, ID1-bHLH heterodimers are unable to bind to the DNA, blocking bHLH-directed transcription, for example, the bHLH-mediated activation of p15, p16, and p21 (Figure adapted from Sikder HA et al. Id proteins in cell growth and tumorigenesis. Cancer Cell 2003. 3: 525-530.)

3.5    ID1  in  Invasion,  Migration,  and  Angiogenesis    ID1 has been implicated in invasion and migration, as well as angiogenesis. ID1 overexpression

in mammary epithelial cells (SCp2) makes these cells partially refractory to differentiation

signals. This results in rapid proliferation, after an initial lag period, and these cells are then able

to invade the basement membrane. Furthermore, antisense targeting ID1 levels results in

decreased cell proliferation, increased differentiation88, and inhibits invasiveness108.

Studies investigating the effect of ID1 knockout mice on the ability to form new blood vessels

have shown that ID1 expression is required for normal embryogenesis87. Moreover, in addition

to the aforementioned ID1/ID3 double knockout mice being embryonic lethal, it has also been

Page 26: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

11

shown that before death all embryos suffered a hemorrhage in the forebrain87. Therefore, these

studies suggest that ID1 is involved in both invasion and angiogenesis.

   ID1  in  Cancer  Biology    

4.1   ID1  expression  in  Primary  Tumour  Cells    Increased expression of ID proteins has been reported in many types of human tumours and

associated with loss of differentiation, enhanced malignancy, and aggressive clinical

behavior109,110. For example, medullary thyroid cancers are associated with high ID1 expression

levels; these tumours exhibit characteristics of loss of cell-growth control, cell proliferation,

dedifferentiation, migration and invasion111. ID1 expression also correlates with loss of p16

expression in melanoma in situ112. Moreover, elevated ID1 expression was found around the

vasculature within metastatic melanoma lesions.

It has been hypothesized that ID proteins may play a role in the development of brain tumours, as

ID proteins are known to be involved in neurogenesis and neural differentiation. Studies have

found ID gene expression to be downregulated during astrocyte differentiation113. Furthermore,

ID1 expression has a positive correlation with tumour grade in astrocytomas114. Levels of ID1

protein expression correlate with less-differentiated phenotypes, high malignant potential, and

poor clinical outcome in cervical115, breast108 and prostate cancer116. Therefore, these studies

taken together would suggest that ID1 may serve as a potential diagnostic/prognostic marker, as

well as an attractive therapeutic target.

4.2   Multimodal  Activation  of  ID1  in  Cancer    ID protein expression is high in stem and progenitor cells, and then typically downregulated

during differentiation. However, ID expression is known to be re-activated in cancer cells,

evidenced by many tumour types displaying high expression levels of ID proteins. Consequently,

there are a number of contributing factors implicated in the activation of ID1 in cancer, including

mediation via oncogenic pathways and the deregulation of protein stability.

ID genes serve as targets of upstream oncogenic events. Aberrantly high levels of ID protein

expression in cancer are often a consequence of transcriptional induction by oncoproteins such as

MYC, RAS, SRC, and by growth factor-directed signals such as transforming growth factor-β

Page 27: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

12

(TGF-β) and bone morphogenetic proteins (BMPs)117,118,119,120. For example, activation of RAS

signaling results in increased ID1118 and ID3121 expression via the transcriptional activation of

early growth response 1 (EGR1). ID1 and ID3 also serve as downstream targets of MYC, and

induction of ID1 by MYC appears to be required for breast cancer cells to enter S phase in

vitro122. However, in many instances, the mechanisms of ID gene activation are unclear. The

combination of multiple genetic and epigenetic events is required to establish aberrant levels of

ID proteins necessary for tumour progression and maintenance. For example, in non-small-cell

lung cancer, ID1 is induced by a number of signaling pathways123,124, which as a result promote

cell proliferation, epithelial-to-mesenchymal transition (EMT), metastasis, and chemoresistance

in vitro and in xenograft models125.

ID proteins are short-lived proteins with a half-life of approximately 10-20 minutes, and are

targeted for degradation by a variety of E3 ubiquitin ligases in the ubiquitin-proteasome

pathway126. For example, the E3 ubiquitin ligase, SMAD ubiquitylation regulatory factor 2

(SMURF2), targets ID1 and ID3 for degradation during cellular senescence127 (Figure 3).

Perturbation of the ubiquitin-proteasome pathway is associated with the accumulation of ID

proteins in cancer. This can be achieved by deubiquitylases (DUBs), which oppose ubiquitin

ligase activity by removing ubiquitin moieties, thus increasing substrate stability. Therefore, both

E3 ligases and DUBs are implicated in the regulation of ID protein accumulation. Another

mechanism of ID protein stabilization involves the alteration of the ubiquitin-specific protease 1

(USP1), a specific DUB for ID1, ID2, and ID3 in osteosarcoma cells and mesenchymal stem

cells. USP1 overexpression in osteosarcoma cell lines sustains the accumulation of ID proteins,

which are responsible for conferring the stem cell-like properties in these cells128.

Page 28: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

13

Stem%cells%orCancer%cells

Deubiquitylases(USP1) ID1

Ub

Differentiationor%senescence

E3%ubiquitin%ligases

(SMURF2)ID1

Ub

ID1$protein$stabilization

ID1$protein$degradation

Figure 3. ID1 protein stabilization and degradation. The E3 ubiquitin ligase, SMURF2, ubiquitinates ID1 during cellular senescence, targeting the protein for degradation. Deubiquitylases, such as USP1, can perturb the ubiquitin-proteasome pathway by removing the ubiquitin moieties on ID1, resulting in ID1 protein stabilization.

4.3   ID1  in  Tumorigenesis    ID1 was first linked to tumorigenesis in a 1999 study that showed constitutive expression of ID1

in keratinocytes ultimately lead to cell immortalization129. This was due to several key changes

in other cellular processes, including the induction of cell proliferation, inhibition of cellular

senescence and differentiation, and extension of life span. Numerous studies have since been

undertaken to elucidate the role of ID1 in both oncogenesis and tumorigenesis.

As previously mentioned, overexpression of ID1 is partly mediated by bona-fide oncogenes,

which in turn can activate or inhibit a number of key oncogenic signaling pathways. For

example, overexpression of ID1 in prostate cancer cells was found to induce serum-independent

cell growth through inactivation of the p16/RB tumour suppressor pathway130. A study

investigating colon tumours found that gain-of-function mutations in TP53, which are potentially

pro-tumorigenic, leads to protein stabilization and this is associated with ID1 overexpression131.

ID1 gene expression has been largely implicated in tumour development. In a transgenic mouse

model, targeted ID1 expression to thymocytes lead to the development of T cell lymphomas,

following aberrant T cell development and massive apoptosis106. Moreover, tumour development

also occurred when ID1 expression was targeted to B-lymphocytes and intestinal epithelia89. ID

proteins also inhibit programmed cell death and promote tumour cell survival, further

contributing to tumour progression. High ID1 levels are associated with upregulation of anti-

Page 29: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

14

apoptotic and pro-survival factors (i.e. NF-kB)132,133 and inhibition of pro-apoptotic signals (i.e.

p21)134. Furthermore, studies have shown that silencing ID proteins in cancer cells can induce

programmed cell death135,136.

4.4    ID1  in  Tumour  Invasion  and  Metastasis    ID1 expression correlates with disease progression in a number of cancers. A major component

of tumour progression involves invasion of cancer cells into the adjacent or distal regions,

resulting in tumour metastasis. One potential mechanism by which IDs influence metastatic

ability occurs at the transcriptional level, whereby it is hypothesized that increased ID1

expression increases migratory features by inhibiting bHLH-mediated transcription of anti-

metastatic genes such as semaphorin 3F (SEMA3F)137.

ID proteins are involved in metastatic colonization by inducing EMT at the primary site. Matrix

metalloproteinases (MMPs) are a protein family that are heavily involved in the EMT process via

regulating the degradation of the basement membrane and remodeling of the extracellular

matrix138. During cancer progression, MMPs are found to be overexpressed in cancer cells,

including MMP2, MMP9, and MMP14139. Moreover, overexpression of both ID1 and MMP

directly correlate with invasiveness in breast cancer cells140. In vivo studies revealed that high

levels of ID1 expression are associated with disease progression and tumour invasion in

endometrial141 and breast142 carcinomas. Conversely, downregulation of ID1 using siRNA

treatment results in decreased invasion and metastatic ability of breast cancer cells108.

Furthermore, a study investigating global expression analysis in human breast cancer identified

ID1 as a key component of its metastatic signature143. ID1 is also implicated in invasion and

metastasis in lung carcinoma. In an in vivo study, ID1 +/- mice implanted with Lewis lung

carcinoma cells showed an increase in survival (approximately 2-fold) and displayed

significantly fewer metastasis87.

4.5   ID1  in  Tumour  Angiogenesis  Increased expression of ID proteins in cancer cells has been implicated in the promotion of

tumour angiogenesis. ID proteins induce expression of pro-angiogenic factors144 that increase

endothelial cell proliferation and migration145,146. The involvement of ID proteins in tumour

angiogenesis was demonstrated in tumour models derived from ID1-ID3 double null mice, which

Page 30: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

15

displayed significant loss of tumour vasculature87,147,144. This finding suggests that ID1/ID3

inactivation in tumour endothelium may have a potential therapeutic benefit.

Moreover, ID1 in combination with ID3 have been shown to be essential during cancer

progression for tumour-associated angiogenesis. A study found that tumour xenografts with an

ID1/ID3 knockout show decreased tumour growth, loss of metastasis, and impaired

neovasculature of the tumour87. Furthermore, ectopic expression of ID1 resulted in significant

increase in vascular endothelial growth factor (VEGF) secretion, a downstream target of the ID1

protein, in prostate cancer cells148. This lead to the promotion of the growth and tube formation

of endothelial cells. Conversely, inactivation of ID1 via siRNA resulted in decrease in VEGF

gene transactivation and protein secretion148.

4.6   The  Role  of  ID1  in  Cancer  Stem  Cells    Cancer stem cells are identified by their ability to proliferate and self-renew, they are

multipotent, and have the capacity to initiate tumours. The function of ID1 in CSCs is

extensively studied in malignant glioma. In an orthotopic model of brain cancer, conditional

deletion of ID1, ID2, and ID3 in tumour cells decreased the glioma stem cell population, blocked

tumour growth, and extended overall survival in mice149. Furthermore, the TGF- β signaling

pathway is known to activate ID1 and ID3 proteins in glioma stem cells150. A study found that

inhibiting the TGF- β receptor and silencing ID1/ID3 results in abrogation of the glioma stem

cell population both in vitro and in vivo150. Finally, a somewhat controversial study using

experimental mouse models of high-grade glioma showed that cells with high ID1 expression

had high self-renewal capacity in vitro and tumorigenic ability in vivo. However, cells with low

ID1 expression had low self-renewal potential in vitro, but demonstrated even more robust

tumorigenicity151. These results can potentially be explained by the existence of a cell population

that is the counterpart to CSCs, which are known as transit-amplifying cells. Therefore, it has

been proposed that targeting both cell populations will be critical to delivering effective

treatment to GBM patients.

4.7   ID1  in  Contributing  to  Treatment  Failure    ID1 has also been implicated in treatment failure and tumour recurrence in various types of

cancer. The progression of breast and prostate cancer typically involves the development of

Page 31: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

16

hormone refractory disease, which is the main reason for treatment failure. A study shows that

transfection of ID1 into androgen-dependent prostate cancer cells resulted in an androgen-

independent phenotype, whereby these transfected cells showed a decrease in responsiveness to

androgen stimulation152. These cells also mimicked androgen-independent prostate cancer cells

found in recurrent prostate cancer patients, following hormonal ablation therapy152. Furthermore,

in a study investigating clinical patient tumour samples, ID1 was found to be highly upregulated

in tumours from recurrent prostate cancer patients compared to metastatic prostate cancer

patients153. Therefore, these studies would suggest that ID1 may play a role in treatment failure

and tumour recurrence.

4.8   ID1  in  Glioblastoma    ID1 has been identified as a key player in modulating glioma cell invasiveness. A study found

that ID1 protein expression correlated with the magnitude of glioma cell invasion; specifically,

the high ID1-expressing U251 GBM cell line showed a substantial increase in cell invasion

compared to cell lines with low ID1 expression154. Furthermore, ID1 knockdown resulted in a

dramatic reduction in glioblastoma cell invasion, which corresponded to a decrease in the key

EMT regulator Snail, as well as active forms of MT1-MMP154. Finally, in an orthotopic model of

human GBM, ID1 genetic knockdown resulted in a significant increase in overall survival,

demonstrating a median increase in survival of 20 days154.

ID1 has also been implicated in the GBM stem cell population. ID1 is enriched in glioma stem

cell fractions (CD133+), and co-localizes with the stemness marker, SOX2, in primary GBM-

derived cultures154. Furthermore, ID1 knockdown resulted in inhibition in self-renewal potential,

a significant reduction in neurosphere growth, and decreased expression of glioma stem cell

markers154. In another study, ID1 and ID3 were identified as the key gene responses to TGF- β

inhibitors, known to target the glioma-initiating cell (GIC) population150. Inhibition of the TGF-

β pathway decreases the ID1high/CD44high GIC population by repressing ID1/ID3 levels, resulting

in inhibition of tumour initiation150. Taken together, these results suggest that ID1 expression

promotes a stem-like phenotype in human GBM, and serves as an attractive therapeutic target.

As previously mentioned, USP1 is a ubiquitination-specific protease involved in ID1 protein

stability. A study revealed that USP1 is highly expressed in GBM, and also promotes ID1

stability in glioblastoma cells155. USP1 inhibition resulted in decreased survival and clonogenic

Page 32: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

17

growth in the glioma stem cell population155. Furthermore, targeting of USP1 either via shRNA-

mediated knockdown or its specific chemical inhibitor pimozide enhanced radiosensitivity of

GBM cells, and significantly prolonged survival of tumour-bearing mice155.

  ID1  as  a  Therapeutic  Target  

5.1   IDs  in  Cancer  Therapy    ID proteins are attractive targets for cancer therapy due to a number of reasons. Firstly, ID

proteins mediate the activity of many important genes that are involved in regulating

tumorigenesis and cancer progression. Secondly, ID proteins are not expressed in most mature

adult tissues, however, they are reactivated in cancer cells; this confers an advantage for systemic

therapy by achieving selective targeting and less toxicity. Finally, only a partial reduction of ID

levels is sufficient to significantly reduce tumour invasion and metastasis.

The therapeutic benefit of targeting ID proteins has previously been demonstrated in various

tumour cell types. For example, ID1/ID3 double knockout mice show defective endothelial-cell

vasculature in tumour xenografts87, and could not support the growth and metastasis of three

different tumours. Moreover, studies have found that a decrease in ID protein levels via siRNA

treatment leads to decreased tumour aggressiveness, invasion, and metastasis110,156.

Current therapeutic approaches to target IDs can be grouped into two main categories. The first

approach is aimed at extinguishing ID gene expression by delivering an ID-specific siRNA

molecule in vivo. For example, in a mouse model, an si-ID1 was fused to a peptide (ID1-PCAO)

known to localize specifically to tumour neovasculature157. Systemic delivery of ID1-PCAO in

breast cancer and lung carcinoma mouse models resulted in increased intra-tumour hemorrhage,

hypoxia, and inhibited tumour growth and metastasis158. Importantly, these studies have found

that ID inhibition does not result in deleterious general or organ-specific effects. The second

common approach is aimed at targeting protein-protein interaction properties of ID proteins via

specific peptides. For example, ID-binding peptides were designed according to the amino acid

sequence of myoblast determination protein 1 (MYOD1), a bHLH protein159. These peptides

possess a high affinity for ID1, thus interfering with the interaction between ID1 and MYOD1,

causing a proliferative block in cancer cells159.

Page 33: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

18

5.2    ID1:  Potential  Therapeutic  Target  in  Combination  with  Chemotherapy  

Chemotherapy is a widely used treatment against metastatic cancer, as it is capable of inducing

cellular apoptosis. Unfortunately, many cancer cell types are capable of developing drug

resistance, which ultimately results in disease recurrence. Recurrent tumours are usually highly

resistant to further chemotherapy treatments and are associated with a more aggressive

phenotype. Therefore, we must elucidate the mechanisms that underlie drug resistance in order to

improve the efficacy of chemotherapy.

Several studies have demonstrated the potential role of ID1 in chemotherapeutic resistance.

Ectopic expression of ID1 in both prostate160 and nasopharyngeal carcinoma (NPC)161 cells was

able to confer resistance to the chemotherapeutic agent, Taxol. In NPC cells, ID1 expression was

found to be associated with decreased sensitivity to Taxol-induced apoptosis161. Furthermore, in

prostate cancer cells, ID1 downregulation via siRNA treatment sensitized the cells to Taxol160.

Finally, a study that found combined expression of ID1 and ID3 increases both self-renewal and

tumour initiation in colon cancer stem cells, also revealed that silencing ID1/ID3 sensitized those

cells to the chemotherapeutic agent, oxilaplatin66. Therefore, although the role of ID1 in

therapeutic resistance in glioblastoma has yet to be explored, these studies would suggest that

ID1 may serve as a potential therapeutic target in combination with chemotherapy treatment in

GBM patients.

  Hypothesis    There is an unmet need for better therapeutic targets in glioblastoma due to its heterogeneous

clinical behavior and limited treatment strategies. While multi-modality treatment results in

immediate disease control, tumour recurrence is invariable and most patients succumb to their

disease within one year. Chemotherapeutic resistance is a fundamental cause of tumor recurrence

in GBM. ID1 has been shown to be involved in chemotherapeutic resistance in other cancers,

and ID1 ablation has resulted in enhanced chemosensitivity and prolonged survival in an in vivo

model. It is my hypothesis that ID1 expression mediates glioblastoma chemoresistance and

identifies the cell population that drives tumour recurrence; and is a potential therapeutic target

to enhance the chemotherapeutic effect in GBM.

Page 34: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

19

Aim I: Investigate the interplay between ID1 expression and response to TMZ in glioblastoma

Aim II: Investigate the effect of ID1 knockout on GBM biology and overall survival in a

xenograft model

Aim III: Determine whether ID1 inhibition enhances the response to TMZ in a mouse xenograft

model of GBM

Page 35: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

20

Chapter  2      

Methods    

  Methods  

7.1   Cell  Culture  Immortalized glioblastoma cell lines (U87, U118, U251, U373, and A172) from American Type

Culture Collection (ATCC), and primary glioblastoma cancer cell lines (GliNS1 and H818) were

used to study the effects of temozolomide and ID1 inhibition in vitro. Immortalized lines were

grown in Dulbecco’s modified eagle media (DMEM) (Wisent) supplemented with 10% FBS, and

1% penicillin/streptomycin (Wisent). GliNS1 and H818 were grown as adherent cells and

spheres, respectively, in Neurocult NS-A Basal Medium (Human Stem Cell Technologies),

supplemented with L-glutamine (2mM) (Invitrogen), antibiotic/antimycotic (1X) (Invitrogen),

N2 supplement (1%) (Gibco), B27 supplement (2%) (Gibco), 75ng/mL Bovine Serum Albumin

(BSA) (Sigma), human epidermal growth factor (hEGF) (20ng/ml) (Sigma) and human basic

fibroblast growth factor (hFGF) (20ng/ml) (Sigma). All cell lines were incubated at 37°C and 5%

CO2.

7.2   Clinical  Chemotherapeutic  Agents  Temozolomide (Sigma) and Cisplatin (Sigma) were reconstituted in dimethyl sulfoxide (DMSO)

prior to treatment of cells at designated concentrations.

7.3   Immunocytochemistry    Briefly, 1x105 GliNS1 cells were seeded on poly-L-ornithine (Sigma)/laminin (Sigma) coated

plates to aid in attachment. The following day cells were treated with either vehicle or incubated

with TMZ (25 or 100 uM) for a 7-day period, at which point the viable cells were cytospun onto

slides. Cells were then fixed with 4% paraformaldehyde, permeated with 0.5% Triton-X, and

blocked with 5% BSA at 37°C for 10 minutes. Slides were then incubated with primary antibody

overnight at 4°C. Primary antibody concentration was as follows: ID1 (1:500, SantaCruz). After

incubation, slides were washed with PBS and incubated with a fluorescent conjugated antibody

specific for the primary antibody for 1h at room temperature in a dark environment (Alexa 594

Page 36: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

21

conjugated antibody 1:500; Life Technologies). Slides were mounted on DAPI containing

VECTASHIELD Mounting Medium (Vectorlabs, H-1200). Images were captured using a

spinning disk confocal microscope.

7.4   Alamar  Blue    The alamarBlue Assay (ThermoFisher) was used to evaluate the effect of temozolomide on cell

viability by incorporating a fluorometric/colorimetric growth indicator based on detection of

metabolic activity. Specifically, this assay was used to determine the half maximal inhibitory

concentration (IC50) in various GBM cell lines. Briefly, in a 96-well format, 2000 cells/well

were seeded in 5x replicates of U87, U118, U251, U373, and A172. Cells were allowed to

adhere overnight. The following morning, graded concentrations of TMZ ranging from 0 to

1600uM were added to the wells. On the seventh day, alamarBlue reagent was added (20ul,

amount equal to 10% of the culture volume), and re-incubated at 37°C, 5% CO2 for four hours.

Fluorescence was then measured with excitation wavelength at 560nm and emission wavelength

at 590 nm. IC50 plots were then generated using the corresponding values.

7.5   Western  Blotting    Cells were lysed with standard cell lysis buffer (Cell Signaling) containing protease inhibitors

(Roche). Protein concentration was determined using the bicinechoninic acid (BCA) assay

(Pierce Chemical Co.). Lysates containing 50 ug total protein were loaded onto 12% SDS-PAGE

gels and electrophoresed. Proteins were then transferred onto PVDF membranes (Bio-Rad) using

a semidry transfer apparatus (Bio-Rad). Membranes were probed for varying proteins overnight

in 5% milk. Antibodies were as follows: beta-actin (1:10,000; Cell Signaling), ID1 (1:200;

SantaCruz). After incubation, membranes were washed 3 times for 10 minutes with PBS with

0.1% Tween and incubated with horseradish peroxidase–conjugated antibodies specific for the

primary antibody (Cell Signaling). Binding was detected using Chemiluminescence Reagent Plus

(PerkinElmer).

7.6   Cell  Viability  Counter    To evaluate the effect of temozolomide alone or in combination with ID1 inhibitor on cell

viability, direct cells viability measurements (Vi Cell XR cell counter, Beckman Coulter) were

taken. Briefly, 1x105 U251 cells were seeded in 6-well dishes in triplicate, treated with indicated

Page 37: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

22

concentration of temozolomide alone or in combination with ID1 inhibitor, and counted on day

three or four post-treatment.

7.7   ID1  Silencing    ID1 Smartpool siRNA (Dharmacon) and scrambled RNA control (Dharmacon) were

reconstituted according to manufacturer’s instructions. 100nM of scrambled and ID1 siRNA

were used to treat U251 cells. Cells were incubated with siRNA and scrambled control for a

period of four days, at which point the cells were collected for cell viability and Western blot

analysis.

7.8   Pimozide  (Clinical  ID1  Inhibitor)      Pimozide (Sigma), a chemical ID1 inhibitor, was reconstituted according to manufacturer’s

instructions. 5uM of pimozide was used to treat U251 cells. Cells were incubated with pimozide

for a period of three days, at which point the cells were collected for cell viability and Western

blot analysis.

7.9   Immunohistochemical  Staining  and  Scoring        Paraffin-embedded blocks were cut into 5-µm sections and were dewaxed in xylene, followed by

rehydration in a standard alcohol series (90, 70, 50%). Antigen retrieval was achieved by 20

minutes of pressure cooking in citrate buffer (pH 6.0), followed by blocking for ten minutes

using Universal Blocking Buffer (Dako). The slides were then incubated with ID1 primary

antibody (1:500 dilution, SantaCruz) overnight at 4°C. Detection used biotinylated secondary

antibodies for 1 hr, the ABC reagent kit (Vector Labs), and DAB chromagen (Vector Labs).

Slides were washed with PBS three times after each step. Sections were then dehydrated using

increasing concentrations of ethanol (50, 70, 90%), followed by a brief washing in xylene.

Finally, the slides were mounted in Permount (Fisher Scientific Inc.). Immunoexpression of ID1

protein was quantified with 3d Histech Panoramic Image Analysis Software, which uses a

colorimetric algorithm to calculate the percentage of positive pixels over a designated tissue area,

defined as relative mask area (rMA).

Page 38: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

23

7.10  CRISPR/Cas9  System          To establish an ID1-knockout (ID1-KO) cell line, U251 cells were transfected via

electroporation with plasmid containing sgRNA targeted at ID1 packaged into lentiviral vector

(Sigma). Two plasmids containing sgRNA targeted at ID1 with different seed sequence were

used. The two sgRNA sequences were: ACAGCGAGCGGTGCGGGCGAGG and

AGGCCGGCAAGACAGCGAGCGG. The lentivector used was a pLV-U6g-EPCG vector,

containing the 22 base-pair sequence and cas9 endonuclease with a puromycin and GFP element.

The cells that integrated ID1-sgRNA were selected using puromycin selection. After five weeks

of selection, cells were isolated using single cells FACS in a 96-well plate. The individual

colonies were expanded to establish ID1 knockout cell lines.

7.11  Colony  Forming  Assay          Briefly, 30,000 cells were seeded in 6-well dishes in triplicate, and treated with temozolomide.

These cells were treated for one week, at which point the media was changed, effectively

removing TMZ. The cells were allowed to continue to grow for one week in normal culture

media, at which point the media was removed, cells were washed with PBS, fixed with 4%

Paraformaldehyde (Electron Microscopy Sciences), and stained with crystal violet solution for

one hour. Cells were washed and colonies were counted.

7.12  Evaluation  of  ID1  Knockout  in  an  In  Vivo  Model  of  Glioblastoma  

Stereotactic-guided intracranial implantation in NOD/SCID/Gamma (NSG) immunodeficient

mice were performed by injecting 2.5x105 U251 control or U251 ID1-KO cells into the frontal

cortex (Coordinates: X=-1.0, Y=1.5, Z=2.4, with Bregma serving as reference point). Both cell

lines are luciferase-tagged allowing the use of bioluminescence imaging to monitor tumour

growth. Mice were sacrificed when they reached a humane endpoint (exemplar indicators

include rapid body weight loss and hunched or abnormal posture). Animal use protocol (AUP-

0293) was approved by the Toronto Centre for Phenogenomics (TCP) animal care committee

(ACC).

Page 39: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

24

7.13  Evaluation  of  ID1  Inhibition  in  Combination  With  Temozolomide  Chemotherapy  in  an  In  Vivo  Model  of  Glioblastoma  

Drugs were administered by IP injections into NSG immunodeficient mice implanted with

2.5x105 U251-luciferase cells. Experimental cohorts included Saline (vehicle), pimozide (15

mg/kg) daily for three 5-day cycles, temozolomide (40 mg/kg) once a day for two 5-day cycles,

or pimozide alone once a day for one 5-day cycle followed by a combination of daily pimozide

and temozolomide for two 5-day cycles. The mice were sacrificed as previously described, using

the same aforementioned AUP approved by TCP.

7.14  Bioluminescence  Imaging  Briefly, mice were put under anesthesia at 2% Isoflurane levels and injected with 0.15mL of

Luciferin (15mg/ml) via IP injection. After the mouse had been injected with Luciferin for 3

minutes, it was scanned to assess highest total flux; this is done repeatedly until there is no

longer an increase in signal. Living Image 4.3.1 software was used to analyze the scans and

quantify total flux. The color scale was adjusted to be the same for each mouse so that

comparisons could be made between experimental groups.

7.15  Quantitative  Real-­time  Polymerase  Chain  Reaction  Briefly, RNA was extracted from GBM cells post-TMZ treatment, and its complementary DNA

was synthesized according to manufacturer’s instructions (Invitrogen). Real-time PCR was

performed using Taqman probes (Aplplied Biosystems), according to the manufacturer’s

recommendations. Reactions were carried out using StepOnePlus Real-Time PCR System

(Applied Biosystems) and results were expressed as fold change calculated by the ΔΔ Ct method

relative to the control sample. GAPDH was used as an internal normalization control.

7.16  Polymerase  Chain  Reaction  Amplification  PCR reaction for sequencing was conducted using the following primers:

chr7:+108096350: Forward: 5’-AGCACAGGTGTACGCACTTC-3’

Reverse: 5’-GTGACACTGGGTCCTGCTC-3’

Page 40: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

25

chr1:+236445511: Forward: 5’-TGTGGGAGTTGTGGTCCTG-3’

Reverse: 5’-GCCGAAGCCATCTCTACAAG-3’

chr2:-40006264: Forward: 5’-AGGCTCTCCGTCTCTCTCAC-3’

Reverse: 5’-GAAAGGAATGGTGGCTTCTC-3’

PCR amplification step was performed in a 20ul reaction volume and consisting of 11.35ul

nuclease free water, 2ul forward primer, 2ul reverse primer, 0.25ul template, 0.4ul HotStar

HiFidelity DNA Polymerase (Qiagen), and 4ul 5x PCR Buffer (Qiagen). Mixture solution was

amplified by PCR machine (Bio-Rad). Thermal cycle programmed for 5 minutes at 95°C as

initial denaturation, followed by 35 cycles of 15 sec at 94°C for denaturation, 1 min at 51-58°C

as annealing, 1 min at 72°C for extension, final extension at 72 °C for 10 min, and indefinitely at

4°C at end of PCR cycling. PCR products were examined by electrophoresis at 100 V for 30

minutes in a 1% (w/v) agarose gel in 1 x TAE buffer. The marker used DNA ladder 1 kb.

Electrophoresis gel was than visualized in UV light.

7.17  Statistical  Analysis    All experiments were done in triplicate. Mean and SDV were used where appropriate. Two

tailed T-tests and Linear regression analyses were used. Statistics were completed with

GraphPad Prism 6.0. *, **, *** denotes significance of p<0.05, p<0.01, p<0.001. Error bars

represent SDV or SEM.

Page 41: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

26

Chapter  3      

Results  

  Results  

8.1   ID1  Expression  Correlates  with  Response  to  Temozolomide  Treatment  in  Glioblastoma    

ID1 has previously been shown to be involved in chemotherapeutic resistance in other types of

cancers. We hypothesized that there is a relationship between ID1 expression and response to

TMZ-chemotherapy in glioblastoma. To test this hypothesis, first we evaluated ID1 protein

expression in primary and recurrent GBM patient tumour samples using a tissue microarray

(TMA), which was constructed from formalin-fixed-paraffin-embedded GBM samples obtained

from St. Michael’s Hospital Pathology Department. Criteria for TMA inclusion were: (1) patient

received TMZ-chemotherapy after primary tumour resection and (2) patient experienced tumour

recurrence. Immunohistochemical analysis demonstrated that there was a significant negative

correlation (r = -0.6779; p<0.05) between fold-change in ID1 expression from the primary to

recurrent tumour and latency to recurrence (Figure 4A), whereby patients with increased ID1

expression following chemotherapy had a shorter latency to recurrence (Figure 4B). These data

suggest that ID1 expression negatively correlates with progression-free survival in glioblastoma.

To test this correlation, we sought to determine if ID1 expression is proportional to TMZ

sensitivity in GBM cells in vitro. Drug response curves to TMZ were determined based on IC50

(the half maximal inhibitory concentration) using 5 immortalized GBM cell lines (U251, U87,

U118, U373, A172) with differential ID1 expression. We found that ID1 expression correlates

with TMZ resistancy in GBM cells. For example, U118 is a low ID1-expressing cell line and

highly sensitive to TMZ (IC50 value= 501uM); whereas, U373 is a high ID1-expressing cell line

and highly resistant to TMZ (IC50 value= 1959uM) (Figure 5A). IC50 curves for U118 and

U373 further highlight the significant difference in TMZ resistancy between the two cell lines

(Figure 5B), along with their corresponding microscopic images (Figure 5C).

Page 42: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

27

Primary'Tumour Recurrent'Tumour

Patie

nt'3

Figure 4. Increased ID1 expression following chemotherapy correlates with decreased progression-free survival in glioblastoma human patient tumour samples. (A) Immunohistochemistry was used to measured ID1 expression in primary and recurrent patient tissue specimens (n=9); staining was quantified and the fold-change in ID1 expression was calculated. Increased ID1 expression post-chemotherapy correlates with a quicker recurrence. (B) An example of immunohistochemical staining of a patient tissue specimen who experienced short progression-free survival (187 days) showing increased ID1 expression in recurrent GBM tumour compared to the primary sample. * Denotes significance of p<0.05.

B

A

!1

0

1

2

3

4

5

6

7

8

0 100 200 300 400 500Fold/ch

ange/in/ID

1/expressio

n/fro

m/prim

ary/

to/recurrent/tum

our/(rM

A)

Progression!free/ Survival/(#of/days)

r/=/!0.6779*

Page 43: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

28

ID1

0

500

1000

1500

2000

2500

U87 U118 U251 U373 A172

IC50-value-(u

M)

GBM-Cell-line

β;actin

U118$(7'day$TMZ$treatment)

U373$(7'day$TMZ$treatment)

CTL TMZ$100uM

CTL TMZ$100uM

Figure 5. ID1 expression correlates with resistance to TMZ in vitro. (A) Drug response curves to TMZ were determined based on the half maximal inhibitory concentration in 5 GBM cell lines with differential ID1 expression (U87, U118, U251, U373, A172). Baseline ID1 expression correlates with TMZ resistancy in GBM cells. (B) Exemplar IC50 curves (logarithmic) comparing a low-ID1 expresser, TMZ-sensitive cell line (U118) and high-ID1 expresser, TMZ-resistant cell line (U373); (C) corresponding microscopic images depicting the differential effect of 100uM of TMZ (7-day treatment) on U118 and U373. Images taken at 10X magnification at day 7 post-TMZ treatment.

0

0.2

0.4

0.6

0.8

1

1.2

0 50 200

600

1000

1400

Fold  change  in  Cell  V

iability

TMZ  Treatment  (uM)

Log.    (U118)

Log.    (U373)

A

B C

Page 44: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

29

8.2   ID1  Expression  Increases  Following  Temozolomide  Treatment  

Having previously established a correlation between ID1 expression and TMZ resistancy, we

next wanted to evaluate the effect of TMZ-chemotherapy on ID1 expression in vitro. We

hypothesized that TMZ has an effect of ID1 expression. To test this hypothesis, the primary

GBM cancer cell line GliNS1 was treated with graded concentrations of TMZ (0, 25, 100 uM)

for 7 days, at which point the viable cells were cytospun onto slides and immunocytochemistry

(ICC) was performed, staining for ID1 expression. It is important to emphasize that the viable

cells in the TMZ-treated groups are those that survived TMZ chemotherapy, therefore

representing the chemoresistant population. The control group (DMSO) exhibited very few cells

expressing ID1, with relatively weak intensity; conversely, in the TMZ-treated groups, the

majority of cells expressed ID1, with increased intensity (Figure 6A). The results from this

initial experiment would suggest that TMZ is either inducing ID1 expression in GBM cells or

selecting for the ID1-expressing cells.

We wanted to further study the effect of TMZ on ID1 expression in GBM cells by characterizing

the protein levels at various time points. Western blot analysis was used to measure ID1 protein

expression at 1, 3, 5, or 7 days post-TMZ treatment (100uM) in U251 and H818. ID1 protein

expression increased following TMZ treatment, peaking ~day 3 and remaining elevated to day 7

(Figure 6B). These results support the previous findings that TMZ exposure results in increased

ID1 expression in GBM cells. To determine if elevation in ID1 expression was due to increased

ID1 gene transcription, we performed comparative QT-PCR to measure ID1 mRNA expression

at 1, 3, 5, or 7 days post-TMZ treatment (100uM) in U251. We found that ID1 expression was

variable over the time course and did not remain stably increased up until day 7 (Figure 6C).

Therefore, we did not observe the same trend at the mRNA level as we did at the protein level.

All these findings taken together would suggest that TMZ is not inducing ID1 expression at the

genetic level, but rather selecting for the ID1-expressing cells.

 

Page 45: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

30

0

0.5

1

1.5

2

2.5

CTL 1D 3D 5D 7D

Fold0change0in0ID10mRNA0

expression

#days0post0TMZFtreatment

CTL$$$$$$1D$$$$$$$$3D$$$$$$$$5D$$$$$$$$$7D

#days$post$TMZ4treatment

β4actin

ID1

β4actin

ID1H818

U251

 

 

 

 

 

 

 

 

Figure 6. ID1 protein expression increased following TMZ-treatment in GBM cells. (A) Immunocytochemistry analysis of the effect of TMZ on ID1 expression in GliNS1. ID1 expression is represented in red, and DAPI is represented in blue. TMZ exposure (25 or 100 uM) results in increased ID1 expression. (B) Western blot (U251 and H818) and (C) comparative QT-PCR (U251) analysis of the effect of TMZ (100uM) on ID1 expression at various time points (1, 3, 5, 7 days post-TMZ treatment). ID1 protein expression increased post-TMZ treatment, peaking ~Day 3 and remaining increased up until Day 7; however, we did not observe the same effect at the mRNA level. Scale bar represents 100um. Error bars represent the ±SD of triplicate measurements and significance was determined by comparing to untreated cells using t-test.

A

C B

Page 46: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

31

GliNS1

ID1

β*actin

Ctl 10µM 25µM 25µM 100µM

Cisplatin7(48h) TMZ7(72h)

U251

ID1

β*actin

Additionally, since it is known that TMZ is not the only chemotherapeutic agent used today, we

asked whether increased ID1 expression is a TMZ-specific response? Cisplatin is a

chemotherapy drug that induces programmed cell death by causing the DNA strands to cross-

link. TMZ employs a different mechanism to induce apoptosis, therefore Cisplatin was selected

for comparison purposes in the following study. U251 and GliNS1 cells were treated with either

Cisplatin or TMZ for 48 or 72 hours, respectively. Western blot analysis demonstrated that

Cisplatin treatment does not result in increased ID1 protein expression in GBM cells, unlike

TMZ treatment (Figure 7). Therefore, these results suggest that the increase in ID1 expression

observed post-treatment is TMZ-specific.

 

Figure 7. Increased ID1 expression is post-treatment is TMZ-specific. Western blot analysis of the effect of TMZ (25 or 100 uM) compared to Cisplatin (10 or 25 uM) on ID1 expression in U251 and GliNS1. Cisplatin treatment does not result in increased ID1 protein expression in GBM cells, unlike TMZ treatment.

Page 47: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

32

8.3   ID1  Inhibition  Sensitizes  GBM  Cells  to  TMZ-­Chemotherapy  Treatment  

Our previous results demonstrate an interplay between ID1 expression and response to TMZ-

treatment. We next wanted to determine whether ID1 inhibition could enhance the cytotoxic

effect of TMZ-chemotherapy. To address this question, we employed a knockdown study using a

pooled siRNA targeting ID1 in U251 GBM cells. The U251 cell line was selected because it was

previously identified as a high basal ID1-expressing, resistant cell line. U251 cells were first

treated with graded concentrations of si-ID1 (25, 50, 75, 100 nM) for optimization purposes. The

optimal concentration of si-ID1 was determined to be 100nM, as it achieved the greatest

knockdown of ID1 protein expression and was not cytotoxic to the cells (Figure 8A). U251 cells

were then either pre-treated with si-ID1 (100nM) followed by TMZ treatment (25, 50, 100 uM)

or treated with TMZ alone for 4 days. We observed a significant reduction in cell density in the

combined si-ID1 and TMZ-treatment group compared to the TMZ-alone condition (Figure 8B).

Furthermore, we found a decrease in the number of viable cells in U251 cells pre-treated with si-

ID1 followed by TMZ treatment compared to TMZ treatment alone (p<0.0001); western blot

confirms ID1 knockdown in si-ID1 treatment groups (Figure 8C). Moreover, there was no

significant difference in cell viability between the U251 control and si-ID1 (100nM) conditions,

confirming that the siRNA-mediated ID1-knockdown alone does not have an effect on cell

viability. These results demonstrate that ID1 inhibition increases GBM cell sensitivity to TMZ-

chemotherapy treatment.

Page 48: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

33

TreatmentCondition

CTL-----scram--si25---si50---si75----si100CTL------nM----nM----nM-----nM

ID1

β9actin

CTL scramble,CTL si,100nM

TMZ,25uM

si+TMZ,25uM

TMZ,50uM

si+TMZ,50uM

TMZ,100uM

si+TMZ,100uM

ID1

β%actin

0

2

4

6

8

10

12

scramble/CTL

CTL si/100nM TMZ/25uM si+TMZ/25uM

TMZ/50uM si+TMZ/50uM

TMZ/100uM

si+TMZ/100uM

Cell/Viability/

(viable/cells/#cells/seeded)

Treatment/Condition

***

******

Figure 8. ID1 inhibition (via siRNA) increases GBM cell sensitivity to TMZ-chemotherapy treatment. (A) Western blot analysis of the optimal si-ID1 concentration to be used for subsequent experiments in U251. si-ID1 of 100nM achieved the greatest ID1 knockdown at the protein level. (B) Microscopic images of the differential treatment conditions, depicting fewer cells in si-ID1+TMZ groups compared to TMZ alone. (C) Cell viability decreased in U251 cells pre-treated with si-ID1 followed by TMZ treatment (25, 50, 100 uM) compared to TMZ treatment alone. Western blot analysis confirms ID1 knockdown in si-ID1 treatment groups. Scale bar represents 100um. Error bars represent the ±SD of triplicate measurements and significance (***) was determined by comparing combined si-ID1 + TMZ-treated to TMZ-treated alone using t-test (p<0.001).

A B

C

Page 49: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

34

8.4   ID1  Knockout  Reduces  Colony  Formation  Capacity  following  TMZ-­Treatment  

8.4.1   Development  of  an  ID1-­knockout  cell  line    

Our in vitro findings that siRNA-mediated knockdown of ID1 potentiated the cytotoxic effect of

TMZ in glioma cells prompts consideration that complete ID1-knockout could similarly enhance

the cytotoxic effect of TMZ in vitro. Two limitations of using an siRNA approach are that it only

provides a transient effect, and difficulties arise in the delivery of siRNA to the brain. To study

the effect of long-term ID1 inhibition on glioma cell biology we employed the CRISPR/Cas9

system and developed a GBM cell line with a stable ID1 knockout. After establishing potential

ID1 knockout (ID1-KO) clones through a rigorous multi-step process in U251 GBM cell line, we

screened these clones for complete ID1-KO at the protein level using Western blot, and

subsequently sent for sanger sequencing to validate ID1 mutation. The screen identified several

clones that achieved ID1 knockout at the protein level, including two clones that we named ID1-

KO 14 and 19; we also selected a clone that did not show ID1 knockout at the protein level, that

we named Clone-33, to serve as an experimental control. (Figure 9A). Sanger sequencing

validated that these two ID1-KO clones harbour a mutation in the region targeted by the sgRNA

within the ID1 gene (Figure 9B-C). ID1-KO 14 and 19 clones were therefore used to establish

cell lines for subsequent experimental use.

A potential concern when employing the CRISPR/Cas9 system is causing a mutation in a non-

targeted region. Using a bio-informatics database, CRISPR Design (http://crispr.mit.edu), we

identified the top off-targets against the sgRNA used in our ID1 knockout model (Table 1).

Sanger sequencing validated that there is no mutation in three of the five top off-targets (Figure

10). Therefore, the results found in subsequent experiments are most likely not due to off-target

effects in our system.

Page 50: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

35

ID1$KO'14

ID1$KO'19

sgRNA PAM(sequence

β"actin

ID1

12,,,,13,,,,,14,,,,15Clone,#

β"actin

ID1

32,,,,33,,,,34,,, 37Clone,#

18,,,,,19,,,,20,,,,21

β"actin

ID1

Clone,#

Figure 9. Development of an ID1-knockout line using CRISPR/Cas9. (A) Western blot analysis of a screen to identify clones with ID1-knockout at the protein level. Clone-14 and Clone-19 achieved a complete knockout of ID1, whereas Clone-33 did not. ID1-KO 14 and 19 clones were used to establish cell lines for subsequent experimental use, and Clone-33 was selected for use as a control. (B) Pictorial depiction of the sgRNA used to target the ID1 sequence, followed by the PAM sequence required by Cas9 to identify the position for DNA cleavage. (C) Sanger sequencing validated ID1-KO 14 and 19 harbor a mutation in the region targeted by the sgRNA within the ID1 gene, a one nucleotide deletion depicted as a red tick upstream of the PAM sequence.

A B

C

Page 51: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

36

ID1$KOWT

Off$target/1/chr7:+108096350/

Off#target)5)chr2:#40006264)

ID1#KOWT

ID1$KOWT

Off$target/4/chr1:+236445511/

Table 1: Top off-target hits in CRISPR/Cas9 ID1-knockout system

sequence score mismatches UCSC

gene

locus

GAAAGCGTGGGGTGCGGGCG

CGG

2.4 3MMs

[3:8:10]

chr7:+10809635

0

AACACCGGGGGGTGCGGGCG

GGG

1.3 3MMs

[5:8:10]

chr1:+23644551

1

TAATGCGAGAGGTGCGGGCG

CGG

1.3 3MMs

[3:4:10]

NR_02810

2

chr2:-40006264

Figure 10. Three top off-target hits in the CRISPR/Cas9 ID1-knockout model display no mutation in the target region. Sanger sequencing validated there is no mutation in the target regions (chr7:+108096350; chr1:+236445511; chr2:-40006264); the target sequences display perfect alignment between ID1-KO and wild-type (WT), with no insertions or deletions.

Page 52: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

37

8.4.2   ID1-­knockout  decreases  colony  formation  following  treatment  with  TMZ    

Using our established stable ID1-knockout cell lines, we next wanted to ask whether ID1

knockout enhances the effect of TMZ in vitro? To address this question, we treated Clone-33

control, ID1-KO 14 and 19 U251 GBM cells with TMZ for either 3 or 7 days, at which point we

measured cell viability. We found no significant difference in the number of viable cells in ID1-

KO 14 and 19 compared to the control, at either time points following TMZ treatment (Figure

11A). We also found that ID1-knockout did not result in decreased cell viability compared to the

control in the no treatment conditions at 3 and 7 days (Figure 11B).

Following from our initial experiments investigating the role of ID1 in GBM tumour recurrence

in patient tumour samples, we next wanted to determine whether knocking out ID1 has an effect

on colony formation in vitro. To address this question, we assessed the ability of GBM cells that

possessed ID1 knockout to form colonies following TMZ treatment. Clone-33 control, or ID1-

KO 14 and 19 cells were treated with TMZ (100uM) for 7 days, followed by 7 days of no

treatment. We identified colonies at this time point (day 14) using crystal violet staining. ID1-

KO 14 and 19 showed significantly less colony formation following TMZ treatment compared to

control (Figure 11C). These results reveal that knocking out ID1 significantly reduces the ability

of U251 glioma cells to form colonies following TMZ treatment, suggesting that ID1 inhibition

could impair the ability of GBM tumours to recur following chemotherapy.

Page 53: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

38

Clone&33(Control ID1&KO(14 ID1&KO(19

0

1

2

3

4

5

6

7

8

Clone/330Control

D3/TMZ

ID1/KO014

D3/TMZ

ID1/KO019

D3/TMZ

Cell0Viability

(viable0cells/#cells0seeded)

Condition

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

Clone/330Control

D7/TMZ

ID1/KO014

D7/TMZ

ID1/KO019

D7/TMZ

Cell0Viability

(viable0cells/#cells0seeded)

Condition

0

1

2

3

4

5

6

7

Clone.33/Control/D3.NT

ID1.KO/14/D3.NT

ID1.KO/19/D3.NT

Cell/Viability

(viable/cells/#cells/seeded)

Condition

0

10

20

30

40

50

60

70

Clone/33/Control/D7.NT

ID1.KO/14/D7.NT

ID1.KO/19/D7.NT

Cell/Viability

(viable/cells/#cells/seeded)

Condition

Figure 11. ID1-knockout reduces colony formation capacity following treatment with temozolomide. (A) No significant difference in the number of viable cells between ID1-KO 14 and 19 compared to Clone-33 control, following TMZ-treatment for 3 or 7 days. (B) No significant decrease in cell viability in ID1-KO clones compared to control, following no treatment for 3 or 7 days. (C) Crystal violet staining depicts the difference in ability to form colonies post-TMZ treatment in Clone-33 control compared to ID1-KO 14 and 19 cells. The cells were treated with TMZ (100uM) for 7 days, followed by 7 days of no treatment. ID1-knockout cells show a significant reduction in colony formation following TMZ-treatment compared to the control condition. Error bars represent the ±SD of triplicate measurements. D3 = 3 days post-TMZ treatment; D7 = 7 days post-TMZ treatment; NT = no treatment.

A

C

B

Page 54: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

39

8.5   ID1  Knockout  Delays  GBM  Tumour  Initiation/Progression  and  Increases  Overall  Survival  

ID1-KO showed a profound effect on glioma cell colony formation following TMZ treatment in

vitro. Our next step was to investigate how ID1 knockout effects GBM biology in vivo.

Specifically, we wanted to determine the effect of knocking out ID1 on GBM tumour growth. To

do so, we developed orthotopic xenograft tumours in NSG mice using control U251 and U251

ID1-KO glioma cells. NSG mice were injected intracranially with U251-luciferase control (n=8)

or ID1-KO 14-luciferase (n=9) cells. Tumour initiation and progression were monitored using

bioluminescence imaging. U251 control mice quickly developed glioblastoma tumours, with all

of the mice showing tumour formation by post-implantation day (PID) 9. Conversely, ID1-KO

14 mice did not all show signs of tumour formation until PID 20 (Figure 12A). Furthermore, the

tumours of U251 control mice progressed rapidly, with some mice developing leptomeningeal

dissemination by PID 13. Conversely, ID1-KO 14 mice showed very slow tumour progression.

Bioluminescence imaging demonstrated a significant difference in total photon flux between

U251 control and ID1-KO 14 at 1-, 2-, and 3-weeks post-injection (p<0.001) (Figure 12B). We

additionally wanted to determine if ID1-KO effected survival outcomes. Mice were allowed to

reach end-point in both groups. We found that the ID1-KO 14 mice survived 2.27 times longer

than the U251 control mice (p<0.001) (Figure 13). The average survival time in the control

group was 21-days post-injection, whereas the average survival time in the ID1-KO 14 group

was 47-days post-injection (Table 2). These results suggest that ID1 plays a critical role in

tumour initiation and progression. Further, ID1 knockout has a significant impact on overall

survival in glioblastoma.

Page 55: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

40

clone #

14 1w

k

Control 1

wk

clone #

14 2w

ks

Control 2

wks

clone #

14 3w

ks

Control 3

wks

clone #

14 4w

ks

clone #

14 5w

ks

clone #

14 6w

ks0

200000

400000

600000

800000

1000000

Weeks after cell inoculation

Tota

l Pho

ton

Flux

clone #14 1wk Control 1wkclone #14 2wks Control 2wksclone #14 3wks Control 3wksclone #14 4wksclone #14 5wksclone #14 6wks

Total&Photon&Flux

Weeks&after&Cell&Inoculation

1 2 3 4 5 6

ID19KO&14

U251&Control

*** ***

***

U251%Control%Xenograft ID13KO%14%Xenograft

Figure 12. ID1-knockout delays glioblastoma tumour initiation and progression. (A) ID1-KO 14 mice show delayed tumour initiation and very slow tumour progression compared to U251 control mice, which show quick GBM tumour development and rapid tumour progression as determined by bioluminescent imaging. Red indicates high cell density while purple represents low cell density. By day-20 post-injection, all U251 control mice demonstrate substantial tumour mass and some have developed leptomeningeal metastasis; whereas the ID1-KO 14 mice continue to display minimal tumour progression. (B) Signal progression of flux activity from U251 cell xenografts and ID1-KO 14 cell xenografts demonstrate a significant difference in total photon flux between the two cohorts 1-, 2-, and 3-weeks after cell inoculation. Error bars represent ±SEM and significance (***) was determined by comparing ID1-KO 14 to U251 control using t-test (p<0.001).

A

B

Page 56: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

41

0 20 40 60 800

50

100

Time (days)

Perc

ent s

urviv

al

Survival of Data for ID1 KO clone #14

U251 Control (n=8)U251 ID1 KO clone#14 (n=9)

ID1$KO'14'(n=9)

U251'Control'(n=8)

Table 2: Survival outcomes in ID1-knockout in vivo study

Cohort #1 Cohort #2

U251 control Survival Time (days) ID1-KO 14 Survival Time (days)

5086 20 5081 38

5087 20 5082 51

5088 20 5083 43

5089 20 5084 52

5090 22 5085 41

5091 20 5094 48

5092 25 5095 56

5093 20 5096 40

5097 58

Average survival time: 21 days Average survival: 47 days***

*** (p<0.001)

Figure 13. ID1-knockout increases overall survival time in glioblastoma. Kaplan-Meier analysis showed a significant difference in survival time between the two cohorts; ID1-KO 14 mice survived 2.27 times longer than the U251 control mice (p<0.001).

Page 57: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

42

8.6   Pimozide  Enhances  the  Effect  of  TMZ-­Treatment  and  Provides  a  Therapeutic  Advantage  in  Glioblastoma  

The sum of our prior work suggested that ID1 inhibition could be of multi-factorial benefit to

patients with GBM. As our final step, we sought to evaluate the potential clinical application of

our work. We reviewed the literature to identify a clinically translatable inhibitor of ID1.

Through this review we identified pimozide, an FDA approved drug that is currently used as an

antipsychotic to treat patients with Tourette’s Disorder. Pimozide chemically inhibits ID1 by

targeting ID1 degradation, and is capable of crossing the blood-brain-barrier. We decided to

evaluate the utility of pimozide in combination with TMZ-chemotherapy in glioblastoma. U251

GBM cells were first treated with graded concentrations of pimozide (2.5, 5, 7.5, 10 uM) for

optimization purposes. The optimal concentration of pimozide selected for subsequent

experimental use was 5 uM, as it was not cytotoxic to the cells, but resulted in decreased ID1

protein levels (Figure 14A). U251 cells were then either pre-treated with pimozide followed by

TMZ treatment or treated with TMZ alone for 3 days. We found that cell viability decreased in

U251 cells pre-treated with pimozide followed by TMZ treatment (50 uM) compared to TMZ

treatment alone (p<0.01) (Figure 14B). Western blot analysis confirms ID1 inhibition in

pimozide treated groups. These results highlight the potential clinical relevance of this work, as

pimozide treatment enhances the effect of TMZ in GBM cells.

We next wanted to determine whether pimozide provides a therapeutic advantage in

glioblastoma when combined with TMZ-chemotherapy. To address this question, NSG mice

were injected intracranially with U251 cells that were luciferase-tagged. Following tumour

formation as confirmed by bioluminescence imaging, mice were treated with vehicle (n=5),

pimozide only (n=5), TMZ only (n=5), or pimozide + TMZ (n=5) for 2 weeks. Tumour

progression was monitored with serial bioluminescence imaging. Mice treated with pimozide

alone showed similar tumour progression compared to control (vehicle-treated) mice. These

results suggest that pimozide treatment has no effect on glioblastoma initiation and development

(Figure 15A). Mice treated with TMZ alone showed slight tumour regression immediately

following TMZ; however, this cohort showed evidence of tumour recurrence at 5 days following

completion of TMZ treatment. Mice treated with a combination of pimozide + TMZ showed the

same slight tumour regression immediately following TMZ; however, 5 days following the

completion of TMZ treatment, these mice continued to show a decrease in tumour growth

Page 58: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

43

0

20

40

60

80

100

120

CTL pimo.5uM pimo.10uM

Cell.Viability.

(viable.cells/#cells.seeded)

Treatment.Condition

0

20

40

60

80

100

120

CTL pimo.5uM TMZ.50uM pimo.5uM.+TMZ.50uM

Cell.Viability.

(Viable.cells/#cells.seeded)

Treatment.Condition

ID1

ßGactin

**

(Figure 15B). These data suggest that pimozide combined with TMZ treatment delays tumour

recurrence in vivo compared to TMZ treatment alone. Moreover, these findings further validate

the potential clinical application of ID1 inhibition in combination with TMZ in the treatment of

glioblastoma patients.

Figure 14. Pimozide (small molecule inhibitor that targets ID1 degradation) increases GBM cell sensitivity to TMZ-treatment. (A) Cell viability analysis of the optimal pimozide concentration to be used for subsequence experiments in U251 cells. Pimozide 5 uM was not cytotoxic to the cells in comparison to 10 uM, and so was selected for further use. (B) Cell viability decreased in U251 cells pre-treated with pimozide followed by TMZ treatment (50uM) compared to TMZ treatment alone. Western blot confirms ID1 inhibition in pimozide treated groups. Error bars represent the ±SD of triplicate measurements and significance (**) was determined by comparing combined pimozide + TMZ-treated to TMZ-treated alone using t-test (p<0.01).

A

B

Page 59: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

44

Figure 15. Pimozide enhances the effect of TMZ-treatment and provides a therapeutic advantage in glioblastoma. (A) Pimozide-treated mice displayed similar tumour progression compared to vehicle control mice, showing rapid tumour growth and leptomeningeal metastasis. Pimozide treatment provides no therapeutic advantage on its own. (B) TMZ-treated mice showed a dramatic increase in tumour growth 5 days post-TMZ treatment; whereas, pimozide + TMZ-treated mice showed a decrease in tumour growth 5 days post-TMZ treatment determined by bioluminescent imaging. Pimozide combined with TMZ treatment delays tumour recurrence in vivo compared to TMZ treatment alone. Red indicates high cell density while purple represents low cell density.

A

B

Page 60: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

45

Chapter  4      

Discussion  and  Future  Directions    

  Discussion  and  Future  Directions  

9.1   Glioblastoma:  Discovering  an  Ideal  Therapeutic  Target  The primary aim of our work was to elucidate the role of ID1 in glioblastoma treatment

resistance and tumour recurrence, and to determine whether ID1 represents a therapeutic target in

this tumour. As outlined in the introduction, ID1 overexpression has been reported in many

cancers and is associated with malignant potential and aggressive clinical behavior162. It has

previously been demonstrated that there is a positive correlation between ID1 expression and

tumour grade in astrocytomas, with highest ID1 expression found in glioblastoma154. Precisely,

Soroceanu et al used immunohistochemistry (IHC) of TMAs containing glioma samples with

various tumour grades and found a gradual increase of ID1 positive samples from normal, to

grade II astrocytoma, grade III astrocytoma, and finally grade IV astrocytoma (glioblastoma)154.

Also, ID1 has previously been found to be highly upregulated in tumours from recurrent prostate

cancer patients, suggesting that ID1 overexpression may be responsible for the development of

treatment resistance153. In our study, immunohistochemical analysis demonstrated that GBM

patients with increased ID1 expression following chemotherapy experienced a shorter latency to

recurrence. This suggests that ID1 expression negatively correlates with progression-free

survival in GBM. Although ours is the first study to our knowledge that describes a correlation

between ID1 expression and latency to recurrence in GBM, these previous findings support that

ID1 may be a potential therapeutic target in glioblastoma treatment resistance and tumour

recurrence.

We corroborated our clinical data by further characterizing the interplay between ID1 expression

and response to temozolomide treatment in vitro. We observed that ID1 expression correlates

with TMZ resistancy in five GBM cell lines with differential baseline expression, in which high

basal ID1 expression was associated with increased resistancy to temozolomide. These findings

are in accordance with a previous report that found ectopic ID1 expression conferred resistance

to the chemotherapeutic agent Taxol in prostate and nasopharyngeal carcinoma cells160,161. Taken

Page 61: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

46

together, ID1 expression appears to be associated with a more aggressive, chemotherapy-

resistant phenotype in many cancer types, including GBM.

Having established a relationship between ID1 expression and response to treatment in both

human patient tumour samples and GBM cell lines, we wanted to further explore the effects of

TMZ-chemotherapy on ID1 expression in vitro. Using immunocytochemistry (ICC) and Western

blot analysis we observed that TMZ exposure results in an increase in ID1 levels in GBM cells.

Initially we believed that this was due to transcriptional changes resulting in increased ID1 gene

expression. However, when we examined the effect of TMZ on ID1 expression at the mRNA

level using QT-PCR, we saw no significant change. These results would suggest that TMZ is not

inducing changes in ID1 at the genetic level, but potentially altering ID1 via post-transcriptional

or post-translational modifications, or enriching for ID1-positive cells. Campos et al found that

TMZ treatment decreases the diversity of tumour subclasses by selecting for the most resistant

subtypes, ultimately diminishing tumour heterogeneity163. We postulate that TMZ may be

selecting for ID1-positive cells, as this subgroup may represent the chemoresistant population in

GBM tumours. Another potential explanation for the observed increase in ID1 protein expression

following TMZ treatment is protein stabilization. USP1 is a ubiquitin-specific protease known to

rescue several proteins from ubiquitination-mediated protein degradation, including ID1. Lee et

al demonstrated that USP1 is highly expressed in primary human glioblastoma and promotes ID1

stability in GBM cells155. Therefore, it can be proposed that TMZ may exert its effect on USP1

and consequently ID1 protein stability, resulting in the observed increase in ID1 protein

expression post-TMZ treatment in GBM cells.

Interestingly, the increase in ID1 seen with TMZ exposure is specific to TMZ and not a more

general response to DNA damage. We did not find the same effect on ID1 expression when

U251 and GliNS1 GBM cells were treated with cisplatin, another chemotherapeutic agent. It is

known that TMZ and cisplatin employ differing mechanisms to induce DNA damage: cisplatin

cross-links DNA, whereas TMZ methylates guanine residues in the DNA molecule. Therefore,

the effect of TMZ treatment on ID1 expression may be attributed to a specific apoptotic

mechanism that does not involve chemical cross-linking of DNA strands, which is considered to

be more toxic.

Page 62: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

47

9.2   Treating  Glioblastoma  in  vitro:  ID1  Inhibition  Enhances  the  Effect  of  Temozolomide  

We found that ID1 inhibition using siRNA increases GBM cell sensitivity to TMZ-chemotherapy

treatment, as demonstrated by decreased cell viability in U251 cells pre-treated with si-ID1

followed by TMZ treatment compared to TMZ treatment alone. Similar to our results, Zhang et

al demonstrated that ID1 inhibition via siRNA resulted in sensitization of prostate cancer cells to

Taxol treatment160. This effect was mediated by JNK activation, a pathway that has previously

been implicated in the inhibition of chemotherapeutic drug-induced apoptosis in cancer cells.

Our results are further supported by the finding that silencing ID1 and ID3 sensitized colon

cancer stem cells to the chemotherapeutic agent, oxilaplatin66. Taken together, ID1 inhibition

enhances the cytotoxic effect of chemotherapy agents in multiple cancer types.

We also evaluated the effect of ID1 inhibition on TMZ sensitivity using pimozide, an FDA

approved drug that chemically inhibits ID1. Pimozide is known as a small-molecule inhibitor

that is capable of crossing the blood-brain-barrier, thus making it clinically applicable for

glioblastoma treatment. As with siRNA-mediated knockdown of ID1, pimozide sensitized U251

GBM cells to TMZ-treatment, resulting in decreased cell viability following TMZ therapy. Lee

at al have also demonstrated that pimozide enhances the therapeutic efficacy of irradiation in

GBM cells155. They found that pimozide combined with the radiometric drug neocarzinostatin

(NSC) resulted in more persistent DNA damage in comparison to NSC alone155.

We employed the CRISPR/Cas9 system to study the effect of long-term ID1 inhibition by

developing a GBM cell line with a stable ID1 knockout. There are many advantages to using this

system in comparison to other genome editing technologies that have previously been established

for gene and cell therapy. For example, the CRISPR/Cas9 system allows simple nuclease

construction and in vitro testing, good targeting efficiency, and low time investment and cost, in

comparison to meganucleases, zinc finger nucleases, and TALENs164. For these reasons, we

decided to utilize the the CRISPR/Cas9 system in our study. We found that knocking out ID1 in

glioma cells significantly reduces their ability to form colonies following TMZ-treatment. Our

data is corroborated by a previous study that showed ID1 inhibition combined with irradiation

significantly decreased clonogenic regrowth compared to irradiation alone in GBM cells155.

Page 63: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

48

Taken together, these results further support that ID1 may be implicated in GBM tumour

recurrence following cytotoxic therapy.

9.3   Treating  Glioblastoma  in  vivo:  Targeting  ID1  Provides  a  Therapeutic  Advantage  

Currently, there have been no studies showing an effective response to ID1 inhibition in

combination with TMZ-treatment in an in vivo model of glioblastoma. After obtaining promising

in vitro results showing that ID1 knockdown or knockout exhibited a significant effect on TMZ

efficacy, we proceeded to test the effectiveness in vivo. However, we first wanted to examine the

effect of knocking out ID1 on GBM biology, without combined TMZ treatment. To do so, we

implanted NSG mice with either U251-luciferase control or ID1-KO-luciferase GBM cells and

monitored tumour growth. We observed rapid tumour initiation and progression in our control

mice; whereas, the ID1-KO mice experienced severely delayed tumour initiation and minimal

tumour progression over the same period of time. Furthermore, the ID1-KO mice demonstrated a

significant increase in survival time, 2.27 times longer than the U251 control mice. Similar to our

findings, Soroceanu et al demonstrated that treatment with the nontoxic cannabinoid,

cannabidiol, significantly reduced ID1 expression and tumour growth in an in vivo model of

GBM154. Specifically, cannabidiol decreased the tumour area by 95%; and 1 of the 5 treated mice

showed no tumour cells in any of the brain regions analyzed154. This study also found that

cannabidiol significantly inhibits glioblastoma dispersal using an ex vivo model154. Additionally,

previous studies have demonstrated the therapeutic benefit of targeting ID proteins in tumour

xenografts. For example, ID1/ID3 double knockout mice could not support tumour growth and

metastasis in three different tumour models87. Our results, taken together with previous findings,

would suggest that ID1 plays a critical role in tumour initiation and progression, as well as

invasion and overall survival in glioblastoma.

Finally, we examined the efficacy of combining TMZ treatment and ID1 inhibition in an in vivo

model of GBM. We treated NSG mice implanted with U251 GBM cells with either saline

(vehicle control), pimozide, TMZ, or pimozide + TMZ. We found that pre-treatment with

pimozide followed by TMZ resulted in prolonged tumour regression following treatment

compared to TMZ alone. The TMZ alone cohort displayed signs of tumour recurrence 5 days

after treatment, whereas the combined treatment group continued to demonstrate a decrease in

Page 64: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

49

tumour growth at the same time point following completion of treatment. These data suggest that

combined therapy with pimozide and TMZ has a prolonged inhibitory effect on tumour

recurrence in vivo compared to TMZ alone, providing a therapeutic advantage. Our study

suggests that patients will derive a therapeutic advantage by combining ID1 inhibition with

standard of care treatment in GBM.

9.4   Conclusions  Our study provides in vitro and in vivo evidence that ID1 inhibition enhances the the efficacy of

temozolomide treatment in glioblastoma. Our findings are significant for two reasons. First, as

the vast majority of glioblastoma patients experience chemoresistance, the identification of

therapies that can prolong time to relapse is of paramount importance. Second, we demonstrated

that there is a significant survival benefit with ID1 knockout alone in a GBM xenograft model.

This finding demonstrates that ID1 is critical to glioma biology, and its targeting may have

benefit to patients with GBM independent of its effects on chemotherapy. While our results are

encouraging, more work is needed to evaluate the optimal combination of treatments in GBM.

For example, pimozide has been combined with irradiation in a previous study, and with

temozolomide in our current study; however, the combination of all three treatments should be

evaluated in the future using in vitro and in vivo models of GBM. Finally, since we are proposing

the use of pimozide in glioblastoma patients, more information about the drug’s long-term

effects, specifically when combined with toxic agents, should be taken into consideration if this

drug is to be used in the clinic in the future.

9.5   Future  Directions    Regarding the effect of temozolomide on ID1 expression in vitro, a better understanding of the

mechanism should be obtained as to what is causing this observed increase in ID1 protein

expression following treatment. We have identified four potential hypotheses to explain the

effect of TMZ on ID1 levels: (1) TMZ selects for ID1-positive glioma cells; (2) TMZ induces

ID1 gene expression; (3) TMZ enhances ID1 stability by post-translational modifications; and

(4) TMZ results in decreased ID1 degradation. To assess whether TMZ is selecting for ID1-

positive cells or inducing ID1 gene expression, we would employ a reporter gene and promoter-

reporter, respectively. One potential way that TMZ could enhance ID1 stability via post-

translational modifications is by increasing the half-life of ID1. To address this possibility, we

Page 65: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

50

would treat GBM cells with cycloheximide, an inhibitor of protein synthesis that is used as an

experimental tool to determine the half-life of a protein, following either treatment with TMZ or

no treatment. Western blot analysis would then be used to measure ID1 protein levels at various

time points following treatment with cycloheximide. The major pathway of protein degradation

uses ubiquitin as a marker that targets nuclear and cytosolic proteins for proteolysis. To address

the hypothesis that TMZ results in decreased ID1 degradation, we would employ an in vitro

ubiquitination assay on GBM cells following treatment with TMZ or no treatment.

To further understand the function of ID1 and how it might mediate resistance, we could perform

ID1-pulldown followed by mass spectrometry to identify the binding partners of ID1,

specifically post-TMZ treatment. This will allow us to gain insight into potential pathways that

may be activated upon treatment with TMZ. For example, ID1 may be binding to a key player in

a DNA damage pathway following TMZ-treatment, thus resulting in its increased expression. In

our study we established a correlation between ID1 expression and TMZ resistancy, and also

demonstrated that ID1 inhibition sensitizes GBM cells to temozolomide. The next step would be

to evaluate the effect of ID1 overexpression on TMZ sensitivity, whereby we hypothesize that

ectopic ID1 expression will result in increased resistancy to treatment. Finally, combining

pimozide with other conventional therapies such as both radio- and chemo-therapy should be

evaluated in an in vivo setting.

Page 66: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

51

References  

1. Ostrom, Q. T. et al. CBTRUS Statistical Report: Primary Brain and Central Nervous

System Tumors Diagnosed in the United States in 2008-2012. Neuro-Oncol. 17 Suppl 4,

iv1–iv62 (2015).

2. Ohgaki, H. & Kleihues, P. Population-based studies on incidence, survival rates, and

genetic alterations in astrocytic and oligodendroglial gliomas. J. Neuropathol. Exp. Neurol.

64, 479–489 (2005).

3. Thumma, S. R. et al. Effect of pretreatment clinical factors on overall survival in

glioblastoma multiforme: a Surveillance Epidemiology and End Results (SEER) population

analysis. World J. Surg. Oncol. 10, 75 (2012).

4. Burnet, N. G., Jefferies, S. J., Benson, R. J., Hunt, D. P. & Treasure, F. P. Years of life lost

(YLL) from cancer is an important measure of population burden--and should be

considered when allocating research funds. Br. J. Cancer 92, 241–245 (2005).

5. Stupp, R. et al. Radiotherapy plus concomitant and adjuvant temozolomide for

glioblastoma. N. Engl. J. Med. 352, 987–996 (2005).

6. Ohgaki, H. et al. Genetic pathways to glioblastoma: a population-based study. Cancer Res.

64, 6892–6899 (2004).

7. Bondy, M. L. et al. Brain tumor epidemiology: consensus from the Brain Tumor

Epidemiology Consortium. Cancer 113, 1953–1968 (2008).

8. Hottinger, A. F. & Khakoo, Y. Update on the management of familial central nervous

system tumor syndromes. Curr. Neurol. Neurosci. Rep. 7, 200–207 (2007).

9. Gu, J., Liu, Y., Kyritsis, A. P. & Bondy, M. L. Molecular epidemiology of primary brain

tumors. Neurother. J. Am. Soc. Exp. Neurother. 6, 427–435 (2009).

10. Easaw, J. C. et al. Canadian recommendations for the treatment of recurrent or progressive

glioblastoma multiforme. Curr. Oncol. Tor. Ont 18, e126–136 (2011).

Page 67: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

52

11. Louis, D. N. et al. The 2007 WHO classification of tumours of the central nervous system.

Acta Neuropathol. (Berl.) 114, 97–109 (2007).

12. Theeler, B. J., Yung, W. K. A., Fuller, G. N. & De Groot, J. F. Moving toward molecular

classification of diffuse gliomas in adults. Neurology 79, 1917–1926 (2012).

13. Dolecek, T. A., Propp, J. M., Stroup, N. E. & Kruchko, C. CBTRUS statistical report:

primary brain and central nervous system tumors diagnosed in the United States in 2005-

2009. Neuro-Oncol. 14 Suppl 5, v1–49 (2012).

14. Rasheed, B. K. et al. Chromosome 10 deletion mapping in human gliomas: a common

deletion region in 10q25. Oncogene 10, 2243–2246 (1995).

15. Kim, D. H., Mohapatra, G., Bollen, A., Waldman, F. M. & Feuerstein, B. G. Chromosomal

abnormalities in glioblastoma multiforme tumors and glioma cell lines detected by

comparative genomic hybridization. Int. J. Cancer 60, 812–819 (1995).

16. Vranová, V. et al. Screening of genomic imbalances in glioblastoma multiforme using high-

resolution comparative genomic hybridization. Oncol. Rep. 17, 457–464 (2007).

17. Collins, V. P. Brain tumours: classification and genes. J. Neurol. Neurosurg. Psychiatry 75

Suppl 2, ii2–11 (2004).

18. von Deimling, A. et al. Comprehensive allelotype and genetic anaysis of 466 human

nervous system tumors. J. Neuropathol. Exp. Neurol. 59, 544–558 (2000).

19. Nishizaki, T. et al. Investigation of genetic alterations associated with the grade of

astrocytic tumor by comparative genomic hybridization. Genes. Chromosomes Cancer 21,

340–346 (1998).

20. Mohapatra, G. et al. Genetic analysis of glioblastoma multiforme provides evidence for

subgroups within the grade. Genes. Chromosomes Cancer 21, 195–206 (1998).

21. Ohgaki, H. & Kleihues, P. Genetic pathways to primary and secondary glioblastoma. Am. J.

Pathol. 170, 1445–1453 (2007).

Page 68: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

53

22. Yin, D. et al. High-resolution genomic copy number profiling of glioblastoma multiforme

by single nucleotide polymorphism DNA microarray. Mol. Cancer Res. MCR 7, 665–677

(2009).

23. Burton, E. C. et al. Genetic aberrations defined by comparative genomic hybridization

distinguish long-term from typical survivors of glioblastoma. Cancer Res. 62, 6205–6210

(2002).

24. Shinojima, N. et al. Prognostic value of epidermal growth factor receptor in patients with

glioblastoma multiforme. Cancer Res. 63, 6962–6970 (2003).

25. Wong, A. J. et al. Increased expression of the epidermal growth factor receptor gene in

malignant gliomas is invariably associated with gene amplification. Proc. Natl. Acad. Sci.

U. S. A. 84, 6899–6903 (1987).

26. Beroukhim, R. et al. Assessing the significance of chromosomal aberrations in cancer:

methodology and application to glioma. Proc. Natl. Acad. Sci. U. S. A. 104, 20007–20012

(2007).

27. Mellinghoff, I. K. et al. Molecular determinants of the response of glioblastomas to EGFR

kinase inhibitors. N. Engl. J. Med. 353, 2012–2024 (2005).

28. Phillips, H. S. et al. Molecular subclasses of high-grade glioma predict prognosis, delineate

a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 9, 157–

173 (2006).

29. Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines

human glioblastoma genes and core pathways. Nature 455, 1061–1068 (2008).

30. Verhaak, R. G. W. et al. Integrated genomic analysis identifies clinically relevant subtypes

of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1.

Cancer Cell 17, 98–110 (2010).

31. Hegi, M. E. et al. MGMT gene silencing and benefit from temozolomide in glioblastoma.

N. Engl. J. Med. 352, 997–1003 (2005).

Page 69: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

54

32. Newlands, E. S., Stevens, M. F., Wedge, S. R., Wheelhouse, R. T. & Brock, C.

Temozolomide: a review of its discovery, chemical properties, pre-clinical development

and clinical trials. Cancer Treat. Rev. 23, 35–61 (1997).

33. Newlands, E. S. et al. Phase I trial of temozolomide (CCRG 81045: M&B 39831: NSC

362856). Br. J. Cancer 65, 287–291 (1992).

34. Stevens, M. F. et al. Antitumor activity and pharmacokinetics in mice of 8-carbamoyl-3-

methyl-imidazo[5,1-d]-1,2,3,5-tetrazin-4(3H)-one (CCRG 81045; M & B 39831), a novel

drug with potential as an alternative to dacarbazine. Cancer Res. 47, 5846–5852 (1987).

35. Denny, B. J., Wheelhouse, R. T., Stevens, M. F., Tsang, L. L. & Slack, J. A. NMR and

molecular modeling investigation of the mechanism of activation of the antitumor drug

temozolomide and its interaction with DNA. Biochemistry (Mosc.) 33, 9045–9051 (1994).

36. Tisdale, M. J. Antitumor imidazotetrazines--XV. Role of guanine O6 alkylation in the

mechanism of cytotoxicity of imidazotetrazinones. Biochem. Pharmacol. 36, 457–462

(1987).

37. Tsang, L. L., Farmer, P. B., Gescher, A. & Slack, J. A. Characterisation of urinary

metabolites of temozolomide in humans and mice and evaluation of their cytotoxicity.

Cancer Chemother. Pharmacol. 26, 429–436 (1990).

38. Patel, M., McCully, C., Godwin, K. & Balis, F. M. Plasma and cerebrospinal fluid

pharmacokinetics of intravenous temozolomide in non-human primates. J. Neurooncol. 61,

203–207 (2003).

39. Drabløs, F. et al. Alkylation damage in DNA and RNA--repair mechanisms and medical

significance. DNA Repair 3, 1389–1407 (2004).

40. Wedge, S. R. & Newlands, E. S. O6-benzylguanine enhances the sensitivity of a glioma

xenograft with low O6-alkylguanine-DNA alkyltransferase activity to temozolomide and

BCNU. Br. J. Cancer 73, 1049–1052 (1996).

Page 70: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

55

41. Kyrtopoulos, S. A. et al. DNA adducts and the mechanism of carcinogenesis and

cytotoxicity of methylating agents of environmental and clinical significance. Cancer

Detect. Prev. 21, 391–405 (1997).

42. Mojas, N., Lopes, M. & Jiricny, J. Mismatch repair-dependent processing of methylation

damage gives rise to persistent single-stranded gaps in newly replicated DNA. Genes Dev.

21, 3342–3355 (2007).

43. Cejka, P. et al. Methylation-induced G(2)/M arrest requires a full complement of the

mismatch repair protein hMLH1. EMBO J. 22, 2245–2254 (2003).

44. D’Atri, S. et al. Involvement of the mismatch repair system in temozolomide-induced

apoptosis. Mol. Pharmacol. 54, 334–341 (1998).

45. Horton, J. K. & Wilson, S. H. Hypersensitivity phenotypes associated with genetic and

synthetic inhibitor-induced base excision repair deficiency. DNA Repair 6, 530–543 (2007).

46. Silber, J. R. et al. O6-methylguanine-DNA methyltransferase-deficient phenotype in human

gliomas: frequency and time to tumor progression after alkylating agent-based

chemotherapy. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 5, 807–814 (1999).

47. Gerson, S. L. Clinical relevance of MGMT in the treatment of cancer. J. Clin. Oncol. Off. J.

Am. Soc. Clin. Oncol. 20, 2388–2399 (2002).

48. Gerson, S. L. MGMT: its role in cancer aetiology and cancer therapeutics. Nat. Rev. Cancer

4, 296–307 (2004).

49. Zhang, J., Stevens, M. F. G., Laughton, C. A., Madhusudan, S. & Bradshaw, T. D.

Acquired resistance to temozolomide in glioma cell lines: molecular mechanisms and

potential translational applications. Oncology 78, 103–114 (2010).

50. Wiewrodt, D. et al. MGMT in primary and recurrent human glioblastomas after radiation

and chemotherapy and comparison with p53 status and clinical outcome. Int. J. Cancer 122,

1391–1399 (2008).

Page 71: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

56

51. Friedman, H. S. et al. Methylator resistance mediated by mismatch repair deficiency in a

glioblastoma multiforme xenograft. Cancer Res. 57, 2933–2936 (1997).

52. Yip, S. et al. MSH6 mutations arise in glioblastomas during temozolomide therapy and

mediate temozolomide resistance. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 15,

4622–4629 (2009).

53. Wood, R. D., Mitchell, M., Sgouros, J. & Lindahl, T. Human DNA repair genes. Science

291, 1284–1289 (2001).

54. Trivedi, R. N., Almeida, K. H., Fornsaglio, J. L., Schamus, S. & Sobol, R. W. The role of

base excision repair in the sensitivity and resistance to temozolomide-mediated cell death.

Cancer Res. 65, 6394–6400 (2005).

55. Aguilar-Quesada, R. et al. Modulation of transcription by PARP-1: consequences in

carcinogenesis and inflammation. Curr. Med. Chem. 14, 1179–1187 (2007).

56. Lockett, K. L., Snowhite, I. V. & Hu, J. J. Nucleotide-excision repair and prostate cancer

risk. Cancer Lett. 220, 125–135 (2005).

57. Alvino, E. et al. A single cycle of treatment with temozolomide, alone or combined with

O(6)-benzylguanine, induces strong chemoresistance in melanoma cell clones in vitro: role

of O(6)-methylguanine-DNA methyltransferase and the mismatch repair system. Int. J.

Oncol. 29, 785–797 (2006).

58. Bradshaw, T. D. et al. Mechanisms of acquired resistance to 2-(4-Amino-3-

methylphenyl)benzothiazole in breast cancer cell lines. Breast Cancer Res. Treat. 110, 57–

68 (2008).

59. Longley, D. B. & Johnston, P. G. Molecular mechanisms of drug resistance. J. Pathol. 205,

275–292 (2005).

60. Lapidot, T. et al. A cell initiating human acute myeloid leukaemia after transplantation into

SCID mice. Nature 367, 645–648 (1994).

Page 72: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

57

61. Bonnet, D. & Dick, J. E. Human acute myeloid leukemia is organized as a hierarchy that

originates from a primitive hematopoietic cell. Nat. Med. 3, 730–737 (1997).

62. Hope, K. J., Jin, L. & Dick, J. E. Acute myeloid leukemia originates from a hierarchy of

leukemic stem cell classes that differ in self-renewal capacity. Nat. Immunol. 5, 738–743

(2004).

63. Auffinger, B. et al. Conversion of differentiated cancer cells into cancer stem-like cells in a

glioblastoma model after primary chemotherapy. Cell Death Differ. 21, 1119–1131 (2014).

64. Bleau, A.-M. et al. PTEN/PI3K/Akt pathway regulates the side population phenotype and

ABCG2 activity in glioma tumor stem-like cells. Cell Stem Cell 4, 226–235 (2009).

65. Beier, D. et al. Efficacy of clinically relevant temozolomide dosing schemes in

glioblastoma cancer stem cell lines. J. Neurooncol. 109, 45–52 (2012).

66. O’Brien, C. A., Pollett, A., Gallinger, S. & Dick, J. E. A human colon cancer cell capable of

initiating tumour growth in immunodeficient mice. Nature 445, 106–110 (2007).

67. Ricci-Vitiani, L. et al. Identification and expansion of human colon-cancer-initiating cells.

Nature 445, 111–115 (2007).

68. Al-Hajj, M., Wicha, M. S., Benito-Hernandez, A., Morrison, S. J. & Clarke, M. F.

Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. U. S. A.

100, 3983–3988 (2003).

69. Eramo, A. et al. Identification and expansion of the tumorigenic lung cancer stem cell

population. Cell Death Differ. 15, 504–514 (2008).

70. Schatton, T. et al. Identification of cells initiating human melanomas. Nature 451, 345–349

(2008).

71. Singh, S. K. et al. Identification of human brain tumour initiating cells. Nature 432, 396–

401 (2004).

Page 73: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

58

72. Galli, R. et al. Isolation and characterization of tumorigenic, stem-like neural precursors

from human glioblastoma. Cancer Res. 64, 7011–7021 (2004).

73. Sanai, N., Alvarez-Buylla, A. & Berger, M. S. Neural stem cells and the origin of gliomas.

N. Engl. J. Med. 353, 811–822 (2005).

74. Chen, J. et al. A restricted cell population propagates glioblastoma growth after

chemotherapy. Nature 488, 522–526 (2012).

75. Weller, M., Cloughesy, T., Perry, J. R. & Wick, W. Standards of care for treatment of

recurrent glioblastoma--are we there yet? Neuro-Oncol. 15, 4–27 (2013).

76. Marucci, G. et al. Pathological spectrum in recurrences of glioblastoma multiforme.

Pathologica 107, 1–8 (2015).

77. Burger, P. C. et al. Computerized tomographic and pathologic studies of the untreated,

quiescent, and recurrent glioblastoma multiforme. J. Neurosurg. 58, 159–169 (1983).

78. De Bonis, P. et al. The influence of surgery on recurrence pattern of glioblastoma. Clin.

Neurol. Neurosurg. 115, 37–43 (2013).

79. van Nifterik, K. A. et al. Genetic profiling of a distant second glioblastoma multiforme after

radiotherapy: Recurrence or second primary tumor? J. Neurosurg. 105, 739–744 (2006).

80. Kim, J. et al. Spatiotemporal Evolution of the Primary Glioblastoma Genome. Cancer Cell

28, 318–328 (2015).

81. Andor, N., Harness, J. V., Müller, S., Mewes, H. W. & Petritsch, C. EXPANDS: expanding

ploidy and allele frequency on nested subpopulations. Bioinforma. Oxf. Engl. 30, 50–60

(2014).

82. Omuro, A. M. P., Faivre, S. & Raymond, E. Lessons learned in the development of targeted

therapy for malignant gliomas. Mol. Cancer Ther. 6, 1909–1919 (2007).

83. Omuro, A. & DeAngelis, L. M. Glioblastoma and other malignant gliomas: a clinical

review. JAMA 310, 1842–1850 (2013).

Page 74: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

59

84. Lasorella, A., Uo, T. & Iavarone, A. Id proteins at the cross-road of development and

cancer. Oncogene 20, 8326–8333 (2001).

85. Massari, M. E. & Murre, C. Helix-loop-helix proteins: regulators of transcription in

eucaryotic organisms. Mol. Cell. Biol. 20, 429–440 (2000).

86. Benezra, R., Davis, R. L., Lockshon, D., Turner, D. L. & Weintraub, H. The protein Id: a

negative regulator of helix-loop-helix DNA binding proteins. Cell 61, 49–59 (1990).

87. Lyden, D. et al. Id1 and Id3 are required for neurogenesis, angiogenesis and vascularization

of tumour xenografts. Nature 401, 670–677 (1999).

88. Desprez, P. Y., Hara, E., Bissell, M. J. & Campisi, J. Suppression of mammary epithelial

cell differentiation by the helix-loop-helix protein Id-1. Mol. Cell. Biol. 15, 3398–3404

(1995).

89. Sun, X. H. Constitutive expression of the Id1 gene impairs mouse B cell development. Cell

79, 893–900 (1994).

90. Yan, W. et al. High incidence of T-cell tumors in E2A-null mice and E2A/Id1 double-

knockout mice. Mol. Cell. Biol. 17, 7317–7327 (1997).

91. Norton, J. D. ID helix-loop-helix proteins in cell growth, differentiation and tumorigenesis.

J. Cell Sci. 113 ( Pt 22), 3897–3905 (2000).

92. Zebedee, Z. & Hara, E. Id proteins in cell cycle control and cellular senescence. Oncogene

20, 8317–8325 (2001).

93. Ruzinova, M. B. & Benezra, R. Id proteins in development, cell cycle and cancer. Trends

Cell Biol. 13, 410–418 (2003).

94. Campuzano, S. Emc, a negative HLH regulator with multiple functions in Drosophila

development. Oncogene 20, 8299–8307 (2001).

Page 75: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

60

95. Morrow, M. A., Mayer, E. W., Perez, C. A., Adlam, M. & Siu, G. Overexpression of the

Helix-Loop-Helix protein Id2 blocks T cell development at multiple stages. Mol. Immunol.

36, 491–503 (1999).

96. Wice, B. M. & Gordon, J. I. Forced expression of Id-1 in the adult mouse small intestinal

epithelium is associated with development of adenomas. J. Biol. Chem. 273, 25310–25319

(1998).

97. Cai, L., Morrow, E. M. & Cepko, C. L. Misexpression of basic helix-loop-helix genes in the

murine cerebral cortex affects cell fate choices and neuronal survival. Dev. Camb. Engl.

127, 3021–3030 (2000).

98. Yokota, Y. Id and development. Oncogene 20, 8290–8298 (2001).

99. Volpert, O. V. et al. Id1 regulates angiogenesis through transcriptional repression of

thrombospondin-1. Cancer Cell 2, 473–483 (2002).

100. Hara, E. et al. Id-related genes encoding helix-loop-helix proteins are required for G1

progression and are repressed in senescent human fibroblasts. J. Biol. Chem. 269, 2139–

2145 (1994).

101. Barone, M. V., Pepperkok, R., Peverali, F. A. & Philipson, L. Id proteins control growth

induction in mammalian cells. Proc. Natl. Acad. Sci. U. S. A. 91, 4985–4988 (1994).

102. Alani, R. M., Young, A. Z. & Shifflett, C. B. Id1 regulation of cellular senescence through

transcriptional repression of p16/Ink4a. Proc. Natl. Acad. Sci. U. S. A. 98, 7812–7816

(2001).

103. Ohtani, N. et al. Opposing effects of Ets and Id proteins on p16INK4a expression during

cellular senescence. Nature 409, 1067–1070 (2001).

104. Everly, D. N., Mainou, B. A. & Raab-Traub, N. Induction of Id1 and Id3 by latent

membrane protein 1 of Epstein-Barr virus and regulation of p27/Kip and cyclin-dependent

kinase 2 in rodent fibroblast transformation. J. Virol. 78, 13470–13478 (2004).

Page 76: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

61

105. Tanaka, K. et al. Expression of Id1 results in apoptosis of cardiac myocytes through a

redox-dependent mechanism. J. Biol. Chem. 273, 25922–25928 (1998).

106. Kim, D., Peng, X. C. & Sun, X. H. Massive apoptosis of thymocytes in T-cell-deficient Id1

transgenic mice. Mol. Cell. Biol. 19, 8240–8253 (1999).

107. Parrinello, S. et al. Id-1, ITF-2, and Id-2 comprise a network of helix-loop-helix proteins

that regulate mammary epithelial cell proliferation, differentiation, and apoptosis. J. Biol.

Chem. 276, 39213–39219 (2001).

108. Fong, S. et al. Id-1 as a molecular target in therapy for breast cancer cell invasion and

metastasis. Proc. Natl. Acad. Sci. U. S. A. 100, 13543–13548 (2003).

109. Perk, J., Iavarone, A. & Benezra, R. Id family of helix-loop-helix proteins in cancer. Nat.

Rev. Cancer 5, 603–614 (2005).

110. Fong, S., Debs, R. J. & Desprez, P.-Y. Id genes and proteins as promising targets in cancer

therapy. Trends Mol. Med. 10, 387–392 (2004).

111. Kebebew, E., Treseler, P. A., Duh, Q. Y. & Clark, O. H. The helix-loop-helix transcription

factor, Id-1, is overexpressed in medullary thyroid cancer. Surgery 128, 952–957 (2000).

112. Polsky, D., Young, A. Z., Busam, K. J. & Alani, R. M. The transcriptional repressor of

p16/Ink4a, Id1, is up-regulated in early melanomas. Cancer Res. 61, 6008–6011 (2001).

113. Andres-Barquin, P. J., Hernandez, M. C. & Israel, M. A. Id4 expression induces apoptosis

in astrocytic cultures and is down-regulated by activation of the cAMP-dependent signal

transduction pathway. Exp. Cell Res. 247, 347–355 (1999).

114. Vandeputte, D. A. A. et al. Expression and distribution of id helix-loop-helix proteins in

human astrocytic tumors. Glia 38, 329–338 (2002).

115. Schindl, M. et al. Overexpression of Id-1 protein is a marker for unfavorable prognosis in

early-stage cervical cancer. Cancer Res. 61, 5703–5706 (2001).

Page 77: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

62

116. Ouyang, X. S., Wang, X., Lee, D. T. W., Tsao, S. W. & Wong, Y. C. Over expression of

ID-1 in prostate cancer. J. Urol. 167, 2598–2602 (2002).

117. Lasorella, A. et al. Id2 is critical for cellular proliferation and is the oncogenic effector of

N-myc in human neuroblastoma. Cancer Res. 62, 301–306 (2002).

118. Tournay, O. & Benezra, R. Transcription of the dominant-negative helix-loop-helix protein

Id1 is regulated by a protein complex containing the immediate-early response gene Egr-1.

Mol. Cell. Biol. 16, 2418–2430 (1996).

119. Gautschi, O. et al. Regulation of Id1 expression by SRC: implications for targeting of the

bone morphogenetic protein pathway in cancer. Cancer Res. 68, 2250–2258 (2008).

120. Ying, Q. L., Nichols, J., Chambers, I. & Smith, A. BMP induction of Id proteins suppresses

differentiation and sustains embryonic stem cell self-renewal in collaboration with STAT3.

Cell 115, 281–292 (2003).

121. Bain, G. et al. Regulation of the helix-loop-helix proteins, E2A and Id3, by the Ras-ERK

MAPK cascade. Nat. Immunol. 2, 165–171 (2001).

122. Swarbrick, A. et al. Regulation of cyclin expression and cell cycle progression in breast

epithelial cells by the helix-loop-helix protein Id1. Oncogene 24, 381–389 (2005).

123. Schuller, H. M. Is cancer triggered by altered signalling of nicotinic acetylcholine

receptors? Nat. Rev. Cancer 9, 195–205 (2009).

124. Improgo, M. R., Tapper, A. R. & Gardner, P. D. Nicotinic acetylcholine receptor-mediated

mechanisms in lung cancer. Biochem. Pharmacol. 82, 1015–1021 (2011).

125. Ponz-Sarvisé, M. et al. Inhibitor of differentiation-1 as a novel prognostic factor in NSCLC

patients with adenocarcinoma histology and its potential contribution to therapy resistance.

Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 17, 4155–4166 (2011).

126. Bounpheng, M. A., Dimas, J. J., Dodds, S. G. & Christy, B. A. Degradation of Id proteins

by the ubiquitin-proteasome pathway. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 13,

2257–2264 (1999).

Page 78: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

63

127. Kong, Y., Cui, H. & Zhang, H. Smurf2-mediated ubiquitination and degradation of Id1

regulates p16 expression during senescence. Aging Cell 10, 1038–1046 (2011).

128. Williams, S. A. et al. USP1 deubiquitinates ID proteins to preserve a mesenchymal stem

cell program in osteosarcoma. Cell 146, 918–930 (2011).

129. Alani, R. M. et al. Immortalization of primary human keratinocytes by the helix-loop-helix

protein, Id-1. Proc. Natl. Acad. Sci. U. S. A. 96, 9637–9641 (1999).

130. Ouyang, X. S. et al. Id-1 stimulates serum independent prostate cancer cell proliferation

through inactivation of p16(INK4a)/pRB pathway. Carcinogenesis 23, 721–725 (2002).

131. Wilson, J. W. et al. Expression of Id helix-loop-helix proteins in colorectal adenocarcinoma

correlates with p53 expression and mitotic index. Cancer Res. 61, 8803–8810 (2001).

132. Kim, H. et al. Id-1 regulates Bcl-2 and Bax expression through p53 and NF-kappaB in

MCF-7 breast cancer cells. Breast Cancer Res. Treat. 112, 287–296 (2008).

133. Lin, J. et al. Inhibitor of differentiation 1 contributes to head and neck squamous cell

carcinoma survival via the NF-kappaB/survivin and phosphoinositide 3-kinase/Akt

signaling pathways. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 16, 77–87 (2010).

134. Sharma, P., Patel, D. & Chaudhary, J. Id1 and Id3 expression is associated with increasing

grade of prostate cancer: Id3 preferentially regulates CDKN1B. Cancer Med. 1, 187–197

(2012).

135. Ling, M.-T., Kwok, W.-K., Fung, M. K., Xianghong, W. & Wong, Y.-C. Proteasome

mediated degradation of Id-1 is associated with TNFalpha-induced apoptosis in prostate

cancer cells. Carcinogenesis 27, 205–215 (2006).

136. Mern, D. S., Hoppe-Seyler, K., Hoppe-Seyler, F., Hasskarl, J. & Burwinkel, B. Targeting

Id1 and Id3 by a specific peptide aptamer induces E-box promoter activity, cell cycle arrest,

and apoptosis in breast cancer cells. Breast Cancer Res. Treat. 124, 623–633 (2010).

137. Coma, S. et al. Id2 promotes tumor cell migration and invasion through transcriptional

repression of semaphorin 3F. Cancer Res. 70, 3823–3832 (2010).

Page 79: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

64

138. Egeblad, M. & Werb, Z. New functions for the matrix metalloproteinases in cancer

progression. Nat. Rev. Cancer 2, 161–174 (2002).

139. Shiomi, T. & Okada, Y. MT1-MMP and MMP-7 in invasion and metastasis of human

cancers. Cancer Metastasis Rev. 22, 145–152 (2003).

140. Desprez, P. Y. et al. A novel pathway for mammary epithelial cell invasion induced by the

helix-loop-helix protein Id-1. Mol. Cell. Biol. 18, 4577–4588 (1998).

141. Takai, N., Miyazaki, T., Fujisawa, K., Nasu, K. & Miyakawa, I. Id1 expression is

associated with histological grade and invasive behavior in endometrial carcinoma. Cancer

Lett. 165, 185–193 (2001).

142. Lin, C. Q. et al. A role for Id-1 in the aggressive phenotype and steroid hormone response

of human breast cancer cells. Cancer Res. 60, 1332–1340 (2000).

143. Minn, A. J. et al. Genes that mediate breast cancer metastasis to lung. Nature 436, 518–524

(2005).

144. Ruzinova, M. B. et al. Effect of angiogenesis inhibition by Id loss and the contribution of

bone-marrow-derived endothelial cells in spontaneous murine tumors. Cancer Cell 4, 277–

289 (2003).

145. Fontemaggi, G. et al. The execution of the transcriptional axis mutant p53, E2F1 and ID4

promotes tumor neo-angiogenesis. Nat. Struct. Mol. Biol. 16, 1086–1093 (2009).

146. Jin, X. et al. EGFR-AKT-Smad signaling promotes formation of glioma stem-like cells and

tumor angiogenesis by ID3-driven cytokine induction. Cancer Res. 71, 7125–7134 (2011).

147. Lyden, D. et al. Impaired recruitment of bone-marrow-derived endothelial and

hematopoietic precursor cells blocks tumor angiogenesis and growth. Nat. Med. 7, 1194–

1201 (2001).

148. Ling, M.-T. et al. Overexpression of Id-1 in prostate cancer cells promotes angiogenesis

through the activation of vascular endothelial growth factor (VEGF). Carcinogenesis 26,

1668–1676 (2005).

Page 80: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

65

149. Niola, F. et al. Mesenchymal high-grade glioma is maintained by the ID-RAP1 axis. J.

Clin. Invest. 123, 405–417 (2013).

150. Anido, J. et al. TGF-β Receptor Inhibitors Target the CD44(high)/Id1(high) Glioma-

Initiating Cell Population in Human Glioblastoma. Cancer Cell 18, 655–668 (2010).

151. Barrett, L. E. et al. Self-renewal does not predict tumor growth potential in mouse models

of high-grade glioma. Cancer Cell 21, 11–24 (2012).

152. Ling, M.-T. et al. Id-1 expression induces androgen-independent prostate cancer cell

growth through activation of epidermal growth factor receptor (EGF-R). Carcinogenesis

25, 517–525 (2004).

153. Li, H., Gerald, W. L. & Benezra, R. Utilization of bone marrow-derived endothelial cell

precursors in spontaneous prostate tumors varies with tumor grade. Cancer Res. 64, 6137–

6143 (2004).

154. Soroceanu, L. et al. Id-1 is a key transcriptional regulator of glioblastoma aggressiveness

and a novel therapeutic target. Cancer Res. 73, 1559–1569 (2013).

155. Lee, J.-K. et al. USP1 targeting impedes GBM growth by inhibiting stem cell maintenance

and radioresistance. Neuro-Oncol. 18, 37–47 (2016).

156. Tsuchiya, T. et al. Targeting Id1 and Id3 inhibits peritoneal metastasis of gastric cancer.

Cancer Sci. 96, 784–790 (2005).

157. Henke, E. et al. Peptide-conjugated antisense oligonucleotides for targeted inhibition of a

transcriptional regulator in vivo. Nat. Biotechnol. 26, 91–100 (2008).

158. de Candia, P. et al. Angiogenesis impairment in Id-deficient mice cooperates with an Hsp90

inhibitor to completely suppress HER2/neu-dependent breast tumors. Proc. Natl. Acad. Sci.

U. S. A. 100, 12337–12342 (2003).

159. Chen, C.-H., Kuo, S.-C., Huang, L.-J., Hsu, M.-H. & Lung, F.-D. T. Affinity of synthetic

peptide fragments of MyoD for Id1 protein and their biological effects in several cancer

cells. J. Pept. Sci. Off. Publ. Eur. Pept. Soc. 16, 231–241 (2010).

Page 81: MA thesis revised - University of Toronto T-Space · Angela Celebre Master of Science Laboratory Medicine and Pathobiology University of Toronto 2016 Abstract$ Glioblastoma (GBM)

66

160. Zhang, X., Ling, M.-T., Wang, X. & Wong, Y. C. Inactivation of Id-1 in prostate cancer

cells: A potential therapeutic target in inducing chemosensitization to taxol through

activation of JNK pathway. Int. J. Cancer 118, 2072–2081 (2006).

161. Cheung, H. W., Ling, M., Tsao, S. W., Wong, Y. C. & Wang, X. Id-1-induced Raf/MEK

pathway activation is essential for its protective role against taxol-induced apoptosis in

nasopharyngeal carcinoma cells. Carcinogenesis 25, 881–887 (2004).

162. Ling, M.-T., Wang, X., Zhang, X. & Wong, Y.-C. The multiple roles of Id-1 in cancer

progression. Differ. Res. Biol. Divers. 74, 481–487 (2006).

163. Campos, B., Olsen, L. R., Urup, T. & Poulsen, H. S. A comprehensive profile of recurrent

glioblastoma. Oncogene (2016). doi:10.1038/onc.2016.85

164. Maeder, M. L. & Gersbach, C. A. Genome-editing Technologies for Gene and Cell

Therapy. Mol. Ther. J. Am. Soc. Gene Ther. 24, 430–446 (2016).


Recommended