182
IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN A PACLITAXEL-RESISTANT MODEL OF TRIPLE NEGATIVE BREAST CANCER VERA KATHERINA KOHLBAUER NATIONAL UNIVERSITY OF SINGAPORE 2016

IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

IDENTIFICATION OF NOVEL ONCOGENIC

PATHWAYS IN A PACLITAXEL-RESISTANT

MODEL OF TRIPLE NEGATIVE BREAST

CANCER

VERA KATHERINA KOHLBAUER

NATIONAL UNIVERSITY OF SINGAPORE

2016

Page 2: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS

IN A PACLITAXEL-RESISTANT MODEL OF TRIPLE

NEGATIVE BREAST CANCER

VERA KATHERINA KOHLBAUER

(M.Sc., University of Salzburg, AUSTRIA)

A THESIS SUBMITTED

FOR THE DEGREE OF DOCTOR OF PHILOSOPHY

YONG LOO LIN SCHOOL OF MEDICINE,

DEPARTMENT OF PHYSIOLOGY

NATIONAL UNIVERSITY OF SINGAPORE

2016

Page 3: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

Declaration

I hereby declare that the thesis is my original work and it has been written by me in

its entirety. I have duly acknowledged all the sources of information which have been

used in the thesis.

The thesis has also not been submitted for any degree in any university previously.

Vera Katherina Kohlbauer

7 January 2016

Page 4: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

i

Acknowledgements

First and foremost I would like to express my gratitude to my Ph.D. supervisor, Prof.

Yu Qiang, for his expert advice and encouragement throughout my Ph.D. studies.

Without his guidance and scientific knowledge, my Ph.D. would not have been

possible.

I am extremely grateful to Agency for Science, Technology and Research, A*STAR,

for sponsoring all of my Ph.D. studies as well as local and international conferences.

Furthermore, my gratitude goes to the Genome Institute of Singapore which has

provided me an inspiring and conductive environment to do my research and pursue

my project.

I would like to thank all members of the Cancer Therapeutics & Stratified Oncology

1 lab for their encouragement, constructive criticism and friendship. It was a pleasure

to work with them and I could not have asked for better colleagues. I especially have

to mention Michelle Pek and Puay Leng Lee who accompanied me throughout my

Ph.D. program. They have a special place in my heart for tolerating all my nonsense

and being a constant source of mental support. I cannot go without mentioning Gokce

Oguz, Maryam Jameelah, and Joanna Tan whose friendship and companionship mean

a lot to me and made my last 4 years so much better.

I would also like to thank Dr. Zhi Xiong who always had an open door for me to talk

about any problems or doubts I might have experienced during my studies.

Finally, I could not have completed this project without the constant love, support and

understanding of my parents and brothers.

I am but a child playing with pebbles by the sea, while vast oceans of truth lie undiscovered before me.

- Sir Isaac Newton –

Page 5: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

ii

Table of Contents

Acknowledgements ........................................................................................................ i

Table of Contents .......................................................................................................... ii

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

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

List of Abbreviations ................................................................................................... xi

List of Publications .................................................................................................... xiv

CHAPTER 1: INTRODUCTION ................................................................................. 1

1.1 Breast Cancer ................................................................................................ 2

1.1.1 Molecular profiling – genetic classification of breast cancers .............. 3

1.1.2 Hormone receptor status – breast cancer subtypes ............................... 4

1.1.2.1 Therapeutic implications ................................................................... 5

1.1.2.2 Endocrine therapy for hormone receptor positive breast cancer ....... 6

1.1.2.3 Treatment for HER2/neu+ breast cancers ......................................... 7

1.1.3 Triple Negative Breast Cancer .............................................................. 8

1.1.3.1 Cellular origin of TNBC ................................................................. 10

1.1.3.2 Treatment strategies ........................................................................ 11

1.1.3.3 Recurrence ...................................................................................... 16

1.2 Mitotic inhibitors ........................................................................................ 18

1.2.1 Paclitaxel (Taxol®) .............................................................................. 19

1.2.2 Mode of action .................................................................................... 20

1.2.3 Mitotic arrest ....................................................................................... 20

1.2.4 Mitotic catastrophe .............................................................................. 22

1.3 Chemo-resistance ........................................................................................ 24

1.3.1 Cancer stem cells ....................................................................................... 25

1.3.2 Mechanisms of chemo-resistance .............................................................. 26

1.3.3 Resistance to microtubule disrupting drugs ............................................... 29

1.4 Aurora Kinase Signaling ............................................................................. 32

1.4.1 Aurora Kinases in cell cycle ............................................................... 32

1.4.2 The Spindle Assembly Checkpoint ..................................................... 35

1.4.3 Paclitaxel-induced activation of the SAC ........................................... 37

1.4.4 Aurora Kinases in Cancer ................................................................... 37

1.5 Protein Kinase C Signaling ......................................................................... 40

1.5.1 Structure and isoforms of PKCs................................................................. 40

Page 6: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

iii

1.5.2 Activation of PKC isoforms and downstream signaling ............................ 41

1.5.3 PKCs in Cancer .......................................................................................... 43

1.5.4 PKC in Breast Cancer ................................................................................ 45

1.5.4. PKC-β ....................................................................................................... 45

1.5.5. The role of PKC-β in cell cycle regulation ............................................... 47

1.6 Aims and objectives of the study ................................................................ 49

CHAPTER 2: MATERIALS AND METHODS .................................................... 50

2.1. Cell culture and treatments .............................................................................. 51

2.2. Cryopreservation of cell lines .......................................................................... 51

2.3. Generation of Paclitaxel-resistant cell lines ..................................................... 52

2.4. Transfection of small interfering RNA ............................................................ 52

2.5. EC50 calculation ............................................................................................... 53

2.6. Cell viability assay ........................................................................................... 53

2.7. Flow Assisted Cytometry Analysis (FACS) .................................................... 54

2.8. Phospho-Histone H3 (Ser28) assay.................................................................. 54

2.9. Caspase 3 assay ................................................................................................ 55

2.10. Anchorage-independent Methylcellulose assay ............................................. 55

2.11. Tumorsphere assay......................................................................................... 56

2.12. 3D Matrigel anchorage-independent growth assay ........................................ 57

2.13. RNA extraction .............................................................................................. 57

2.14. cDNA conversion and quantitative RealTime-PCR (RT-PCR) ..................... 57

2.15. Microarray gene expression profiling ............................................................ 59

2.16. Protein extraction ........................................................................................... 59

2.17. Western blotting ............................................................................................. 60

2.18. H-score calculation ........................................................................................ 61

2.19. Statistical analysis .......................................................................................... 61

CHAPTER 3: RESULTS ......................................................................................... 62

3.1. Generation and characterization of Paclitaxel-resistant TNBC cell lines ........ 63

3.2. Gene expression profiling of SUM159PT Paclitaxel-resistant cell lines ......... 68

3.2.1. The NF-κB network .................................................................................. 69

3.3. Kinase inhibitor screen to identify Paclitaxel-sensitizing compounds ............ 74

3.4. Validation of top hits from the kinase inhibitor screen .................................... 78

Page 7: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

iv

3.5. The PKC-β and PKA inhibitors sensitize resistant cells and induce apoptosis in

the presence of Paclitaxel ........................................................................................ 83

3.6. The PKC-β inhibitor Enzastaurin sensitizes chemo-resistant cells to Paclitaxel

in anchorage-dependant and –independent conditions ........................................... 87

3.7. Enzastaurin reduces tumorsphere growth in Paclitaxel-resistant TNBC cell

lines in the presence of Paclitaxel ........................................................................... 92

3.8. PKC-α inhibitors are not capable of re-sensitizing resistant cells to Paclitaxel

................................................................................................................................ 94

3.9. PKC-β expression and protein levels in breast cancer vs. normal breast

samples .................................................................................................................... 98

3.10. Mechanism of PKC-β signaling in resistance and upon sensitization ......... 101

3.11. GSK3β inhibitors recapitulate the effect of re-sensitization by Enzastaurin 104

3.12. Paclitaxel-resistant cell lines exhibit cross-resistance to other

chemotherapeutic compounds ............................................................................... 109

3.13. Enzastaurin can restore mitotic arrest in resistant cells upon treatment with

Paclitaxel ............................................................................................................... 113

3.13.1. Paclitaxel-induced mitotic arrest and cell death in sensitive cell lines . 113

3.13.2. Paclitaxel-resistant cells over come mitotic arrest by down-regulating

Aurora Kinases.................................................................................................. 117

3.14. Aurora B levels negatively correlate with Paclitaxel sensitivity in a panel of

basal-like breast cancer cell lines .......................................................................... 123

3.15. Targeted siRNA knock-down of Aurora B inhibits re-sensitization of

Paclitaxel-induced mitotic arrest ........................................................................... 125

CHAPTER 4: DISCUSSION ................................................................................. 129

4.1. Enzastaurin: mechanism of Paclitaxel-sensitization ...................................... 130

4.2. Aurora Kinase B expression levels as a biomarker for Paclitaxel-sensitivity in

basal-like breast cancer ......................................................................................... 133

4.3. Aurora Kinase B regulation ........................................................................... 134

4.4. Mcl-1 degradation by GSK3β in the presence of Paclitaxel .......................... 136

CHAPTER 5: CONCLUSION ............................................................................... 138

5.1 Conclusion ...................................................................................................... 139

5.2 Significance of findings and future prospects ................................................. 141

REFERENCES ........................................................................................................ 144

Page 8: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

v

Summary

Triple Negative Breast Cancer (TNBC) is generally more aggressive that other

subtypes and shows early lymph node involvement. These cancers are characterized

by the lack of expression of the estrogen receptor, progesterone receptor, and the

epidermal growth factor receptor HER2/neu and, as such, don’t respond to endocrine

therapies targeting hormone receptors, or therapies with monoclonal antibodies

inhibiting downstream signaling of HER2/neu. Although they respond very well to

neoadjuvant chemotherapy, patients often suffer from relapse and distant metastasis,

coupled with resistance to chemotherapeutic drugs. In this study, we sought to

identify pathways critical for resistance and apply pharmacological methods to

sensitize cancer cells to chemotherapy.

We generated Paclitaxel-resistant TNBC cell lines, as Paclitaxel is a

chemotherapeutic agent often used in first line treatment of TNBC patients. The

TNBC cell lines, SUM159PT and MB231, were exposed to increasing concentrations

of Paclitaxel, subsequently, showed an approximately 1000-fold higher EC50 to

Paclitaxel, compared to their parental chemo-sensitive counterparts. We then used a

pharmacological approach to identify pathways which play an important role and

whose inhibition can sensitize resistant cells. Using a kinase inhibitor screen with 180

small molecule inhibitors targeting various kinases, we identified PKC-β inhibitors to

play an important role in re-sensitization. Upon validation, we found that PKC-β

inhibitors can successfully sensitize resistant TNBC cell lines to Paclitaxel, and

treatment with the PKC-β inhibitor Enzastaurin resulted in resistant cells undergoing

apoptosis in the presence of Paclitaxel. PKC-α inhibitors, on the other hand, were not

able to induce such a phenotype.

Paclitaxel induces cell death of cancer cells by stabilizing microtubules, which results

in mitotic arrest and subsequent cell death of parental, chemo-sensitive cells.

Paclitaxel-resistant cells, however, are able to surpass mitotic arrest in the presence of

Page 9: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

vi

Paclitaxel. Interestingly, treatment with Enzastaurin was able to restore mitotic arrest

resulting apoptosis of the resistant cell lines. It has been shown that activation of the

Spindle Assembly Checkpoint (SAC) is a crucial event in Paclitaxel-induced cell

death, and one of the main players in this checkpoint is Aurora Kinase B.

Interestingly, Aurora Kinase B expression levels correlate with Paclitaxel-sensitivity

only in basal-like breast cancer cell lines but not in cell lines of the luminal subtype.

As expected, we found that the resistant cell lines had lower basal levels of Aurora

Kinase B as compared to the parental cell lines, even in the presence of Paclitaxel.

However, treatment with Enzastaurin could restore Aurora Kinase B expression

which resulted in mitotic arrest and subsequent cell death in the presence of

Paclitaxel. As expected, knock-down of Aurora B inhibited cell death in the resistant

cell lines upon combination treatment.

Taken together, this study shows that the small molecule inhibitor Enzastaurin can

successfully restore Paclitaxel-sensitivity in Paclitaxel-resistant TNBC cell lines.

Enzastaurin has been well studied in clinical trials and shows low toxicity in patients,

therefore using this inhibitor in combination with Paclitaxel is a potential therapeutic

strategy in treating patients with TNBC suffering from metastatic and recurrent

disease.

Page 10: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

vii

List of Tables

Table 2.13 Primers used for quantitative RealTime-PCR of target genes

Page 11: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

viii

List of Figures

Chapter 1

Figure 1.1 Distribution of breast cancer subtypes diagnosed in women in Atlanta

(USA) from 2003-2004

Figure 1.2 Intrinsic subtypes of Triple Negative Breast Cancer

Figure 1.3 Model of the human mammary epithelial hierarchy linked to cancer

subtype

Figure 1.4 Survival after a diagnosis of breast cancer

Figure 1.5 Cell fate in response to anti-mitotic drug treatment

Figure 1.6 Schematic representation of human Aurora A, B and C

Figure 1.7 Localization of Aurora kinase A and B together with PLK1 in the dividing

cell

Figure 1.8 Schematic sequence of Protein Kinase C (PKC) isozymes indicating the

domain structure of the PKC subfamilies and their activators

Chapter 3

Figure 3.1.1 Generation of Paclitaxel-resistant TNBC cell lines

Figure 3.1.2 Increase in the cancer stem cell population in Paclitaxel-resistant cell

lines

Figure 3.2.1 Changes in functions and pathways in Paclitaxel-resistant TNBC cell

lines

Figure 3.2.2 The NF-κB network in Paclitaxel-resistant cell lines

Figure 3.2.3 SUM159PT Paclitaxel-resistant cells are equally sensitive to NF-κB

inhibitors as compared to the parental cell line

Figure 3.3.1 Kinase inhibitor screen identifies PKC-β inhibitors to be able to re-

sensitize resistant cells

Figure 3.3.2 Top hit of the kinase inhibitor screen

Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen

Figure 3.4.2 PKC-β and PKA inhibitors can inhibit colony formation of Paclitaxel-

resistant cell lines in the presence of Paclitaxel

Figure 3.5.1 PKC-β and PKA inhibitors induce Caspase 3 activation in the presence

of Paclitaxel

Figure 3.5.2 PKC-β and PKA inhibitors increase SubG1 fraction in Paclitaxel-

resistant cell lines in the presence of Paclitaxel

Page 12: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

ix

Figure 3.6.1 The PKC-β inhibitor Enzastaurin sensitizes resistant cells to Paclitaxel in

a dose-dependent manner

Figure 3.6.2 Enzastaurin inhibits 3D colony growth in resistant cell lines in the

presence of Paclitaxel

Figure 3.6.3 Enzastaurin treatment drastically reduces Paclitaxel EC50 in SUM159PT

resistant cell lines

Figure 3.7 Enzastaurin in combination with Paclitaxel inhibits tumorsphere growth in

Paclitaxel-resistant cells

Figure 3.8.1 PKC-α inhibitors are not able to sensitize Paclitaxel-resistant cells in the

same manner as Enzastaurin

Figure 3.8.2 PKC-α inhibitors have no effect on colony formation of SUM159PT

Paclitaxel-resistant cells in the presence of Paclitaxel

Figure 3.9 PKC-β is highly expressed in breast cancer compared to normal breast

tissue in patient samples and cell lines

Figure 3.10 Western blot analysis of down-stream signaling of Enzastaurin treatment

on PKC-β, GSK3β and Mcl-1 in SUM159PT and MB231

Figure 3.11.1 Western blot analysis of the effect of GSK3β inhibitors in MB231

parental and resistant cells

Figure 3.11.2 A set of GSK3β inhibitors reduce colony formation in the resistant cells

in the presence of Paclitaxel

Figure 3.11.3 GSK3β activation leads to increased cell death in the resistant cells in

the presence of Paclitaxel

Figure 3.12.1 Paclitaxel-resistant cells exhibit cross-resistance to Vincristine which

can be reversed with Enzastaurin

Figure 3.12.2 Paclitaxel-resistant cells exhibit cross-resistant to Adriamycin

Figure 3.13.1 Paclitaxel induces mitotic arrest and subsequent cell death in MB231

parental cells

Figure 3.13.2 Up-regulation of Aurora Kinase B is crucial for Paclitaxel-induced cell

death

Figure 3.13.3 Enzastaurin restores levels of Aurora B in resistant cell lines upon

treatment with Paclitaxel

Figure 3.13.4 Various Aurora Kinase inhibitors can rescue cell death upon

combination treatment of Paclitaxel with Enzastaurin in Paclitaxel-resistant cell lines

Figure 3.13.5 Aurora Kinase B inhibition rescues cell death upon re-sensitization of

Paclitaxel-resistant cells by Enzastaurin

Page 13: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

x

Figure 3.14 Aurora Kinase B expression negatively correlates with Paclitaxel

sensitivity in a panel of basal breast cancer cell lines

Figure 3.15.1 Aurora Kinase B is crucial for Paclitaxel-induced inhibition of cell

viability in parental and resistant SUM159PT and MB231 cell lines

Figure 3.15.2 Aurora Kinase B abolished Paclitaxel-induced mitotic arrest

Chapter 4

Figure 4.1 Increased expression of ABC transporters in Paclitaxel-resistant

SUM159PT cell lines

Figure 4.2 Major regulators of Aurora Kinase B

Page 14: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

xi

List of Abbreviations

ABC ATP-binding cassette domain

ABCB1 ATP-Binding Cassette, Sub-family B, Member 1

ADCC Antibody-Dependent Cellular Cytotoxicity

ADR Adriamycin

AI Aromatase Inhibitor

Akt Protein Kinase B (PKB)

ALDH Aldehyde Dehydrogenase

APC Allophycocyanin

APC/C Anaphase-Promoting Complex/Cyclosome

ASCO American Society of Clinical Oncology

ATP Adenosin Triphosphate

AURKA/AURKB Aurora Kinase A/B gene

Bcl-2 B-Cell Lymphoma 2

Bcl-XL B-Cell Lymphoma-extra large

BRCA1/2 Breast Cancer 1/2, early onset

BTK Bruton’s Tyrosine Kinase

BUB1 Mitotic Checkpoint Serine/Threonine Kinase

CDK Cyclin-Dependent Kinase

CENP-E Centrosome-Associated Protein E

CHK2 Checkpoint Kinase 2

CK5/6, 14, 17 Cytokeratin 5/6, 14, 17

CPC Chromosomal Passenger Complex

CRISPR Clustered Regularly Interspaced Short Palindromic

Region

CSC Cancer Stem Cell

CTG CellTiterGlo®

CTGF Connective Tissue Growth Factor

DAG Diacylglycerol

DCIS Ductal Carcinoma In Situ

DFS Distant metastasis-Free Survival

DLBCL Diffuse Large B-Cell Lymphoma

DMSO Dimethyl Sulfoxide

DNA Deoxyribonucleic Acid

EGF Epidermal Growth Factor

EGFR Epidermal Growth Factor Receptor

EMT Epithelial-Mesenchymal Transition

EpCAM Epithelial Cellular Adhesion Molecule

ER Estrogen Receptor

ERK Extracellular signal-Regulated Kinase

ESA Epithelial Specific Antigen

FAK Focal Adhesion Kinase

Page 15: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

xii

FBW7 F-Box and WD repeat domain-containing 7

FDA Food and Drug Administration

FITC Fluorescein Isothiocyanate

FLT3 Fms-related Tyrosine Kinase 3

GDP/GFP Guanosine Di-/Triphosphate

GOBO Gene expression-based Outcome for Breast cancer

Online

GPCR G-Protein Coupled Receptor

GSK3β Glycogen Synthase Kinase 3β

HER2, ERBB2 Receptor Tyrosine-Protein Kinase erbB-2

Hh Hedgehog

HR Homologous Recombination

IKK Inhibitor of IκB Kinase

INCENP Inner Centromere Protein

INT Tetrazolium

IPA Ingenuity® Pathway Analysis

JNK Jun-terminal Protein Kinase

LiCl Lithium Chloride

LOH Loss Of Heterozygosity

MAD1/2 MAX Dimerization Protein 1/2

MAP Microtubule-Associated Protein

MAPK, MEK Mitogen-Activated Protein Kinase

MBC Metastatic Breast Cancer

MCC Mitotic Checkpoint Complex

Mcl-1 Myeloid Cell Leukemia 1

MDM2 Mouse Double Minute 2 homolog

MDR1 Multidrug Resistance Protein 1

MSK1 Mitogen and Stress-activated Protein Kinase

MYC V-Myc Avian Myelocytomatosis Viral Oncogene

Homolog

mRNA Messenger RNA

NF-κB Nuclear Factor-κB

NCCN National Comprehensive Cancer Network

NCI National Cancer Institute

ORR Overall Response Rate

OS Overall Survival

P-gp P-glycoprotein

p-H3 Phospho-Histone H3

PARP Poly(ADP-ribose) polymerase

pCR Pathological Complete Response

PCR Polymerase Chain Reaction

PDGFR Platelet-Derived Growth Factor Receptor

PDK1 Pyruvate Dehydrogenase Kinase, isozyme 1

Page 16: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

xiii

PFS Progression-Free Survival

PI3K Phosphoinositide 3-Kinase

PKA Protein Kinase A

PI Propidium Iodide

PKC Protein Kinase C

PLC Phspholipase C

PLK1 Polo-Like Kinase 1

PR Progesterone Receptor

PRKCB Protein Kinase C β gene

p53, p63, p38 Protein53/63/38

PTEN Phosphatase and Tensin Homolog

RACK Receptor for Activated Kinase

RICK Receptor for Inactivated Kinase

RKIP Raf Kinase Inhibitory Protein

RNA Ribonucleic Acid

RFS Relapse-Free Survival

SAC Spindle Assembly Checkpoint

SASP Senescence Associated Secretory Phenotype

SCID Severe Combined Immunodeficiency

SERM Selective Estrogen Receptor Modulatir

TAZ Transcriptional Co-activator with PDZ-binding motif

TGF-β Transforming Growth Factor β

TNBC Triple Negative Breast Cancer

TNKS1/2 Tankyrase 1/2

VCR Vincristine

VEGF Vascular Endothelial Growth Factor

Page 17: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

xiv

List of Publications

1. Zhen Ning Wee, Siti Maryam J. M. Yatim, Vera K Kohlbauer, Min Feng,

Jian Yuan Goh, Yi Bao, Puay Leng Lee, Songjing Zhang, Pan Pan Wang,

Elgene Lim, Wai Leong Tam, Yu Cai,Henrik J Ditzel, Dave S. B. Hoon, Ern

Yu Tan & Qiang Yu, 2015, IRAK1 is a therapeutic target that drives breast

cancer metastasis and resistance to paclitaxel, Nat Commun. 6:8746

2. Conference paper: Vera K Kohlbauer & Qiang Yu, Identification of novel

oncogenic pathways in a Paclitaxel-resistant model of Triple Negative Breast

Cancer. Submitted for:

EACR Conference Series 2015, Cancer Genomics (July 2015),

Cambridge, UK

Cell Symposia: Hallmarks of Cancer: Asia (November 2014),

Beijing, China

Gordon Research Conferences: Cell Growth & Proliferation (June

2013), Vermont, USA

Page 18: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

1

CHAPTER 1: INTRODUCTION

Page 19: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

2

1.1 Breast Cancer

Breast cancer is the most common cancer among women (1 in 3 cancers diagnosed)

and the second most common cause of cancer deaths. According to the American

Cancer Society ACS (Cancer Facts & Figures 2015), it is estimated that in the United

States 231,840 women will be diagnosed with invasive breast cancer and 40,730

women will die from it in 2015. Breast cancer arises from ductal or lobular tissues

and is confined to these two regions at the initial stage of development and is

therefore termed in situ carcinoma. At this stage, the cancer is treatable (surgical

removal) with highly positive outcomes. However, if it is not detected it becomes

invasive and infiltrates surrounding tissue leading to cancers with aggressive

phenotypes with a high rate of metastasis and poor outcome.

In general, breast cancer is more common among women of older age, but most cases

diagnosed in younger women (age 35 and below) are of high grade, estrogen receptor

positive (Gonzalez-Angulo et al., 2005) and hereditary. About 40% of all familial

breast cancer cases are caused by genetic predispositions from mutations in high

penetrance genes such as BRCA1 and BRCA2, whereas as low prevalence genes like

TP53, CHK2 or PTEN (Kenemans et al., 2008) account for the rest. Mammographic

screening was introduced to women in order to detect breast cancers early on and

improve outcome, but there is much controversy around this topic, since many studies

achieved different results. Some studies reported a decrease in breast cancer mortality

(Gotzsche and Olsen, 2000) with regular screening in randomized trials, whereas

others claim that regular screening comes with such high cost (length bias, lead-time

bias and over-diagnosis), that they are not justifiable (Berry, 2013).

Breast cancers are very heterogenous with substantial morphological, pathological

and genetic variation. Therefore, classifying them into different subtypes provides

important implications for diagnosis and therapy. This can be done based on gene

Page 20: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

3

expression profiles of different breast cancers, or based on their hormone receptor

status.

1.1.1 Molecular profiling – genetic classification of breast cancers

Microarrays enable gene expression profiling of thousands of genes in different

cancers and allow us to create molecular profiles of a tumor. Perou et al. examined

the expression levels of 8,102 genes in 65 breast cancer samples and classified then

into different intrinsic subtypes: Luminal, Basal-like, HER2+ and Normal breast

(Perou et al., 2000). Later it was found that the Luminal subtype can be further

divided into Luminal A and Luminal B. Luminal breast cancers are characterized by a

high expression of hormone receptors and hormone receptor-associated genes,

HER2+ cancers have a high expression of HER2 and a low expression of the estrogen

receptor (ER), and are more likely of high grade. Basal-like cancers manifest with a

high expression of basal epithelial genes, but low expression of ER and HER2, and

generally come with poor prognosis.

Notably, the most important clinical implication of breast cancer subtyping is for

prognosis and treatment. To date, there are various genomic assays available for

breast cancer, which give information on the type of cancer and the therapy which

will be most beneficial. Such platforms include Oncotype DX®, MammaPrint® and

Mammostrat®. Oncotype DX is a diagnostic test to estimate disease recurrence in

breast cancer patients and if they will benefit from chemotherapy after surgery. It is

for women with early stage, estrogen receptor (ER) positive breast cancer and takes

into account the gene expression levels of 21 genes (16 cancer genes and 5 reference

genes). It also calculates the recurrence risk for DCIS (Ductal Carcinoma In Situ).

Clinical studies are still ongoing with Oncotype DX. MammaPrint is based on the

expression levels of 70 genes in the Amsterdam 70-gene breast cancer signature to

calculate the risk if a patient will develop metastases if given chemotherapy. It has

Page 21: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

4

been FDA-approved for early stage, lymph node negative, hormone receptor-positive

or -negative breast cancer and is carried out on paraffin-embedded or fresh tumor

samples. The Mammostrat test uses the score of 5 genes based on their expression

levels and assigns patients into different risk groups (high, moderate, low) to measure

their risk of recurrence. So far, only Oncotype DX has been included in the NCCN

and ASCO treatment guidelines.

1.1.2 Hormone receptor status – breast cancer subtypes

Although gene expression profiling of breast cancer can be used for predicitive and

prognostic purposes, it is still expensive and not widely available yet. Other than gene

expression profiling, the status of two hormone receptors can also be used to

distinguish different breast cancer subtypes. The different subtypes vary in the

expression status of the estrogen receptor (ER) and progesterone receptor (PR), and

the epidermal growth factor (EGF) receptor HER2/neu. Breast cancer can be

classified into four different subtypes based on their receptor expression status:

1. Luminal A: ER+ and/or PR+, HER2/neu-

2. Luminal B: ER+ and/or PR+, HER2/neu+

3. HER2+: ER- and PR-, HER2/neu+

4. Triple Negative Breast Cancer (TNBC): ER-, PR- and HER2/neu-

Out of these four, Luminal A is the most common subtype with 66.9%, followed by

TNBC (18.8%), Luminal B (8.3%), and HER2/neu+ (6.0%) (Lund et al., 2010).

Figure 1.1 shows the distribution of breast cancer subtypes in women diagnosed with

breast cancer from 2003-2004 where information on receptor status was available at

time of diagnosis.

Page 22: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

5

Furthermore, incidence rates for TNBC were higher in black women compared to

white women, and TNBC is more common among younger women (age 35 and

below) (Carvalho et al., 2010).

1.1.2.1 Therapeutic implications

Subtyping breast cancer according to ER, PR and HER2/neu has therapeutic

implications since endocrine therapies can be used to treat hormone receptor positive

tumors and HER2/neu can be blocked with monoclonal antibodies. TNBC, on the

other hand, does not respond to any endocrine therapies or treatment with monoclonal

antibodies, and the predominant form of treatment to this day is chemotherapy. In

general, hormone receptor positive patients have a higher rate of survival compared to

patients lacking hormone receptor expression (Fisher et al., 1988, Crowe et al., 1991)

and adjuvant hormonal/ chemotherapeutic treatment can further increase survival rate

in these patients (Fisher et al., 2004).

Figure 1.1 Distribution of breast cancer subtypes diagnosed in women in Atlanta (USA)

from 2003-2004. Adapted from Lund et al. (Lund et al., 2010).

Page 23: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

6

1.1.2.2 Endocrine therapy for hormone receptor positive breast cancer

Estrogen receptor signaling plays an important role in the normal development and

growth of the breast tissue. The crucial role of estrogen in breast cancer was

discovered over 100 years ago, when mortality rates for breast cancer patients

decreased after their ovaries had been removed.

ER+ breast cancers are dependent on positive signaling from activated ERs and when

this signaling is shut off, it can lead to reduction of hormone-dependent proliferation

and cell death. The ER status can be seen as a biomarker for responsiveness to

endocrine therapies. Therapies can either target ER signaling directly using pure

antagonists and selective estrogen receptor modulators (SERMs), or indirectly using

aromatase inhibitors (AIs) which block estrogen production.

There are two estrogen receptors, ERα and ERβ, that share 56% similarity in ligand

binding domains, and nearly identical DNA binding domains. However, they possess

different gene expression profiles and only a minority of genes regulated by ERβ is

also regulated by ERα. (Grober et al., 2011). ERβ expression is more dominant in

mammary tissues. It is expressed in epithelial and stromal cells, while ERα

expression is only found in a subset of epithelial cells. However, ERα is upegulated in

a large number of breast cancers and is linked to prognosis and response to hormone

therapy, in contrast to ERβ expression (Roger et al., 2001, Zhao et al., 2008). The

most commonly used antagonist targeting ERα is Tamoxifen, which binds to the

receptor without activating it. Tamoxifen is a selective estrogen receptor modulator as

it can act as an agonist in other tissues, such as the endometrium. It is standard

therapy for ER+ tumors in premenopausal women and on occasion used in

postmenopausal women. In addition to Tamoxifen, AIs are also effective in treating

ER+, early stage breast cancers (Harvey et al., 1999). The aromatase enzyme is

needed to convert androgen to estrogen and inhibition of its activity inhibits estrogen

Page 24: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

7

production in the ovaries, thereby starving ERs of estrogen. AIs, such as anastrozole

and letrozole, have been approved for treatment of ER+ tumors in postmenopausal

women.

1.1.2.3 Treatment for HER2/neu+ breast cancers

Patients diagnosed with HER2/neu+ breast cancer show low overall survival (OS)

and relapse-free survival (RFS), and poor prognosis. Overexpression of HER2 leads

to increased activation of the MAPK and PI3K/Akt pathways, which leads to

increased cell proliferation and survival (Yarden and Sliwkowski, 2001). Many of

these patients respond well to treatment with Tratuzumab (Herceptin) which is a

humanized monoclonal antibody targeting the extracellular juxtamembrane domain

IV of HER2. Trastuzumab leads to a decrease in HER2 signaling followed by

inhibition of cell proliferation, while simultaneously inducing antibody-dependent

cellular cytotoxicity (ADCC) as it can activate immune effector cells (natural killer

cells) (Yamauchi et al., 2011, Spector and Blackwell, 2009). Furthermore, it can

inhibit the MAPK and PI3K pathways and inhibit angiogenesis (Nahta, 2012).

Despite it’s positive effects for patients with primary HER2+ breast cancer, many

tumors show resistance to single-agent treatment with Trastuzumab with a low

median survival of 9 months (Nahta, 2012). Trastuzumab in combination with

chemotherapy in an adjuvant setting can increase disease-free survival for patients

with early stage breast cancer (Romond et al., 2005). Metastatic breast cancer (MBC)

also shows a low response rate of 12-34% to Trastuzumab treatment as a single agent.

However, clinical trials have shown that combination treatment of Trastuzumab with

chemotherapy (Paclitaxel or Docetaxel) can increase response rates and OS (Esteva et

al., 2002). Similar effects were achieved when combined with other chemotherapeutic

agents such as cisplatin, carboplatin or ionizing radiation (Slamon et al., 2001).

Page 25: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

8

This thesis focuses on TNBC, so we will describe this subtype in more detail in the

next part of the introduction.

1.1.3 Triple Negative Breast Cancer

TNBCs are more aggressive than other subtypes of breast cancer in terms of relapse

rates, tumor grade, and lymph node involvement (Haffty et al., 2006). Moreover, less

than 30% of patients diagnosed with metastatic TNBC have a 5 year survival rate due

to the lack of effective targeted therapies (Dent et al., 2007).

Approximately 70%-80% of all Basal-like breast cancers are also triple negative and

more than 80% of TNBC are basal-like, therefore these 2 terms are often used

interchangeably to describe the same breast cancer subtype (Weigelt et al., 2010). The

group of TNBCs can also encompass tumors that are of : (1) the claudin-low subtype,

which have a high percentage of cells that have stem-like properties and show

features of EMT; (2) the interferon-rich subgroup which has better prognosis than

TNBC tumors; and (3) the normal-like subgroup with a high content of normal and

stromal cells (Sotiriou and Pusztai, 2009). In addition, approximately 20% of basal-

like tumors express ER or HER2/neu (Turner et al., 2010).

Various immunohistochemical panels have been proposed to distinguish basal-like

subtypes, such as: (1) lack of the ER, PR and HER2/neu (TNBC); (2) expression of at

least one high molecular weight/basal cytokeratin (CK5/6, CK14, CK17); (3) lack of

ER and HER2/neu expression together with the expression of CK5/6 and/or EGFR

(Nielsen et al., 2004); (4) lack of ER, PR and HER2/neu expression together with

expression of CK5/6 and/or EGFR (Cheang et al., 2008). Despite the presence of

various different definitions of basal-like breast cancer, they have distinct clinical

Page 26: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

9

presentation, histological features, response to chemotherapy, sites of distant relapse,

and outcome.

Even among TNBC cancers there is great heterogeneity. Lehmann et al. analyzed

gene expression profiles of 587 TNBC breast cancer cases from 21 databases and

performed clustering analysis (Lehmann et al., 2011). He found that, depending on

the gene expression profiles, the TNBC cases could be further classified into six

different subtypes, which are listed in the table below.

Subtype Gene expression profile

Basal-like 1 (BL-1) High in the expression of genes involved in cell cycle progression,

cell division, and DNA damage response pathway

Basal-like 2 (BL-2) High in the expression of genes involved in cell cycle progression,

cell division, and growth factor signaling

Immunomodulatory

(IM)

High in the expression of genes involved in immune processes and

cell signaling

Mesenchymal (M) High in the expression of genes involved in motility and

extracellular matrix

Mesenchymal stem-

like (MSL)

High in the expression of genes involved in motility, extracellular

matrix, and growth factor signaling

Luminal androgen

receptor (LAR)

High in the expression of genes involved in hormonally regulated

pathways

However, basal-like breast cancers and TNBC share many similarities in terms of

clinical prognosis and therapeutic responses (Badve et al., 2011). These cancers show

lymph node involvement, high rate of recurrence, and are biologically more

aggressive (Thike et al., 2010). Interestingly, basal-like breast cancers respond well to

neoadjuvant chemotherapy with higher pathological complete response (pCR)

compared to other subtypes (Lin et al., 2010). However, rates of distant metastasis

and recurrence are higher for these patients.

Figure 1.2 Intrinsic subtypes of Triple Negative Breast Cancer

Six subtypes of Triple Negative Breast Cancer based on gene expression profiling (Peddi et al.,

2012, Lehmann et al., 2011).

Page 27: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

10

Although the overlap between basal-like breast cancer and TNBC is not complete,

many papers still use these terms interchangeably. Therefore, I will summarize the

findings for both in a collective manner.

1.1.3.1 Cellular origin of TNBC

The fact that basal-like breast cancer cells possess breast stem cell-like properties

gives rise to the question whether these tumors arise from breast stem cells. To date

there are many reports on the epithelial hierarchy in the human breast where an

undifferentiated ER-/PR-/HER2/neu- mammary stem cell (MaSC), capable of self

renewal and differentiation, gives rise two committed progenitors: the myoepithelial

and luminal progenitors (Figure 1.3). The myoepithelial progenitor then gives rise to

mature myoepithelial cells that surround the luminal epithelium and contact the

basement membrane. The luminal progenitor can be ER- or ER+ and will give rise to

mature ductal and alveolar cells which line the lumen of the mammary gland and are

also ER- or ER+ (Lim et al., 2009). Two different cell surface markers can be used to

identify different subpopulations of cells in the breast: CD49f and EpCAM. When

performing FACS analysis on highly purified subpopulations of MasSC/bipotent cells

(CD49f+/EpCAM-), committed luminal progenitors (CD49f+/EpCAM+) and mature

ER+/luminal cells (CD49f-/EpCAM+), it was found that the gene signature of the

luminal progenitors was very similar to the basal-like gene signature (Lim et al.,

2009). Together with the findings that BRCA1-mutated pre-neoplastic breast tissues,

which have a >80% chance to develop into basal-like breast cancer, show an

increased population of luminal progenitor cells, these findings suggest that the cell

of origin of sporadic and BRCA1-mutated basal-like breast cancer is the luminal

progenitor (Figure 1.3).

Page 28: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

11

1.1.3.2 Treatment strategies

Patients with TNBC do not benefit from endocrine therapies targeting ER and PR, or

therapies using monoclonal antibodies to target HER2/neu, as they lack expression of

these specific hormone and EGF receptors. The mainstay of treatment until now is

surgery and chemotherapy, or a combination of both. Some newer studies have

identified certain receptors which play a role in tumorigenesis and progression of

TNBC, and which could be potential targets for targeted therapies. Such receptors are

EGFR and c-Kit, where it was found that c-Kit is highly expressed in 31% of basal-

like tumors and 11% of non-basal-like tumors (Nielsen et al., 2004). Moreover, 66%

of patients triple negative tumor cells and basal-like tumor cells have been shown to

Figure 1.3 Model of the human mammary epithelial hierarchy linked to cancer subtype

Subpopulations of normal breast tissue and potential cells of origin for the intrinsic subtypes;

various breast cancer subtypes molecularly compared to subpopulations from normal breast

tissue; defining expression patterns of luminal, mesenchymal or claudin-low, and basal-like

cells (Prat and Perou, 2009).

Page 29: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

12

express EGFR, suggesting a therapeutic role for targeting EGFR (Reis-Filho et al.,

2005). Despite the promising preclinical data, clinical studies combining cetuximab,

an anti-EGFR antibody, with carboplatin in stage IV triple negative breast cancer

patients, showed that less than 20% of patients showed inhibition of the EGFR

pathway, suggesting alternative activation mechanisms (Carey et al., 2012).

We will look closer into the therapies available for TNBC patients.

Chemotherapy

Anthracyclines Anthracyclines are the most common chemotherapeutic drugs used

for the treatment of TNBC patients. These compounds intercalate between base pairs

and inhibit DNA and RNA synthesis preventing replication. They also inhibit

topoisomerase II, an enzyme involved in the relaxation of supercoiled DNA, leading

to DNA double strand breaks, which leads to activation of the DNA damage response

signaling cascade (Nitiss, 2009). If DNA repair fails, the cells will undergo apoptosis.

More recent studies have found that anthracyclines also induce histone eviction from

chromatin and subsequent deregulation of the DNA damage response and DNA repair

(Pang et al., 2013). Many studies show that TNBC patients are sensitive to

anthracycline-containing therapies. The clinical response to therapy containing

doxorubicin (an anthracycline) and cyclophosphamide, was higher in TNBC patients

compared with non-TNBC patients in terms of pCR, although overall outcome (OS

and DMFS) was still worse in comparison with non-TNBC patients (Carey et al.,

2007).

Taxanes TNBCs show a high rate of genomic instability, due to defective pathways,

such as mutations in BRCA1 (discussed above), therefore, using Taxanes may benefit

these patients. Taxanes are spindle poisons which stabilize microtubules, and it was

found that TNBC and HER2/neu+ patients are more sensitive to doxorubicin and

Paclitaxel when used in a neoadjuvant setting as compared to luminal- and normal-

Page 30: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

13

like breast cancer patients (Rouzier et al., 2005). The addition of Paclitaxel to

cyclophosphamide and doxorubicin could also enhance DMFS and OS in node

positive, as well as TNBC and HER2/neu+ patients (Hayes et al., 2007). Several

studies show improved outcome for patients with the addition of a taxane to the

standard chemotherapy routine. One study showed great benefit with the addition of

docetaxel in TNBC patients receiving conventional 6 cycles of FEC (5-Fluorouracil,

epirubicine, cyclophosphamide) (Martin et al., 2010), while another study found that

weekly paclitaxel is much more effective than paclitaxel every 3 weeks (Sparano et

al., 2008).

Platinum agents The mechanism of action for platinum agents, such as carboplatin

and cisplatin, is intra- and inter-strand cross-linking of double-stranded DNA, thereby

preventing formation of the replication fork and production of double strand breaks.

Because TNBCs often harbor BRCA1 mutations or silencing, the DNA repair

cascade is not fully functional and the cells will undergo cell death (Hastak et al.,

2010). One study showed that neoadjuvant cisplatin as a single agent in TNBC

patient showed a pCR of 22% (50% of patients had a partial response) (Silver et al.,

2010). Another study found that 9 out of 10 patients with BRCA1 mutations achieved

pCR after neoadjuvant therapy with cisplatin (Byrski et al., 2009). Cisplatin

sensitivity is mediated by a p63-dependent tumor survival pathway in TNBC cells

(Leong et al., 2007) and, importantly, cisplatin yielded a higher pCR rate in patients

with p63+ tumors (Rocca et al., 2008). In a meta-analysis of randomized-controlled

trials of platinum agents for TNBC patients it was found that platinum-based

therapies yielded significantly better response rates and pCR rates compared to

therapies without platinum (Guan et al., 2015).

Combination of platin with a taxane-based primary chemotherapy has also shown to

be effective in patients with locally advanced breast cancer (Frasci et al., 2009).

Neoadjuvant cisplatin combined with paclitaxel and epirubicin in a weekly schedule

Page 31: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

14

led to a 65% pCR. Moreover, when cisplatin was used in combination with epirubicin

and 5-Fluorouracil a complete clinical response of 88% was achieved (Sirohi et al.,

2008). Therefore, when patients show resistance or high toxicity to therapies with

anthracyclines, platinum-based therapies are a good alternative.

Targeted therapies

PARP inhibitors Poly(ADP-ribose) polymerase (PARP) is an enzyme involved in

DNA repair. Upon single strand breaks, PARP is recruited and its activation leads to

single strand repair. However, when PARP is inhibited, the repair system can no

longer be activated, leading to a stalling of the replication fork. The stalled replication

fork will collapse and induce a double strand break in the DNA, which can be

repaired by homologous recombination (HR). If the replication restarts, then the cell

will survive. In BRCA-deficient cells where homologous recombination is impaired,

the double strand breaks cannot be repaired, subsequently leading to cell death

(Livraghi and Garber, 2015).

The rationale for using PARP inhibitors in the treatment of TNBC comes from the

observations that breast cancers with mutated BRCA show impairments of the HR

pathway (Tutt et al., 2005) and some sporadic TNBCs harbor BRCA1 mutations

(Turner et al., 2004). Others lack BRCA1 mutations but may show genetic profiles

that are similar to tumors arising in BRCA1 carriers (van Beers et al., 2005), often

times because they have defective BRCA1 pathways. This is partly due to CpG island

methylation of the BRCA1 promoter and subsequent deactivation. Promoter

methylation of one allele is found in 11-14% of sporadic breast cancers (Jones and

Baylin, 2002) and often the second allele is lost due to loss of heterozygosity (LOH).

This leads to the absence of BRCA1 expression and an inhibition of the pathway.

Phase I and II clinical trials revealed that treatment with PARP inhibitors lead to

improved outcomes as monotherapy in breast cancers with BRCA1/2 mutation, as

Page 32: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

15

well as in ovarian cancers, and pancreatic and prostate cancers with confirmed

BRCA1/2 mutations (Fong et al., 2009, Kaufman et al., 2015). Even in combination

therapy, PARP inhibitors showed promising results especially when given together

with platin-based agents in BRCA-related breast cancers (Balmana et al., 2014).

Moreover, there are randomized phase III studies ongoing to test PARP inhibitors vs.

single agent chemotherapy in breast cancer patients with metastatic disease and

BRCA1/2 germline mutations (https://clinicaltrials.gov/ct2/show/NCT01905592,

https://clinicaltrials.gov/ct2/

show/NCT01945775, https://clinicaltrials.gov/ct2/show/NCT02163694).

EGFR inhibitors EGFR is often over-expressed in TNBC, therefore it is one of the

targets in treatment for these patients. However, the results have not been positive. In

a more recent phase II randomized study, metastatic TNBC patients were treated with

a chimeric monoclonal antibody targeting EGFR alone (cetuximab), or in

combination with carboplatin (Carey et al., 2012), however, there were no significant

differences in PFS and both treatment groups showed rapid disease progression.

Despite these results, there are still phase I and II clinical trials ongoing with

cetuximab in combination with cytotoxic agents in metastatic TNBC.

VEGF inhibitors VEGF was found to be higher expressed in TNBC compared with

non-TNBC tumors, and that therapy with a recombinant humanized monoclonal

antibody targeting VEGF, bevacizumab, could cause tumor suppression (Yadav et al.,

2014). However, clinical trials were not conclusive. Although some trials showed

improved outcome in terms of ORR and PFS when adding bevacizumab to Paclitaxel

in first-line treatment for TNBC patients (Gray et al., 2009), or in addition with

docetaxel where it lead to an improvement in PFS (Miles et al., 2010), trials for the

use in second-line therapy for metastatic TNBC showed improved RR and PFS, but

not OS (http://meetinglibrary.asco.org/content/78329-102). Moreover, patients

receiving bevacizumab-based treatments in first-line treatment experienced high

Page 33: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

16

toxicity, hence its approval has been withdrawn due to a modest risk-benefit ratio.

There is still one ongoing trial using bevacizumab in combination with an

anthracycline-taxane-based chemotherapy in TNBC patients (Gerber et al., 2013).

Although 40.1% patients showed pCR with improved DFS, only long-term studies

will show whether patients experience OS.

Tyrosine kinase inhibitors Other inhibitors targeting various kinases, such as c-Kit

and other tyrosine kinases are being validated. However, the effect of c-Kit inhibitors,

such as imatinib, in patients with TNBC and basal-like tumors is not clear as there are

problems with the specificity of the antibodies in immunohistochemical staining in

patient samples. Tyrosine kinases are also over-expressed in breast cancer metastatic

disease, however, the use of tyrosine kinase inhibitors proves to lead to more positive

outcomes in HER2/neu+ breast cancer patients (Ryan et al., 2008). Regardless,

tyrosine kinase inhibitors are still in clinical trials as monotherapy and combination

therapy.

1.1.3.3 Recurrence

Patients with basal-like and HER2/neu+ breast cancers respond well to neoadjuvant

chemotherapy composed of paclitaxel- and doxorubicin-containing chemotherapeutic

agents, as compared to patients with other subtypes of breast cancer, in terms of pCR

(Rouzier et al., 2005). Despite high response rates, many of these patients undergo

relapse. Indeed, a study found that when compared to non-TNBC patients, patients

harboring TNBC tumors treated with neoadjuvant chemotherapy showed higher pCR

rates, but decreased 3-year PFS and 3-year OS. Moreover, these patients suffered

increased visceral metastasis and shorter post recurrence survival (Liedtke et al.,

2008). Despite having decreased survival rates in the first 3 to 5 years after diagnosis

(Figure 1.4 (A)), patients with TNBC or basal-like tumors actually suffer less distant

relapse after this time, as compared to patients with other breast cancer subtypes

Page 34: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

17

(Figure 1.4 (B)) (Foulkes et al., 2010, Tischkowitz et al., 2007). 10 years after

diagnosis, ER+ patients are more likely to suffer relapse than ER- patients.

Recurrence is often due to and accompanied by chemo-resistance to conventional

chemotherapeutic agents. Some cells survive chemotherapy and can survive long-

term and maintain cell viability and lead to recurrent tumors. We will discuss

mechanisms of chemo-resistance in later chapters in more detail.

Figure 1.4 Survival after a diagnosis

of breast cancer

A. Survival rate of 3744

patients according to

subtype. 639 women with

TNBC subtype were divided

into 2 groups: those with and

without basal markers

(cytokeratin 5, EGFR). (Data

from (Cheang et al., 2008)).

B. Hazard rates for distant

recurrence of TNBC and

non-TNBC patients. (Data

from (Dent et al., 2007).

Page 35: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

18

1.2 Mitotic inhibitors

Mitotic inhibitors prevent cells from undergoing mitosis by disrupting microtubules,

which are the main components of the mitotic spindle. Microtubules are crucial for a

normal cell division as they bind to the kinetochores of sister chromatids and ensure

proper segregation and migration of chromosomes during anaphase.

Microtubules are made up of α- and β-tubulin dimers which bind to GTP on the (+)-

end of the microtubules. Once bound, the GTP hydrolyzes to GDP through contact

with the dimers. The stability of microtubules is dependent on whether the β-tubulin

dimer binds to a GDP or GTP on the microtubule. Dimers which are bound to GTP

lead to microtubule assembly, whereas dimers bound to GDP tend to fall apart. The

dynamic stability/instability is therefore dependent on the GTP/GDP bound to

microtubules. The binding of microtubules to kinetochores is closely monitored by

the Spindle Assembly Checkpoint (SAC), a cell cycle surveillance pathway, which

can forestall further cell cycle progression in case of any misalignment (Foley and

Kapoor, 2013). During the cell cycle, microtubules are assembled from the

centrosome and then bind to the chromosomes once they have aligned along the

metaphase plate. When the cell has passed through the SAC, the microtubules are

disassembled into single dimers again. Correct microtubule assembly and

disassembly is a requirement for proper cell division.

Mitotic inhibitors target microtubule dynamics, thereby disrupting cell division, and

can either be microtubule-stabilizing or –destabilizing, depending on their effects on

microtubule dynamics. Microtubule-destablizing agents inhibit microtubule

polymerization and inhibit the formation of the mitotic spindle at high concentrations.

Most of these agents bind to either of two domains on tubulin: the “vinca” domain

(vinca alkaloids such as vinblastine, vincristine, vinorelbine) or the “colchicines”

domain (colchicines and its analogs). Microtubule-stabilizing drugs, on the other

Page 36: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

19

hand, stabilize microtubules at high concentrations and inhibit progression into the

anaphase. Such drugs include taxanes (Paclitaxel and Docetaxel), the epothilones, and

ixabepilone.

1.2.1 Paclitaxel (Taxol®)

Taxanes (complex diterpenes) are produced by plants of the genus Taxus (yews),

while some are now being synthesized artificially. The two main groups of taxanes

are Paclitaxel (Taxol®), which is derived from Taxus Brevifolia, and Docetaxel

(Taxotere), which is a semisynthetic analogue of Taxol (extracted from Taxus

Baccata (Fauzee, 2011). The empirical formula for Taxol is C47H51NO14, it has a

molecular weight of 853.9 and is insoluble in water (highly lipophilic).

The antitumor activity of Taxol was first confirmed in 1977 in a mouse model of

melanoma B16, and subsequently in mammary, lung and colon tumors. In 1984, the

National Cancer Institute (NCI) started a Phase I clinical trial of Taxol against various

cancer types. In 1992 it was FDA approved for ovarian cancer and shortly after for

use against breast cancer (Seidman, 1994). Since then, Taxol has been proven

effective against a number of solid tumors, including ovarian cancer, lung cancer,

breast cancer, colon cancer, sarcoma, multiple myeloma, AIDS-related Kaposi’s

sarcoma, and acute leukemias (Kumar et al., 2010, Sparano et al., 2008, Gill et al.,

1999, Rowinsky et al., 1989). Moreover, the antitumor effect of Taxol is increased

when it is used in combination with other chemotherapeutic drugs, such as

doxorubicin and various anthracyclines (Sledge et al., 2003, Kaufman, 1999), or

specific inhibitors, such as trastuzumab (Herceptin) in breast cancer (Klos et al.,

2003), or the Notch inhibitor in platinum-resistant ovarian cancer (Groeneweg et al.,

2014).

Page 37: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

20

1.2.2 Mode of action

All drugs targeting microtubules bind to the β-tubulin subunit, and they all have

distinct binding sites, dependent on whether they stabilize or disrupt the microtubules.

Taxol binds to the β-subunit on three different peptides: amino acids 1-31 (Rao et al.,

1994), 217-231 (Rao et al., 1995), and 277-293 (Rao et al., 1999). It binds to the

microtubule (it cannot bind to single tubulin dimers such as colchicines and vinca

alkaloids) and enables microtubule polymerization in the absence of GTP. In

addition, it protects microtubules from destabilization (Orr et al., 2003). At low

concentrations, Taxol suppresses microtubule dynamics since it does not bind to all

available binding sites. At high concentrations, however, it reduces the number of

tubulin dimers available to assemble microtubules by stabilizing microtubule

polymers which leads to a mitotic arrest.

At low concentrations, where only a small part of the Taxol-binding sites are

occupied on the microtubules, it suppresses microtubule dynamics, whereas at high

concentrations, it increases polymer mass and induces microtubule bundle formation,

which has become a hallmark of Taxol binding (Schiff and Horwitz, 1980).

1.2.3 Mitotic arrest

Cancer cells are especially susceptible to treatment with Taxol as they undergo a

rapid cell cycle.

Microtubule stabilization leads to cell cycle arrest in the mitotic (M) phase. Cells

trapped in the M phase can undergo different fates, depending on the type and

concentration of drug used (Gascoigne and Taylor, 2009) and the strength of the SAC

signaling (Rieder and Maiato, 2004). After a mitotic arrest, cells can divide

unequally, undergo cell death during mitosis, or exit the mitotic arrest without

division. However, if they exit without division, they can arrest in the interphase or

Page 38: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

21

undergo apoptosis, or they can pass through the cell cycle, which leads to tetraploidy.

The variation in response can also be seen among cells of the same origin. Gascoigne

et al. have summarized the different cell fates after treatment with anti-mitotic drugs

(Gascoigne and Taylor, 2009), as seen in Figure 1.5.

In a normal cell cycle, activation of the SAC will prevent Cyclin B1 degradation,

which is a requirement for M phase exit. Cyclin B1 is a protein involved in the

regulation of mitosis. Its level is highest during the mitotic phase and cells can only

progress through this phase after Cyclin B1 degradation. It was found, that in

mammalian cells treated with anti-mitotic drugs, despite SAC activation, Cyclin B1 is

slowly degraded which enables cells to exit mitosis, this is also called “mitotic

slippage” (Brito and Rieder, 2006). The different rates of Cyclin B1 degradation may

explain the variation in response to mitotic arrest. Mammalian cells that have an

activated SAC and do not exit mitosis will undergo a mitotic catastrophe. It has been

shown that targeting Cyclin B1 sensitizes breast cancer cells to Taxol (Androic et al.,

Figure 1.5 Cell fate in response to anti-mitotic drug treatment

Mitotic arrest after Paclitaxel treatment leads to chronic SAC activation and mitotic arrest, after

which they undergo one of several fates. (Derived from (Gascoigne and Taylor, 2009)).

Page 39: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

22

2008), presumably because they will enter mitotic slippage which is dependent on

Cyclin B1 proteolysis (Brito et al., 2008).

1.2.4 Mitotic catastrophe

Until now, there is no clear cut definition of what a mitotic catastrophe really is. It

has been described by some as an aberrant form of mitosis (Ianzini and Mackey,

1997), or a delayed form of reproductive death (Waldman et al., 1996), but in most

cases it will eventually lead to cell death. Many events can lead to a mitotic

catastrophe such as DNA damage, ‘mitotic slippage”, or defects in mitosis, such as

mitotic arrest. Cell death following mitotic arrest can be through apoptosis or

necrosis.

Apoptosis following mitotic arrest requires activation of the SAC and mitotic

slippage (Tao et al., 2005). Although this relation is not entirely clear, it has been

found that BUB1 mediates caspase-independent cell death after mitotic arrest and

determines the cell’s fate (Niikura et al., 2007). BUB1 is bound to the kinetochores of

sister chromatids during mitosis (with a peak in the G2/M phase) to ensure correct

chromosome alignment and plays a major role in the formation and regulation of the

SAC. Together with BUB1, there are 13 other proteins that play a role in the SAC

(such as Aurora kinase B, MAD1, MAD2, BUBR1, BUB3 etc.). Aurora B is required

for the activation of the SAC and cells which have been depleted of Aurora B, fail to

undergo mitotic arrest, even in the presence of chromosome misalignment or

detachment from microtubules (Hauf et al., 2003). Taxol treatment causes a reduction

of inter-kinetochore tension due to microtubule stabilization and this leads to an

increase of BUBR1 and BUB1 at the kinetochores (Gorbsky and Ricketts, 1993) and,

moreover, Taxol-mediated mitotic arrest is dependent on Aurora B (Ditchfield et al.,

2003, Hauf et al., 2003) and the presence of MAD2 at kinetochores (Waters et al.,

Page 40: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

23

1998). Survivin plays an important role in the maintenance of SAC and Aurora B

localization (Okada et al., 2004).

Several studies have shown that Taxol-induced apoptosis is caspase-dependent, as

shown in the human colon carcinoma cell line HCT116 (Llovera et al., 2012) and in

the Non-Small Cell Lung Cancer cell line NCI-H460 (Huisman et al., 2002). One

possible mechanism is the involvement of death receptors which activate Fas-

associated death domains and Caspase-8, leading to Bid cleavage and the disruption

of mitochondria. This is similar to many other DNA-damaging agents which induce a

caspase-dependent apoptotic response. It was found that wild-type p53, activated

after DNA damage, can inhibit mitotic catastrophe (Roninson et al., 2001) and many

tumor cells are deficient for wild-type p53, making them susceptible to mitotic

inhibitors (Gascoigne and Taylor, 2009).

Page 41: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

24

1.3 Chemo-resistance

Chemo-resistance is one of the major challenges in the treatment of TNBC patients,

as chemotherapy is still the mainstay of treatment and recurrence rates with poor

outcome are high. Recurrence following chemotherapy can be attributed to the fact

that some cancer cells survive chemotherapy and are able to maintain viability and

survive long-term by undergoing cellular senescence and autophagy, as compared to

apoptosis. Cells undergoing senescence have exited the cell cycle and lost their

proliferative capacity, however, they maintain viability and metabolic activity (Saab,

2010). By exiting the cell cycle, these cells are protected from the cytotoxic effects of

chemotherapeutic agents. Moreover, it has been found that the senescence pathway

plays a role in breast carcinogenesis by promoting tumor progression through

stimulation of the neighboring cells with the senescence-associated secretory

phenotype (SASP) (Pare et al., 2013). It was found that senescent cells produce

lactate which is implicated in the tumor-stroma co-evolution. Breast cancer patients

with long-term survival were found to have lower levels of lactate in tumor biopsies

as compared with patients that died of cancer recurrence (Cao et al., 2012). On the

other hand, autophagy is a type II programmed cell death involved in cellular

homeostasis and protein and organelle recycling (Nishida et al., 2008). Its role in cell

survival/cell death is controversial: it can either be activated as part of the survival

pathway, or as part of the cell death process. Its role in cancer is even more

controversial where it has positive and negative effects on tumor cell survival (Notte

et al., 2011, Mathew et al., 2007). It is dependent on the type of tumor, tumor stage

and extent of the insult. Especially in TNBC cells compared to other breast cancer

cells, expression of the autophagy-related microtubule-associated protein, beclin-1,

light-chain (LC) 3A and LC3B, are high, with low expression in the stroma (Choi et

al., 2013). Moreover, LC3A is associated with tumor progression and poor outcome

in TNBC (Zhao et al., 2013). It was found that the effect of paclitaxel treatment can

Page 42: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

25

be enhanced when inhibiting autophagy. Also, positive effects were seen after

treatment with an autophagy inhibitor combined with ER stress aggravators in TNBC

(Thomas et al., 2012).

1.3.1 Cancer stem cells

Chemo-resistance accounts for up to 90% of drug failures in patients with metastatic

cancer (Longley and Johnston, 2005). There are theories which suggest that a certain

population of cells survive chemotherapy, these are cells with properties similar to

stem cells and are responsible for tumor initiation and recurrence. These cells have

been called cancer stem cells (CSCs) (Dalerba et al., 2007a) as they have properties

similar to normal stem cells such as self-renewal and differentiation. The first

evidence of CSCs was reported in 1994 by Lapidot et al. in acute myelogenous

leukemia (AML) where they found cell surface markers by which they could select

for CSCs (CD34+ and CD38-) and, more importantly, these cells could give rise to

leukemic growth in severe combined immunodeficiency (SCID) mice (Lapidot et al.,

1994). CSCs of different origin express different cell surface markers and it was

discovered that breast cancer tumor-initiating CSCs are ESA+/CD44+/CD24-/low. Cells

selected based on these markers are capable of self renewal and differentiation (Al-

Hajj et al., 2003). Since then various CSCs populations in different solid tumors have

been identified based on cell surface markers such as CD133, EpCAM and CD90, as

well as aldehyde dehydrogenase (ALDH) activity (Yang et al., 2008, Dalerba et al.,

2007b, Singh et al., 2003).

CSCs have been linked to tumor expansion and relapse through their ability to escape

chemotherapy treatment. Many studies suggest that CSCs have adopted various

mechanisms of chemo-resistance and, although there is an overlap between chemo-

resistance and CSCs, not all cells which have acquired resistance are also classified as

CSCs.

Page 43: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

26

1.3.2 Mechanisms of chemo-resistance

Two mechanisms are thought to contribute to chemo-resistance: instrinsic and

extrinsic resistance. Intrinsic resistance originates in the cancer cells itself which

gives them the capability to withstand chemotherapy (Wilson et al., 2006), whereas

extrinsic resistance is acquired by cells over a period of time via genetic and

epigenetic mutations in genes as a result of chemotherapy treatment (Gillet and

Gottesman, 2010). We will discuss some of the established mechanisms of chemo-

resistance in closer detail.

Multidrug resistance The main mechanism of resistance is a high expression of

membrane efflux pumps of the ATP binding cassette (ABC) family of transporters.

The most important family member involved in drug resistance is the ABC subfamily

B member 1 (ABCB1), also known as P-glycoprotein, or multidrug resistance protein

1 (MDR1), which is capable of actively pumping Hoechst dye. It is a glycosylated

membrane-associated enzyme which exports a number of different substrates. It has

been found to be expressed in many different tissues such as liver, kidney, intestines,

placenta and blood-brain-barrier, and hematologic and solid tumors. (Goldstein et al.,

1989). In the placenta and blood-brain-barrier its function is to protect from toxins,

whereas in other tissues it functions as an efflux pump for xenobiotics, toxins and

other drugs. In addition, ABCB1 transporters were found to be strongly expressed in

the neurons of the hippocampus formation, and expression was further up-regulated

upon induction of a status epilepticus in rat brains (Pardridge et al., 1997). In breast

cancer, it was found that EGF could increase phosphorylation of ABCB1 to enhance

multidrug resistance activity (Yang et al., 1997).

More importantly, ABCB1 is also expresses in certain stem cells. Chaudhary and

Robinson discovered that CD34+ hematopoietic stem cells show a high expression of

ABCB1(Chaudhary and Roninson, 1991). One study used ABCB1-deficient mouse

Page 44: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

27

models to study the function of ABC transporters and suggested that efflux pumping

of xenobiotics which limits the success of chemotherapy may not be their only

function. One of their main functions is a protection from apoptosis of stem cells.

ABCB1 induces a stress-induced regeneration genetic program in normal tissues and

is activated in cancer cells due to cancer therapy (Israeli et al., 2005).

These results reveal an association between ABCB1 expression and stem cells, and,

in addition, between ABCB1 and cancer cells. This suggests that high ABCB1

expression could be linked to CSCs and also to chemo-resistance, as these

transporters can efflux vincas and taxances, effectively reducing their intracellular

drug concentration (Fojo and Menefee, 2005). In 1982, 6 years after the first link

between ABCB1 and chemo-resistance was established, many compounds were

developed which could inhibit ABCB1 and restore chemo-sensitivity in vitro. Despite

some promising results with cancer cells and tissue, they proved ineffective in clinical

trials due to many different reasons: low selectivity for ABCB1; poor potency; high

toxicity; and diverse interactions with other anticancer drugs (Crowley et al., 2010).

Another ABC transporter with importance in chemo-resistance is ABCG2. It was

found to be especially highly expressed in progenitor cells/reactive ductules in the

human liver, with a possible function of protection from cytotoxic agents (Borst and

Elferink, 2002). It is often associated with ABCB1 as it has very similar functions in

terms if efflux pumping of cytotoxic compounds, but in contrast to ABCB1 which

characterizes proliferating cells, ABCG2 is believed to be a marker for quiescent cells

(Ueda et al., 2005).

Aldehyde dehydrogenase Another functional marker of CSCs which confers

resistance to chemotherapeutics is Aldehyde dehydrogenase (ALDH). ALDH1 is a

cytosolic enzyme which oxidizes aldehydes and converts them into carboxylic acids

(Ikawa et al., 1983). There are 16 different ALDH isoforms and each isoforms has

Page 45: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

28

specific functions in different types of cancer. ALDH1A3 has been found to be

significantly associated with distant metastasis, disease-free survival and overall

survival in breast cancer patients (Qiu et al., 2014). Since ALDH has been linked to

normal stem and progenitor cells, its role in CSCs has also been extensively studied.

Acute myeloid leukemic cells with elevated ALDH engraft better in NOD/SCID mice

compared to cells with low ALDH activity (Cheung et al., 2007). And as little as 20

breast cancer cells selected with an ALDH marker together with CD24-/CD44+

markers could form tumors (Ginestier et al., 2007).

In addition to its function in CSCs, ALDH plays a role in chemo-resistance.

Resistance to cyclophosphamide, a chemotherapeutic agent often used in first-line

treatment for TNBC patients, is accompanied by high levels of ALDH and could be

reversed by inhibition of ALDH in cyclophosphamide-resistant L1210 leukemic cell

lines (Hilton, 1984). It has since then been shown to be important in

cyclophosphamide-resistance in medulloblastoma (Friedman et al., 1992) and other

cancer systems, thereby confirming the importance of ALDH in chemo-resistance and

CSCs (Sreerama and Sladek, 1997). ALDH1A1 activity is also linked to gemcitabine-

resistance in human pancreatic adenocarcinoma. Ongoing studies with inhibitors

targeting ALDH activity or influencing ALDH expression can help to overcome

chemo-resistance in CSCs (Sullivan et al., 2010, Cortes-Dericks et al., 2014, Hellsten

et al., 2011). ALDH is an established marker for chemo-resistance, although its exact

function is not clear (Januchowski et al., 2013).

We will discuss mechanisms of resistance to microtubule-disrupting drugs more in

detail as this thesis is focusing on Paclitaxel-resistance.

Page 46: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

29

1.3.3 Resistance to microtubule disrupting drugs

It is important to understand the specific mechanism of resistance to such agents in

order to overcome and develop new and more effective drugs. Resistance can be the

result of changes in the pharmacodynamics of these microtubule-disrupting agents

such as cellular efflux and metabolization, ineffective target interaction, and

resistance to apoptotic signals.

Alterations in microtubules Changes in the targets of microtubule-disrupting agents

contribute to resistance. Such changes can include changes in the conformation of

tubulin dimers/microtubules so that the drugs are unable to bind, and changes in the

GTPase activity needed for the synthesis of microtubules. Many proteins play a role

in microtubule dynamics and can contribute to resistance through alterations in their

expression levels, cellular localization, and post-translational modifications. These

include various microtubule-associated proteins (MAPs) (MAP1, MAP2, MAP4, tau,

STOP), and others like survivin which counteracts the effect of microtubule

destabilizing agents (Cheung et al., 2009), FHit which interacts with microtubules

and acts as a tumor suppressor (Chaudhuri et al., 1999) and stathmin which alters

drug binding and inhibits progression into the M-phase (Cheung et al., 2009). Low

expression of protein tau, which has the same tubulin binding site as Paclitaxel, is

associated with Paclitaxel-sensitivity, as seen in preclinical studies. Similarly, high

tau expression in ER+ breast cancer confers resistance to taxane-based chemotherapy

and sensitivity to hormone therapy (Smoter et al., 2011).

There are 13 different α- and β-tubulin isotypes involved in building of microtubules.

The composition of the microtubules and the types of tubulin involved play a role in

resistance. β-tubulin III has been shown to be associated with reduced taxane

sensitivity in several types of tumors including lung, breast and ovarian cancers (Seve

and Dumontet, 2008), and taxane-resistance has been attributed to increased levels of

Page 47: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

30

βII and βV tubulins (Bhattacharya and Cabral, 2009, Haber et al., 1995).

Interestingly, resistance to taxanes is not the only role for β-tubulin III. It is also a

survival factor which can increase cancer progression independent of drug treatments

in lung cancer (McCarroll et al., 2010).

In addition to altered expression of tubulin, somatic mutations in β-tubulin also

contribute to taxane resistance. In paclitaxel-resistant ovarian cancer cell lines,

mutations at nucleotides 810 and 1092 of the HM40 isotype of β-tubulin lead to

impaired paclitaxel-driven polymerization (Giannakakou et al., 1997).

Resistance to apoptotic signaling Signaling downstream of microtubules also plays a

role in resistance. P53 is involved in a complex mechanism which was believed to

play a crucial role due to the fact that it can arrest the cell cycle and allow DNA

repair. It was observed that by inactivating p53, normal and human murine fibroblasts

could be sensitized to taxol (Wahl et al., 1996). However, it was later observed that

more often than not, mutant p53 lead to drug resistance to various agents due to a

disabled apoptotic machinery (Delia et al., 1996, Varna et al., 2009). In contrast, it

was later published that p53 is not involved in resistance to Paclitaxel or Docetaxel in

breast cancer (Noguchi, 2006). These conflicting results regarding the involvement of

p53 in resistance render it an ineffective marker for taxane resistance.

Apart from p53, other apoptotic regulators can influence taxane sensitivity. One such

effector is the pro-survival protein B-cell lymphoma-extra large (Bcl-XL). Using a

small molecule inhibitor targeting Bcl-XL, the cytotoxic effects of various

chemotherapeutic agents, including Paclitaxel, could be enhanced in a NSCLC cell

line (Shoemaker et al., 2006). In addition, certain microRNAs were also shown to

play a role. One such example is of miR-125b which could confer resistance to

Paclitaxel through suppression of the activity of the pro-apoptotic BAK1 in various

breast cancer cell lines (Zhou et al., 2010). Another microRNA with a role in

Page 48: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

31

resistance is miR-148a, which sensitizes prostate cancer cell lines to Paclitaxel by

inhibiting the expression of mitogen and stress-activated protein kinase (MSK1)

(Fujita et al., 2010).

Resistance to other oncogenic signaling pathways Since chemo-resistance plays a

major role in the treatment of TNBC patients, many studies have been conducted to

unravel the importance of various pathways in conferring resistance. One such

pathway found to play a role in resistance to Taxol is the Hippo pathway. TAZ

(transcriptional co-activator with PDZ-binding motif) is a major component of the

Hippo-LATS pathway and has been found to be elevated in human breast cancer cells

and responsible for Taxol resistance (Lai et al., 2011). TAZ activates the

Cyr61/CTGF signaling pathway which modifies the Taxol response in breast cancer

cells. However, it is not clear how the activation of this pathway leads to increased

resistance.

Page 49: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

32

1.4 Aurora Kinase Signaling

The Aurora Kinases are a family of evolutionary conserved serine/threonine kinases

that play an important role and regulate many processes during cell division. They

control the centrosome and nuclear cycles, they play crucial roles in chromosome

condensation, spindle dynamics, kinetochore-microtubule interactions, chromosome

orientation, and establishment of the metaphase plate. There are three Aurora

Kinases, Aurora A, B and C, that are very similar especially in the carboxy-terminal

catalytic domain, but they differ in the length and sequence of their amino-terminal

(Figure 1.6). All three kinases have an activating T-loop, a destruction box (D-box)

which leads to its destruction at the end of mitosis, and in addition Aurora A requires

phosphorylation of its D-box activating domain (A-box) for its degradation.

1.4.1 Aurora Kinases in cell cycle

Interestingly, the three Aurora kinases have distinct functions and subcellular

locations despite their similarities. While Aurora A associates with the spindle poles

during cell division regulating mitotic entry, centrosome maturation and separation,

and spindle pole polarity, Aurora B is an important member of the Chromosomal

Figure 1.6 Schematic representation of human Aurora A, B and C

The numbers indicate their size in terms of amino acids and their percentage of sequence

identities. Also shown are the kinase domain (green), the activating T-loops (yellow), the

destruction box (D-box, blue) and the D-box activating domain (DAD, red). (Figure adopted

from Keen N. et al (Keen and Taylor, 2004)).

Page 50: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

33

Passenger Complex (CPC) together with INCENP, Survivin and Borealin/DasraB. It

localizes to the chromosomal centromeres during mitotic entry but will relocate to the

microtubules at the spindle equator during anaphase. When the cell undergoes

cytokineses, Aurora B accumulates at the spindle midzone and the cell cortex at the

cleavage furrow, and subsequently concentrates at the midbody. In addition, Aurora

B plays a crucial role for correct chromosome alignment and segregation, it regulates

the kinetochore function. Moreover, it is required for correct spindle checkpoint

function and cytokinesis.

Equally important for cell division, is Polo-like kinase 1 (PLK1), one of the five

members of the Polo-like kinase family. Inhibitors of PLK1 lead to mitotic arrest with

a monopolar or disorganized spindle, and the cells will eventually undergo mitotic

arrest (Steegmaier et al., 2007). All Polo-like kinases possess a highly conserved

polo-box domain (PBD) in the carboxy-terminal end, which can serve as a docking-

motif that brings the kinase into close proximity of its substrates (Park et al., 2010).

PLK1 can bind its substrates in a phospho-independent manner (Archambault et al.,

2008), or after it has undergone a priming phosphorylation by Cyclin-dependent

kinase 1 (CDK1) or PLK1 itself (Elia et al., 2003). Once bound, the substrates will

recruit PLK1 to the centrosome, kinetochore or the spindle midzone. It is not

completely clear how it regulates spindle formation, but it is well known that PLK1

plays a role in centrosome maturation and stable kinetochore-microtubule

attachments (Archambault and Glover, 2009). The effect of PLK1 inhibition is

similar to inhibition of the Aurora kinases, suggesting that they might co-regulate

certain mitotic processes. PLK1 and Aurora A play a crucial role in mitotic entry.

They co-localize at the centrosomes, but then PLK1 also localizes to kinetochores in

the prometa- and metaphase. Aurora B, however, is important in the prophase where

it localizes to the arms of the chromosome and the inner centromere, whereas in the

prometa- and metaphases it localizes solely to the inner centromeres. Aurora B co-

Page 51: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

34

localizes with PLK1 on the central spindle, the cortex and midbody in the anaphase

and telophase where they regulate cytokinesis. Aurora A is degraded in the anaphase

and telophase, although a small portion remains at the central spindle and midbody,

but its function is unclear.

Figure 1.7 Localization of

Aurora kinase A and B

together with PLK1 in the

dividing cell

Aurora kinase B is localized

to the chromosome arms and

inner centromere in the

prophase, whereas in the

prometaphase and metaphase

it is only found at the inner

centromere. In anaphase and

telophase it co-localizes with

PLK1 on the central spindle,

cortex and midbody where it

regulates cytokinesis. (Figure

adopted from Lens S.M.A. et

al.. (Lens et al., 2010)).

Page 52: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

35

1.4.2 The Spindle Assembly Checkpoint

Aurora B, along with the CPC, stabilizes the mitotic spindle (Gassmann et al., 2004)

by phosphorylating Stathmin/Op18 and the kinsein-13 microtubule-destabilizing

protein (MCAK), two proteins involved in the chromatin-driven spindle assembly

pathway (Ohi et al., 2004, Gadea and Ruderman, 2006).

The main components of the spindle assembly checkpoint (SAC) are MAD1, MAD2,

BUBR1, BUB1, BUB3, MPS1, and Aurora B, and it is activated during cell division

where it inhibits anaphase as long as the kinetochores are not attached to the

microtubules. Only when all kinetochores are stably bound, the SAC is satisfied and

inhibition of the anaphase is alleviated. Three of the SAC proteins, MAD2, BUBR1

and BUB3, as well as CDC20, make up the mitotic checkpoint complex (MCC),

which binds the ubiquitin ligase anaphase-promoting complex/cyclosome (APC/C),

inhibiting its ubiquitin ligase ability on securin and cyclin B (Tang et al., 2001, Fang

et al., 1998). Securin is the inhibitor of a protease called separase which is required to

cleave the cohesion complex which binds the sister chromatids, while cyclin B

activates CDK1, inhibiting exit from mitotsis. CDC20 is a co-factor of APC/C and by

keeping it in check, the SAC can prolong the prometaphase until all the kinetochores

are attached to the microtubules and are bi-oriented along the metaphase plate.

Aurora B plays an important role in the SAC as it regulates kinetochore assembly.

Kinetochores are complex structures comprising of at least 80 different proteins at the

centromere of each sister chromatid (Cheeseman and Desai, 2008, Santaguida and

Musacchio, 2009), and they link the chromosomes to the microtubule polymers of the

mitotic spindle. The inner kinetochore is tightly associated with the centrosome DNA

throughout the cell cycle, whereas the outer kinetochore, which interacts with the

microtubules, is a dynamic structure only present during cell division. For correct

chromosome segregation, each sister chromatid has to attach to microtubules

Page 53: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

36

generated from opposite spindle poles through its kinetochores. This is called a

amphitelic configuration, or bi-orientation. If the kinetochores are not correctly

attached, they can have a monotelic configuration (only one of the chromatids is

attached to microtubules), syntelic configuration (both chromatids are attached to the

same microtubule generated from one spindle pole), or a merotelic configuration (at

least one chromatid is attached to both microtubules generated by opposite spindle

poles). The role of Aurora B is to ensure correct kinetochore-microtubule attachments

by destabilizing incorrect attachments. Correct amphitelic attachments are stabilized

by tension through the mitotic spindles (Nicklas and Koch, 1969), whereby Aurora B

is an important tension sensor at centromeres and kinetochores (Biggins and Murray,

2001, Tanaka et al., 2002). Inhibition of Aurora B using small molecule inhibitors or

inhibitory antibodies has been shown to stabilize incorrect attachments to a single

spindle pole (Hauf et al., 2003). More recently, a “spatial separation” model has been

proposed which explains how Aurora B is able to sense tension. In this model,

tension sensing depends on the localization of Aurora B. When bi-oriented

kinetochores are separated, Aurora B at the inner centromere is separated from its

outer kinetochore substrates (Tanaka et al., 2002).

Several mechanisms contribute to the deactivation of the SAC. During the process of

“stripping”, MAD1 and MAD2 are removed from the kinetochores and this seems to

be a crucial step in deactivation (Maldonado and Kapoor, 2011). This process is

mediated by the minus-end microtubule motor dynein (Howell et al., 2001). Also

important is the disassembly of the inhibitory complexes in order to free Cdc20 so

that it can activate APC/C, thereby allowing it to ubiquinitylate securin and cyclin B,

allowing entry into anaphase and mitotic exit.

Page 54: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

37

1.4.3 Paclitaxel-induced activation of the SAC

Paclitaxel stabilizes microtubules, thereby reducing the tension on the kinetochores

from mitotic spindles in bi-oriented sister chromatids. This process turns on the SAC

thereby stopping the progression of the cell cycle. Moreover, it was found that MAD2

is required to maintain taxol-mediated mitotic arrest (Shannon et al., 2002). CENP-E

is a microtubule plus-end-directed motor that is crucial for chromosome alignment

and when CENP-E is absent, unattached kinetochores are unable to sustain SAC

activity. However, the addition of taxol disrupts tension and can override CENP-E

absence and activate SAC activity (Wood et al., 2008). However, siRNA knock-down

experiments revealed that the absence of Aurora B compromised checkpoint arrest in

the Paclitaxel-treated colon cancer cell line DLD1 (Ditchfield et al., 2003). Moreover,

Aurora B knock-down also led to absence of microtubule-kinetochore interactions,

suggesting that Aurora B plays role in correct kinetochore attachments. In addition to

its role in destabilizing improper microtubule attachments, Aurora B also plays a

direct role in the SAC.

1.4.4 Aurora Kinases in Cancer

It was discovered that Aurora A and Aurora B are over-expressed in primary breast

cancer samples (Sen et al., 1997) and colon tumor samples (Warner et al., 2003),

suggesting that they are associated with tumorigenesis. Aurora A is most consistently

associated with cancer and its role is well studied. It has been found to be over-

expressed in many different kinds of cancer, such as colon, breast, bladder, liver,

gastric and pancreatic cancer (Sakakura et al., 2001, Goepfert et al., 2002) and its

over-expression is significantly associated with high grade tumors and poor prognosis

(Jeng et al., 2004). There is a correlation between Aurora A over-expression and

aneuploidy in gastric cancer. Aneuploidy is associated with poor outcome in a

number of malignancies, and in gastric and papillary thyroid cancer it is a metastatic

Page 55: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

38

marker (Sasaki et al., 1999, Sturgis et al., 1999). Aurora A over-expression is also

associated with centrosome amplification which leads to bipolar mitotic spindle

defects, chromosomal segregation deficiency and aneuploidy. Centrosomal

abnormalities have been shown to arise in early stages of tumor development and

expand as the tumor progresses (Pihan et al., 2001), especially in brain, breast, lung,

colon and prostate cancer (Pihan et al., 1998, Lingle and Salisbury, 1999). However,

so far no direct link has been found between Aurora A over-expression and

centrosomal abnormalities, although they are often associated with each other.

Aurora A has also been reported to play an important role in transformation. It

phosphorylates and regulates the breast cancer-associated gene product BRCA1

(Ouchi et al., 2004), and it can regulate telomerase activity through c-Myc in ovarian

and breast epithelial carcinomas (Yang et al., 2004). Moreover, it interacts with p53

at multiple levels. It directly phosphorylates p53 at Ser315 mediating degradation of

p53 by MDM2 in cancer cells, and at Ser215 thereby inactivating its transcriptional

activity (Katayama et al., 2004, Liu et al., 2004). TP53 is the most common gene

mutated in a number of human cancers, whereby the most common mutations lead to

a complete loss or inactivity of the p53 protein (Kandoth et al., 2013). This shows the

importance of Aurora A in cancer, especially in transforming processes.

Aurora B has been found to be over-expressed in cancers such as breast, kidney,

colorectal, prostate and lung cancer, and high levels of Aurora B lead to multi-

nucleation and polyploidy in human cells (Tatsuka et al., 1998). Moreover, it was

found that high levels of Aurora B correlate with late stages of colorectal cancer

(Katayama et al., 1999). It may also play a role in carcinogenesis as high levels lead

to chromosome lagging in the metaphase, errors in chromosome segregations and

cytokinesis (Ota et al., 2002). Although Aurora B alone is not able to transform cells,

it potentiates H-Ras(G12V)-induced transformation (Kanda et al., 2005). During

mitosis, half of the Aurora B forms a complex with INCENP, Survivin and Borealin,

Page 56: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

39

whereas the other half forms a complex with INCENP, but it is not clear which of

these complexes is involved in the potentiation of oncogenic Ras.

Aurora C, however, is a chromosome messenger protein and only expressed in the

testis and not somatic cells. It has been found to be up-regulated in cancer cell lines

such as HepG2, HuH7, MDA-MD-453 and HeLa cells, although its role is unclear

(Kimura et al., 1999).

Page 57: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

40

1.5 Protein Kinase C Signaling

Protein Kinase C (PKC) has become an attractive target for drug discovery in the past

20 years as it functions as the receptor for the tumor promoter phorbol ester

(Castagna et al., 1982). It has gained even more interest since it was discovered to

play a role, not only in cancer, but also in other diseases such as diabetes, ischemic

heart disease and hear failure, autoimmune diseases, Parkinson’s disease,

Alzheimer’s and many other diseases.

1.5.1 Structure and isoforms of PKCs

PKC is a family of at least 12 highly related but distinct serine/threonine kinases

which play a role in signaling pathways involved in proliferation, differentiation,

angiogenesis and apoptosis, as well as transcription, translation, and cell-cell contact.

The different isoforms can be grouped into 3 classes depending on the number of

conserved regions (C1-C4) and the number of variable regions (V1-V5) (Khalil,

2010). The C1 region contains cystein-rich zinc finger-like motifs and the recognition

site for Diacylglycerol (DAG), phosphatidylserine and phorbol ester. The C2 region

of some isoforms contains the binding site for Ca2+, and the C3 and C4 regions

constitute the ATP- and substrate-binding lobes (Figure 1.8). Due to the PKC

isoforms containing different conserved and variable regions, they are activated

through different processes. The conventional PKC isoforms (α, βI, βII and γ) contain

all conserved regions (C1-C4) and all variable regions (V1-V5) and can be activated

by DAG, phoshatidylserine, phorbol ester and Ca2+. The novel PKCs (δ, ε, η and θ)

lack the C2 region and are therefore not affected by Ca2+, and the atypical PKCs (ζ

and λ/ι) have only one cysteine-rich zinc finger-like motif and can only be activated

by phosphatidylserine. Phospholipase C (PLC) is an enzyme which, when activated

by G-protein coupled receptors (GPCR), can generate DAG and 1,4,5-triphosphate

(IP3) by hydrolyzing phosphatidylinositol-4,5-biphosphate (PIP2). IP3 then binds to

Page 58: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

41

Ca2+positive channels on the endoplasmic reticulum which release Ca2+ into the

cytoplasm. PKC can bind to Ca2+, translocate to the cell membrane and interact with

DAG. A fourth class of PKCs, the PKN subfamily, is distinctly different to the

previous 3 classes in that it has 3 tandem coiled motifs, known as HR1 domains,

which can bind to RhoA and play a role in Rho GTPase regulation (Flynn et al.,

1998).

Due to their highly homologous kinase domains and variable regulatory domains, the

PKC isoforms can activate a number of different cellular signaling pathways.

Therefore, targeting this pathway remains challenging and the question to ask should

not be “what” activity to target, but “where”.

1.5.2 Activation of PKC isoforms and downstream signaling

PKCs contain an autoinhibitory pseudosubstrate. When PKC is inactive, it is folded

in such a way that its kinase region is bound to its own pseudosubstrate. Upon

phosphorylation of serine/threonine residues by any of its activators, PKC can unfold

and bind its true substrate. Stimulation of PLC increases the DAG levels at the

Figure 1.8 Schematic sequence of Protein Kinase C (PKC) isozymes indicating the domain

structure of the PKC subfamilies and their activators

(Figure adopted from Cosentino-Gomes D. et al. (Cosentino-Gomes et al., 2012)).

Page 59: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

42

plasma membrane, followed by the re-localization and activation of various PKC

isoforms. There are some requirements for the activation of PKC isoforms: regulation

of their intrinsic kinase domain activity through a priming phosphorylation of the

activation loop by PDK1; and phosphorylation (or charged residues) at the

hydrophobic sites within the C-terminal tail through mTORC2 (Facchinetti et al.,

2008, Ikenoue et al., 2008). Once activated, the C-terminal tail will fold back and

occupy the hydrophobic pocket to stabilize the active conformation. A recently

identified player in the activation of cPKCs and nPKCs is heat shock protein-90

(HSP90) (Gould et al., 2009). In order for the PKCs to mature so that they can be

phosphorylated by PDK1 and mTORC2, HSP90 and the co-chaperone Cdc37 need to

bind to a molecular clamp in the kinase domain formed by a conserved PXXP motif

in the carboxyl-terminal tail. If this binding is not possible, PKCs are unable to be

phosphorylated and will be degraded. Once HSP90 and Cdc37 have bound, PKCs can

be phosphorylated by PDK1, a step necessary for the processing of PKCs to their

fully phosphorylated forms.

When PKC binds to the membrane, it undergoes a conformational change to expose

its kinase binding region (Newton, 2003) and can activate downstream signaling

pathways, such as the MEK/ERK and PI3K/Akt (Cai et al., 1997, Balendran et al.,

2000). Different PKC isoforms show different subcellular localization depending on

the tissue type. The Receptors for Inactivated Kinases (RICKs) and the Receptors for

Activated Kinases (RACKs) bind PKC in an isozyme-specific manner and are

responsible for the translocation of PKC to the different cellular compartments

(Mochly-Rosen and Gordon, 1998). Phorbol esters can activate PKC by promoting its

translocation to the cell membrane, and this effect can be inhibited by

glycoglycerolipid analogues, which block its translocation and activation of the

downstream signaling pathways MAPK and FAK, which are involved in adhesion,

migration and proliferation (Colombo et al., 2011). Further downstream signaling of

Page 60: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

43

PKC includes the modulation of membrane structure events, transcription regulation,

receptor desensitization, and immune response (Nishizuka, 1992).

It has been shown that the PKC isoforms α, βI and γ can phosphorylate GSK3β at

Serine 9 and that this renders GSK3β inactive (Goode et al., 1992). This in turn

inhibits the phosphorylation of c-Jun at its inhibitory phosphorylation site, thereby

allowing it to interact with its DNA binding site. GSK3β therefore acts as a cytosolic

bridge between PKC and c-Jun.

1.5.3 PKCs in Cancer

The discovery of the PKC activator, phorbol ester, has led to the belief that the

activation of PKC can promote carcinogen-induced tumorigenesis (Griner and

Kazanietz, 2007). It has functions related to cell survival, proliferation, invasion,

migration, angiogenesis, apoptosis and drug resistance. However, targeting PKC has

been unsuccessful so far as it is an unpredictable target. In some cancers it can act as

an oncogene, whereas it can have tumor suppressing functions in others. PKC-δ, for

example, can promote tumor progression in lung and pancreatic cancers under certain

conditions (Symonds et al., 2011), whereas it can act as a tumor suppressor in many

cell types (Reyland, 2007). In colon cancer cell lines, both loss and over-expression

of PKC-ζ could decrease tumorigenocity (Ma et al., 2013, Luna-Ulloa et al., 2011);

and PKC-α can induce (Wu et al., 2013) and suppress (Gwak et al., 2009)

proliferation of colon cancer cells. Moreover, PKCs can promote apoptosis through

different pathways. These include secretion of TNFα and TRAIL in prostate cancer

cells by PKC-δ which triggers an autocrine apoptotic loop; induction of cytochrome c

release and activation of Caspase 3 by PKC-δ in colon and prostate tumor-derived

cell lines. On the other hand, PKC-δ can also confer resistance to apoptotic signals by

activation of the Akt pathway and modulation of NF-κB-dependent gene expression

(Diaz Bessone et al., 2011). PKCs not only affect apoptosis in cancer but also

Page 61: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

44

migration, invasion and tumor promotion. Over-expression of PKC-α can lead to

increased anchorage-independent growth, tumorigenicity and metastasis; over-

expression of PKC-βII enhances invasiveness through Ras and MEK signaling

(Zhang et al., 2004). It has been shown that PKC is able to phosphorylate Raf-1, but

this can only stimulate its autophosphorylation, not its activity towards MEK-1

(Macdonald et al., 1993). However, some studies on prostate cancer have found that

phosphorylation of PKC could inhibit the Raf Kinase Inhibitory Protein (RKIP)

thereby increasing Raf-1 activity. Activation of this pathway is linked with metastasis

in prostate cancer (Keller et al., 2004).

Recently, Gong et al. found opposing roles for conventional and novel PKC isoforms

in the activation and regulation of the Hippo-YAP pathway (Gong et al., 2015). This

pathway plays an important role in development by modulating cell number, cell

death and cell differentiation, and deregulation can be the cause for many types of

cancer (Yu and Guan, 2013). Important players of the Hippo pathway are the MST1/2

and LATS1/2 kinases, whereby LATS1/2 is phosphorylated by MST1/2. LATS1/2

can then phosphorylate and inhibit the transcriptional co-activators YAP and TAZ.

Moreover, it has been discovered that extracellular signals act through GPCRs to

regulate the Hippo-YAP pathway, and PKC is one of the main downstream actors of

GPCRs, linking the Hippo and PKC pathways. Interestingly, they found cell type-

specific roles for PKC in regulating the Hippo pathway. While the cPKCs can

dephosphorylate and activate YAP, the nPKCs phosphorylate YAP, thereby

inhibiting it. These effects occur through the activation/inhibition of LATS by the

different isotypes.

The diverse effects of PKC signaling on cancer cells could be the reason why PKC

inhibitors have been ineffective in clinical trials. Zhang et al. recently reported the

results of a meta-analysis of controlled trials using PKC inhibitors combined with

chemotherapy or chemotherapy alone in patients with non-small cell lung cancer

Page 62: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

45

(Zhang et al., 2015). They found that PKC inhibitors even decreased the response

rates (RR 0.79) and disease control rates (RR 0.90) with no significant difference in

progression free survival and overall survival. This raises the question of whether loss

or over-expression of PKC isozymes can promote carcinogenesis and promote tumor

progression. Although phorbol esters are tumor promoters and considered PKC

activators, it is well known that chronic treatment of cells with Phorbol esters can

deplete PKC isozymes, rather than lead to hyperactivation of the PKC signaling

pathway. This might explain the unexpected results of clinical trials.

1.5.4 PKC in Breast Cancer

Various PKC isoforms are involved in normal mammary development, such as in the

control and activation of mitogenic/apoptotic signaling. Their expression and

localization are modulated during mammary gland development and differentiation.

Moreover, an over-expression of PKC isoforms has been reported in breast tumors

and breast cancer cell lines (Jarzabek et al., 2002). PKC-α, for example, has been

found by some to be over-expressed in human breast cancer cells and tumor samples

(Lahn et al., 2004), and others have found it to be down-regulated (Kerfoot et al.,

2004). Moreover, PKC-α was found to play a role in breast CSCs as its expression

positively correlates with aggressive TNBC, and inhibiting PKC-α could specifically

target CSCs with little effect on non-CSCs (Tam et al., 2013). Furthermore, PKC-α

was shown to suppress c-Jun phosphorylation in ER+ breast cancer cells, leading to a

down-regulation of ER-α expression. It is, therefore, a potential therapeutic target for

treating ER+ Tamoxifen-resistant breast cancer (Kim et al., 2014).

1.5.4. PKC-β

PKC-β has two major splice variants: PKC-βI and PKC-βII. Although it has been

found that both isoforms have slightly different functions, it is complicated to

Page 63: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

46

interpret their roles since many early studies did not differentiate between the

different splice variants.

PKC-β has gained importance in breast cancer since the discovery of its role in

angiogenesis, and breast cancer tumorigenesis in rodent models and humans.

Especially in TNBC, the PKC-β inhibitor, LY379196, can inhibit growth of various

TNBC cell lines in vitro (Li and Weinstein, 2006). Although PKC-β drives tumor

growth and proliferation of cancer cells, over-expression can impair the tumorigenic

and metastatic potential of tumor-derived murine mammary cell lines, suggesting an

inhibitory role for PKC-β (Grossoni et al., 2009). In addition to growth inhibition,

over-expression could also revert a transformed phenotype by leading to re-

expression of fibronectin and extracellular matrix glycoproteins.

A recent study analyzed 8% of PKC mutations identified in human cancers and found

that the majority of these mutations are loss-of-function mutations, rather than gain-

of-function mutations (Antal et al., 2015). Moreover, they found that by correcting a

loss-of-function in PKC-β using Clustered Regularly-Interspaced Short Palindromic

Repeats (CRISPR) they could reduce tumor growth in a xenograft model of a patient-

derived colon cancer cell line. This study suggests that PKC isoforms act as tumor

suppressors rather than oncogenes, and that clinical studies are inconclusive as they

are focusing on diminishing PKC, rather than attempting to restore PKC function in

patients.

Despite these conflicting results, PKC-β inhibitors have also shown positive results in

breast cancer patients in preclinical and clinical studies. The potent PKC-β inhibitor,

Enzastaurin, could inhibit tumor growth and angiogenesis in human tumor

xenografts. Likewise, Phase II clinical trials involving Enzastaurin in patients with

high-grade gliomas and lymphomas have shown promising results, and prove the

Page 64: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

47

safety for this inhibitor in terms of effectiveness and toxicity (Sledge and Gokmen-

Polar, 2006).

In this review we will focus mainly on the PKC-β isoform as this thesis explores the

effect of the PKC-β inhibitor Enzastaurin on Paclitaxl-sensitivity in breast cancer

cells.

1.5.5. The role of PKC-β in cell cycle regulation

The fact that PKCs are downstream of various growth factors led to the idea that they

have an effect on mitotic signals. Indeed, PKCs can influence cell cycle, but

depending on various cell internal and external factors, they can have positive or

negative effects on cell cycle progression (Black, 2010). The effect of the PKCs is

dependent on the timing and duration of PKC activation, the specific PKC isoform

involved, the cellular context, cell type, and the signaling environment. PKCs can

influence the activation of various cell cycle regulatory proteins, such as cyclins,

cyclin-dependent kinases (CDKs), and CDK inhibitors, whereby p21Waf/Cip1 and cyclin

D1 seem to be the key targets.

The role of PKC-β in cell cycle is largely stimulatory. It has been shown to have

effects on the G1 phase through phosophorylation and stimulation of CAK activity

(Acevedo-Duncan et al., 2002), promotion of Rb phosphorylation (Suzuma et al.,

2002), and enhancement of cyclin D1 expression in breast cancer cells (Li and

Weinstein, 2006). It is also critical for nuclear lamina disassembly during the G2/M

transition by phosphorylating lamin B in K562 erythroleukemia cells (Thompson and

Fields, 1996). Most importantly, PKC-β has been shown to regulate microtubule

function. It was found that PKC can phosphorylate the microtubule-associated

protein, tau, at Serine8 in peptide 369 (Correas et al., 1992). Tau proteins stabilize

microtubules and promote tubulin assembly, however, both of these functions are

reduced when it is phosphorylated by PKC at its tubulin binding domain. Further

Page 65: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

48

phosphorylation by PKC on other Serine phosphorylation sites may even further

inhibit its tubulin binding capacity.

Page 66: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

49

1.6 Aims and objectives of the study

One of the major complications in the treatment of TNBC patients is the high rate of

relapse after an initial positive response to a combination of different types of

chemotherapeutic agents. Many patients suffer from residual disease and the relapse

rate is high. A retrospective analysis of 269 TNBC patients found that the 5-year

disease-free survival was only 68.2% with most patients suffering recurrent disease in

the first 3 years following diagnosis (Ovcaricek et al., 2011). Once the patients

present with metastatic or relapsed disease, the survival time rarely exceeds 1 year.

The reason for this is that relapsed disease is often resistant to chemotherapy, and

combined with the lack of effective targeted therapies, the treatment options are

limited.

Our aim was therefore to generate a model of Paclitaxel-resistance in TNBC which

mimics the situation in a patient who develops chemo-resistant recurrent disease after

chemotherapy treatment. Applying a top-down approach to our model we then sought

to identify pathways which play a crucial role in resistance and are more highly

activated in resistant compared to parental cell lines. At the same time we aimed to

discover compounds that can revert resistance and sensitize resistant cell lines to

Paclitaxel which can then be translated into a clinical setting.

Page 67: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

50

CHAPTER 2: MATERIALS AND METHODS

Page 68: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

51

2.1. Cell culture and treatments

MDA-MB231 cell line was obtained from American Type Culture Collection ATCC

(Manassas, VA) and SUM159PT was obtained from Asterand Bioscience

(Hertfordshire, UK). MDA-MB231 and its resistant derivatives were cultured in

Dulbecco’s modified Eagle’s medium (DMEM) (Gibco®) supplemented with 10%

fetal bovine serum (FBS) and 5000U/mL penicillin/streptomycin (Invitrogen).

SUM159PT and its resistant derivatives were cultured in Ham’s F-12 Nutrient Mix

medium with L-Glutamine (Gibco®) and supplemented with 5% FBS, 10mM HEPES

(Gibco®), 5ng/ml Insulin, 1μg/ml hydrocortisone and 5000U/mL

penicillin/streptomycin. All cell lines were maintained at 37°C with 5% CO2 in a

humidified atmosphere. In addition, the resistant cell lines were maintained in

different concentrations of Paclitaxel: 500nM, 1uM, 50nM and 75nM for SUM159PT

500nM-TR and 1uM-TR, and MB231 50nM-TR and 75nM-TR, respectively. For all

cell-based assays, the cells were seeded without Paclitaxel and treatment was started

24 hours later.

2.2. Cryopreservation of cell lines

Cell lines were kept long-term in a liquid nitrogen tank (at -196°C) and short-term

(~6 months) at -80°C. To recover cell lines, they were thawed at 37°C and neutralized

with 8ml complete media. They were then spun down at 1000rpm for 3 mins and the

cell pellet was resuspended in 10ml complete medium and seeded in a 75cm2 flask.

Cells were grown and allowed to stabilize for 1 week before they were used for

experiments. Cells were used for approximately 25 passages, after which they were

discarded and new vials of frozen cells were recovered. To freeze down cells, they

were trypsinized, neutralized with complete medium and spun down at 1000rpm for 3

mins. Cells from one 150cm2 flask (80% confluency) were then resuspended in 5ml

freezing medium (90% FBS and 10% DMSO) and 1ml aliquots were transferred into

Page 69: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

52

cryovials. For the freezing process, cryovials were placed into a Mr. Frosty cylinder

(NALGENE) which was filled with 99% isopropanol prior to transferring it into the -

80°C freezer. After one week, the cells were transferred to the liquid nitrogen tank.

2.3. Generation of Paclitaxel-resistant cell lines

Two TNBC cell lines were used to generate Paclitaxel-resistant derivatives,

SUM159PT and MDA-MB231. Prior to treatment, the EC50 to Paclitaxel was

measured for both cell lines. Cells were seeded in 6-well plates at a concentration of

0.5x105 cells/ml, 2ml per well and the treatment was started the next day at half the

concentration of the EC50. Once the surviving cells had recovered and started

proliferating again, the cells were passaged into a new plate and the Paclitaxel

concentration was doubled. This cycle continued until the final Paclitaxel

concentrations were achieved for all four resistant cell lines (Figure 1A). For each of

the two cell lines, 2 resistant derivatives were generated: SUM159PT 500nM-TR and

1uM-TR, and MDA-MB231 50nM-TR and 75nM-TR, growing in 500nM, 1μM,

50nM and 75nM Paclitaxel, respectively. The resistant cell lines were constantly

maintained in medium supplemented with Paclitaxel.

2.4. Transfection of small interfering RNA

siRNA transfection of cell lines was conducted using Lipofectamine RNAiMAX

(Invitrogen) according to the manufacturer’s protocol. Target-specific siRNA and

non-targeting control siRNA were purchased from Sigma Aldrich. Two target

sequences were used to knock-down AURKB: 1. siAurB (Pre) is pre-designed by

Sigma Aldrich (SASI_Hs01_00076963); 2. siAurB is custom designed according to

the sequence used by Ditchfield et al. (Ditchfield et al., 2003): 5’-

AACGCGGCACUUCACAAUUGA-3’. Non-targeting control used is MISSION®

siRNA Universal Negative Control #1 (SIC001) (Sigma Aldrich). For siRNA

transfection, 4μL siRNA (stock concentration 10μM) and 4μL RNAiMAX were

Page 70: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

53

diluted in 100μL Opti-MEM medium (Invitrogen). After 5 minutes of incubation,

both solutions were combined and incubated for another 20 minutes at room

temperature. Before adding the transfection mixture onto the cells which were seeded

in 6-well plates 18 hours earlier (1x105 cells/well), medium was changed to 800μL

fresh complete medium. The end concentration of the siRNA used is 40nM. 24 hours

later, the cells were then trypsinized and re-seeded for further down-stream assays.

2.5. EC50 calculation

To measure the resistance of cell lines to various chemotherapeutic drugs, the half

maximal effective concentration (EC50) of the drugs was calculated in those cell lines.

This value gives the concentration of a drug, which induces a response halfway

between the baseline and the maximum. It is used to measure the potency of a drug

and thereby resembles the sensitivity/resistance of cells to the drug. To measure the

EC50 of various drugs in different cell lines, a cell viability assay was used (see Cell

viability assay). The cells were treated 24 hours after seeding with 9 different drug

concentrations and a control. As the cell lines had different sensitivity to the chemo-

drugs, different concentrations were used: 1) Paclitaxel and Vincristine: working

concentrations of 10μM, 5μM, 1μM, 500nM, 100nM, 10nM, 1nM, 0.1nM, 0.01nM;

2) Adriamycin: 50μM, 10μM, 5μM, 1μM, 100nM, 10nM, 1nM, 0.1nM, 0.01nM. Cell

viability was measured after 4 days treatment and GraphPad Prism was used to plot a

sigmoidal-dose response curve (three-parameter logistic equation) to measure the

EC50 concentration of the chemo-drugs.

2.6. Cell viability assay

To measure the cell viability of cells in response to treatment, 1250 cells were seeded

per well in a volume of 100μl in a white, opaque 96-well plate. After 24 hours, the

media was changed to fresh media containing the drugs in the working concentration.

Cell viability was measured every day for 5 days, or after 4 days of treatment, using

Page 71: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

54

the CellTiter-Glo® Luminescent Cell Viability Assay (Promega) according to the

manufacturer’s instructions.

2.7. Flow Assisted Cytometry Analysis (FACS)

1x105 cells were seeded in 2ml complete medium per well in 6-well plates. 24 hours

later, the drugs were added into the wells, and the cells were harvested 4 days after

treatment. The cells were trypsinized, neutralized with complete medium and spun

down at 1000rpm for 3 mins. The cell pellet was washed once with 1.5ml ice-cold

phosphate-buffered saline (PBS) (Invitrogen) and then resuspended in 1ml ice-cold

70% ethanol. The samples were kept at 4°C for at least one hour and maximum 7

days. To stain the fixed cells, they were washed once with PBS and the cell pellet was

treated with RNase for 5 mins at room temperature (RT). Then 400μl of a 50μg/ml

Propidium Iodide (PI) solution was added to each sample, and they were incubated in

the dark at RT for 30 mins. The stained cells were analyzed for their DNA content by

FACS with FACSCalibur (Beckton Dickinson Instrument). The percentage of cell

death was defined as the SubG1 population quantified using the CellQuest software

(Beckton Dickinson).

2.8. Phospho-Histone H3 (Ser28) assay

The cells were treated for 24 hours after which they were washed with PBS and fixed

in 1ml ice-cold PBS for at least one hour but not more than 7 days. They were then

washed twice with PBS, blocked in 1ml blocking buffer (PBS, 0.1% Tween-20) for

15 mins on ice, washed with 1ml staining buffer (PBS, 1% FBS) and transferred to a

1.5ml Eppendorf tube. The cells were spun down at 2000rpm for 2 mins and the

supernatant was vacuumed, leaving approximately 40μl with the cell pellet. The cells

were incubated with 10μl anti-Phospho-Histone H3 (Ser28) antibody (Cell Signaling

Technology, #9713) for 45 mins in the dark at room temperature. The cells were then

washed once with PBS, incubated with 50μl RNase for 5 mins, followed by 400μl

Page 72: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

55

Propidium Iodide (50μg/ml) for 30 mins in the dark. The samples were analyzed

using FACS.

2.9. Caspase 3 assay

Caspase 3 is a marker for cells undergoing apoptosis and consists of a heterodimer of

17 and 12kDa subunits derived from a 32kDa proenzyme. The FITC-conjugated anti-

active Caspase 3 antibody was used to detect cells with activated Caspase 3 by flow

cytometry (BD Biosciences, #559341). The cells were seeded at a concentration of

1x105 cells/well in 2ml/well of 6-well plates and subjected to different treatment

groups 24 hours later. 24 hours after treatment, the cells were trypsinized and stained

with the FITC-conjugated anti-active Caspase 3 antibody according to the

manufacturer’s instructions. In short, the cells were washed with cold PBS once and

fixed in Cytofix/Cytoperm solution for 20 mins. They were then washed twice with

the Perm/Wash buffer and incubated with the FITC-conjugated anti-active Caspase 3

antibody for 1 hour. Then they were washed with the Perm/Wash buffer once and

analyzed by flow cytometry using the FACSCalibur. The amount of cells with active

Caspase 3 was determined using the CellQuest software.

2.10. Anchorage-independent Methylcellulose assay

The wells of an opaque 96-well plate with clear bottoms were coated with BactoTM

Agar before being used for this assay. To do so, the agar was dissolved in PBS to a

concentration of 3% and boiled until it was dissolved completely. It was then mixed

with complete medium to a final concentration of 0.6% and 80μl were used to coat

the bottom of each well. The plates were kept in the incubator (37°C, 5% CO2) for at

least 45 mins before use. The Methylcellulose powder (4,000 cP viscosity, 2% in

H2O; Sigma-Aldrich, St. Louis, MO) was autoclaved and dissolved in autoclaved

H2O to make a 2% stock solution. The cells were resuspended in fresh medium and

the cell concentration was measured. Consequently, the cells were diluted to a

Page 73: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

56

concentration of 2x104cells/ml in 30ml complete media, and 10ml of 2%

Methylcellulose solution was added. The final cell concentration attained was

1.5x104cells/ml in complete media with 0.5% Methylcellulose. 1ml aliquots were

made to which the drugs were added and 100μl were seeded per well. Subsequently,

the cells were allowed to grow for 13 days in the incubator before they were stained

with 80μl of a 40μg/ml p-Iodonitrotetrazolium Violet (INT) solution and the colonies

were counted and analyzed using the GelCountTM automatic plate scanner (Oxford

OPTRONIX) and the GelCount Version 0.025.1 software (Oxford OPTRONIX).

Plates were scanned at 1200 dpi.

2.11. Tumorsphere assay

To measure the number of tumor-initiating cells, a tumorsphere assay was performed.

The tumorsphere medium used to grow breast cancer tumorspheres (mammospheres)

consists of 90ml MammoCultTM Basal Medium (Human) (STEMCELLTM

Technologies, Vancouver, CA), supplemented with 10ml MammoCultTM

Proliferation Supplements (Human) (STEMCELLTM Technologies), 200μl heparin

solution (stock 0.2%), and 200μl hydrocortisone (stock 250μg/ml). Cells were

trypsinized, neutralized with complete medium and spun down at 1000rpm for 3

mins. The cell pellet was washed once with 5ml PBS, resuspended in 3ml

tumorsphere media, and the cell count was determined. 5,000 cells were seeded in

one well of a low-attachment 96-well plate in a volume of 2ml and placed in the

incubator. After 24 hours, the drugs were added into the wells without changing the

medium. Three days later, the wells were topped-up with 1ml of fresh tumorsphere

medium, and after 7 days 300μl of a 40μg/ml INT solution were added per well and

incubated over night to stain the tumorspheres. The INT end concentration is 4μg/ml.

The number of tumorspheres was measured the next day using the GelCountTM

automatic plate scanner and the GelCount software. Plates were scanned at 1200 dpi

and the colony detection algorithm was optimized for each cell line individually.

Page 74: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

57

2.12. 3D Matrigel anchorage-independent growth assay

An 8-well chamber slide (#384118, BD Biosciences) was pre-coated with 45μl of

7.6mg/ml growth factor-reduced matrigel (#356231, BD Biosciences) for 30 mins at

37°C. 5x103cells in a volume of 400μl in complete media with 150μg/ml matrigel

containing the different concentrations of the drugs were seeded into each well. The

cells were placed in the incubator and media was replaced with fresh media after 4

days. Phase contrast images were taken of the cells at different time points.

2.13. RNA extraction

Cell pellets were collected by trypsinizing monolayer cells, and the cell pellet was

washed once with ice-cold PBS. The cells were resuspended in 700μl Qiazol

(Invitrogen) to lyse the cells, after which 200μl chloroform was added and the

mixture was centrifuged at 13,000rpm for 15 mins at 4°C to separate RNA, DNA and

other cellular components. After centrifugation, the top clear layer was transferred

into a fresh 1.5ml eppendorf tube, followed by the addition of 540μl 100% ethanol.

The miRNeasy mini kit (Qiagen) was used to purify the RNA. In short, the samples

were transferred to the column and the RNA was allowed to bind to the membrane

via centrifugation. The columns were washed once with 700μl RW1 buffer, and twice

with 500μl RPE buffer. Subsequently, the RNA was eluted from the column with

50μl RNAse-free water. The concentration of the RNA was determined using a

NanoDrop ND-1000.

2.14. cDNA conversion and quantitative RealTime-PCR (RT-PCR)

To transcribe RNA into single-stranded complementary DNA (cDNA), a High

Capacity cDNA Reverse Transcription Kit (Applied Biosystems) was used. Briefly,

750ng of RNA were diluted in 25μl RNAse-free water. Then a master mix was

prepared with 5μl RT buffer, 5μl random primers, 2μl dNTP mix, 2.5μl

Page 75: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

58

MultiScribeTM reverse transcriptase, and 10.5μl RNAse-free water, which was added

to the previously prepared RNA. A PCR was then performed in a thermo cycler at

25°C for 10 mins, followed by 37°C for 2 hours. To perform quantitative RealTime-

PCR, 0.44μl of the cDNA (15ng/μl) was added to 3.96μl RNAse-free water.

Concurrently, 0.4μl gene-specific primer mix (10μM) was added to 5μl of 2X master

mix from KAPA SYBR® FAST qPCR Kit (Kapa Biosystems). The two reactions

were then mixed together and subjected to quantitative RealTime-PCR. The reaction

mix was amplified and quantified with PRISM 7900 Sequence Detection System

(Applied Biosystems). 18S or Actin were used as internal controls. All the reactions

were analyzed using Applied Biosystems PRISM 7500 Fast Real-Time PCR system

in 96-well plate format.

Table 2.13 Primers used for quantitative RealTime-PCR of target genes

Page 76: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

59

2.15. Microarray gene expression profiling

To control the quality of the mRNA used and to detect mRNA degradation, Agarose

gel electrophoresis was performed on a 1% Agarose gel. The microarray

hybridization was performed using the Illumina Gene Expression Sentrix BeadChip

HumanRef-8_V2 (San Diego, CA). In short, 500ng of RNA was used to perform

cDNA conversion and further processed into double-stranded cDNA. The purified

cDNA was used to generate biotinylated cRNA, which was likewise purified before

further processing. The biotinylated cRNA was hybridized onto the BeadChip,

washed, and stained with streptavidin-Cy3. The BeadChip was scanned using the

Illumina BeadArray Reader and the images were stored with indicated barcodes.

Illumina GenomeStudioTM was used to analyse the scanned images and the output

was used for further analysis with GeneSpringGXTM (Agilent Technology). The data

were analyzed by selecting Illumina single color as the experimental type and quartile

normalization was performed for the readings. Fold changes in gene expression of the

resistant cells lines were analyzed by pair-wise comparisons to the respective parental

cell lines.

2.16. Protein extraction

For total protein extraction, the cells were trypsinized, neutralized with complete

medium and spun down at 1200rpm for 3 mins and washed once with 1.5ml ice-cold

PBS. The volume of the cell pellet was estimated and resuspended in 5x the volume

of radioimmunoprecipitation assay (RIPA) lysis buffer (50nM Tris-HCl pH7.4, 1mM

EDTA, 150mM NaCl, 1% Igepal CA630, 0.5% sodium deoxycholate, 1mM

Na3VO4, 20mM NaF, 1mM PMSF, and complete protease inhibitor (Roche)). The

samples were kept on ice for 30 mins and vortexed every 5 mins for 10 seconds, and

then sonicated 3x 5 seconds at 20% maximum force, after which they were spun

down at 13,200rpm for 15 mins at 4°C. The supernatant containing the protein was

Page 77: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

60

transferred into a fresh eppendorf tube and the concentration was estimated with the

DC Protein Assay (Bio-Rad) using BSA to generate a standard curve, and measured

using the Tecan XflourTM software.

2.17. Western blotting

A 10% or 12% SDS-PAGE gel was used to separate protein samples (20-30ug),

which were then transferred onto a PVDF membrane (Milipore) at 23V for 70 mins

using the Trans-Blot SD Semi-Dry transfer cell (Bio-Rad). The membranes

containing the protein were blocked with 5% non-fat milk (Bio-Rad) or 5% bovine

serum albumin (BSA) for 2 hours, or overnight. The primary antibodies were diluted

to their recommended working concentrations using the blocking buffer and

incubated on the membranes overnight, or for 2 hours RT, according to the

manufacturer’s instructions. The membranes were washed 3 times for 5 mins using

the washing buffer and incubated with the HRP-conjugated secondary antibody

diluted in blocking buffer for 1 hour at RT. The membranes were incubated with the

chemiluminescent ECL Substrate Kit (GE Healthcare) for 5 mins, and the signal was

subsequently detected using the ChemiDocTM Imaging System (BIO-RAD). The blots

were probed with primary antibodies against PKC-β (GTX113252) purchased from

GeneTex; phospho-PKC-βI (Thr641) (sc-101776), PKC (sc-80) and MCL-1 (sc-819)

purchased from Santa Cruz Biotechnology Inc.; Aurora B/AIM1 (#3094S), phospho-

Aurora A/B/C (T288/232/398) (#2914S), phospho-C-Raf (Ser289/296/301) (#9431P),

phospho-GSK3β (Ser9) (#5558S) and cleaved PARP (#9541S) purchased from Cell

Signaling Technology®; and β-Actin purchased from Sigma Aldrich.

Page 78: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

61

2.18. H-score calculation

H-score was used as a semi-quantitative analysis to describe the extent of

immunohistological staining of patient tumor samples. The formula used is the

following:

Samples used to calculate H-score are derived from The Human Protein Atlas

(www.proteinatlas.org). Patient samples made available from this database were

stained with 2 different anti-PKC-β antibodies and grouped into low, medium and

high intensity staining. According to this grouping, the H-score was calculated for

normal breast tissue and breast cancer samples.

2.19. Statistical analysis

Statistical analyses of XY and bar graphs were performed using the GraphPad Prism

software. Experimental readings were performed in duplicates, triplicates or 6

replicates and a Student’s T-test was used to generate p-values for measuring

significance between measurements. All statistical tests and graphs are generated with

GraphPad Prism Version 6.

Page 79: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

62

CHAPTER 3: RESULTS

Page 80: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

63

3.1. Generation and characterization of Paclitaxel-resistant TNBC cell

lines

To study mechanisms of acquired chemo-resistance in Triple Negative Breast Cancer

(TNBC) cell lines and, subsequently, find ways to revert resistance we generated

chemo-resistant cell lines. We used Paclitaxel-resistance as a model as it is used in

first line treatment for patients harboring triple negative breast tumors. To generate

this model we used the two TNBC cell lines, SUM159PT and MDA-MB-231

(hereafter named MB231). The cell lines were seeded in a 2D culture and treated with

a Paclitaxel starting concentration of ½ of the EC50 concentration which leads to a

reduction of cell viability and apoptosis in approximately 30% of the cells. As soon as

the remaining cells had stabilized and started proliferating again, the Paclitaxel

concentration was doubled (Figure 3.1.1 (A)). After approximately 5 months, we had

generated two resistant derivatives for each of the two TNBC cell lines, stably

growing in different Paclitaxel concentrations. For SUM159PT, the resistant cell lines

grow in 500nM and 1μM Paclitaxel, and are termed 500nM-TR and 1μM-TR. The

MB231 resistant cell lines grow in 50nM and 75nM Paclitaxel, and are therefore

called 50nM-TR and 75nM-TR, respectively. The parental MB231 cell lines show a

lower intrinsic resistance to Paclitaxel, therefore the resistant cell lines cannot be

stably grown in higher concentrations. To measure resistance, we calculated the EC50

of Paclitaxel in these cell lines using a cell viability assay. The EC50 is the

concentration of a drug which gives a response halfway between the baseline and the

maximum. As seen in Figure 3.1.1 (B) and (C), the resistant cell lines show ~1000-

fold and 300-fold higher EC50 value compared to their chemo-sensitive counterparts

in SUM159PT and MB231, respectively.

Page 81: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

64

Figure 3.1.1 Generation of Paclitaxel-resistant

TNBC cell lines

A. Paclitaxel-sensitive parental SUM159PT

and MB231 cell lines were treated with

½ of the Paclitaxel EC50. After initial

reduction in cell viability and cell death,

the remaining cells were further cultured

and the Paclitaxel concentration was

doubled. This cycle was repeated until 2

resistant derivatives were derived from

each cell line: SUM159PT 500nM-TR

and 1μM-TR; and MB231 50nM-TR and

75nM-TR

B. Paclitaxel EC50 for both parental and the

resistant cell lines was measured using

cell viability assay. The cells were

treated 24 hours after seeding and cell

viability was measured after 4 days

treatment (n=6). The EC50 was calculated

using Sigmoidal dose-response and

plotting the non-linear regression curve

fit (Error bars show ±SD, n=6)

C. Bar graph depicting the increase in

Paclitaxel EC50 in the resistant compared

to the parental cell lines, as measured

with cell viability assay.

B

A

C

Page 82: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

65

As we know, chemo-resistance has been linked to cancer stem cells (CSCs), as these

are believed to confer resistance through mechanisms such as up-regulation of anti-

apoptotic proteins such as Bcl-2, and high expression of ABC transporters for

effective efflux pumping. Therefore, we examined the population of CSCs in the

resistant cell lines which can be identified by the expression of specific cell surface

markers. Breast CSCs are characterized by a high expression of CD44 and a low

expression of CD24. FACS analysis revealed a 3-fold increase in the CSC population

in the SUM159PT resistant cell lines, and a 3- and 5-fold higher amount in the

MB231 50nM-TR and 75nM-TR cell lines, respectively (Figure 3.1.2 (A) and (B)).

These results are in concordance with other studies showing an increase in the CSC

population in chemo-resistant ovarian and colorectal cancer cell lines (Cole et al.,

2014, Dallas et al., 2009). We performed a tumorsphere assay in order to validate the

increase in CSCs in SUM159PT parental and resistant cell lines (Figure 3.1.2 (C)).

Surprisingly, the tumorspheres generated by the SUM159PT resistant cell lines show

a different morphology compared to the parental cell lines. While the parental cell

lines build round distinct spheres, the spheres generated by the resistant cell lines

seem to be aggregating. It can be ruled out that these are merely simple cell

aggregates as they cannot be dissociated easily by pipetting and need to be

trypsinized to generate single cells, a characteristic of spheres with intact cell-cell

interactions (Manuel Iglesias et al., 2013). In addition, the high expression of the cell

surface marker CD44 can make the cells more “sticky”, so the spheres are able to

clump together.

Page 83: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

66

A

B

C D

Page 84: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

67

When we checked for the expression of the breast CSCs markers, CD44 and CD24,

we found that the expression of CD24 was reduced in the parental SUM159PT cell

line compared to the resistant cell lines, whereas there was an increased expression of

CD44 in the resistant cell lines. These results validate our FACS analysis, which

identified an increase in the CD44+/CD24- CSCs population in the resistant

compared to the parental cell lines.

Figure 3.1.2 Increase in the cancer stem cell population in Paclitaxel-resistant cell lines

A. FACS analysis of SUM159PT and MB231 parental and resistant cell lines. The cells

were stained with APC- and FITC-conjugated CD24 and CD44 antibodies,

respectively.

B. Bar graph depicting the percentage of CD24-/CD44+ population in parental and

resistant cell lines as determined by FACS analysis (A).

C. Images of tumorspheres of SUM159PT parental, 500nM-TR and 1μM-TR cell lines

after 7 days growth in anchorage-independent conditions.

D. RealTime-PCR was performed on SUM159PT parental and resistant cell lines to

determine mRNA expression of CD44 and CD24. mRNA expression values were

normalized against 18S and fold change values were plotted. (Error bars show ± SD,

n=3).

Page 85: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

68

3.2. Gene expression profiling of SUM159PT Paclitaxel-resistant cell lines

To gain insights into the differential regulation of genes and pathways in the resistant

derivatives of SUM159PT compared to the parental cells, we performed microarray

analysis. After analyzing the microarray data with GeneSpringGXTM, we extracted

the list of genes significantly up-regulated in the 500nM-TR and 1uM-TR compared

to the parental cell line (fold change > 2; FDR<0.05) and analyzed them with

Ingenuity Pathway Analysis® (IPA). We examined the functions and pathways that

these genes are involved in and plotted the predicted Activation Z-score for each

pathway. This score represents the bias in gene regulation and predicts whether this

pathway is activated (Activation Z-score > 2) or inhibited (Activation Z-score < -2).

We found that genes up-regulated in both cell lines lead to an increased activation of

pathways and functions such as Cell Death and Survival, Cellular Movement,

Cellular Assembly and Organization, and Cellular Development, Growth and

Proliferation, among others (Figure 3.2.1). In contrast, increased activation of these

genes leads to an inhibition of apoptotic and cell death pathways. These findings are

not surprising as the resistant cell lines are maintained in Paclitaxel and are under

constant survival pressure, although they have acquired resistance. This means that

cell survival and proliferative pathways are up-regulated, while apoptotic pathways

are down-regulated. Moreover, resistant cell lines show an increase in microtubule

dynamics, a consequence of the mechanism of action of Paclitaxel, which stabilizes

microtubules, in order to maintain an intact cell cycle and chromosome segregation.

This comes hand in hand with an activation of cell cycle progression pathways, and

increased invasive and migratory properties of the resistant cell lines.

Page 86: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

69

3.2.1. The NF-κB network

We then wanted to determine the gene networks which are more highly activated in

the resistant cell lines compared to the parental cell line. IPA analysis lets us

determine these networks and ranks them according to a score calculated by their

algorithm. This score is calculated based on the p-value which takes into account the

number of Focus Genes in a certain network. The p-value is calculated based on the

probability of finding f or more Focus Genes in a set of n genes randomly selected

from the Global Molecular Network and the size of the network to approximate its

relevancy to the list of Focus Genes. Fisher’s exact test is used to calculate the p-

Figure 3.2.1 Changes in functions and pathways in Paclitaxel-resistant TNBC cell lines

Microarray analysis was used to determine gene expression changes in the SUM159PT

resistant compared to the parental cell lines. After quantile normalization, significantly over-

expressed genes (FDR<0.05, fold change>2) common in both resistant compared to parental

cell lines (581 genes) were analyzed using Ingenuity Pathway Analysis®. The figure shows

significantly altered pathways and functions according to the Activation z-score, which is

dependent on the number of genes whose expression is significantly up-regulated in the gene

sets for each specific function/pathway.

Page 87: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

70

value which in turn is used to determine the p-score. This score is defined as:

p-score= -log10(p-value). The top 5 networks with the highest scores when comparing

gene expression values in the SUM159PT resistant compared to the parental cell lines

are shown in Figure 3.2.2 (A). We zoomed into the highest ranked network and found

that most of the genes involved belong to the NF-κB complex (Figure 3.2.2 (B)). We

extracted the normalized gene expression values from the GeneSpring analysis of the

genes from Network 1 which are NF-κB-related genes and validated them using

RealTime-PCR (Figure 3.2.2 (C)). We found that, indeed, most of the genes are more

highly expressed in the resistant compared to the parental cell lines, but expression

values were not as drastic as in the microarray data.

Page 88: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

71

NF-κB complex

Figure 3.2.2 The NF-κB network

in Paclitaxel-resistant cell lines

A. IPA was used to determine

enriched networks in

SUM159PT 500nM-TR

and 1μM-TR compared to

the parental cell lines.

Genes selected for the

analysis were commonly

up-regulated in both

resistant cell lines and had

a fold change cut-off=2 and

FDR<0.05.

B. The top network with the

highest score has the NF-

κB complex at the centre.

C. Gene expression values of

genes which play a role in

NF-κB signaling and are

found in the top network.

Gene expression values

from microarray analysis

shown are after quantile

normalization and relative

to the parental cell line

(fold change>2,

FDR<0.05). RealTime-

PCR analysis is normalized

to gene expression values

of SUM159PT parental

cells. 18S was used to

normalize mRNA

expression values (Error

bars show ±SD, n=3).

A

B

C

Page 89: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

72

It has been shown that the NF-κB pathway plays an important role in chemo-

resistance and inhibition of this pathway (using disulfiram and copper) sensitizes

breast CSCs to Paclitaxel (Yip et al., 2011). To verify its role in our model of

Paclitaxel-resistance in TNBC cell lines, we used various NF-κB inhibitors to test

whether this pathway is more important for cell survival in the SUM159PT 500nM-

TR and 1μM-TR resistant cell lines compared to the parental cell line. We treated the

cells for 4 days with 3 IKK inhibitors: 1. BAY 11-7082, an inhibitor of the IκB kinase

(IKK) α subunit, IKKα, and of phosphorylation of cytokine-inducible IκBα

(IC50=10μM); 2. PHA-408, a potent inhibitor of IKKβ (IC50=40nM); 3. PS-1145, a

selective IKK inhibitor (IC50=100nM). The IKK kinase complex is made of the two

kinases IKKα and IKKβ, where IKKα plays a role in the non-canonical NF-κB

pathway and IKKβ in the canonical NF-κB pathway. As a negative control, to

determine whether the effect of treatment is specific to the NF-κB inhibitors, we

included a MEK inhibitor (PD0325901, IC50=0.33nM), and a PDK1 inhibitor

(BX795, IC50=6nM). We found that the resistant cell lines were only slightly more

sensitive to the NF-κB inhibitors compared to the parental cell line at high

concentrations, although the parental cell line also responded to the inhibitors in a

dose-dependent manner (Figure 3.2.3). The two inhibitors used as negative controls,

MEK and PDK1 Inhibitors, showed a dose-dependent effect in the resistant and

parental cell lines, although the resistant cell lines are more resistant compared to the

parental cell line.

These results don’t fit our hypothesis to identify activation of pathways specific to

Paclitaxel-resistant cell lines. This could possibly be because the NF-κB pathway is

important in TNBC, even without Paclitaxel-resistance, which is why various NF-κB

inhibitors are only slightly effective in the resistant cell lines. We therefore decided to

use a different approach to find pathways that play a role in Paclitaxel-resistance and

to find ways to revert resistance.

Page 90: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

73

Figure 3.2.3 SUM159PT Paclitaxel-resistant cells are equally sensitive to NF-κB inhibitors

as compared to the parental cell line

SUM159PT parental and 500nM-TR and 1μM-TR resistant cell lines were treated for 4 days

with different concentrations for all inhibitors, after which cell viability was measured. All

measurements are normalized to the DMSO control and the percentage is plotted. (Error bars

show ±SD, n=3).

Page 91: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

74

3.3. Kinase inhibitor screen to identify Paclitaxel-sensitizing compounds

To identify compounds that can re-sensitize resistant TNBC cells to Paclitaxel, we

performed a kinase inhibitor screen using 180 small molecule inhibitors targeting

different kinases. This screen was used to detect synthetic lethal drug combinations

with Paclitaxel, specific for the resistant cell lines.

The cells were seeded in 96 well plates, treated 24 hours later and after 4 days

treatment, cell viability was measured. The cells were treated either with the inhibitor

alone, or in combination with Paclitaxel (Figure 3.3.1 (A)). We then calculated the Z-

score for each treatment group for each resistant cell line separately. Z-score

calculations take into account the mean of a population (in this case the mean of all

the cell viability measurements for each treatment group (inhibitor vs. inhibitor +

Paclitaxel) for one cell line, and assigns each single measurement a score, depending

on whether it is above or below the population mean. Scores > 0 mean that the cell

viability is higher, whereas scores < 0 mean that cell viability is lower than the

population mean. When Z-scores of the inhibitor single treatment vs. inhibitor +

Paclitaxel treatment are plotted on a scatter plot (Figure 3.3.1 (B)), distinct

populations can be identified. The inhibitors most interesting to us are the ones

clustering in the II. Quadrant. These inhibitors show no effect on cell viability as

single treatment but significantly reduced cell viability in the combination treatment.

Inhibitors in the III. Quadrant also have an effect on the resistant cell lines as single

treatment and are, therefore, not interesting to us as they are not targeting pathways

specific to Paclitaxel-resistance. Inhibitors clustering in the I. Quadrant have no effect

as single treatment or in combination with Paclitaxel and we also eliminated these.

Many of the inhibitors in the II. Quadrant are PKC and PKC-β inhibitors, suggesting

a strong role for the PKC pathway in Paclitaxel-resistance.

Page 92: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

75

Figure 3.3.1 Kinase inhibitor screen identifies PKC-β inhibitors to be able to re-sensitize

resistant cells

A. Schema for performing the kinase inhibitor screen. The cells were seeded in 96-well

plates and treated with 180 small molecule inhibitors targeting different kinases 24

hours later. All the inhibitors were used at 5μM unless otherwise stated (Figure 3.3.2).

One set was treated with the inhibitor alone, whereas another set was treated with the

inhibitor in combination with Paclitaxel. Cell viability was measured after 4 days of

treatment. Cell viability measurements were normalized to the DMSO control.

B. After normalization, the Z-score was calculated for each inhibitor by determining the

whole population mean for each cell line separately. The Z-score of single treatment

was then plotted against the Z-score for combination treatment on a scatter plot. The

inhibitors in the II. Quadrant are those with no effect as single treatment, but reduce

cell viability when combined with Paclitaxel.

A

B

Page 93: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

76

To calculate whether the difference of single treatment vs. combination treatment was

significant for each compound, we used a Student’s t-test to calculate p-values. We

assumed that the cell viability measurements of the 4 resistant cell lines (SUM159PT

500nM-TR and 1uM-TR, MB231 50nM-TR and 75nM-TR) are replicates in the two

different treatment groups: inhibitor alone, and inhibitor + Paclitaxel. After

calculating the p-value between the two treatment groups, and thereby identifying

those inhibitors that have no effect when given as a single treatment but lead to

reduced cell viability in combination with Paclitaxel, we ranked the inhibitors

accordingly (Figure 3.3.2). As shown, some of the top hits included inhibitors

targeting proteins which have been known to play a role in chemo-resistance, such as

p38 in colorectal cancer (Grossi et al., 2014), the hedgehog pathway (Hh) in breast

cancer and prostate cancer (Chai et al., 2013, Singh et al., 2012), EGFR in ovarian

cancer cell lines (Servidei et al., 2008), the MEK5/ERK5 pathway, whose over-

expression was found to be associated with poor survival in breast cancer patients

treated with chemotherapy (Miranda et al., 2015), and BTK which, when inhibited,

can sensitize leukemic cells to chemotherapeutic drugs (Uckun et al., 2002).

Page 94: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

77

Figure 3.3.2 Top hits of the kinase inhibitor screen

The inhibitors were ranked according to the p-value calculating the significant difference in cell

viability of inhibitor alone vs. combination treatment in the resistant cell lines. The cell

viability measurements in the 4 resistant cell lines (SUM159PT 500nM-TR and 1μM-TR, and

MB231 50nM-TR and 75nM-TR) for each treatment group were taken as replicates. Using this

method the inhibitors were ranked according to how significantly they could reduce cell

viability in combination with Paclitaxel, while having minimal effect as single agent. Cell

viability measurements are color-coded with red depicting low, and green depicting high

values. All inhibitors were used at 5μM unless stated otherwise.

Page 95: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

78

3.4. Validation of top hits from the kinase inhibitor screen

For further validation, we decided to focus on novel targets in chemo-resistance such

as Kit/PDGFR, TNKS1/2, PKC-β, and FLT3, in addition to some of the previously

published targets (p38 MAPK, Hh, BTK, PKA), which serve to validate our own

screen, but also whose role specifically in breast cancer chemo-resistance is not clear.

We first validated 9 inhibitors (highlighted in yellow) which target the

aforementioned kinases with a cell viability assay at 2 different concentrations each

(1μM and 5μM). As shown in Figure 3.4.1 (A) and (B), all the inhibitors lead to a

reduction in cell viability in all the 4 resistant cell lines when used in combination

with Paclitaxel, but have no effect as a single agent. In addition, they also have no

effect in SUM159PT and MB231 parental cell lines. Cell viability is normalized to

Day 0, which means that if the bar drops below CTG Signal=0, it is suggestive of

apoptosis. In general, the SUM159PT resistant cells are more sensitive to the

combination treatment, with more inhibitors leading to apoptosis, compared to the

MB231 resistant cell lines. This can be linked to high Paclitaxel EC50 in these cell

lines.

Page 96: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

79

The only inhibitors which were able to reduce cell viability below 0 in combination

with Paclitaxel in all the 4 resistant cell lines are the PKC-β inhibitors (LY333531

and Enzastaurin), as well as the PKA inhibitor (H-89) (except in MB231 50nM-TR).

Since the PKA inhibitor H-89 was also one of the top hits of the kinase screen with a

significant p-value calculation, and PKA has been previously reported to play a role

in chemo-resistance in breast cancer cells through regulation of the expression of the

Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen

Validation of the 9 top hits in SUM159PT (A) and MB231 (B) parental and both resistant cell

lines. The cells were treated with inhibitor alone (at 1μM and 5μM each) or in combination

with Paclitaxel and cell viability was measured on Day 0 and Day 4. Day 4 measurements were

normalized to Day 0 and values were plotted on the bar graph. All the inhibitors had no or

minimal effect as single agent in the parental or resistant cell lines but could successfully

sensitize resistant cells to Paclitaxel when compared to the Paclitaxel treatment alone. (Error

bars indicate ±SD, n=6).

A B

A B

Page 97: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

80

MDR1 protein, we also used this inhibitor for the following validation assay (Cvijic

and Chin, 1997).

We performed a Methylcellulose assay which measures colony formation in

anchorage-independent conditions. We found that both PKC-β inhibitors (LY333531

and Enzastaurin) and the PKA inhibitor (H-89) given at 1μM and 5μM each, led to a

significant decrease in the number of colonies in combination with Paclitaxel, but not

as single treatment (Figure 3.4.2 (A) and (B)). Additionally, the effect was slightly

stronger with the PKC-β inhibitors compared to the PKA inhibitor at low

concentrations, as seen for the SUM159PT resistant cells. Interestingly, the resistant

cell lines showed no change or a slight increase in colony formation in the Paclitaxel

compared to the DMSO treatment groups, suggesting that the resistant cells, to some

degree, rely on Paclitaxel for cell growth Paclitaxel treatment alone can even enhance

colony formation.

Page 98: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

81

Figure 3.4.2 PKC-β and PKA inhibitors can inhibit colony formation of Paclitaxel-

resistant cell lines in the presence of Paclitaxel

Validation of the PKC-β (LY333531, Enzastaurin) and PKA (H-89) inhibitors in SUM159PT

(A) and MB231 (B) in an anchorage-independent Methylcellulose assay. The cells were seeded

in a Methylcellulose assay and treated for 13 days before the colonies were stained with INT

o/n. The plates were then scanned using a GelCount and colonies/well were counted. The

inhibitors could significantly reduce colony formation in the resistant cell lines in combination

with Paclitaxel but only had a slight or no effect when used a single agent. (Error bars show

±SD, n=6)

A B

Page 99: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

82

For subsequent experiments, a Paclitaxel treatment control was added for all the

parental cell lines where they were treated with a very low dose of Paclitaxel. This

dose was derived from the cell survival of the resistant cell lines in their maintenance

doses of Paclitaxel. For SUM159PT, the 500nM-TR cell line is cultured in 500nM

Taxol, and according to the Sigmoidal curve formula when calculating the EC50,

98.9% of the cells survive. We then calculated how much Paclitaxel is needed for the

parental cells, so that 98.9% survive. For SUM159PT parental cells, 98.9% cell

survival is achieved with 0.73nM Paclitaxel. For MB231 parental cells the Paclitaxel

dose used is 0.46nM, derived from cell survival of the 50nM-TR cell line in 50nM

Paclitaxel.

Page 100: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

83

3.5. The PKC-β and PKA inhibitors sensitize resistant cells and induce

apoptosis in the presence of Paclitaxel

To test whether the reduction in cell viability after combination treatment is really

due to an induction of cell death, we performed 2 different apoptotic assays: Caspase

3 and Propidium Iodide (PI) staining. Both assays are flow cytometry-based, where

the former detects cells with activated Caspase 3, and the latter detects the apoptotic

cell population based on DNA fragmentation. For these assays we used the PKC-β

inhibitor Enzastaurin and the PKA inhibitor H-89. We first performed a Caspase 3

assay and found that single treatment with either of the inhibitors had little to no

effect on the activation of Caspase 3 in any of the resistant cell lines (Figure 3.5.1).

However, in combination with Paclitaxel we observed a significant increase in

Caspase 3+ cells, whereby the combination with Enzastaurin yielded the greatest

amount of apoptotic cells in all 4 resistant cell lines. The combination treatment with

the PKA inhibitor and Paclitaxel only resulted in a mild increase in Caspase 3+ cells.

As expected, Paclitaxel treatment alone could not increase the amount of Caspase 3+

cells in the resistant cell lines. Moreover, the parental cell lines showed a slight

increase in Caspase 3+ cells after single treatment with Enzastaurin alone, but there

was no further increase when Paclitaxel was added to the treatment.

Page 101: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

84

We then performed PI staining to validate these results in the resistant cell lines. We

treated the cells for 4 days with either the inhibitors alone or in combination with

Paclitaxel. The cells were then fixed and stained with PI which intercalates into the

Figure 3.5.1 PKC-β and PKA inhibitors induce Caspase 3 activation in the presence of

Paclitaxel

Measurement of Caspase 3+ cells after treatment with PKC-β and PKA inhibitors at different

concentrations with and without Paclitaxel in SUM159PT (A) and MB231 (B) parental and

resistant cell lines. The Paclitaxel concentration used for SUM159PT and MB23 parental cells

is 0.73nM and 0.46nM, respectively, whereas for the resistant cells, the maintenance

concentration was used. The cells were treated for 4 days, after which they were fixed with

70% ethanol for 1 day and then stained with a FITC-conjugated anti-Caspase 3 antibody. FACS

was performed to detect FITC-positive cells (n=1).

A B

Page 102: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

85

DNA, thereby allowing us to measure the DNA content of the cells. A normal 2N

DNA content means that the cells are in the G1 phase, a DNA content of 4N means

that the cells are in the G2/M phase, and a DNA content less than 2N suggests that

the cells have undergone DNA fragmentation due to apoptosis. This is called the

SubG1 population which can be measured using this assay and represents the

population of apoptotic cells. As can be seen in Figure 3.5.2, single inhibitor

treatment did not lead to apoptosis, whereas the combination with Paclitaxel lead to a

significant increase in the amount of apoptotic cells in the resistant cell lines in a

dose-dependent manner. Interestingly, the percentage of cells undergoing apoptosis

was greater when they were treated with the PKC-β inhibitor Enzastaurin as

compared to the PKA inhibitor in all 4 cell lines.

A B

Page 103: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

86

We therefore decided to focus on PKC-β as a target for chemo-resistance in triple

negative breast cancer cells. For one, in the top 40 hits of the kinase inhibitor screen,

3 target hits were PKC-β inhibitors (LY333531, Enzastaurin and LY317615), and one

was a pan-PKC inhibitor (Sotrastaurin) (Figure 3.4). This is strongly indicative of the

importance for the role of PKC-β in chemo-resistance. Secondly, it has a significant

effect in reducing cell viability and anchorage-independent growth in resistant cells,

and induces high levels of apoptosis in combination with Paclitaxel. And thirdly,

PKC-β inhibitors are the most advanced in clinical trials out of all the validated hits,

and have undergone successful Phase III clinical trials for complications of diabetes

(Danis and Sheetz, 2009), as well as various pre-clinical and clinical trials for

treatment of patients with different hematological and non-hematological cancers,

such diffuse large B-cell lymphoma (DLBCL) (Robertson et al., 2007), non-small cell

lung cancer (Oh et al., 2008), metastatic colorectal cancer (Glimelius et al., 2010),

and recurrent high-grade gliomas (Kreisl et al., 2010). Although the responses have

been mixed and negative so far, Phase II studies were conducted which used

Enzastaurin in combination with Paclitaxel and Carboplatin as first-line treatment for

ovarian cancer to increase progression-free survival (Vergote et al., 2013).

Enzastaurin is an oral drug with high safety in patients, also in combination with

chemotherapeutic drugs. This PKC-β-specific inhibitor was advanced for clinical

development due to its anti-angiogenic activity in human tumor xenografts , where it

leads to decreased VEGF expression and microvessel density (Keyes et al., 2004).

Figure 3.5.2 PKC-β and PKA inhibitors increase SubG1 fraction in Paclitaxel-resistant cell

lines in the presence of Paclitaxel

Measurement of SubG1 fraction upon treatment with PKC-β and PKA inhibitors at different

concentrations with and without Paclitaxel in SUM159PT (A) and MB231 (B) resistant cell

lines. The cells were treated for 4 days with Enzastaurin and H-89 alone or in combination with

Paclitaxel, and subsequently fixed with 70% ethanol for 1 day. They were then stained with

Propidium Iodide for 30 mins before they were analyzed by FACS. Both inhibitors were able to

increase SubG1 fraction in the resistant cell lines upon Paclitaxel treatment, whereas Paclitaxel

alone is not sufficient to induce cell death. (n=1).

Page 104: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

87

3.6. The PKC-β inhibitor Enzastaurin sensitizes chemo-resistant cells to

Paclitaxel in anchorage-dependant and –independent conditions

To look closer into the effect of PKC-β inhibition on chemo-resistance, we focused

on Enzastaurin for further experiments.

We used the SUM159PT parental and 1uM-TR, and MB231 parental and 75nM-TR

cell lines and treated them with different doses of Enzastaurin and measured cell

viability over a course of 4 or 5 days. The Paclitaxel concentration used for

SUM159PT and MB231 parental cell lines was 0.73nM and 0.46nM, respectively,

and the resistant cells were treated with the maintenance dose of Paclitaxel.

Expectedly, both the parental cells showed no effect, even in combination with

Paclitaxel, but for both resistant cell lines, a dose-dependent and time-dependent

effect is seen when Enzastaurin is given in combination with Paclitaxel (Figure

3.6.1). Enzastaurin at 2.5μM is sufficient to significantly decrease cell viability in the

resistant cell lines in the presence of Paclitaxel.

Page 105: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

88

Figure 3.6.1 The PKC-β inhibitor Enzastaurin sensitizes resistant cells to Paclitaxel in a

dose-dependent manner

Measurement of cell viability in SUM159PT (A) and MB231 (B) parental and resistant cell

lines after treatment with Enzastaurin with and without Paclitaxel. The Paclitaxel concentration

used for SUM159PT and MB23 parental cells is 0.73nM and 0.46nM, respectively, whereas for

the resistant cells, the maintenance concentration was used. SUM159PT and MB231 cells were

treated over a period of 4 or 5 days, respectively, with different concentrations of Enzastaurin

with and without Paclitaxel and cell viability was measured each day. Measurements were

normalized to Day 0 and log2 values were plotted. Paclitaxel treatment alone has no effect in

the resistant cell lines but Enzastaurin can sensitize cells to Paclitaxel. (Error bars show ±SD,

n=3)

A

B

Page 106: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

89

To examine colony growth and morphology of the cells upon treatment, we

performed a 3D colony growth assay on Matrigel and found that all the parental and

resistant cell lines grew slightly smaller colonies when treated with Paclitaxel alone

or Enzastaurin alone, despite only using the maintenance concentrations of Paclitaxel

(Figure 3.6.2). This can be explained by the fact that although the cells are resistant to

Paclitaxel, they are still undergoing selective pressure which selects for highly

resistant cells. The combination treatment, on the other hand, has a minimal effect on

the parental cell lines, but can dramatically reduce colony growth in the resistant cell

lines. Moreover, for both SUM159PT and MB231, the size of the colonies grown by

the resistant cells is smaller compared to the parental cells, which can be explained by

their slightly slower growth rate.

Figure 3.6.2 Enzastaurin inhibits 3D colony growth in resistant cell lines in the presence

of Paclitaxel

Measurement of 3D growth in SUM159PT (A) and MB231 (B) parental and resistant cell lines

after treatment with Enzastaurin and Paclitaxel. The cells were seeded on Matrigel, treated the

same day and imaged after 9 days of treatment.

A

B

Page 107: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

90

We then went on to calculate the degree of re-sensitization through combination

treatment by measuring the Paclitaxel EC50 as determined by cell viability in

SUM159PT parental and resistant cell lines. The cells were treated for 4 days with

increasing concentrations of Paclitaxel together with Enzastaurin at 2 different

concentrations (1μM and 2.5μM) and cell viability was measured 4 days later. As

expected, there was no change in the EC50 of Paclitaxel in the parental cell lines when

Enzastaurin was combined with Paclitaxel, whereas Enzastaurin treatment drastically

reduced EC50 in the resistant cell lines (Figure 3.6.3). Enzastaurin at both

concentrations was able to restore EC50 similar to that in the parental cell line,

although a slight dose-dependent effect can be seen in both resistant cell lines. At a

concentration of 2.5μM, Enzastaurin is potent enough to reduce the EC50 by

approximately 500-fold in both the resistant cell lines (4.7μM to 13.5nM in 500nM-

TR, and 4.9μM to 13.3nM in 1μM-TR), which is similar to the EC50 of Paclitaxel in

the parental cell line (3.8nM).

Page 108: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

91

Figure 3.6.3 Enzastaurin treatment drastically reduces Paclitaxel EC50 in SUM159PT

resistant cell lines

A. Paclitaxel EC50 measurements in SUM159PT parental, 500nM-TR and 1μM-TR cell

lines upon Enzastaurin treatment. The cells were treated for 24 hours with increasing

concentrations of Paclitaxel in combination with Enzastaurin at 1μM and 2.5μM, after

which cell viability was measured. The EC50 was calculated using Sigmoidal dose-

response and plotting the non-linear regression curve fit (Error bars show ±SD, n=6).

B. Bar graph depicting the EC50 values of Paclitaxel in SUM159PT 500nM-TR and

1μM-TR as determined by previous experiment (n=1)

A

B

Page 109: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

92

3.7. Enzastaurin reduces tumorsphere growth in Paclitaxel-resistant

TNBC cell lines in the presence of Paclitaxel

To test whether the combination treatment has any effect on the cancer stem cell side-

population, we performed a tumorsphere assay. To do so, we first seeded the

SUM159PT and MB231 resistant cells in a 2D culture and treated them with

Paclitaxel and Enzastaurin as single or combination treatment. Four days later, the

dead cells were washed away and the remaining surviving cells were seeded for a

tumorsphere assay and grown for 7 days. As seen in Figure 3.7, single treatment in

both the resistant lines had no significant effect on the number of tumorspheres, in

contrary, Paclitaxel treatment alone even enhanced the amount of tumorspheres in

SUM159PT 1μM-TR. This is due to the selection the cells undergo in the presence of

Paclitaxel. The combination treatment, however, successfully eliminated tumorsphere

growth as seen by a significant reduction in tumorsphere number compared to the

Paclitaxel treatment group. These results are especially interesting since the cancer

cells are believed to be the ones surviving chemotherapy and leading to metastasis

and disease recurrence. Finding a way to target these cells is a major challenge in

overcoming cancer. We show here that the combination treatment of Paclitaxel with

the PKC-β inhibitor Enzastaurin can successfully eliminate this side population.

Page 110: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

93

Figure 3.7 Enzastaurin in combination with Paclitaxel inhibits tumorsphere growth in

Paclitaxel-resistant cells

Measurement of tumorsphere growth in SUM159PT 1μM-TR and MB231 75nM-TR cell lines.

The cells were pre-treated with either single or combination treatment of Paclitaxel and

Enzastaurin for 4 days in 2D culture. The surviving cells were then seeded in ultra-low

attachment conditions for 7 days. They were then stained with INT o/n and the number of

tumorspheres was counted using a GelCount. (Error bars show ±SD, n=2).

Page 111: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

94

3.8. PKC-α inhibitors are not capable of re-sensitizing resistant cells to

Paclitaxel

It has been shown before that PKC-α plays an important role in breast cancer stem

cells and targeting PKC-α can specifically target breast CSCs (Tam et al., 2013). This

is due to a switch from EGFR to PDGFR signaling in CSCs which leads to the PKC-

α-dependent activation of FRA1. To test wether re-sensitization of Paclitaxel-resistant

cells is isoform-specific, we tested different PKC-α inhibitors in an attempt to achieve

the same re-sensitization effect as with the PKC-β inhibitors. We treated SUM159PT

and MB231 parental and resistant cells with 2 different PKC-α inhibitors and

measured cell viability over the course of 5 days. The two inhibitors we used, Gö-

6976 and Ro-32-0432, inhibit PKC-α with an approximately 3-fold lower IC50

compared to PKC-β (Gö-6976: 2.3nM vs 6.2nM, Ro-32-0432: 9nM vs 28nM). Both

inhibitors had no significant effect as single treatment in both parental and resistant

cells in SUM159PT and MB231 (Figure 3.8.1). In addition, the parental cells were

not effected by the combination treatment, whereas the resistant cells showed only a

minor effect. MB231 75nM-TR showed no significant reduction in cell viability

when PKC-α inhibitors were combined with Paclitaxel, but SUM159PT 1μM-TR

exhibited reduced cell viability with only one of the inhibitors (Ro-32-0432).

Page 112: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

95

Figure 3.8.1 PKC-α inhibitors are not able to sensitize Paclitaxel-resistant cells in the

same manner as Enzastaurin

Measurement of cell viability in SUM159PT (A) and MB231 (B) parental and resistant cell

lines after treatment with the PKC-α inhibitors, Gö-6976 and Ro-32-0432, and Paclitaxel. The

Paclitaxel concentrations used were the maintenance concentrations for the resistant cell lines

and corresponding concentrations for the parental cell lines. The cells were treated with 2

different PKC-α inhibitors at different concentrations and cell viability was measured each day.

The measurements were plotted relative to measurements on Day 0. (Error bars show ±SD,

n=6).

A

B

Page 113: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

96

We then tested these inhibitors together with another PKC-α inhibitor, Ro-31-8220,

in anchorage-independent conditions. SUM159PT parental and both resistant cell

lines were seeded for a Methylcellulose assay, treated for 13 days after which they

were stained in order to count the number of colonies. In both resistant cell lines,

500nM-TR and 1μM-TR, there was no significant difference between inhibitor single

treatment and in combination with Paclitaxel (Figure 3.8.2). Ro-31-8220 inhibited

colony formation only in the parental cell lines at a high concentration, as single

treatment and in combination with Paclitaxel. The parental cells grew slightly less

colonies when Paclitaxel was added.

Page 114: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

97

These results show that the effect of re-sensitization in Paclitxel-resistant TNBC cell

lines is isoform-specific. Inhibitors specifically targeting PKC-β can significantly

reduce cell viability in 2D and 3D assays, and induce apoptosis in the presence of

Paclitaxel, whereas PKC-α-specific inhibitors show no significant effects. Although

PKC signaling is very broad and many PKC isoforms share similar functions, there

are differences in the downstream targets of the various isoforms which can account

for their different functions in Paclitaxel-resistance and re-sensitization. PKC-α

promotes invasion of breast cancer cells and its expression correlates with tumor

grade and poor prognosis (Lonne et al., 2010), but it has not been linked with chemo-

resistance, which is consistent with our results.

Figure 3.8.2 PKC-α inhibitors have no effect on colony formation of SUM159PT

Paclitaxel-resistant cells in the presence of Paclitaxel

Measurement of colony formation in anchorage-independent conditions in a Methylcellulose

assay of SUM159PT parental and resistant cell lines. The Paclitaxel concentrations used are the

maintenance concentrations for the resistant cell lines and 0.73nM for the parental cell line. The

cells were seeded for the assay and simultaneously treated with the inhibitors, Ro-31-8220, Gö-

6976 and Ro-32-0432, at 1μM, 2.5μM and 5μM. The cells were stained with INT after 13 days

and the colonies were counted. (Error bars show ±SD, n=6).

Page 115: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

98

3.9. PKC-β expression and protein levels in breast cancer vs. normal

breast samples

To expand the results we derived from experiments on cell lines to patient samples,

we explored 2 different publicly available online databases. The Human Protein Atlas

portal combines knowledge about the human proteome derived mainly from

antibody-based methods combined with transcriptomics analysis. Higher protein

levels of PKC-β were detected in ductal carcinoma and lobular carcinoma tissue

samples compared to normal breast tissue (Figure 3.9 (A)). To quantify the intensity

of the staining we then calculated the H-score for two groups: normal breast tissue vs.

breast cancer. The H-score is calculated from the intensity of staining from each

sample (low, medium and high) and the amount of samples of each staining intensity

in the combined group (see MATERIALS AND METHODS 2.17). We found that the

H-score was higher for the breast cancer samples compared to the normal breast

tissue, strongly suggesting that PKC-β plays a role in carcinogenesis in breast cancer.

Activation of PKC-β depends on PDK1, PLCγ, as well as DAG and Ca2+ and takes

place at the cell membrane, after which it translocates to the cytoplasm where it can

phosphorylate downstream targets such as GSK3β. Recent studies have shown that

PKC isoforms in TNBC cancers can relocate to the nucleus upon activation by TGFβI

and IL-1β, however, it is not clear what the exact mechanism behind this

translocation and the function of PKC in the nucleus is (Paul et al., 2014).

To get a clearer picture about the importance of PRKCB (PKC-β gene) in the

development of breast cancer, we examined mRNA expression in Breast Ductal

Carcinoma In Situ (DCIS) and normal breast tissue using Oncomine, a database of

cancer transcriptome profiles. DCIS is a type of non-invasive breast cancer where

ductal cells have undergone transformation but have not spread beyond the milk ducts

and have not invaded into the surrounding breast tissue. Often, DCIS progresses to

Page 116: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

99

invasive breast cancer later on. As seen in Figure 3.9 (B), PRKCB mRNA levels were

significantly higher in Breast DCIS compared to normal breast in the Ma Breast 4

dataset. We also examined the expression levels of GSK3B, a direct downstream

target of PRKCB, which follows the same trend as PRKCB and also has significantly

higher mRNA levels in Breast DCIS. We then wanted to explore PRKCB expression

in the stroma of invasive ductal breast tumors, as stroma is an important player in

tumor formation, invasion and metastasis. The bulk of stroma surrounding tumors are

cancer-associated fibroblasts (CAFs) that are activated by cancer cells and have been

shown to induce EMT in breast cancer cells through paracrine TGF-β signaling (Yu

et al., 2014). In addition, one study found that PKC-β is up-regulated in the tumor

microenvironment in mouse mammary tumor virus-polyoma middle T-antigen

(MMTV-PyMY) induced mammary tumorigenesis (Wallace et al., 2014). When PKC-

β was diminished in the stromal compartment, the mice formed smaller tumors with

diminished collagen deposition. In concordance with these results, we found that

PRKCB was also increased in the stroma of invasive ductal breast carcinoma patient

samples compared to normal breast samples in the Karnoub dataset (Karnoub et al.,

2007).

These results suggest that PKC-β plays an important role in the development of breast

cancer, as well as in established ductal and lobular carcinoma.

Page 117: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

100

Figure 3.9 PKC-β is highly expressed in breast cancer compared to normal breast tissue

in patient samples and cell lines

A. The Human Protein Atlas. Patient samples (n=6) were stained with anti-PKC-β

antibodies in normal breast tissue, and ductal and lobular carcinoma of the breast

(n=28). Sample staining is shown for each group. The H-score was calculated

according to the staining intensity given by the database. To calculate the H-score the

staining intensity of each sample (1,2 or 3 for increasing intensity) was multiplied

with the percentage of samples in each intensity group and all 3 values were added

together. This calculation was performed for normal breast tissue and breast cancer

samples separately.

B. Oncomine. PRKCB and GSK3B expression are shown in Breast DCIS (GSK3B and

PRKCB: n=11) compared to normal breast tissue (GSK3B and PRKCB: n=14) (Ma

Breast dataset), and in the stroma of invasive ductal breast carcinoma (n=7) compared

to normal breast tissue (n=15) (Karnoub Breast dataset).

A B

Page 118: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

101

3.10. Mechanism of PKC-β signaling in resistance and upon sensitization

To understand how the inhibition of PKC-β can re-sensitize cells to Paclitaxel once

they have acquired resistance, we examined the modulation of signaling pathways

upon PKC-β inhibition. We first treated MB231 parental cells with increasing

concentrations of Enzastaurin for 24 and 48 hours and found no change in the total

protein levels of PKC-β, but phosphorylation of GSK3β on Serine 9 (GSK3βSer9) was

diminished (Figure 3.10 (A)). GSK3β is directly phosphorylated by PKC-β (Goode et

al., 1992), and is a reliable pharmacodynamic marker for Enzastaurin activity

(Miranda et al., 2015). Serine 9 phosphorylation leads to inactivation of its kinase

activity, therefore PKC-β inhibits GSK3β activity. Phosphorylation of PKCs is not a

reliable marker for its activation and activity as they don’t require phosphorylation of

the enzyme. Rather translocation of the protein to different cellular compartments

induced by upstream signaling is a reliable marker for monitoring its activity (Rosse

et al., 2010).

When we subjected SUM159PT and MB231 parental and resistant cell lines to single

treatment with 2.5μM Enzastaurin or combination treatment with Paclitaxel, we

found similar results. We examined phosphorylation of PKC-β at Thr641 which

mediates its binding to Hsp70 thereby regulating its stability and phosphorylation.

Thr641 is also needed for the catalytic function and autophosphorylation of PKC-β.

Protein levels of Thr641-phosphorylated PKC-β remained unchanged in all treatment

groups, but GSK3βSer9 was diminished upon Enzastaurin treatment in all cell lines

(Figure 3.10 (B)). Moreover, GSK3βSer9 basal levels were slightly lower in the

resistant cell lines compared to the parental cell lines in the DMSO treatment groups,

suggesting that GSK3β is over-activated and could be directly involved in chemo-

resistance as part of the PKC-β signaling cascade. We examined Mcl-1 protein levels

in order to deterimine its role in apoptosis of resistant cell lines upon combination

Page 119: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

102

treatment. It has been published earlier that the pro-survival protein Mcl-1 plays a

crucial role in regulating apoptosis upon treatment with spindle poisons (Wertz et al.,

2011). Mcl-1 is directly phosphorylated by GSK3β on Serine 159 (Maurer et al.,

2006) and when phosphorylated, it can interact with the tumor-suppressor FBW7, the

substrate-binding component of a ubiquitin-ligase complex, which leads to its

ubiquitinylation and degradation. As seen below, Mcl-1 is degraded only in the

resistant cell lines when treated with Paclitaxel and Enzastaurin. Interestingly,

treatment with Enzastaurin alone has no effect on Mcl-1 levels in either the parental

or resistant cell lines.

These results show that Enzastaurin acts by inhibiting the inhibitory Serine 9

phosphorylation on GSK3β which activates GSK3β, and in turn leads to Mcl-1

degradation only when Paclitaxel is present in the treatment group.

Page 120: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

103

Figure 3.10 Western blot analysis of down-stream signaling of Enzastaurin treatment on

PKC-β, GSK3β and Mcl-1 in SUM159PT and MB231

Cells were treated for 24 hours and then harvested for western blot analysis.

A. MB231 parental cells were subjected to treatment with increasing concentrations of

Enzastaurin. At high concentrations, total PKC-β levels were slightly reduced, but

GSK3βSer9, a pharmacodynamic marker for Enzastaurin activity, was completely

diminished in all treatment groups.

B. SUM159PT and MB231 parental and both resistant cells were subjected to 24 hour

treatment with Enzastaurin and Paclitaxel as single treatment or in combination.

A

B

Page 121: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

104

3.11. GSK3β inhibitors recapitulate the effect of re-sensitization by

Enzastaurin

We used a GSK3β inhibitor and an activator to investigate whether the inhibition of

GSK3βSer9 downstream of PKC-β can also re-sensitize resistant cells to Paclitaxel, or

if it is merely a side-effect of Enzastaurin treatment. SB216763 has the same effect on

GSK3β Serine 9 phosphorylation as Enzastaurin. It acts as a GSK3β “activator” and

diminishes GSK3βSer9 phosphorylation. Lithium Chloride (LiCl), on the other hand,

has the opposite effect and inhibits GSK3β by increasing Serine 9 phosphorylation

levels. The mechanism by which LiCl inhibits GSK3β is unclear but there are 2

proposed mechanisms: 1) it is a competitive inhibitor with regards to Mg2+; 2) it

inhibits potassium deprivation, which would otherwise lead to dephosphorylation of

GSK3β (Kramer et al., 2012). We treated MB231 parental and 75nM-TR resistant

cells with these inhbititors alone or together with Paclitaxel for 24 hours to examine

GSK3βSer9 levels. As seen in the western blot below, SB216763 successfully inhibits

GSK3β phosphorylation in both cell lines, whereas LiCl leads to increased levels of

phosphorylated GSK3β (Figure 3.11.1). These results are consistent with previously

published data (Tan et al., 2005). Other activating phosphorylation sites of GSK3β

show no change after treatment with the inhibitors alone or in combination with

Paclitaxel. Mcl-1 levels remain unchanged in all the treatment groups in the parental

cell lines, whereas they are slightly reduced in the combination treatment groups in

the 75nM-TR cell lines, more so in the SB216763 + Paclitaxel treatment group.

Page 122: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

105

We then performed an anchorage-independent Methylcellulose assay to examine the

effect of the GSK3β inhibitors in 3D growth conditions in SUM159PT parental and

1μM-TR, and MB231 parental and 75nM-TR cell lines. The cells were treated for 13

days with 4 different inhibitors: 3 GSK3β “activators” (BIO, A1070722 and

SB216763) at 2.5μM and 5μM each, and one GSK3β inhibitor (LiCl) at 1mM and

2mM. The colonies were then stained with INT and counted.

In SUM159PT and MB231 parental cell lines, none of the inhibitors showed a

decrease in colony formation in the combination treatment groups compared to single

treatment with the inhibitors alone (Figure 3.11.2). However, SUM159PT 1μM-TR

cell line showed significant dose-dependent reduction in colony formation when

treated with the 3 GSK3β “activators” in the presence of Paclitaxel, whereas LiCl, the

true GSK3β inhibitor, had no effect on colony formation, even when Paclitaxel was

added. The results for MB231 75nM-TR cell line are not so conclusive because the

Figure 3.11.1 Western blot analysis of the effect of GSK3β inhibitors in MB231 parental

and resistant cells

MB231 parental and 75nM-TR cell lines were treated with SB216763 (5μM) and LiCl (2mM)

alone or in combination with Paclitaxel for 24 hours and then harvested for western blot

analysis. Paclitaxel was used at 0.46nM for the parental and 75nM for the resistant cell line.

SB216763 diminished GSK3βSer9, similar to Enzastaurin, whereas LiCl slightly increased

protein levels.

Page 123: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

106

Paclitaxel only treated group showed reduced colony formation compared to the

DMSO treatment group, which is unexpected. This could be because in general, the

MB231 cells are not able to grow and form distinct colonies when seeded together

with Methylcellulose on the Bacto-Agar. The colonies formed by these cells are

“loose” and the cells seem to disperse, so the software used to count the colonies may

not give reliable results.

These results shows that GSK3β “activators”, which have the same effect on

GSK3βSer9 as Enzastaurin, are also able to sensitize resistant cells to Paclitaxel.

To determine wether the treatment with GSK3β compounds can induce apoptosis in

the presence of Paclitaxel, we treated SUM159PT 1μM-TR and MB231 75nM-TR

Figure 3.11.2 A set of GSK3β inhibitors reduce colony formation in the resistant cells in the

presence of Paclitaxel

Anchorage-independent Methylcellulose assay was used to determine colony formation in

SUM159PT parental and 1μM-TR, and MB231 parental and 75nM-TR cell lines after treatment

with various GSK3β inhibitors. The cells were treated for 13 days with different inhibitors alone

or in combination with Paclitaxel and, subsequently, stained with INT o/n before the colonies

were counted. (Error bars show ±SD, n=6).

Page 124: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

107

cell lines with either SB216763 or LiCl alone or in combination with Paclitaxel for 4

days and performed cell cycle analysis using PI staining. Since we know that

Enzastaurin in combination with Paclitaxel can induce cell death in both cell lines, as

determined by an increase in the SubG1 population, we used this as our positive

control. The combination of SB216763 and Paclitaxel has a very similatr effect in the

SUM159PT resistant cell lines where the amount of cell death is almost identical to

the positive control (Figure 3.11.3 (A)). Surprisingly, the MB231 resistant cell line

showed a much more modest increase in the SubG1 population compared to

Paclitaxel treatment alone. One reason could be that SB216763 was not able to

completely deplete GSK3βSer9 protein levels in this cell line, as determined by

western blot analysis (see Figure 3.11.1). As expected, LiCl does not increase cell

death in combination with Paclitaxel in either of the cell lines when compared to the

Paclitaxel treatment group alone.

Although the amount of SubG1 in the GSK3β “activator” + Paclitaxel treatment

group in MB23 75nM-TR is much lower than the Enzastaurin + Paclitaxel treatmen

group, the number of floating cells in the wells, which represent cells that are most

likely undergoing apoptosis, are similar (Figure 3.11.3 (B)).

Page 125: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

108

This data suggests that GSK3β plays an important role in chemo-resistance and

activation of GSK3β can re-sensitize cells to Paclitaxel. This could be the reason for

the effectiveness of Enzastaurin in re-sensitization, as GSK3β is directly downstream

of PKC-β and is phosphorylated by PKC-β.

Figure 3.11.3 GSK3β activation leads to increased cell death in the resistant cells in the

presence of Paclitaxel

A. Measurement of SubG1 fraction in the SUM159PT and MB231 resistant cell lines

after 4 days of treatment with SB216763 (5μM) and LiCl (2mM) as single treatment

or in combination with Paclitaxel. The Paclitaxel concentration used is 1μM and

75nM for the SUM159PT and MB231 resistant cell lines, respectively. (Error bars

show ±SD, n=3).

B. Microscope images of different treatment groups in SUM159PT and MB231 cell lines

before harvesting for cell cycle analysis analysis.

A B

Page 126: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

109

3.12. Paclitaxel-resistant cell lines exhibit cross-resistance to other

chemotherapeutic compounds

Vincristine (VCR) is a spindle poison that belongs to the same class of

chemotherapeutic drugs as Paclitaxel, although its effect on microtubule stability is

opposite. While Paclitaxel binds to tubulin and promotes microtubule stabilization,

VCR binds to tubulin dimers, thereby inhibiting microtubule assembly. Both drugs

lead to cell cycle arrest in the mitotic phase, but cells treated with VCR arrest in the

prometaphase. Due to the missing spindle tubules in these cells, the kinetochores

remain unattached and the SAC remains inactive, leading to mitotic arrest (Rudner

and Murray, 1996).

When tested whether Paclitaxel-resistant cells showed cross-resistance to VCR, we

found that, indeed, the resistant cells lines are also more resistant to VCR, compared

to the parental cell lines, as measured by their EC50 (Figure 3.12.1 (A)). SUM159PT

Paclitaxel-resistant cell lines exhibit a 1000-fold increase in the EC50 compared to the

parental cell lines (3.62μM vs. 3.17nM), whereas the shift in EC50 of VCR is less

drastic in the MB231 resistant cells compared to parental (23.21nM vs. 3.01nM). In

both resistant cell lines, however, Enzastaurin is able to restore VCR-sensitivity and

reduce the EC50 similar to those of the parental cells.

To examine whether Enzastaurin can induce apoptosis in the resistant cell lines upon

VCR treatment, we treated cells with increasing concentrations of VCR and measured

the amount of SubG1 population as a measure of apoptosis (Figure 3.12.1 (B)).

Although VCR alone could induce small amounts of apoptosis, there is a significant

increase in apoptotic cells in both resistant cell lines when Enzastaurin is added to

VCR treatment. Therefore, Enzastaurin is able to restore VCR-sensitivity in the

resistant cell lines by inducing apoptosis.

Page 127: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

110

We also included another chemotherapeutic drug, Adriamycin, which is not a spindle

poison and targets cells through a different mechanism, in order to determine whether

re-sensitization with Enzastaurin is dependent on mitotic arrest. It is routinely used

for the treatment of a number of different cancers such as breast, gastric and ovarian

cancers, sarcoma and mutiple myeloma (Cortes-Funes and Coronado, 2007, Weiss,

Figure 3.12.1 Paclitaxel-resistant cells exhibit cross-resistance to Vicristine which can be

reversed with Enzastaurin

A. Paclitaxel EC50 measured by cell viability assay in SUM159PT parental and 1μM-TR,

and MB231 parental and 75nM-TR cell lines. The cells were treated for 4 days before

cell viability was measured. (Error bars show ±SD, n=6)

B. Percentage of SubG1 fractions after 4 days of treatment in SUM159PT 1μM-TR and

MB231 75nM-TR cell lines with different concentrations of Vincristine (0.1μM, 1μM

and 5μM for SUM159PT 1μM-TR, and 1nM, 10nM and 100nM for MB231 75nM-TR)

with and without Enzastaurin (2.5μM). (Error bars show ±SD, n=3, **: p-value <0.01,

***: p-value<0.001, ****: p-value <0.0001).

A

B

Page 128: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

111

1992). Adriamycin (ADR) is an anthracycline, it intercalates into the DNA and

inhibits topoisomerase II, an enzyme that detangles and relaxes DNA supercoils

(Tewey et al., 1984). Another mechanism of action has been proposed which includes

the generation of free radicals that cause damage to the cellular membranes and DNA

(Gewirtz, 1999). By inhibiting topoisomerase II, the DNA chain which has been

broken for replication, will not be released and the cells are unable to undergo

mitosis.

Paclitaxel-resistant SUM159PT and MB231 cells also show higher resistance to ADR

compared to the parental cell lines, as determined by an approximate 1000-fold

increase in the EC50 to ADR (Figure 3.12.2 (A)). However, Enzastaurin is only partly

able to restore ADR-sensitivity in SUM159PT, but not in the MB231 resistant cell

line. When we determined the SubG1 fraction in single and combination treatment

groups, we found that althoug ADR can reduce ADR EC50 in SUM159PT resistant

cell line, it is unable to induce significantly higher levels of apoptosis in combination

with Paclitaxel (Figure 3.12.2 (B)). In MB231 75nM-TR, the increase in apoptotic

cells is also not sighnificant in the combination vs. single treatment group. This is

likely due to the difference in their chemo-toxic effects. This data suggests that

Enzastaurin is only able to significantl restore sensitivity for drugs that induce mitotic

arrest as part of their cytotoxic mechanism.

Page 129: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

112

Figure 3.12.2 Paclitaxel-resistant cells exhibit cross-resistance to Adriamycin

A. Paclitaxel EC50 measured by cell viability assay in SUM159PT parental and 1μM-TR,

and MB231 parental and 75nM-TR cell lines. The cells were treated for 4 days before

cell viability was measured. (Error bars show ±SD, n=6).

B. Percentage of SubG1 fractions after 4 days of treatment in SUM159PT 1μM-TR and

MB231 75nM-TR cell lines with different concentrations of Adriamycin (0.1μM,

1μM and 5μM) with and without Enzastaurin (2.5μM). (Error bars show ±SD, n=3, *:

p-value<0.05).

A

B

Page 130: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

113

3.13. Enzastaurin can restore mitotic arrest in resistant cells upon

treatment with Paclitaxel

In order for Paclitaxel to be effective, the cells must undergo cell division and reach

mitosis. Paclitaxel then leads to stabilization of mitotic microtubules and,

subsequently, mitotic arrest. It has been found that Paclitaxel-induced cell death is

dependent on the activation of the spindle assembly checkpoint (SAC), and the

suppression of Mad2 and BubR1, two important checkpoint genes, results in higher

resistance to Paclitaxel (Sudo et al., 2004).

3.13.1. Paclitaxel-induced mitotic arrest and cell death in sensitive cell lines

To validate these results in our cell lines, we treated MB231 parental cell lines with

Paclitaxel and used an anti-phospho-Histone H3 (p-H3) antibody to detect cells in the

mitotic phase. We found that upon treatment with Paclitaxel for 24 hours, 16.87% of

the cells were arrested in the mitotic phase compared to only 1.5% in untreated

conditions, and at 48 and 72 hours, this population decreased dramatically,

accompanied by an increase in the number of multinucleated cells (Figure 3.13.1

(A)). This can be explained by the fact that cells arrested in the mitotic phase can

undergo different fates. One consequence of mitotic arrest is mitotic cell death, but

cells that can overcome mitotic arrest can slip into the next interphase (mitotic

slippage), or undergo one round of aberrant mitosis. As correct mitosis is not

possible, the cells become tetraploid and will eventually undergo senescence or cell

death (Vitale et al., 2011). Additionally, we observed an increase in the number of

apoptotic cells from 24 to 72 hours determined by the SubG1 population, indicating

that most of the tetraploid cells and those arrested in the mitotic phase, do in fact

undergo cell death later on (Figure 3.13.1 (B)).

Page 131: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

114

To look closer into the mechanism of cell death upon treatment with Paclitaxel, we

treated SUM159PT and MB231 parental cells with increasing concentrations of

Figure 3.13.1 Paclitaxel induces mitotic arrest and subsequent cell death in MB231 parental

cells

A. MB231 parental cells were treated for 24, 48 and 72 hours with 50nM Paclitaxel before

they were harvested and stained with Propidium Iodide and anti-pH3 antibody. FACS

analysis was performed measuring DNA content and intensity of p-H3 staining.

B. MB231 parental cells were treated for 24, 48 and 72 hours with 50nM Paclitaxel, then

harvested, fixed and stained with Propidium Iodide. FACS analysis was performed to

measure SubG1 population in the different treatment groups. (n=1).

C. Bar graph depicting percentages of SubG1 after Paclitaxel treatment as determined by

the previous experiment.

B

A

DMSO

Paclitaxel

50nM

DMSO

Paclitaxel

50nM

C

Page 132: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

115

Paclitaxel and performed western blot to detect changes in signaling pathways. The

cells were harvested at 24 hours when most of the affected cells are still arrested in

the mitotic phase. We found that Aurora B levels are increased, especially in

SUM159PT cells, which is due to the activation of the SAC in the mitotic phase

(Figure 3.13.2 (A)). It is believed that unattached kinetochores lead to an

accumulation of Aurora B at the centromeres of chromosomes in the metaphase plate

and Aurora B is stabilized when there is a lack of tension from the microtubules

between the sister chromatids (Musacchio and Salmon, 2007). This is consistent with

the fact that Paclitaxel arrests cells in the mitotic phase inhibiting tension on the

chromatids. Furthermore, levels of phosphorylated C-Raf are higher in the Paclitaxel

treated samples, and it has been shown that C-Raf phosphorylation is a requirement

for Paclitaxel-induced apoptosis (Blagosklonny et al., 1996). C-Raf, when activated,

can post-translationally phsophorylate Mcl-1, which leads to its ubiquitination and

subsequent proteasome-mediated degradation. Mcl-1 downregulation correlates with

increased levels of Poly(ADP-ribose) polymerase (PARP) cleavage, one of the main

cleavage targets of Caspase 3 and a marker for cells undergoing apoptosis.

We then went on to determine whether the inhibition of Aurora B could abolish

Paclitaxel-induced cell death. To do so, we treated SUM159PT and MB231 parental

cells with the Aurora B inhibitor VX-680, a potent and highly selective Aurora

Kinase inhibitor. As expected, we found that treatment with VX-680 could abolish

cell death in Paclitaxel-treated cells up to appromximately 70% in the MB231

parental cells (Figure 3.13.2 (B)). The effect in SUM159PT cells is milder but

remains notable with approximately 40% reduction. As seen in the microscope

images, Paclitaxel treated cells evidently undergo cell death with an increase in the

number of floating cells, whereas the addition of VX-680 can rescue apoptosis

(Figure 3.13.2 (C)).

Page 133: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

116

These results show that Aurora Kinases B is upregulated upon Paclitaxel treatment of

parental Paclitaxel-sensitive cells, and that inhibition of Aurora Kinases partly

inhibits Paclitaxel-induced cell death.

Figure 3.13.2 Up-regulation of Aurora Kinase B is crucial for Paclitaxel-induced cell

death

A. SUM159PT and MB231 parental cells were treated with increasing concentrations of

Paclitaxel (Taxol) for 24 hours and downstream signaling was determined using

western blot analysis.

B. SUM159PT and MB231 parental cells were treated with 50nM Paclitaxel alone or in

combination with the Aurora Kinase inhibitor (VX-680) at 1μM for 4 days and then

harvested for FACS analysis. The SubG1 fraction was determined to detect the

amount of cell death. (n=1).

C. Microscope images of cells before they were harvested for FACS analysis.

A

B C

Page 134: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

117

3.13.2. Paclitaxel-resistant cells over come mitotic arrest by down-regulating

Aurora Kinases

We then investigated the effect of Paclitaxel and Enzastaurin treatment on signaling

in the resistant cells, especially in terms of Aurora Kinase activation. As seen in

Figure 3.13.3, we found that C-Raf is increasingly phosphorylated upon combination

treatment, more so in SUM159PT resistant cells. As expected, this treatment group

also shows increased levels of cleaved PARP, indicative of apoptosis. Aurora B

levels are lower in both resistant cell lines compared to their parental counterpart.

However, combination treatment of Enzastaurin and Paclitaxel is able to restore

Aurora B levels, whereas single treatment alone has no effect. These findings are

similar to Aurora A, however the increase in protein levels upon combination

treatment are not as striking in MB231 resistant cell lines. Moreover, Aurora A levels

in the MB231 resistant cell line are similar to the parental cell line. These results

suggest that Enzastaurin treatment can restore SAC activation and mitotic arrest in

the resistant cell lines in the presence of Paclitaxel, by restoring accumulation and

stabilization of Aurora Kinases, especially Aurora Kinase B.

Page 135: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

118

Figure 3.13.3 Enzastaurin restores levels of Aurora Kinase B in resistant cell lines upon

treatment with Paclitaxel

SUM159PT parental and 1μM-TR, and MB231 parental and 75nM-TR cell lines were treated

with Paclitaxel or Enzastaurin (2.5μM) alone, and in combination. Paclitaxel was used at

0.73nM and 1μM for SUM159PT parental and resistant cell lines, and 0.46nM and 75nM for

MB231 parental and resistant cell lines. Cells were harvested after 24 hours and levels of

protein were detected using western blot analysis.

A. Protein levels detected by western blotting. The blots were incubated with antibodies

targeting Serine 388-phosphorylated C-Raf, cleaved PARP, Aurora kinase A and B.

B. Quantification of Aurora kinase B bands detected by western blotting. Image Lab 4.1

software was used to quantify bands relative to the DMSO control of the parental cell

line for both SUM159PT and MB231.

A

B

Page 136: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

119

We then dived further into elucidating the function of Aurora Kinases in re-

sensitizing resistant cells to Paclitaxel. To do so, we screened various Aurora Kinase

inhibitors in the resistant cell lines in order to determine whether these can rescue

cells when treated with Paclitaxel and Enzastaurin. If the addition of an Aurora

Kinase inhibitor to the combination treatment can inhibit cell death in resistant cell

lines, the we can conclude that Aurora Kinases are required for the re-sensitization

effect. This, in turn, suggests that cell death upon combination treatment is a result of

mitotic arrest induced by Paclitaxel due to Enzastaurin.

We found that out of all the Aurora Kinase inhibitors we used (VX-680, TAK901,

AZD1152-HQPA, MLN8237, and ZM447439), VX-680 could rescue combination

treatment effect in all the 4 resistant cell lines while having minimal effect on cell

viability when used as single treatment (Figure 3.13.4 (A)). In general, all the

inhibitors showed a more significant rescue effect in the MB231 resistant cell lines,

whereas VX-680 had the strongest effect in SUM159PT resistant cells. VX-680

(Tozasertib) is a potent and selective small-molecule inibitor of the Aurora Kinases

with apparent inhibition constants (Ki(app)) of 0.6nM, 18nM and 4.6nm for Aurora

Kinases A, B and C, respectively.

Page 137: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

120

Figure 3.13.4 Various Aurora Kinase inhibitors can rescue cell death upon combination

treatment of Paclitaxel with Enzastaurin in Paclitaxel-resistant cell lines

A. SUM159PT and MB231 resistant cell lines were treated with various Aurora Kinase

inhibitors alone or in combination with Paclitaxel and Enzastaurin. Aurora Kinase

inhibitors were all used at 1μM and 5μM. Cell viability was measured at Day 0 and

Day 4. Dat 4 measurements were normalized to Day 0 measurements and plotted

(Error bars show ±SD, n=6).

B. Table listing Aurora Kinase inhibitors with the respective Ki (inhibitory constant)

concentrations for the different Aurora Kinases (in nM).

B

A

Page 138: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

121

To show that, indeed, the Aurora Kinase Inhibitor VX-680 can inhibit cell cycle

arrest in the resistant cell lines upon combination treatment, we performed p-H3

staining of the different treatment groups at 24, 48 and 72 hours in MB231 75nM-TR.

We found that the resistant cell line does not undergo mitotic arrest in the presence of

Paclitaxel, as do the parental chemo-sensitive cell lines, as shown earlier. However,

when Enzastaurin is added to the Paclitaxel treatment, mitotic arrest is restored in the

resistant cell lines, as seen by an increase in the number of p-H3+ cells from 2.65%

(Paclitaxel treatment only) to 34.51% (combination treatment) (Figure 3.13.5).

Enzastaurin treatment alone has no effect on the mitotic phase. At later time points,

48 and 72 hours, the cells that have arrested in the mitotic phase become

multinucleated, as suggested by a 4N DNA content. They remain pH3-positive,

therefore they are in the mitotic phase, due to the effect of Paclitaxel, which stabilizes

microtubules. We showed earlier that VX-680 can rescue decreased cell viability

upon combination treatment, and here we show that this is due to the inhibition of cell

cycle and mitotic arrest. Addition of VX-680 to the combination group depletes

mitotic arrest upon combination treatment. When the resistant cells fail to activate the

SAC, then they cannot undergo mitotic arrest and, subsequent cell death.

Page 139: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

122

Figure 3.13.5 Aurora Kinase B inhibition rescues cell death upon re-sensitization of

Paclitaxel-resistant cells by Enzastaurin

Cells were treated for 24, 48 or 72 hours with 75nM Paclitaxel, 2.5μM Enzastaurin, and 1μM

VX-680 in different combination groups. They were then stained with anti-phospho-H3

antibody and FACS analysis was performed.

Page 140: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

123

3.14. Aurora B levels negatively correlate with Paclitaxel sensitivity in a

panel of basal-like breast cancer cell lines

As shown earlier, Aurora B and partially Aurora A levels are downregulated in the

resistant compared to the parental cell lines, therefore we went on to explore whether

there is a correlation between Aurora Kinase expression and Paclitaxel sensitivity in a

panel of basal and luminal breast cancer cell lines and the normal breast epithelial cell

line MCF10A. Aurora Kinase B (AURKB) and A (AURKA) expression levels were

obtained from the Neve et al. dataset on Gene Expression-Based Outcome for Breast

Cancer Online (GOBO) (Neve et al., 2006), where they performed transcription

profiling of a panel of 51 breast cancer cell lines. We measured Paclitaxel EC50 in all

cell lines using cell viability assay. We then plotted both sets of data on a scatter plot

with AURKB/A expression on the y-axis and Paclitaxel EC50 on the x-axis. As seen

in Figure 3.14, there is no significant correlation between AURKB expression and

EC50 when all the cell lines are combined. However, when we selected only for the

basal breast cancer cell lines, the correlation is highly significant, with an R2

coefficient of determination of 0.8411 (p-value=0.0013). This suggests that Aurora B

plays an important role in Paclitaxel-sensitivity, which can be attributed to its

function in mitotic spindle attachment and mitotic arrest.

We went on to determine whether Aurora A levels also determine sensitivity to

Paclitaxel, as Aurora A plays an important role in early mitosis (prophase) in

regulating correct function of the centrosomes. Interestingly, we found no correltation

between AURKA expression and Paclitaxel EC50 in all of the breast cancer cell lines,

suggesting that Paclitaxel sensitivity is independent of Aurora Kinase A.

Page 141: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

124

Figure 3.14 Aurora Kinase B expression negatively correlates with Paclitaxel sensitivity in

a panel of basal breast cancer cell lines

AURKB/A expression values were obtained from Neve et al. dataset (Neve et al., 2006) from

Gene Expression-Based Outcome for Breast Cancer Online (GOBO). Transcription profiling

was performed using A-AFFY-33-Affymetrix GeneChip Human Genome HG-U133A [HG-

U133A]. Expression values were obtained by performing Robust Micro-Array Average (RMA)

normalization and the values were Log2 transformed. AffyID for AURKB: 209464_at, AffyIDs

for AURKA: 208079_s_at and 204092_s_at. Paclitaxel EC50 was measured using cell viability

assay. Cells were treated for 4 days with increasing concentrations of Paclitaxel (n=6).

Page 142: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

125

3.15. Targeted siRNA knock-down of Aurora B inhibits re-sensitization of

Paclitaxel-induced mitotic arrest

To further examine whether the rescue effect seen with VX-680 is specific to the

Aurora B isoform, we performed siRNA-based knock-down of AURKB. We

knocked-down Aurora B using 2 different siRNA sequences (siAurB (Pre) and

siAurB) in SUM159PT parental and 1μM-TR, and MB231 parental and 75nM-TR

cell lines. 24 hours after knock-down we seeded the cells to measure cell viability

over a course of 5 days with different treatment groups. As seen in the Figure 3.28

(A), both siRNA sequences are able to significantly rescue cell viability in the

MB231 parental cell lines upon Paclitaxel treatment at 5nM and 50nM. However, the

effect is milder in SUM159PT parental cells lines, which is not surprising as the basal

levels of Aurora B mRNA expression are lower in SUM159PT compared to MB231

cell lines (Figure 3.15.1 (C)). This trend can further be explained by Paclitaxel

sensitivity of both cell lines, as SUM159PT is intrinsically more resistant to

Paclitaxel compared to MB231.

In case of the resistant cell lines, siRNA knock-down of Aurora B significantly

rescued cell viability upon combination treatment in SUM159PT 1μM-TR (only

siAurB (Pre)) and MB231 75nM-TR (Figure 3.15.1 (B)). Again, the rescue effect is

stronger in MB231 resistant cell lines compared to SUM159PT resistant cell lines due

to the higher levels of AURKB mRNA (Figure 3.15.1 (C)) . When the mRNA levels

are already very low, as in SUM159PT cells, it is no surprise that the knock-down

effect is weak. In general, resistant cell lines have lower levels of AURKB mRNA

and this is in agreement with the database results showing a negative correltation

between Paclitaxel EC50 and AURKB expression levels (Figure 3.14).

Page 143: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

126

A

B

C

Page 144: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

127

To examine whether this rescue effect is due to the inability of mitotic arrest and SAC

activation as a result of Aurora Kinase B knock-down, we performed cell cycle

analysis. We treated MB231 parental and 75nM-TR cells with combination treatment

for 24 hours after knock-down and performed pH3-staining. Our results replicate our

findings using the Aurora Kinase inhibitor VX-680. SiRNA-based knock-down of

Aurora Kinase B abolished mitotic arrest after Paclitaxel treatment in the parental cell

lines, and after combination treatment in the resistant cell lines (Figure 3.15.2).

Figure 3.15.1 Aurora Kinase B is crucial for Paclitaxel-induced inhibition of cell viability

in parental and resistant SUM159PT and MB231 cell lines

A. siRNA knock-down of Aurora B in SUM159PT and MB231 parental cell lines. Two

siRNA sequences were used to knock-down Aurora B in the cell lines (siAurB (Pre)

and siAurB) at a concentration of 40nM. 24 hours after siRNA treatment, cells were

re-seeded treated with 5nM or 50nM Paclitaxel and cell viability was measured over

a course of 5 days and normalized to the DMSO control. (Error bars show ±SD, n=6,

****:p-values<0.0001).

B. siRNA knock-down of Aurora B in SUM159PT 1μM-TR and MB231 75nM-TR.

Two siRNA sequences were used to knock-down Aurora B at a concentration of

40nM. After 24 hours, the cells were re-seeded and treated with DMSO, or 2.5μM

Enzastaurin + Paclitaxel 1μM or 75nM in SUM159PT 1μM-TR and MB231 75nM-

TR, respectively. Cell viability was measured over a course of 5 days and

measurements were normalized to the DMSO control. (Error bars show ± SD, n=6,

****:p-values<0.0001).

C. Validation of Aurora B knock-down. 48 hours after siRNA treatment, cells were

harvested and Aurora B (AURKB) mRNA levels were determined using Realtime-

PCR.mRNA expression values were normalized to Actin (ACTB) (Error bars show

± SD, n=3).

Page 145: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

128

These results suggest that Enzastaurin can restore Paclitaxel-induced mitotic arrest in

the resistant cell lines dependent on Aurora Kinase B.

Figure 3.15.2 Aurora Kinase B abolished Paclitaxel-induced mitotic arrest

p-H3 staining of MB231 parental and 75nM-TR cell lines after Aurora B siRNA knockdown.

Cells were treated with two different siRNA sequences targeting Aurora Kinase B (siAurB

(Pre) and siAurB) at a concentration of 40nM. 24 hours later, the cells were re-seeded and

treated with Paclitaxel alone (parental) or Paclitaxel + Enzastaurin (resistant) and p-H3 staining

was performed after 24 hours of treatment.

Page 146: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

129

CHAPTER 4: DISCUSSION

Page 147: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

130

4.1. Enzastaurin: mechanism of Paclitaxel-sensitization

The exact mechanism by which Enzastaurin inhibits cell viability is still unclear.

Enzastaurin was originally synthesized as a Protein Kinase C (PKC) β-specific

inhibitor based on the structure of Staurosporine, a natural compound which is a

potent inhibitor of PKC but has limited selectivity for ATP-dependent kinases and

individual PKC isozymes in vitro (Faul et al., 2003). Moreover, PKCs regulate ATP-

binding cassette sub-family B member 1 (ABCB1). Interestingly, it was found that

Enzastaurin could sensitize ABCB1-expressing Vincristine-resistant neuroblastoma

and rhabdomyosarcoma cells, while having minimal effect on the cell viability of

non-resistant cells (Michaelis et al., 2015). It was suggested that Enzastaurin directly

interacts with ABCB1 and inhibits ABCB1 ATPase activity, rather than through

PKC, as determined by docking studies. Moreover, siRNA-targeted knock-down of

PKC-β, but not PKC-α, could inhibit the efflux pumping mechanism of ABCB1.

Other studies show that PKC-α and -β can directly phosphorylate and regulate

ABCB1, although this seems to be cell type specific. It was found that in ovarian

cancer cells PKC-α and PKC-β knock-down could reverse ABCB1-mediated multi

drug resistance and Taxol-resistant cells (Masanek et al., 2002).

In the SUM159PT Paclitaxel-resistant cells lines we have generated, we found a

notable increase in the ABCB1 expression when compared to the parental cells as

determined by microarray analysis (Figure 4.1). Although many of the ABC

transporters show increased expression in the resistant cell lines, ABCB1 expression

is the most distinguished. Therefore, the effect of re-sensitization to Paclitaxel

through Enzastaurin treatment could be due to its direct or indirect inhibition of

ABCB1 activity.

Page 148: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

131

ABCB1, also known a P-glycoprotein (P-gp) or multidrug resistance protein 1

(MDR1), is phosphorylated by PKCs and PKA at the serine residues in its linker

region (Chambers et al., 1994). Some have even suggested that these

phosphorylations are required for the translocation and activation of ABCB1

(Germann et al., 1996). There are 3 PKC-mediated phosphorylation residues at

Ser661, Ser667 and Ser671, and 3 PKA-mediated phosphorylation residues at Ser667,

Ser671 and Ser683, and all of them are in the intracellular linker region anchoring the

ABCB1 protein in the cellular membrane. Interestingly, the 2 classes of inhibitors

which showed the greatest effect in the kinase inhibitor screen on our Paclitaxel-

resistant cell lines were PKC and PKA inhibitors. We decided to focus on PKC-β for

this project as these inhibitors induced slightly higher levels of apoptosis in the cell

lines in combination with Paclitaxel, and PKC-β inhibitors are already being used in

clinical trials. However, the kinase screen revealed an important role for PKA in

Figure 4.1 Increased expression of ABC transporters in Paclitaxel-resistant SUM159PT

cell lines

Microarray analysis was performed to determine gene expression changes in Paclitaxel-

resistant SUM159PT 500nM-TR and 1μM-TR cell lines compared to the parental cell line.

Quantile normalization was performed and after fold change cut-off =2 and FDR<0.05, the list

of significantly over-expressed genes was obtained. The graph shows the log2 gene expression

of various ABC transporters in the 2 resistant cell lines when normalized to the parental cell

line.

Page 149: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

132

Paclitaxel-resistance, similar to PKC-β, which could be due to its role on ABCB1

phosphorylation and activation.

Moreover, the Enzastaurin concentration of 2.5μM, which we have used for all the

experiments in this study, did not lead to a reduction in the phosphorylation levels of

PKC-β (Figure 3.10).

These results suggest that one possible mechanism of Enzastaurin activity is through

inhibition of ABCB1 activity, either directly or indirectly through PKC-β, or a

combination of both. In order to investigate whether Enzastaurin regulates PKC-β,

thereby sensitizing cells, knock-down experiments should be performed. If this is

true, knocking down PKC-β should sensitize resistant cells to Paclitaxel in a similar

manner as Enzastaurin treatment. Similarly, over-expression of PKC-β in parental

chemo-sensitive cells should render the cells more resistant to Paclitaxel.

Page 150: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

133

4.2. Aurora Kinase B expression levels as a biomarker for Paclitaxel-

sensitivity in basal-like breast cancer

We have shown that there is a correlation between Paclitaxel-sensitivity, as

determined by the EC50, and AURKB expression in basal-like, but not luminal-like,

breast cancer. The correlation is significant in 8 different cell lines (p-value=0.0013,

R2=0.8411) (Figure 3.27). Moreover, the Paclitaxel-resistant cells show down-

regulation of Aurora Kinase B in the presence of Paclitaxel, whereas when treated

with Enzastaurin and Paclitaxel, Aurora B levels and Paclitaxel-sensitivity are

restored (Figure 3.24). Also, siRNA-targeted knock-down of Aurora B confers

resistance to Paclitaxel (parental cells) and combination treatment (resistant cells)

(Figure 3.28). These results suggest that levels of Aurora Kinase B determine

Paclitaxel-sensitivity.

Paclitaxel is often used in first line treatment for TNBC in patients with primary

breast cancer due to high positive response to neoadjuvant chemotherapy, but the

relapse rate is high and the patients often suffer from metastatic and recurrent tumors.

In these cases it is crucial to find the right treatment option as these tumors are high

grade and progress quickly.

We propose the use of Aurora B expression levels as a biomarker for Paclitaxel-

sensitivity. The expression levels of Aurora B in the tumor tissue vs. normal breast

tissue can potentially determine the patient response rate to therapy containing

Enzastaurin in combination with a Paclitaxel-containing chemotherapy regimen. This

is especially beneficial for patients, as it allows us to eliminate Paclitaxel-based

therapies in patients with low levels of Aurora B, or to use Enzastaurin in

combination with Paclitaxel to restore Aurora B levels. In addition, we found that

Paclitaxel-resistant cells exhibit cross-resistance to other spindle poisons such as

Vincristine, therefore the use of Aurora B as a biomarker and its implications in

treatment can be extended to other mitotic spindle poisons.

Page 151: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

134

4.3. Aurora Kinase B regulation

We have shown that Aurora Kinase B plays an important role in Paclitaxel-resistance

in our model of TNBC cell lines. Moreover, we showed that Aurora Kinase B levels

negatively correlate with Paclitaxel-sensitivity in a number of basal, but not luminal,

breast cancer cell lines. Our Paclitaxel-resistant cell lines have reduced expression

levels of Aurora Kinase B as compared to the parental cell line and Aurora Kinase B

knock-down inhibits Paclitaxel-induced cell death. This data suggests that modulation

of Aurora Kinase B expression could influence sensitivity to Paclitaxel. There are a

number of proteins which have an impact on Aurora Kinase B function through

phosphorylation and dephosphorylation (Figure 4.3). The major players are INCENP,

survivin and Borealin which are member of the Chromosomal Passenger Complex,

together with Aurora Kinase B.

The two major activators of Aurora Kinase B are TLK1 and Chk1, whose direct

mechanism of Aurora activation are not clear. More interestingly, the two most

important inhibitors of Aurora Kinase B are the protein phosphatases PP1 and PP2A.

Figure 4.2 Major regulator of Aurora Kinase B

Red arrows indicate protein phosphorylation events, whereas protein phosphatases are indicated

in blue (Carmena et al., 2009).

Page 152: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

135

They form complexes with Aurora Kinase B and negatively regulate its kinase

function (Sugiyama et al., 2002).

Although there are no reports of PKC directly regulating Aurora Kinase B function, it

has been found that PKCs phosphorylate α-tubulin in the α/β-tubulin dimer which

results in elongation of microtubules in human breast cells (De et al., 2014). In

MB231 cells with high intrinsic PKC activity, this effect could be suppressed when

the cells were treated with a PKC inhibitor. These results link PKC to microtubules

and suggest that it could interact and influence Aurora Kinase B, however, they found

that PKC-α is most likely responsible for α-tubulin phosphorylation, rather than PKC-

β. Moreover, one study found that PKC-δ plays a role in meiotic spindle organization

in mouse oocytes and is associated with the meiotic spindle, thereby further

strengthening the possibility of PKC-β regulating Aurora Kinase B function through

its association with the mitotic spindle.

It is not clear whether the increase in Aurora B levels observed in the resistant cell

lines after sensitization with Enzastaurin is the reason for the cells being sensitized to

Paclitaxel, or merely a side effect of the mitotic arrest.

Page 153: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

136

4.4. Mcl-1 degradation by GSK3β in the presence of Paclitaxel

Mcl-1 stability is primarily regulated by GSK3β, as it can phosphorylate Mcl-1 at

Ser155, Ser159, and Thr163 (Ding et al., 2007, Maurer et al., 2006). Once Mcl-1 is

phosphorylated, it can be ubiquitinated and will subsequently undergo proteasome-

mediated degradation. Three E3 ubiquitin ligases have been identified which

ubiquitinate Mcl-1: 1) Mule was the first ligase identified to ubiquitinate Mcl-1

(Zhong et al., 2005); 2) β-TrCP ubiquitinates and leads to Mcl-1 degradation which is

dependent on Mcl-1 phosphorylation by GSK3β (Ding et al., 2007); 3) FBW7 is a E3

ubiquitin ligase which ubiquitinates Mcl-1 in a GSK3β phosphorylation-dependent

manner (Wertz et al., 2011). Mcl-1 down-regulation is crucial for apoptosis induced

by mitotic arrest after treatment with spindle poisons. It was shown that inactivation

of FBW7 could increase Mcl-1 levels and resistance to Taxol and Vincristine in

ovarian cancer cell lines. Moreover, FBW7-depleted cells showed an increase in cells

undergoing mitotic slippage and a decrease in cells undergoing apoptosis, when

treated with Taxol or Vincristine.

In agreement with the reports demonstrating the importance of GSK3β in inducing

apoptosis through Mcl-1 upon Paclitaxel treatment, our Paclitaxel-resistant cell lines

show increased levels of Serine 9 phosphorylation of GSK3β, the inhibitory

phosphorylation site. Upon treatment with Enzastaurin, GSK3βSer9 levels are

completely depleted, however, this only leads to Mcl-1 degradation in the presence of

Paclitaxel. One possible reason is that Mcl-1 requires a priming phosphorylation in

order to be recognized by GSK3β as a substrate (Morel et al., 2009). It was shown

that upon cellular stress, the c-Jun-terminal protein kinase (JNK) phosphorylates Mcl-

1 on Thr144, which acts as a priming phosphorylation so that GSK3β can then

phosphorylate Mcl-1 at its own phosphorylation site. It has been shown that

Paclitaxel treatment activates JNK (Wang et al., 1999) and that inhibition of JNK

decreases Paclitaxel-induced apoptosis without changing the cell cycle profile (Wang

Page 154: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

137

et al., 2006). This may explain why only in the combination treatment of Enzastaurin

+ Paclitaxel of our Paclitaxel-resistant cell lines, but not in the single treatment with

Enzastaurin alone, Mcl-1 undergoes degradation which subsequently leads to

apoptosis.

On the other hand, prolonged mitotic arrest leads to Mcl-1 destruction (Harley et al.,

2010), which leads to activation of BAX and BAK and subsequently mitochondrial

outer membrane permeabilization and activation of the caspase cascade and induction

of apoptosis (Youle and Strasser, 2008). Mcl-1 degradation upon mitotic arrest is

crucial for the induction of apoptosis. Degradation is regulated by the APC/CCdc20

complex, a multi-component ubiquitin ligase which is active during mitosis. It has

been shown that phosphorylation of Mcl-1 at Thr92 is mediated by Cdk1/cyclin-B

upon mitotic arrest which then leads to its proteolytic destruction by APC/CCdc20.

During mitosis, Mcl-1 levels gradually decrease and when mitotic arrest is too long

and Mcl-1 levels are too low, the cells will undergo apoptosis. Therefore, Mcl-1

degradation upon combination treatment in our resistant cells is possibly a result of

prolonged mitotic arrest.

These are two possible mechanisms of Mcl-1 degradation during re-sensitization of

Paclitaxel-resistant cells with Enzastaurin. At this point it is unclear if either of the

two possible mechanisms, a combination of both, or even a different mechanism is

responsible for Mcl-1 degradation in the resistant cell lines upon re-sensitization.

Either way, to examine whether Mcl-1 degradation is required for sensitization of

resistant cell lines, Mcl-1 over-expression should be able to block the effect of

Enzastaurin and rescue the sensitization effect of Enzastaurin with Paclitaxel in the

resistant cell lines.

Page 155: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

138

CHAPTER 5: CONCLUSION

Page 156: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

139

5.1 Conclusion

In this study we developed an in vitro model of Paclitaxel resistance in Triple

Negative Breast Cancer by treating the TNBC cell lines, SUM159PT and MB231,

over a long period of time with increasing concentrations of Paclitaxel and

maintaining them in medium containing Paclitaxel. The stable cell lines exhibited

EC50 values for Paclitaxel which were approximately 1000-fold and 300-fold higher

than their parental counterparts in SUM159PT and MB231, respectively. Gene

expression profiling showed that pathways for cell viability, cell survival,

proliferation, invasion, migration and microtubule dynamics were more highly

activated, whereas cell death and apoptotic pathways were less activated in the

resistant compared to the parental cell lines. Although, according to IPA analysis, the

network with the highest score has the NF-κB complex at its core, NF-κB inhibitors

were not selective for the resistant over the parental cell lines. This shows that the

NF-κB pathway is important in both resistant and sensitive cells.

In order to determine pathways which play a crucial role in resistance to Paclitaxel,

we performed a pharmacological screen with 180 small molecule inhibitors targeting

various kinases. The screen revealed PKC-β inhibitors to be effective in sensitizing

resistant cells to Paclitaxel. In the presence of Paclitaxel, the PKC-β inhibitor

Enzastaurin reduces cell viability, colony formation and induces cell death. We found

that the resistant cell lines show reduced levels of Aurora Kinase B, a member of the

spindle assembly checkpoint which plays an important role in Paclitaxel-induced cell

death (Sudo et al., 2004). Although the mechanism of sensitization is not entirely

clear, Enzastaurin restores Aurora Kinase B levels in the resistant cell lines, thereby

reinstating mitotic arrest and subsequent apoptosis when treated with Paclitaxel.

Interestingly, we found an inverse correlation between Aurora Kinase B and

Paclitaxel-sensitivity in basal-like, but not luminal, breast cancer cell lines, which

reinforces the importance of Aurora Kinase B in Paclitaxel-induced cell death.

Page 157: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

140

Knocking-down Aurora Kinase B in the parental Paclitaxel-sensitive cell lines using

siRNA renders them more resistant to Paclitaxel treatment, whereas Aurora Kinase B

knock-down in the resistant cell lines inhibits reduction of cell viability upon

combination treatment of Paclitaxel with Enzastaurin. This shows that Aurora Kinase,

indeed, plays an important role in Paclitaxel-sensitivity. Moreover, knock-down

inhibited mitotic arrest in the presence of Paclitaxel in the parental and Enzastaurin-

treated resistant cell lines.

In summary, a kinase inhibitor screen identified PKC-β inhibitors which can re-

sensitize Paclitaxel-resistant TNBC cell lines by restoring Aurora Kinase B

expression.

Page 158: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

141

5.2 Significance of findings and future prospects

TNBC is the most aggressive subtype of breast cancer. The mainstay of treatment is

chemotherapy which shows positive results in a neoadjuvant setting, however,

approximately 70% of patients will relapse and present with recurrent disease, often

accompanied by chemo-resistance and poor survival outcome. Based on the results

from this study, the PKC-β inhibitor Enzastaurin can potentially be used in the

treatment of TNBC patients suffering from recurrent and metastatic disease.

Especially since recurrent disease is a major issue in the treatment of these patients.

Despite the fact that clinical trials with Enzastaurin and Paclitaxel have proved that it

is safe when administered in combination with chemotherapeutics, the results

regarding improvement of patient survival have been inconclusive. A phase II

double-blind study which was conducted with and without Enzastaurin in

combination with Paclitaxel and Carboplatin as first-line treatment in advanced

ovarian cancer showed a slight increase in PFS for patients which received

Enzastaurin, however it was not significant (Vergote et al., 2013).

To provide proof of concept, in vivo studies using xenograft models should be

conducted. We propose to generate an in vivo mouse model of Paclitaxel resistance.

For this, chemo-sensitive cells are injected orthotopically into the mammary fat pad

of mice, once the tumors have grown they will be treated with Paclitaxel until they

regress. The mice are then monitored for recurrence (approximately 3-4 months) and

subsequently randomized into 4 different treatment groups: Control, Paclitaxel,

Enzastaurin, and combination of Paclitaxel with Enzastaurin. If it is true that

Enzastaurin can sensitize resistant cells to Paclitaxel, then mice in the combination

treatment group should have significant reduction of tumor size and increased

survival advantage compared to mice in other treatment groups. The tumors of mice

in the control, Paclitaxel, and Enzastaurin treatment groups, on the other hand, should

continue to grow.

Page 159: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

142

As we have shown earlier, the Paclitaxel-resistant cell lines we generated show

resistance to other chemotherapeutic compounds: the spindle poison Vincristine; and

Adriamycin. Interestingly, Enzastaurin can greatly sensitize resistant cells to

Vincristine but to a much lesser extent to Adriamycin. This suggests that the

mechanism of sensitization by Enzastaurin is specific to microtubule-disrupting

drugs. Similar to the Paclitaxel resistance mouse model, in vivo mouse models of

Vincristine and Adriamycin resistance should be established to give proof-of-concept

that Enzastaurin can sensitize to Vincristine and Adriamycin. This would greatly

enhance the significance of using Enzastaurin in treating recurrent and chemo-

resistant TNBC patients, as Vincrstine and Adriamycin are 2 drugs which are also

often used in first line treatment of TNBC tumors. More importantly, since the

sensitization effect is stronger for Vincristine, Enzastaurin can potentially be used to

treat cancer recurrence in patients suffering from acute leukemia, Hodgkin’s and

Non-Hodgkin’s lymphoma, neuroblastoma, rhabdomyosarcoma, Ewing’s sarcoma,

multiple myeloma, thyroid cancer, brain tumors since Vincristine is one of the main

drugs used in first-line treatment of these patients.

To measure the true effect of Enzastaurin in increasing Paclitaxel-sensitivity, a

clinical trial should be conducted with patients suffering from residual and recurrent

disease and that have previously been treated with a combination of chemotherapeutic

agents containing Paclitaxel. These cancers are more likely to have developed

mechanisms of Paclitaxel resistance and will respond to treatments comprising of

Paclitaxel and Enzastaurin.

Furthermore, the mechanism of re-sensitization by Enzastaurin needs to be studied in

more detail. Although Enzastaurin was originally developed as a PKC-β inhibitor, it

has been shown to have other targets as well, such as ABCB1 (Michaelis et al., 2015).

Moreover, since the various PKC isoforms are structurally similar, we need to

identify the specific isoforms inhibited by Enzastaurin. Therefore, we need to

Page 160: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

143

specifically knock-out the PKC-β isoform and determine whether this can sensitize

our Paclitaxel-resistant cell lines, similar to Enzastaurin treatment. CRISPR can be

used to knock-out PRKCB, rather than just performing a gene knock-down, which

does not ensure complete elimination of gene function. This is important since

residual gene expression can often be enough to induce down-stream signaling and

will not give us correct information of the function PKC-β in Paclitaxel-resistance.

In addition, as this study shows that there is a negative correlation between Aurora

Kinase B expression and Paclitaxel-sensitivity, Aurora Kinase B levels should be

monitored in patient tumor cells during the course of diagnosis and treatment. The

expression of Aurora Kinase B in the tumor tissue and the tissue surrounding the

tumor should be measured at the time of diagnosis and taken as a reference should the

patient present himself with residual or recurrent disease. If the levels of Aurora

Kinase B have decreased, it can be considered to treat the patient with a combination

of Paclitaxel and Enzastaurin.

Moreover, the strategy of restoring Aurora Kinase B expression to sensitize resistant

cancer cells can be applied to other cancers which show a reduction in Aurora Kinase

B levels as the disease progresses. Therefore, this study not only benefits TNBC

breast cancer patients but those presenting with different types of chemo-resistant,

recurrent cancers.

Page 161: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

144

REFERENCES

Page 162: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

145

ACEVEDO-DUNCAN, M., PATEL, R., WHELAN, S. & BICAKU, E. 2002. Human glioma PKC-iota and PKC-betaII phosphorylate cyclin-dependent kinase activating kinase during the cell cycle. Cell Prolif, 35, 23-36.

AL-HAJJ, M., WICHA, M. S., BENITO-HERNANDEZ, A., MORRISON, S. J. & CLARKE, M. F. 2003. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A, 100, 3983-8.

ANDROIC, I., KRAMER, A., YAN, R., RODEL, F., GATJE, R., KAUFMANN, M., STREBHARDT, K. & YUAN, J. 2008. Targeting cyclin B1 inhibits proliferation and sensitizes breast cancer cells to taxol. BMC Cancer, 8, 391.

ANTAL, C. E., HUDSON, A. M., KANG, E., ZANCA, C., WIRTH, C., STEPHENSON, N. L., TROTTER, E. W., GALLEGOS, L. L., MILLER, C. J., FURNARI, F. B., HUNTER, T., BROGNARD, J. & NEWTON, A. C. 2015. Cancer-associated protein kinase C mutations reveal kinase's role as tumor suppressor. Cell, 160, 489-502.

ARCHAMBAULT, V., D'AVINO, P. P., DEERY, M. J., LILLEY, K. S. & GLOVER, D. M. 2008. Sequestration of Polo kinase to microtubules by phosphopriming-independent binding to Map205 is relieved by phosphorylation at a CDK site in mitosis. Genes Dev, 22, 2707-20.

ARCHAMBAULT, V. & GLOVER, D. M. 2009. Polo-like kinases: conservation and divergence in their functions and regulation. Nat Rev Mol Cell Biol, 10, 265-75.

BADVE, S., DABBS, D. J., SCHNITT, S. J., BAEHNER, F. L., DECKER, T., EUSEBI, V., FOX, S. B., ICHIHARA, S., JACQUEMIER, J., LAKHANI, S. R., PALACIOS, J., RAKHA, E. A., RICHARDSON, A. L., SCHMITT, F. C., TAN, P. H., TSE, G. M., WEIGELT, B., ELLIS, I. O. & REIS-FILHO, J. S. 2011. Basal-like and triple-negative breast cancers: a critical review with an emphasis on the implications for pathologists and oncologists. Mod Pathol, 24, 157-67.

BALENDRAN, A., HARE, G. R., KIELOCH, A., WILLIAMS, M. R. & ALESSI, D. R. 2000. Further evidence that 3-phosphoinositide-dependent protein kinase-1 (PDK1) is required for the stability and phosphorylation of protein kinase C (PKC) isoforms. FEBS Lett, 484, 217-23.

BALMANA, J., TUNG, N. M., ISAKOFF, S. J., GRANA, B., RYAN, P. D., SAURA, C., LOWE, E. S., FREWER, P., WINER, E., BASELGA, J. & GARBER, J. E. 2014. Phase I trial of olaparib in combination with cisplatin for the treatment of patients with advanced breast, ovarian and other solid tumors. Ann Oncol, 25, 1656-63.

BERRY, D. A. 2013. Breast cancer screening: controversy of impact. Breast, 22 Suppl 2, S73-6.

BHATTACHARYA, R. & CABRAL, F. 2009. Molecular basis for class V beta-tubulin effects on microtubule assembly and paclitaxel resistance. J Biol Chem, 284, 13023-32.

BIGGINS, S. & MURRAY, A. W. 2001. The budding yeast protein kinase Ipl1/Aurora allows the absence of tension to activate the spindle checkpoint. Genes Dev, 15, 3118-29.

BLACK, J. D. 2010. PKC and control of the cell cycle. Protein Kinase C in Cancer Signaling and Therapy. Springer.

BLAGOSKLONNY, M. V., SCHULTE, T., NGUYEN, P., TREPEL, J. & NECKERS, L. M. 1996. Taxol-induced apoptosis and phosphorylation of Bcl-2 protein involves c-Raf-1 and represents a novel c-Raf-1 signal transduction pathway. Cancer Res, 56, 1851-4.

BORST, P. & ELFERINK, R. O. 2002. Mammalian ABC transporters in health and disease. Annu Rev Biochem, 71, 537-92.

Page 163: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

146

BRITO, D. A. & RIEDER, C. L. 2006. Mitotic checkpoint slippage in humans occurs via cyclin B destruction in the presence of an active checkpoint. Curr Biol, 16, 1194-200.

BRITO, D. A., YANG, Z. & RIEDER, C. L. 2008. Microtubules do not promote mitotic slippage when the spindle assembly checkpoint cannot be satisfied. J Cell Biol, 182, 623-9.

BYRSKI, T., HUZARSKI, T., DENT, R., GRONWALD, J., ZUZIAK, D., CYBULSKI, C., KLADNY, J., GORSKI, B., LUBINSKI, J. & NAROD, S. A. 2009. Response to neoadjuvant therapy with cisplatin in BRCA1-positive breast cancer patients. Breast Cancer Res Treat, 115, 359-63.

CAI, H., SMOLA, U., WIXLER, V., EISENMANN-TAPPE, I., DIAZ-MECO, M. T., MOSCAT, J., RAPP, U. & COOPER, G. M. 1997. Role of diacylglycerol-regulated protein kinase C isotypes in growth factor activation of the Raf-1 protein kinase. Mol Cell Biol, 17, 732-41.

CAO, M. D., SITTER, B., BATHEN, T. F., BOFIN, A., LONNING, P. E., LUNDGREN, S. & GRIBBESTAD, I. S. 2012. Predicting long-term survival and treatment response in breast cancer patients receiving neoadjuvant chemotherapy by MR metabolic profiling. NMR Biomed, 25, 369-78.

CAREY, L. A., DEES, E. C., SAWYER, L., GATTI, L., MOORE, D. T., COLLICHIO, F., OLLILA, D. W., SARTOR, C. I., GRAHAM, M. L. & PEROU, C. M. 2007. The triple negative paradox: primary tumor chemosensitivity of breast cancer subtypes. Clin Cancer Res, 13, 2329-34.

CAREY, L. A., RUGO, H. S., MARCOM, P. K., MAYER, E. L., ESTEVA, F. J., MA, C. X., LIU, M. C., STORNIOLO, A. M., RIMAWI, M. F., FORERO-TORRES, A., WOLFF, A. C., HOBDAY, T. J., IVANOVA, A., CHIU, W. K., FERRARO, M., BURROWS, E., BERNARD, P. S., HOADLEY, K. A., PEROU, C. M. & WINER, E. P. 2012. TBCRC 001: randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer. J Clin Oncol, 30, 2615-23.

CARMENA, M., RUCHAUD, S. & EARNSHAW, W. C. 2009. Making the Auroras glow: regulation of Aurora A and B kinase function by interacting proteins. Curr Opin Cell Biol, 21, 796-805.

CARVALHO, L. V., PEREIRA, E. M., FRAPPART, L., BONIOL, M., BERNARDO, W. M., TARRICONE, V., TAVTIGIAN, S. & SOUTHEY, M. C. 2010. Molecular characterization of breast cancer in young Brazilian women. Rev Assoc Med Bras, 56, 278-87.

CASTAGNA, M., TAKAI, Y., KAIBUCHI, K., SANO, K., KIKKAWA, U. & NISHIZUKA, Y. 1982. Direct activation of calcium-activated, phospholipid-dependent protein kinase by tumor-promoting phorbol esters. J Biol Chem, 257, 7847-51.

CHAI, F., ZHOU, J., CHEN, C., XIE, S., CHEN, X., SU, P. & SHI, J. 2013. The Hedgehog inhibitor cyclopamine antagonizes chemoresistance of breast cancer cells. Onco Targets Ther, 6, 1643-7.

CHAMBERS, T. C., POHL, J., GLASS, D. B. & KUO, J. F. 1994. Phosphorylation by protein kinase C and cyclic AMP-dependent protein kinase of synthetic peptides derived from the linker region of human P-glycoprotein. Biochem J, 299 ( Pt 1), 309-15.

CHAUDHARY, P. M. & RONINSON, I. B. 1991. Expression and activity of P-glycoprotein, a multidrug efflux pump, in human hematopoietic stem cells. Cell, 66, 85-94.

Page 164: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

147

CHAUDHURI, A. R., KHAN, I. A., PRASAD, V., ROBINSON, A. K., LUDUENA, R. F. & BARNES, L. D. 1999. The tumor suppressor protein Fhit. A novel interaction with tubulin. J Biol Chem, 274, 24378-82.

CHEANG, M. C., VODUC, D., BAJDIK, C., LEUNG, S., MCKINNEY, S., CHIA, S. K., PEROU, C. M. & NIELSEN, T. O. 2008. Basal-like breast cancer defined by five biomarkers has superior prognostic value than triple-negative phenotype. Clin Cancer Res, 14, 1368-76.

CHEESEMAN, I. M. & DESAI, A. 2008. Molecular architecture of the kinetochore-microtubule interface. Nat Rev Mol Cell Biol, 9, 33-46.

CHEUNG, A. M., WAN, T. S., LEUNG, J. C., CHAN, L. Y., HUANG, H., KWONG, Y. L., LIANG, R. & LEUNG, A. Y. 2007. Aldehyde dehydrogenase activity in leukemic blasts defines a subgroup of acute myeloid leukemia with adverse prognosis and superior NOD/SCID engrafting potential. Leukemia, 21, 1423-30.

CHEUNG, C. H., CHEN, H. H., KUO, C. C., CHANG, C. Y., COUMAR, M. S., HSIEH, H. P. & CHANG, J. Y. 2009. Survivin counteracts the therapeutic effect of microtubule de-stabilizers by stabilizing tubulin polymers. Mol Cancer, 8, 43.

CHOI, J., KIM DO, H., JUNG, W. H. & KOO, J. S. 2013. Metabolic interaction between cancer cells and stromal cells according to breast cancer molecular subtype. Breast Cancer Res, 15, R78.

COLE, J. M., JOSEPH, S., SUDHAHAR, C. G. & COWDEN DAHL, K. D. 2014. Enrichment for chemoresistant ovarian cancer stem cells from human cell lines. J Vis Exp, 51891.

CORREAS, I., DIAZ-NIDO, J. & AVILA, J. 1992. Microtubule-associated protein tau is phosphorylated by protein kinase C on its tubulin binding domain. J Biol Chem, 267, 15721-8.

CORTES-DERICKS, L., FROMENT, L., BOESCH, R., SCHMID, R. A. & KAROUBI, G. 2014. Cisplatin-resistant cells in malignant pleural mesothelioma cell lines show ALDH(high)CD44(+) phenotype and sphere-forming capacity. BMC Cancer, 14, 304.

CORTES-FUNES, H. & CORONADO, C. 2007. Role of anthracyclines in the era of targeted therapy. Cardiovasc Toxicol, 7, 56-60.

COSENTINO-GOMES, D., ROCCO-MACHADO, N. & MEYER-FERNANDES, J. R. 2012. Cell Signaling through Protein Kinase C Oxidation and Activation. Int J Mol Sci, 13, 10697-721.

CROWE, J. P., JR., GORDON, N. H., HUBAY, C. A., SHENK, R. R., ZOLLINGER, R. M., BRUMBERG, D. J., MCGUIRE, W. L. & SHUCK, J. M. 1991. Estrogen receptor determination and long term survival of patients with carcinoma of the breast. Surg Gynecol Obstet, 173, 273-8.

CROWLEY, E., MCDEVITT, C. A. & CALLAGHAN, R. 2010. Generating inhibitors of P-glycoprotein: where to, now? Methods Mol Biol, 596, 405-32.

CVIJIC, M. E. & CHIN, K. V. 1997. Regulation of P-glycoprotein expression in cyclic AMP-dependent protein kinase mutants. Cell Growth Differ, 8, 1243-7.

DALERBA, P., CHO, R. W. & CLARKE, M. F. 2007a. Cancer stem cells: models and concepts. Annu Rev Med, 58, 267-84.

DALERBA, P., DYLLA, S. J., PARK, I. K., LIU, R., WANG, X., CHO, R. W., HOEY, T., GURNEY, A., HUANG, E. H., SIMEONE, D. M., SHELTON, A. A., PARMIANI, G., CASTELLI, C. & CLARKE, M. F. 2007b. Phenotypic characterization of human colorectal cancer stem cells. Proc Natl Acad Sci U S A, 104, 10158-63.

DALLAS, N. A., XIA, L., FAN, F., GRAY, M. J., GAUR, P., VAN BUREN, G., 2ND, SAMUEL, S., KIM, M. P., LIM, S. J. & ELLIS, L. M. 2009. Chemoresistant colorectal

Page 165: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

148

cancer cells, the cancer stem cell phenotype, and increased sensitivity to insulin-like growth factor-I receptor inhibition. Cancer Res, 69, 1951-7.

DANIS, R. P. & SHEETZ, M. J. 2009. Ruboxistaurin: PKC-beta inhibition for complications of diabetes. Expert Opin Pharmacother, 10, 2913-25.

DE, S., TSIMOUNIS, A., CHEN, X. & ROTENBERG, S. A. 2014. Phosphorylation of alpha-tubulin by protein kinase C stimulates microtubule dynamics in human breast cells. Cytoskeleton (Hoboken), 71, 257-72.

DELIA, D., MIZUTANI, S., LAMORTE, G., GOI, K., IWATA, S. & PIEROTTI, M. A. 1996. p53 activity and chemotherapy. Nat Med, 2, 724-5.

DENT, R., TRUDEAU, M., PRITCHARD, K. I., HANNA, W. M., KAHN, H. K., SAWKA, C. A., LICKLEY, L. A., RAWLINSON, E., SUN, P. & NAROD, S. A. 2007. Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res, 13, 4429-34.

DIAZ BESSONE, M. I., BERARDI, D. E., CAMPODONICO, P. B., TODARO, L. B., LOTHSTEIN, L., BAL DE KIER JOFFE, E. D. & URTREGER, A. J. 2011. Involvement of PKC delta (PKCdelta) in the resistance against different doxorubicin analogs. Breast Cancer Res Treat, 126, 577-87.

DING, Q., HE, X., HSU, J. M., XIA, W., CHEN, C. T., LI, L. Y., LEE, D. F., LIU, J. C., ZHONG, Q., WANG, X. & HUNG, M. C. 2007. Degradation of Mcl-1 by beta-TrCP mediates glycogen synthase kinase 3-induced tumor suppression and chemosensitization. Mol Cell Biol, 27, 4006-17.

DITCHFIELD, C., JOHNSON, V. L., TIGHE, A., ELLSTON, R., HAWORTH, C., JOHNSON, T., MORTLOCK, A., KEEN, N. & TAYLOR, S. S. 2003. Aurora B couples chromosome alignment with anaphase by targeting BubR1, Mad2, and Cenp-E to kinetochores. J Cell Biol, 161, 267-80.

ELIA, A. E., RELLOS, P., HAIRE, L. F., CHAO, J. W., IVINS, F. J., HOEPKER, K., MOHAMMAD, D., CANTLEY, L. C., SMERDON, S. J. & YAFFE, M. B. 2003. The molecular basis for phosphodependent substrate targeting and regulation of Plks by the Polo-box domain. Cell, 115, 83-95.

ESTEVA, F. J., VALERO, V., BOOSER, D., GUERRA, L. T., MURRAY, J. L., PUSZTAI, L., CRISTOFANILLI, M., ARUN, B., ESMAELI, B., FRITSCHE, H. A., SNEIGE, N., SMITH, T. L. & HORTOBAGYI, G. N. 2002. Phase II study of weekly docetaxel and trastuzumab for patients with HER-2-overexpressing metastatic breast cancer. J Clin Oncol, 20, 1800-8.

FACCHINETTI, V., OUYANG, W., WEI, H., SOTO, N., LAZORCHAK, A., GOULD, C., LOWRY, C., NEWTON, A. C., MAO, Y., MIAO, R. Q., SESSA, W. C., QIN, J., ZHANG, P., SU, B. & JACINTO, E. 2008. The mammalian target of rapamycin complex 2 controls folding and stability of Akt and protein kinase C. EMBO J, 27, 1932-43.

FANG, G., YU, H. & KIRSCHNER, M. W. 1998. The checkpoint protein MAD2 and the mitotic regulator CDC20 form a ternary complex with the anaphase-promoting complex to control anaphase initiation. Genes Dev, 12, 1871-83.

FAUL, M. M., GILLIG, J. R., JIROUSEK, M. R., BALLAS, L. M., SCHOTTEN, T., KAHL, A. & MOHR, M. 2003. Acyclic N-(azacycloalkyl)bisindolylmaleimides: isozyme selective inhibitors of PKCbeta. Bioorg Med Chem Lett, 13, 1857-9.

FAUZEE, N. J. 2011. Taxanes: promising anti-cancer drugs. Asian Pac J Cancer Prev, 12, 837-51.

FISHER, B., JEONG, J. H., BRYANT, J., ANDERSON, S., DIGNAM, J., FISHER, E. R. & WOLMARK, N. 2004. Treatment of lymph-node-negative, oestrogen-receptor-positive breast cancer: long-term findings from National Surgical

Page 166: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

149

Adjuvant Breast and Bowel Project randomised clinical trials. Lancet, 364, 858-68.

FISHER, B., REDMOND, C., FISHER, E. R. & CAPLAN, R. 1988. Relative worth of estrogen or progesterone receptor and pathologic characteristics of differentiation as indicators of prognosis in node negative breast cancer patients: findings from National Surgical Adjuvant Breast and Bowel Project Protocol B-06. J Clin Oncol, 6, 1076-87.

FLYNN, P., MELLOR, H., PALMER, R., PANAYOTOU, G. & PARKER, P. J. 1998. Multiple interactions of PRK1 with RhoA. Functional assignment of the Hr1 repeat motif. J Biol Chem, 273, 2698-705.

FOJO, A. T. & MENEFEE, M. 2005. Microtubule targeting agents: basic mechanisms of multidrug resistance (MDR). Semin Oncol, 32, S3-8.

FOLEY, E. A. & KAPOOR, T. M. 2013. Microtubule attachment and spindle assembly checkpoint signalling at the kinetochore. Nat Rev Mol Cell Biol, 14, 25-37.

FONG, P. C., BOSS, D. S., YAP, T. A., TUTT, A., WU, P., MERGUI-ROELVINK, M., MORTIMER, P., SWAISLAND, H., LAU, A., O'CONNOR, M. J., ASHWORTH, A., CARMICHAEL, J., KAYE, S. B., SCHELLENS, J. H. & DE BONO, J. S. 2009. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med, 361, 123-34.

FOULKES, W. D., SMITH, I. E. & REIS-FILHO, J. S. 2010. Triple-negative breast cancer. N Engl J Med, 363, 1938-48.

FRASCI, G., COMELLA, P., RINALDO, M., IODICE, G., DI BONITO, M., D'AIUTO, M., PETRILLO, A., LASTORIA, S., SIANI, C., COMELLA, G. & D'AIUTO, G. 2009. Preoperative weekly cisplatin-epirubicin-paclitaxel with G-CSF support in triple-negative large operable breast cancer. Ann Oncol, 20, 1185-92.

FRIEDMAN, H. S., COLVIN, O. M., KAUFMANN, S. H., LUDEMAN, S. M., BULLOCK, N., BIGNER, D. D. & GRIFFITH, O. W. 1992. Cyclophosphamide resistance in medulloblastoma. Cancer Res, 52, 5373-8.

FUJITA, Y., KOJIMA, K., OHHASHI, R., HAMADA, N., NOZAWA, Y., KITAMOTO, A., SATO, A., KONDO, S., KOJIMA, T., DEGUCHI, T. & ITO, M. 2010. MiR-148a attenuates paclitaxel resistance of hormone-refractory, drug-resistant prostate cancer PC3 cells by regulating MSK1 expression. J Biol Chem, 285, 19076-84.

GADEA, B. B. & RUDERMAN, J. V. 2006. Aurora B is required for mitotic chromatin-induced phosphorylation of Op18/Stathmin. Proc Natl Acad Sci U S A, 103, 4493-8.

GASCOIGNE, K. E. & TAYLOR, S. S. 2009. How do anti-mitotic drugs kill cancer cells? J Cell Sci, 122, 2579-85.

GASSMANN, R., CARVALHO, A., HENZING, A. J., RUCHAUD, S., HUDSON, D. F., HONDA, R., NIGG, E. A., GERLOFF, D. L. & EARNSHAW, W. C. 2004. Borealin: a novel chromosomal passenger required for stability of the bipolar mitotic spindle. J Cell Biol, 166, 179-91.

GERBER, B., LOIBL, S., EIDTMANN, H., REZAI, M., FASCHING, P. A., TESCH, H., EGGEMANN, H., SCHRADER, I., KITTEL, K., HANUSCH, C., KREIENBERG, R., SOLBACH, C., JACKISCH, C., KUNZ, G., BLOHMER, J. U., HUOBER, J., HAUSCHILD, M., NEKLJUDOVA, V., UNTCH, M. & VON MINCKWITZ, G. 2013. Neoadjuvant bevacizumab and anthracycline-taxane-based chemotherapy in 678 triple-negative primary breast cancers; results from the geparquinto study (GBG 44). Ann Oncol, 24, 2978-84.

GERMANN, U. A., CHAMBERS, T. C., AMBUDKAR, S. V., LICHT, T., CARDARELLI, C. O., PASTAN, I. & GOTTESMAN, M. M. 1996. Characterization of

Page 167: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

150

phosphorylation-defective mutants of human P-glycoprotein expressed in mammalian cells. J Biol Chem, 271, 1708-16.

GEWIRTZ, D. A. 1999. A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics adriamycin and daunorubicin. Biochem Pharmacol, 57, 727-41.

GIANNAKAKOU, P., SACKETT, D. L., KANG, Y. K., ZHAN, Z., BUTERS, J. T., FOJO, T. & PORUCHYNSKY, M. S. 1997. Paclitaxel-resistant human ovarian cancer cells have mutant beta-tubulins that exhibit impaired paclitaxel-driven polymerization. J Biol Chem, 272, 17118-25.

GILL, P. S., TULPULE, A., ESPINA, B. M., CABRIALES, S., BRESNAHAN, J., ILAW, M., LOUIE, S., GUSTAFSON, N. F., BROWN, M. A., ORCUTT, C., WINOGRAD, B. & SCADDEN, D. T. 1999. Paclitaxel is safe and effective in the treatment of advanced AIDS-related Kaposi's sarcoma. J Clin Oncol, 17, 1876-83.

GILLET, J. P. & GOTTESMAN, M. M. 2010. Mechanisms of multidrug resistance in cancer. Methods Mol Biol, 596, 47-76.

GINESTIER, C., HUR, M. H., CHARAFE-JAUFFRET, E., MONVILLE, F., DUTCHER, J., BROWN, M., JACQUEMIER, J., VIENS, P., KLEER, C. G., LIU, S., SCHOTT, A., HAYES, D., BIRNBAUM, D., WICHA, M. S. & DONTU, G. 2007. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell, 1, 555-67.

GLIMELIUS, B., LAHN, M., GAWANDE, S., CLEVERLY, A., DARSTEIN, C., MUSIB, L., LIU, Y., SPINDLER, K. L., FRODIN, J. E., BERGLUND, A., BYSTROM, P., QVORTRUP, C., JAKOBSEN, A. & PFEIFFER, P. 2010. A window of opportunity phase II study of enzastaurin in chemonaive patients with asymptomatic metastatic colorectal cancer. Ann Oncol, 21, 1020-6.

GOEPFERT, T. M., ADIGUN, Y. E., ZHONG, L., GAY, J., MEDINA, D. & BRINKLEY, W. R. 2002. Centrosome amplification and overexpression of aurora A are early events in rat mammary carcinogenesis. Cancer Res, 62, 4115-22.

GOLDSTEIN, L. J., GALSKI, H., FOJO, A., WILLINGHAM, M., LAI, S. L., GAZDAR, A., PIRKER, R., GREEN, A., CRIST, W., BRODEUR, G. M. & ET AL. 1989. Expression of a multidrug resistance gene in human cancers. J Natl Cancer Inst, 81, 116-24.

GONG, R., HONG, A. W., PLOUFFE, S. W., ZHAO, B., LIU, G., YU, F. X., XU, Y. & GUAN, K. L. 2015. Opposing roles of conventional and novel PKC isoforms in Hippo-YAP pathway regulation. Cell Res, 25, 985-8.

GONZALEZ-ANGULO, A. M., BROGLIO, K., KAU, S. W., ERALP, Y., ERLICHMAN, J., VALERO, V., THERIAULT, R., BOOSER, D., BUZDAR, A. U., HORTOBAGYI, G. N. & ARUN, B. 2005. Women age < or = 35 years with primary breast carcinoma: disease features at presentation. Cancer, 103, 2466-72.

GOODE, N., HUGHES, K., WOODGETT, J. R. & PARKER, P. J. 1992. Differential regulation of glycogen synthase kinase-3 beta by protein kinase C isotypes. J Biol Chem, 267, 16878-82.

GORBSKY, G. J. & RICKETTS, W. A. 1993. Differential expression of a phosphoepitope at the kinetochores of moving chromosomes. J Cell Biol, 122, 1311-21.

GOTZSCHE, P. C. & OLSEN, O. 2000. Is screening for breast cancer with mammography justifiable? Lancet, 355, 129-34.

GOULD, C. M., KANNAN, N., TAYLOR, S. S. & NEWTON, A. C. 2009. The chaperones Hsp90 and Cdc37 mediate the maturation and stabilization of protein kinase C through a conserved PXXP motif in the C-terminal tail. J Biol Chem, 284, 4921-35.

Page 168: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

151

GRAY, R., BHATTACHARYA, S., BOWDEN, C., MILLER, K. & COMIS, R. L. 2009. Independent review of E2100: a phase III trial of bevacizumab plus paclitaxel versus paclitaxel in women with metastatic breast cancer. J Clin Oncol, 27, 4966-72.

GRINER, E. M. & KAZANIETZ, M. G. 2007. Protein kinase C and other diacylglycerol effectors in cancer. Nat Rev Cancer, 7, 281-94.

GROBER, O. M., MUTARELLI, M., GIURATO, G., RAVO, M., CICATIELLO, L., DE FILIPPO, M. R., FERRARO, L., NASSA, G., PAPA, M. F., PARIS, O., TARALLO, R., LUO, S., SCHROTH, G. P., BENES, V. & WEISZ, A. 2011. Global analysis of estrogen receptor beta binding to breast cancer cell genome reveals an extensive interplay with estrogen receptor alpha for target gene regulation. BMC Genomics, 12, 36.

GROENEWEG, J. W., DIGLORIA, C. M., YUAN, J., RICHARDSON, W. S., GROWDON, W. B., SATHYANARAYANAN, S., FOSTER, R. & RUEDA, B. R. 2014. Inhibition of notch signaling in combination with Paclitaxel reduces platinum-resistant ovarian tumor growth. Front Oncol, 4, 171.

GROSSI, V., PESERICO, A., TEZIL, T. & SIMONE, C. 2014. p38alpha MAPK pathway: a key factor in colorectal cancer therapy and chemoresistance. World J Gastroenterol, 20, 9744-58.

GROSSONI, V. C., TODARO, L. B., KAZANIETZ, M. G., DE KIER JOFFE, E. D. & URTREGER, A. J. 2009. Opposite effects of protein kinase C beta1 (PKCbeta1) and PKCepsilon in the metastatic potential of a breast cancer murine model. Breast Cancer Res Treat, 118, 469-80.

GUAN, X., MA, F., FAN, Y., ZHU, W., HONG, R. & XU, B. 2015. Platinum-based chemotherapy in triple-negative breast cancer: a systematic review and meta-analysis of randomized-controlled trials. Anticancer Drugs, 26, 894-901.

GWAK, J., JUNG, S. J., KANG, D. I., KIM, E. Y., KIM, D. E., CHUNG, Y. H., SHIN, J. G. & OH, S. 2009. Stimulation of protein kinase C-alpha suppresses colon cancer cell proliferation by down-regulation of beta-catenin. J Cell Mol Med, 13, 2171-80.

HABER, M., BURKHART, C. A., REGL, D. L., MADAFIGLIO, J., NORRIS, M. D. & HORWITZ, S. B. 1995. Altered expression of M beta 2, the class II beta-tubulin isotype, in a murine J774.2 cell line with a high level of taxol resistance. J Biol Chem, 270, 31269-75.

HAFFTY, B. G., YANG, Q., REISS, M., KEARNEY, T., HIGGINS, S. A., WEIDHAAS, J., HARRIS, L., HAIT, W. & TOPPMEYER, D. 2006. Locoregional relapse and distant metastasis in conservatively managed triple negative early-stage breast cancer. J Clin Oncol, 24, 5652-7.

HARLEY, M. E., ALLAN, L. A., SANDERSON, H. S. & CLARKE, P. R. 2010. Phosphorylation of Mcl-1 by CDK1-cyclin B1 initiates its Cdc20-dependent destruction during mitotic arrest. EMBO J, 29, 2407-20.

HARVEY, J. M., CLARK, G. M., OSBORNE, C. K. & ALLRED, D. C. 1999. Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol, 17, 1474-81.

HASTAK, K., ALLI, E. & FORD, J. M. 2010. Synergistic chemosensitivity of triple-negative breast cancer cell lines to poly(ADP-Ribose) polymerase inhibition, gemcitabine, and cisplatin. Cancer Res, 70, 7970-80.

HAUF, S., COLE, R. W., LATERRA, S., ZIMMER, C., SCHNAPP, G., WALTER, R., HECKEL, A., VAN MEEL, J., RIEDER, C. L. & PETERS, J. M. 2003. The small molecule

Page 169: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

152

Hesperadin reveals a role for Aurora B in correcting kinetochore-microtubule attachment and in maintaining the spindle assembly checkpoint. J Cell Biol, 161, 281-94.

HAYES, D. F., THOR, A. D., DRESSLER, L. G., WEAVER, D., EDGERTON, S., COWAN, D., BROADWATER, G., GOLDSTEIN, L. J., MARTINO, S., INGLE, J. N., HENDERSON, I. C., NORTON, L., WINER, E. P., HUDIS, C. A., ELLIS, M. J. & BERRY, D. A. 2007. HER2 and response to paclitaxel in node-positive breast cancer. N Engl J Med, 357, 1496-506.

HELLSTEN, R., JOHANSSON, M., DAHLMAN, A., STERNER, O. & BJARTELL, A. 2011. Galiellalactone inhibits stem cell-like ALDH-positive prostate cancer cells. PLoS One, 6, e22118.

HILTON, J. 1984. Role of aldehyde dehydrogenase in cyclophosphamide-resistant L1210 leukemia. Cancer Res, 44, 5156-60.

HOWELL, B. J., MCEWEN, B. F., CANMAN, J. C., HOFFMAN, D. B., FARRAR, E. M., RIEDER, C. L. & SALMON, E. D. 2001. Cytoplasmic dynein/dynactin drives kinetochore protein transport to the spindle poles and has a role in mitotic spindle checkpoint inactivation. J Cell Biol, 155, 1159-72.

HUISMAN, C., FERREIRA, C. G., BROKER, L. E., RODRIGUEZ, J. A., SMIT, E. F., POSTMUS, P. E., KRUYT, F. A. & GIACCONE, G. 2002. Paclitaxel triggers cell death primarily via caspase-independent routes in the non-small cell lung cancer cell line NCI-H460. Clin Cancer Res, 8, 596-606.

IANZINI, F. & MACKEY, M. A. 1997. Spontaneous premature chromosome condensation and mitotic catastrophe following irradiation of HeLa S3 cells. Int J Radiat Biol, 72, 409-21.

IKAWA, M., IMPRAIM, C. C., WANG, G. & YOSHIDA, A. 1983. Isolation and characterization of aldehyde dehydrogenase isozymes from usual and atypical human livers. J Biol Chem, 258, 6282-7.

IKENOUE, T., INOKI, K., YANG, Q., ZHOU, X. & GUAN, K. L. 2008. Essential function of TORC2 in PKC and Akt turn motif phosphorylation, maturation and signalling. EMBO J, 27, 1919-31.

ISRAELI, D., ZIAEI, S., GONIN, P. & GARCIA, L. 2005. A proposal for the physiological significance of mdr1 and Bcrp1/Abcg2 gene expression in normal tissue regeneration and after cancer therapy. J Theor Biol, 232, 41-5.

JANUCHOWSKI, R., WOJTOWICZ, K. & ZABEL, M. 2013. The role of aldehyde dehydrogenase (ALDH) in cancer drug resistance. Biomed Pharmacother, 67, 669-80.

JARZABEK, K., LAUDANSKI, P., DZIECIOL, J., DABROWSKA, M. & WOLCZYNSKI, S. 2002. Protein kinase C involvement in proliferation and survival of breast cancer cells. Folia Histochem Cytobiol, 40, 193-4.

JENG, Y. M., PENG, S. Y., LIN, C. Y. & HSU, H. C. 2004. Overexpression and amplification of Aurora-A in hepatocellular carcinoma. Clin Cancer Res, 10, 2065-71.

JONES, P. A. & BAYLIN, S. B. 2002. The fundamental role of epigenetic events in cancer. Nat Rev Genet, 3, 415-28.

KANDA, A., KAWAI, H., SUTO, S., KITAJIMA, S., SATO, S., TAKATA, T. & TATSUKA, M. 2005. Aurora-B/AIM-1 kinase activity is involved in Ras-mediated cell transformation. Oncogene, 24, 7266-72.

KANDOTH, C., MCLELLAN, M. D., VANDIN, F., YE, K., NIU, B., LU, C., XIE, M., ZHANG, Q., MCMICHAEL, J. F., WYCZALKOWSKI, M. A., LEISERSON, M. D., MILLER, C. A., WELCH, J. S., WALTER, M. J., WENDL, M. C., LEY, T. J., WILSON, R. K.,

Page 170: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

153

RAPHAEL, B. J. & DING, L. 2013. Mutational landscape and significance across 12 major cancer types. Nature, 502, 333-9.

KARNOUB, A. E., DASH, A. B., VO, A. P., SULLIVAN, A., BROOKS, M. W., BELL, G. W., RICHARDSON, A. L., POLYAK, K., TUBO, R. & WEINBERG, R. A. 2007. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature, 449, 557-63.

KATAYAMA, H., OTA, T., JISAKI, F., UEDA, Y., TANAKA, T., ODASHIMA, S., SUZUKI, F., TERADA, Y. & TATSUKA, M. 1999. Mitotic kinase expression and colorectal cancer progression. J Natl Cancer Inst, 91, 1160-2.

KATAYAMA, H., SASAI, K., KAWAI, H., YUAN, Z. M., BONDARUK, J., SUZUKI, F., FUJII, S., ARLINGHAUS, R. B., CZERNIAK, B. A. & SEN, S. 2004. Phosphorylation by aurora kinase A induces Mdm2-mediated destabilization and inhibition of p53. Nat Genet, 36, 55-62.

KAUFMAN, B., SHAPIRA-FROMMER, R., SCHMUTZLER, R. K., AUDEH, M. W., FRIEDLANDER, M., BALMANA, J., MITCHELL, G., FRIED, G., STEMMER, S. M., HUBERT, A., ROSENGARTEN, O., STEINER, M., LOMAN, N., BOWEN, K., FIELDING, A. & DOMCHEK, S. M. 2015. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol, 33, 244-50.

KAUFMAN, P. A. 1999. Paclitaxel and anthracycline combination chemotherapy for metastatic breast cancer. Semin Oncol, 26, 39-46.

KEEN, N. & TAYLOR, S. 2004. Aurora-kinase inhibitors as anticancer agents. Nat Rev Cancer, 4, 927-36.

KELLER, E. T., FU, Z., YEUNG, K. & BRENNAN, M. 2004. Raf kinase inhibitor protein: a prostate cancer metastasis suppressor gene. Cancer Lett, 207, 131-7.

KENEMANS, P., VERSTRAETEN, R. A. & VERHEIJEN, R. H. 2008. Oncogenic pathways in hereditary and sporadic breast cancer. Maturitas, 61, 141-50.

KERFOOT, C., HUANG, W. & ROTENBERG, S. A. 2004. Immunohistochemical analysis of advanced human breast carcinomas reveals downregulation of protein kinase C alpha. J Histochem Cytochem, 52, 419-22.

KEYES, K. A., MANN, L., SHERMAN, M., GALBREATH, E., SCHIRTZINGER, L., BALLARD, D., CHEN, Y. F., IVERSEN, P. & TEICHER, B. A. 2004. LY317615 decreases plasma VEGF levels in human tumor xenograft-bearing mice. Cancer Chemother Pharmacol, 53, 133-40.

KHALIL, R. A. 2010. Regulation of Vascular Smooth Muscle Function. San Rafael (CA). KIM, S., LEE, J., LEE, S. K., BAE, S. Y., KIM, J., KIM, M., KIL, W. H., KIM, S. W., LEE, J. E.

& NAM, S. J. 2014. Protein kinase C-alpha downregulates estrogen receptor-alpha by suppressing c-Jun phosphorylation in estrogen receptor-positive breast cancer cells. Oncol Rep, 31, 1423-8.

KIMURA, M., MATSUDA, Y., YOSHIOKA, T. & OKANO, Y. 1999. Cell cycle-dependent expression and centrosome localization of a third human aurora/Ipl1-related protein kinase, AIK3. J Biol Chem, 274, 7334-40.

KLOS, K. S., ZHOU, X., LEE, S., ZHANG, L., YANG, W., NAGATA, Y. & YU, D. 2003. Combined trastuzumab and paclitaxel treatment better inhibits ErbB-2-mediated angiogenesis in breast carcinoma through a more effective inhibition of Akt than either treatment alone. Cancer, 98, 1377-85.

KRAMER, T., SCHMIDT, B. & LO MONTE, F. 2012. Small-Molecule Inhibitors of GSK-3: Structural Insights and Their Application to Alzheimer's Disease Models. Int J Alzheimers Dis, 2012, 381029.

Page 171: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

154

KREISL, T. N., KOTLIAROVA, S., BUTMAN, J. A., ALBERT, P. S., KIM, L., MUSIB, L., THORNTON, D. & FINE, H. A. 2010. A phase I/II trial of enzastaurin in patients with recurrent high-grade gliomas. Neuro Oncol, 12, 181-9.

KUMAR, S., MAHDI, H., BRYANT, C., SHAH, J. P., GARG, G. & MUNKARAH, A. 2010. Clinical trials and progress with paclitaxel in ovarian cancer. Int J Womens Health, 2, 411-27.

LAHN, M., KOHLER, G., SUNDELL, K., SU, C., LI, S., PATERSON, B. M. & BUMOL, T. F. 2004. Protein kinase C alpha expression in breast and ovarian cancer. Oncology, 67, 1-10.

LAI, D., HO, K. C., HAO, Y. & YANG, X. 2011. Taxol resistance in breast cancer cells is mediated by the hippo pathway component TAZ and its downstream transcriptional targets Cyr61 and CTGF. Cancer Res, 71, 2728-38.

LAPIDOT, T., SIRARD, C., VORMOOR, J., MURDOCH, B., HOANG, T., CACERES-CORTES, J., MINDEN, M., PATERSON, B., CALIGIURI, M. A. & DICK, J. E. 1994. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature, 367, 645-8.

LEHMANN, B. D., BAUER, J. A., CHEN, X., SANDERS, M. E., CHAKRAVARTHY, A. B., SHYR, Y. & PIETENPOL, J. A. 2011. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest, 121, 2750-67.

LENS, S. M., VOEST, E. E. & MEDEMA, R. H. 2010. Shared and separate functions of polo-like kinases and aurora kinases in cancer. Nat Rev Cancer, 10, 825-41.

LEONG, C. O., VIDNOVIC, N., DEYOUNG, M. P., SGROI, D. & ELLISEN, L. W. 2007. The p63/p73 network mediates chemosensitivity to cisplatin in a biologically defined subset of primary breast cancers. J Clin Invest, 117, 1370-80.

LI, H. & WEINSTEIN, I. B. 2006. Protein kinase C beta enhances growth and expression of cyclin D1 in human breast cancer cells. Cancer Res, 66, 11399-408.

LIEDTKE, C., MAZOUNI, C., HESS, K. R., ANDRE, F., TORDAI, A., MEJIA, J. A., SYMMANS, W. F., GONZALEZ-ANGULO, A. M., HENNESSY, B., GREEN, M., CRISTOFANILLI, M., HORTOBAGYI, G. N. & PUSZTAI, L. 2008. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J Clin Oncol, 26, 1275-81.

LIM, E., VAILLANT, F., WU, D., FORREST, N. C., PAL, B., HART, A. H., ASSELIN-LABAT, M. L., GYORKI, D. E., WARD, T., PARTANEN, A., FELEPPA, F., HUSCHTSCHA, L. I., THORNE, H. J., FOX, S. B., YAN, M., FRENCH, J. D., BROWN, M. A., SMYTH, G. K., VISVADER, J. E. & LINDEMAN, G. J. 2009. Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nat Med, 15, 907-13.

LIN, Y., LIN, S., WATSON, M., TRINKAUS, K. M., KUO, S., NAUGHTON, M. J., WEILBAECHER, K., FLEMING, T. P. & AFT, R. L. 2010. A gene expression signature that predicts the therapeutic response of the basal-like breast cancer to neoadjuvant chemotherapy. Breast Cancer Res Treat, 123, 691-9.

LINGLE, W. L. & SALISBURY, J. L. 1999. Altered centrosome structure is associated with abnormal mitoses in human breast tumors. Am J Pathol, 155, 1941-51.

LIU, Q., KANEKO, S., YANG, L., FELDMAN, R. I., NICOSIA, S. V., CHEN, J. & CHENG, J. Q. 2004. Aurora-A abrogation of p53 DNA binding and transactivation activity by phosphorylation of serine 215. J Biol Chem, 279, 52175-82.

LIVRAGHI, L. & GARBER, J. E. 2015. PARP inhibitors in the management of breast cancer: current data and future prospects. BMC Med, 13, 188.

Page 172: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

155

LLOVERA, L., MANSILLA, S. & PORTUGAL, J. 2012. Apoptotic-like death occurs through a caspase-independent route in colon carcinoma cells undergoing mitotic catastrophe. Cancer Lett, 326, 114-21.

LONGLEY, D. B. & JOHNSTON, P. G. 2005. Molecular mechanisms of drug resistance. J Pathol, 205, 275-92.

LONNE, G. K., CORNMARK, L., ZAHIROVIC, I. O., LANDBERG, G., JIRSTROM, K. & LARSSON, C. 2010. PKCalpha expression is a marker for breast cancer aggressiveness. Mol Cancer, 9, 76.

LUNA-ULLOA, L. B., HERNANDEZ-MAQUEDA, J. G., SANTOYO-RAMOS, P., CASTANEDA-PATLAN, M. C. & ROBLES-FLORES, M. 2011. Protein kinase C zeta is a positive modulator of canonical Wnt signaling pathway in tumoral colon cell lines. Carcinogenesis, 32, 1615-24.

LUND, M. J., BUTLER, E. N., HAIR, B. Y., WARD, K. C., ANDREWS, J. H., OPREA-ILIES, G., BAYAKLY, A. R., O'REGAN, R. M., VERTINO, P. M. & ELEY, J. W. 2010. Age/race differences in HER2 testing and in incidence rates for breast cancer triple subtypes: a population-based study and first report. Cancer, 116, 2549-59.

MA, L., TAO, Y., DURAN, A., LLADO, V., GALVEZ, A., BARGER, J. F., CASTILLA, E. A., CHEN, J., YAJIMA, T., POROLLO, A., MEDVEDOVIC, M., BRILL, L. M., PLAS, D. R., RIEDL, S. J., LEITGES, M., DIAZ-MECO, M. T., RICHARDSON, A. D. & MOSCAT, J. 2013. Control of nutrient stress-induced metabolic reprogramming by PKCzeta in tumorigenesis. Cell, 152, 599-611.

MACDONALD, S. G., CREWS, C. M., WU, L., DRILLER, J., CLARK, R., ERIKSON, R. L. & MCCORMICK, F. 1993. Reconstitution of the Raf-1-MEK-ERK signal transduction pathway in vitro. Mol Cell Biol, 13, 6615-20.

MALDONADO, M. & KAPOOR, T. M. 2011. Constitutive Mad1 targeting to kinetochores uncouples checkpoint signalling from chromosome biorientation. Nat Cell Biol, 13, 475-82.

MANUEL IGLESIAS, J., BELOQUI, I., GARCIA-GARCIA, F., LEIS, O., VAZQUEZ-MARTIN, A., EGUIARA, A., CUFI, S., PAVON, A., MENENDEZ, J. A., DOPAZO, J. & MARTIN, A. G. 2013. Mammosphere formation in breast carcinoma cell lines depends upon expression of E-cadherin. PLoS One, 8, e77281.

MARTIN, M., RODRIGUEZ-LESCURE, A., RUIZ, A., ALBA, E., CALVO, L., RUIZ-BORREGO, M., SANTABALLA, A., RODRIGUEZ, C. A., CRESPO, C., ABAD, M., DOMINGUEZ, S., FLORIAN, J., LLORCA, C., MENDEZ, M., GODES, M., CUBEDO, R., MURIAS, A., BATISTA, N., GARCIA, M. J., CABALLERO, R. & DE ALAVA, E. 2010. Molecular predictors of efficacy of adjuvant weekly paclitaxel in early breast cancer. Breast Cancer Res Treat, 123, 149-57.

MASANEK, U., STAMMLER, G. & VOLM, M. 2002. Modulation of multidrug resistance in human ovarian cancer cell lines by inhibition of P-glycoprotein 170 and PKC isoenzymes with antisense oligonucleotides. J Exp Ther Oncol, 2, 37-41.

MATHEW, R., KARANTZA-WADSWORTH, V. & WHITE, E. 2007. Role of autophagy in cancer. Nat Rev Cancer, 7, 961-7.

MAURER, U., CHARVET, C., WAGMAN, A. S., DEJARDIN, E. & GREEN, D. R. 2006. Glycogen synthase kinase-3 regulates mitochondrial outer membrane permeabilization and apoptosis by destabilization of MCL-1. Mol Cell, 21, 749-60.

MCCARROLL, J. A., GAN, P. P., LIU, M. & KAVALLARIS, M. 2010. betaIII-tubulin is a multifunctional protein involved in drug sensitivity and tumorigenesis in non-small cell lung cancer. Cancer Res, 70, 4995-5003.

Page 173: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

156

MICHAELIS, M., ROTHWEILER, F., LOSCHMANN, N., SHARIFI, M., GHAFOURIAN, T. & CINATL, J., JR. 2015. Enzastaurin inhibits ABCB1-mediated drug efflux independently of effects on protein kinase C signalling and the cellular p53 status. Oncotarget, 6, 17605-20.

MILES, D. W., CHAN, A., DIRIX, L. Y., CORTES, J., PIVOT, X., TOMCZAK, P., DELOZIER, T., SOHN, J. H., PROVENCHER, L., PUGLISI, F., HARBECK, N., STEGER, G. G., SCHNEEWEISS, A., WARDLEY, A. M., CHLISTALLA, A. & ROMIEU, G. 2010. Phase III study of bevacizumab plus docetaxel compared with placebo plus docetaxel for the first-line treatment of human epidermal growth factor receptor 2-negative metastatic breast cancer. J Clin Oncol, 28, 3239-47.

MIRANDA, M., ROZALI, E., KHANNA, K. K. & AL-EJEH, F. 2015. MEK5-ERK5 pathway associates with poor survival of breast cancer patients after systemic treatments. Oncoscience, 2, 99-101.

MOCHLY-ROSEN, D. & GORDON, A. S. 1998. Anchoring proteins for protein kinase C: a means for isozyme selectivity. FASEB J, 12, 35-42.

MOREL, C., CARLSON, S. M., WHITE, F. M. & DAVIS, R. J. 2009. Mcl-1 integrates the opposing actions of signaling pathways that mediate survival and apoptosis. Mol Cell Biol, 29, 3845-52.

MUSACCHIO, A. & SALMON, E. D. 2007. The spindle-assembly checkpoint in space and time. Nat Rev Mol Cell Biol, 8, 379-93.

NAHTA, R. 2012. Pharmacological strategies to overcome HER2 cross-talk and Trastuzumab resistance. Curr Med Chem, 19, 1065-75.

NEVE, R. M., CHIN, K., FRIDLYAND, J., YEH, J., BAEHNER, F. L., FEVR, T., CLARK, L., BAYANI, N., COPPE, J. P., TONG, F., SPEED, T., SPELLMAN, P. T., DEVRIES, S., LAPUK, A., WANG, N. J., KUO, W. L., STILWELL, J. L., PINKEL, D., ALBERTSON, D. G., WALDMAN, F. M., MCCORMICK, F., DICKSON, R. B., JOHNSON, M. D., LIPPMAN, M., ETHIER, S., GAZDAR, A. & GRAY, J. W. 2006. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell, 10, 515-27.

NEWTON, A. C. 2003. Regulation of the ABC kinases by phosphorylation: protein kinase C as a paradigm. Biochem J, 370, 361-71.

NICKLAS, R. B. & KOCH, C. A. 1969. Chromosome micromanipulation. 3. Spindle fiber tension and the reorientation of mal-oriented chromosomes. J Cell Biol, 43, 40-50.

NIELSEN, T. O., HSU, F. D., JENSEN, K., CHEANG, M., KARACA, G., HU, Z., HERNANDEZ-BOUSSARD, T., LIVASY, C., COWAN, D., DRESSLER, L., AKSLEN, L. A., RAGAZ, J., GOWN, A. M., GILKS, C. B., VAN DE RIJN, M. & PEROU, C. M. 2004. Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res, 10, 5367-74.

NIIKURA, Y., DIXIT, A., SCOTT, R., PERKINS, G. & KITAGAWA, K. 2007. BUB1 mediation of caspase-independent mitotic death determines cell fate. J Cell Biol, 178, 283-96.

NISHIDA, K., YAMAGUCHI, O. & OTSU, K. 2008. Crosstalk between autophagy and apoptosis in heart disease. Circ Res, 103, 343-51.

NISHIZUKA, Y. 1992. Intracellular signaling by hydrolysis of phospholipids and activation of protein kinase C. Science, 258, 607-14.

NITISS, J. L. 2009. Targeting DNA topoisomerase II in cancer chemotherapy. Nat Rev Cancer, 9, 338-50.

NOGUCHI, S. 2006. Predictive factors for response to docetaxel in human breast cancers. Cancer Sci, 97, 813-20.

Page 174: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

157

NOTTE, A., LECLERE, L. & MICHIELS, C. 2011. Autophagy as a mediator of chemotherapy-induced cell death in cancer. Biochem Pharmacol, 82, 427-34.

OH, Y., HERBST, R. S., BURRIS, H., CLEVERLY, A., MUSIB, L., LAHN, M. & BEPLER, G. 2008. Enzastaurin, an oral serine/threonine kinase inhibitor, as second- or third-line therapy of non-small-cell lung cancer. J Clin Oncol, 26, 1135-41.

OHI, R., SAPRA, T., HOWARD, J. & MITCHISON, T. J. 2004. Differentiation of cytoplasmic and meiotic spindle assembly MCAK functions by Aurora B-dependent phosphorylation. Mol Biol Cell, 15, 2895-906.

OKADA, H., BAKAL, C., SHAHINIAN, A., ELIA, A., WAKEHAM, A., SUH, W. K., DUNCAN, G. S., CIOFANI, M., ROTTAPEL, R., ZUNIGA-PFLUCKER, J. C. & MAK, T. W. 2004. Survivin loss in thymocytes triggers p53-mediated growth arrest and p53-independent cell death. J Exp Med, 199, 399-410.

ORR, G. A., VERDIER-PINARD, P., MCDAID, H. & HORWITZ, S. B. 2003. Mechanisms of Taxol resistance related to microtubules. Oncogene, 22, 7280-95.

OTA, T., SUTO, S., KATAYAMA, H., HAN, Z. B., SUZUKI, F., MAEDA, M., TANINO, M., TERADA, Y. & TATSUKA, M. 2002. Increased mitotic phosphorylation of histone H3 attributable to AIM-1/Aurora-B overexpression contributes to chromosome number instability. Cancer Res, 62, 5168-77.

OUCHI, M., FUJIUCHI, N., SASAI, K., KATAYAMA, H., MINAMISHIMA, Y. A., ONGUSAHA, P. P., DENG, C., SEN, S., LEE, S. W. & OUCHI, T. 2004. BRCA1 phosphorylation by Aurora-A in the regulation of G2 to M transition. J Biol Chem, 279, 19643-8.

OVCARICEK, T., FRKOVIC, S. G., MATOS, E., MOZINA, B. & BORSTNAR, S. 2011. Triple negative breast cancer - prognostic factors and survival. Radiol Oncol, 45, 46-52.

PANG, B., QIAO, X., JANSSEN, L., VELDS, A., GROOTHUIS, T., KERKHOVEN, R., NIEUWLAND, M., OVAA, H., ROTTENBERG, S., VAN TELLINGEN, O., JANSSEN, J., HUIJGENS, P., ZWART, W. & NEEFJES, J. 2013. Drug-induced histone eviction from open chromatin contributes to the chemotherapeutic effects of doxorubicin. Nat Commun, 4, 1908.

PARDRIDGE, W. M., GOLDEN, P. L., KANG, Y. S. & BICKEL, U. 1997. Brain microvascular and astrocyte localization of P-glycoprotein. J Neurochem, 68, 1278-85.

PARE, R., YANG, T., SHIN, J. S. & LEE, C. S. 2013. The significance of the senescence pathway in breast cancer progression. J Clin Pathol, 66, 491-5.

PARK, J. E., SOUNG, N. K., JOHMURA, Y., KANG, Y. H., LIAO, C., LEE, K. H., PARK, C. H., NICKLAUS, M. C. & LEE, K. S. 2010. Polo-box domain: a versatile mediator of polo-like kinase function. Cell Mol Life Sci, 67, 1957-70.

PAUL, A., GUNEWARDENA, S., STECKLEIN, S. R., SAHA, B., PARELKAR, N., DANLEY, M., RAJENDRAN, G., HOME, P., RAY, S., JOKAR, I., VIELHAUER, G. A., JENSEN, R. A., TAWFIK, O. & PAUL, S. 2014. PKClambda/iota signaling promotes triple-negative breast cancer growth and metastasis. Cell Death Differ, 21, 1469-81.

PEDDI, P. F., ELLIS, M. J. & MA, C. 2012. Molecular basis of triple negative breast cancer and implications for therapy. Int J Breast Cancer, 2012, 217185.

PEROU, C. M., SORLIE, T., EISEN, M. B., VAN DE RIJN, M., JEFFREY, S. S., REES, C. A., POLLACK, J. R., ROSS, D. T., JOHNSEN, H., AKSLEN, L. A., FLUGE, O., PERGAMENSCHIKOV, A., WILLIAMS, C., ZHU, S. X., LONNING, P. E., BORRESEN-DALE, A. L., BROWN, P. O. & BOTSTEIN, D. 2000. Molecular portraits of human breast tumours. Nature, 406, 747-52.

Page 175: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

158

PIHAN, G. A., PUROHIT, A., WALLACE, J., KNECHT, H., WODA, B., QUESENBERRY, P. & DOXSEY, S. J. 1998. Centrosome defects and genetic instability in malignant tumors. Cancer Res, 58, 3974-85.

PIHAN, G. A., PUROHIT, A., WALLACE, J., MALHOTRA, R., LIOTTA, L. & DOXSEY, S. J. 2001. Centrosome defects can account for cellular and genetic changes that characterize prostate cancer progression. Cancer Res, 61, 2212-9.

PRAT, A. & PEROU, C. M. 2009. Mammary development meets cancer genomics. Nat Med, 15, 842-4.

QIU, Y., PU, T., LI, L., CHENG, F., LU, C., SUN, L., TENG, X., YE, F. & BU, H. 2014. The expression of aldehyde dehydrogenase family in breast cancer. J Breast Cancer, 17, 54-60.

RAO, S., HE, L., CHAKRAVARTY, S., OJIMA, I., ORR, G. A. & HORWITZ, S. B. 1999. Characterization of the Taxol binding site on the microtubule. Identification of Arg(282) in beta-tubulin as the site of photoincorporation of a 7-benzophenone analogue of Taxol. J Biol Chem, 274, 37990-4.

RAO, S., KRAUSS, N. E., HEERDING, J. M., SWINDELL, C. S., RINGEL, I., ORR, G. A. & HORWITZ, S. B. 1994. 3'-(p-azidobenzamido)taxol photolabels the N-terminal 31 amino acids of beta-tubulin. J Biol Chem, 269, 3132-4.

RAO, S., ORR, G. A., CHAUDHARY, A. G., KINGSTON, D. G. & HORWITZ, S. B. 1995. Characterization of the taxol binding site on the microtubule. 2-(m-Azidobenzoyl)taxol photolabels a peptide (amino acids 217-231) of beta-tubulin. J Biol Chem, 270, 20235-8.

REIS-FILHO, J. S., MILANEZI, F., CARVALHO, S., SIMPSON, P. T., STEELE, D., SAVAGE, K., LAMBROS, M. B., PEREIRA, E. M., NESLAND, J. M., LAKHANI, S. R. & SCHMITT, F. C. 2005. Metaplastic breast carcinomas exhibit EGFR, but not HER2, gene amplification and overexpression: immunohistochemical and chromogenic in situ hybridization analysis. Breast Cancer Res, 7, R1028-35.

REYLAND, M. E. 2007. Protein kinase Cdelta and apoptosis. Biochem Soc Trans, 35, 1001-4.

RIEDER, C. L. & MAIATO, H. 2004. Stuck in division or passing through: what happens when cells cannot satisfy the spindle assembly checkpoint. Dev Cell, 7, 637-51.

ROBERTSON, M. J., KAHL, B. S., VOSE, J. M., DE VOS, S., LAUGHLIN, M., FLYNN, P. J., ROWLAND, K., CRUZ, J. C., GOLDBERG, S. L., MUSIB, L., DARSTEIN, C., ENAS, N., KUTOK, J. L., ASTER, J. C., NEUBERG, D., SAVAGE, K. J., LACASCE, A., THORNTON, D., SLAPAK, C. A. & SHIPP, M. A. 2007. Phase II study of enzastaurin, a protein kinase C beta inhibitor, in patients with relapsed or refractory diffuse large B-cell lymphoma. J Clin Oncol, 25, 1741-6.

ROCCA, A., VIALE, G., GELBER, R. D., BOTTIGLIERI, L., GELBER, S., PRUNERI, G., GHISINI, R., BALDUZZI, A., PIETRI, E., D'ALESSANDRO, C., GOLDHIRSCH, A. & COLLEONI, M. 2008. Pathologic complete remission rate after cisplatin-based primary chemotherapy in breast cancer: correlation with p63 expression. Cancer Chemother Pharmacol, 61, 965-71.

ROGER, P., SAHLA, M. E., MAKELA, S., GUSTAFSSON, J. A., BALDET, P. & ROCHEFORT, H. 2001. Decreased expression of estrogen receptor beta protein in proliferative preinvasive mammary tumors. Cancer Res, 61, 2537-41.

ROMOND, E. H., PEREZ, E. A., BRYANT, J., SUMAN, V. J., GEYER, C. E., JR., DAVIDSON, N. E., TAN-CHIU, E., MARTINO, S., PAIK, S., KAUFMAN, P. A., SWAIN, S. M., PISANSKY, T. M., FEHRENBACHER, L., KUTTEH, L. A., VOGEL, V. G., VISSCHER, D. W., YOTHERS, G., JENKINS, R. B., BROWN, A. M., DAKHIL, S. R., MAMOUNAS, E. P., LINGLE, W. L., KLEIN, P. M., INGLE, J. N. & WOLMARK, N.

Page 176: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

159

2005. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med, 353, 1673-84.

RONINSON, I. B., BROUDE, E. V. & CHANG, B. D. 2001. If not apoptosis, then what? Treatment-induced senescence and mitotic catastrophe in tumor cells. Drug Resist Updat, 4, 303-13.

ROSSE, C., LINCH, M., KERMORGANT, S., CAMERON, A. J., BOECKELER, K. & PARKER, P. J. 2010. PKC and the control of localized signal dynamics. Nat Rev Mol Cell Biol, 11, 103-12.

ROUZIER, R., PEROU, C. M., SYMMANS, W. F., IBRAHIM, N., CRISTOFANILLI, M., ANDERSON, K., HESS, K. R., STEC, J., AYERS, M., WAGNER, P., MORANDI, P., FAN, C., RABIUL, I., ROSS, J. S., HORTOBAGYI, G. N. & PUSZTAI, L. 2005. Breast cancer molecular subtypes respond differently to preoperative chemotherapy. Clin Cancer Res, 11, 5678-85.

ROWINSKY, E. K., BURKE, P. J., KARP, J. E., TUCKER, R. W., ETTINGER, D. S. & DONEHOWER, R. C. 1989. Phase I and pharmacodynamic study of taxol in refractory acute leukemias. Cancer Res, 49, 4640-7.

RUDNER, A. D. & MURRAY, A. W. 1996. The spindle assembly checkpoint. Curr Opin Cell Biol, 8, 773-80.

RYAN, Q., IBRAHIM, A., COHEN, M. H., JOHNSON, J., KO, C. W., SRIDHARA, R., JUSTICE, R. & PAZDUR, R. 2008. FDA drug approval summary: lapatinib in combination with capecitabine for previously treated metastatic breast cancer that overexpresses HER-2. Oncologist, 13, 1114-9.

SAAB, R. 2010. Cellular senescence: many roads, one final destination. ScientificWorldJournal, 10, 727-41.

SAKAKURA, C., HAGIWARA, A., YASUOKA, R., FUJITA, Y., NAKANISHI, M., MASUDA, K., SHIMOMURA, K., NAKAMURA, Y., INAZAWA, J., ABE, T. & YAMAGISHI, H. 2001. Tumour-amplified kinase BTAK is amplified and overexpressed in gastric cancers with possible involvement in aneuploid formation. Br J Cancer, 84, 824-31.

SANTAGUIDA, S. & MUSACCHIO, A. 2009. The life and miracles of kinetochores. EMBO J, 28, 2511-31.

SASAKI, O., KIDO, K. & NAGAHAMA, S. 1999. DNA ploidy, Ki-67 and p53 as indicators of lymph node metastasis in early gastric carcinoma. Anal Quant Cytol Histol, 21, 85-8.

SCHIFF, P. B. & HORWITZ, S. B. 1980. Taxol stabilizes microtubules in mouse fibroblast cells. Proc Natl Acad Sci U S A, 77, 1561-5.

SEIDMAN, A. D. 1994. Single-agent use of Taxol (paclitaxel) in breast cancer. Ann Oncol, 5 Suppl 6, S17-22.

SEN, S., ZHOU, H. & WHITE, R. A. 1997. A putative serine/threonine kinase encoding gene BTAK on chromosome 20q13 is amplified and overexpressed in human breast cancer cell lines. Oncogene, 14, 2195-200.

SERVIDEI, T., RICCARDI, A., MOZZETTI, S., FERLINI, C. & RICCARDI, R. 2008. Chemoresistant tumor cell lines display altered epidermal growth factor receptor and HER3 signaling and enhanced sensitivity to gefitinib. Int J Cancer, 123, 2939-49.

SEVE, P. & DUMONTET, C. 2008. Is class III beta-tubulin a predictive factor in patients receiving tubulin-binding agents? Lancet Oncol, 9, 168-75.

SHANNON, K. B., CANMAN, J. C. & SALMON, E. D. 2002. Mad2 and BubR1 function in a single checkpoint pathway that responds to a loss of tension. Mol Biol Cell, 13, 3706-19.

Page 177: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

160

SHOEMAKER, A. R., OLEKSIJEW, A., BAUCH, J., BELLI, B. A., BORRE, T., BRUNCKO, M., DECKWIRTH, T., FROST, D. J., JARVIS, K., JOSEPH, M. K., MARSH, K., MCCLELLAN, W., NELLANS, H., NG, S., NIMMER, P., O'CONNOR, J. M., OLTERSDORF, T., QING, W., SHEN, W., STAVROPOULOS, J., TAHIR, S. K., WANG, B., WARNER, R., ZHANG, H., FESIK, S. W., ROSENBERG, S. H. & ELMORE, S. W. 2006. A small-molecule inhibitor of Bcl-XL potentiates the activity of cytotoxic drugs in vitro and in vivo. Cancer Res, 66, 8731-9.

SILVER, D. P., RICHARDSON, A. L., EKLUND, A. C., WANG, Z. C., SZALLASI, Z., LI, Q., JUUL, N., LEONG, C. O., CALOGRIAS, D., BURAIMOH, A., FATIMA, A., GELMAN, R. S., RYAN, P. D., TUNG, N. M., DE NICOLO, A., GANESAN, S., MIRON, A., COLIN, C., SGROI, D. C., ELLISEN, L. W., WINER, E. P. & GARBER, J. E. 2010. Efficacy of neoadjuvant Cisplatin in triple-negative breast cancer. J Clin Oncol, 28, 1145-53.

SINGH, S., CHITKARA, D., MEHRAZIN, R., BEHRMAN, S. W., WAKE, R. W. & MAHATO, R. I. 2012. Chemoresistance in prostate cancer cells is regulated by miRNAs and Hedgehog pathway. PLoS One, 7, e40021.

SINGH, S. K., CLARKE, I. D., TERASAKI, M., BONN, V. E., HAWKINS, C., SQUIRE, J. & DIRKS, P. B. 2003. Identification of a cancer stem cell in human brain tumors. Cancer Res, 63, 5821-8.

SIROHI, B., ARNEDOS, M., POPAT, S., ASHLEY, S., NERURKAR, A., WALSH, G., JOHNSTON, S. & SMITH, I. E. 2008. Platinum-based chemotherapy in triple-negative breast cancer. Ann Oncol, 19, 1847-52.

SLAMON, D. J., LEYLAND-JONES, B., SHAK, S., FUCHS, H., PATON, V., BAJAMONDE, A., FLEMING, T., EIERMANN, W., WOLTER, J., PEGRAM, M., BASELGA, J. & NORTON, L. 2001. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med, 344, 783-92.

SLEDGE, G. W., JR. & GOKMEN-POLAR, Y. 2006. Protein kinase C-beta as a therapeutic target in breast cancer. Semin Oncol, 33, S15-8.

SLEDGE, G. W., NEUBERG, D., BERNARDO, P., INGLE, J. N., MARTINO, S., ROWINSKY, E. K. & WOOD, W. C. 2003. Phase III trial of doxorubicin, paclitaxel, and the combination of doxorubicin and paclitaxel as front-line chemotherapy for metastatic breast cancer: an intergroup trial (E1193). J Clin Oncol, 21, 588-92.

SMOTER, M., BODNAR, L., DUCHNOWSKA, R., STEC, R., GRALA, B. & SZCZYLIK, C. 2011. The role of Tau protein in resistance to paclitaxel. Cancer Chemother Pharmacol, 68, 553-7.

SOTIRIOU, C. & PUSZTAI, L. 2009. Gene-expression signatures in breast cancer. N Engl J Med, 360, 790-800.

SPARANO, J. A., WANG, M., MARTINO, S., JONES, V., PEREZ, E. A., SAPHNER, T., WOLFF, A. C., SLEDGE, G. W., JR., WOOD, W. C. & DAVIDSON, N. E. 2008. Weekly paclitaxel in the adjuvant treatment of breast cancer. N Engl J Med, 358, 1663-71.

SPECTOR, N. L. & BLACKWELL, K. L. 2009. Understanding the mechanisms behind trastuzumab therapy for human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol, 27, 5838-47.

SREERAMA, L. & SLADEK, N. E. 1997. Cellular levels of class 1 and class 3 aldehyde dehydrogenases and certain other drug-metabolizing enzymes in human breast malignancies. Clin Cancer Res, 3, 1901-14.

STEEGMAIER, M., HOFFMANN, M., BAUM, A., LENART, P., PETRONCZKI, M., KRSSAK, M., GURTLER, U., GARIN-CHESA, P., LIEB, S., QUANT, J., GRAUERT, M.,

Page 178: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

161

ADOLF, G. R., KRAUT, N., PETERS, J. M. & RETTIG, W. J. 2007. BI 2536, a potent and selective inhibitor of polo-like kinase 1, inhibits tumor growth in vivo. Curr Biol, 17, 316-22.

STURGIS, C. D., CARAWAY, N. P., JOHNSTON, D. A., SHERMAN, S. I., KIDD, L. & KATZ, R. L. 1999. Image analysis of papillary thyroid carcinoma fine-needle aspirates: significant association between aneuploidy and death from disease. Cancer, 87, 155-60.

SUDO, T., NITTA, M., SAYA, H. & UENO, N. T. 2004. Dependence of paclitaxel sensitivity on a functional spindle assembly checkpoint. Cancer Res, 64, 2502-8.

SUGIYAMA, K., SUGIURA, K., HARA, T., SUGIMOTO, K., SHIMA, H., HONDA, K., FURUKAWA, K., YAMASHITA, S. & URANO, T. 2002. Aurora-B associated protein phosphatases as negative regulators of kinase activation. Oncogene, 21, 3103-11.

SULLIVAN, J. P., SPINOLA, M., DODGE, M., RASO, M. G., BEHRENS, C., GAO, B., SCHUSTER, K., SHAO, C., LARSEN, J. E., SULLIVAN, L. A., HONORIO, S., XIE, Y., SCAGLIONI, P. P., DIMAIO, J. M., GAZDAR, A. F., SHAY, J. W., WISTUBA, II & MINNA, J. D. 2010. Aldehyde dehydrogenase activity selects for lung adenocarcinoma stem cells dependent on notch signaling. Cancer Res, 70, 9937-48.

SUZUMA, K., TAKAHARA, N., SUZUMA, I., ISSHIKI, K., UEKI, K., LEITGES, M., AIELLO, L. P. & KING, G. L. 2002. Characterization of protein kinase C beta isoform's action on retinoblastoma protein phosphorylation, vascular endothelial growth factor-induced endothelial cell proliferation, and retinal neovascularization. Proc Natl Acad Sci U S A, 99, 721-6.

SYMONDS, J. M., OHM, A. M., CARTER, C. J., HEASLEY, L. E., BOYLE, T. A., FRANKLIN, W. A. & REYLAND, M. E. 2011. Protein kinase C delta is a downstream effector of oncogenic K-ras in lung tumors. Cancer Res, 71, 2087-97.

TAM, W. L., LU, H., BUIKHUISEN, J., SOH, B. S., LIM, E., REINHARDT, F., WU, Z. J., KRALL, J. A., BIERIE, B., GUO, W., CHEN, X., LIU, X. S., BROWN, M., LIM, B. & WEINBERG, R. A. 2013. Protein kinase C alpha is a central signaling node and therapeutic target for breast cancer stem cells. Cancer Cell, 24, 347-64.

TAN, J., ZHUANG, L., LEONG, H. S., IYER, N. G., LIU, E. T. & YU, Q. 2005. Pharmacologic modulation of glycogen synthase kinase-3beta promotes p53-dependent apoptosis through a direct Bax-mediated mitochondrial pathway in colorectal cancer cells. Cancer Res, 65, 9012-20.

TANAKA, T. U., RACHIDI, N., JANKE, C., PEREIRA, G., GALOVA, M., SCHIEBEL, E., STARK, M. J. & NASMYTH, K. 2002. Evidence that the Ipl1-Sli15 (Aurora kinase-INCENP) complex promotes chromosome bi-orientation by altering kinetochore-spindle pole connections. Cell, 108, 317-29.

TANG, Z., BHARADWAJ, R., LI, B. & YU, H. 2001. Mad2-Independent inhibition of APCCdc20 by the mitotic checkpoint protein BubR1. Dev Cell, 1, 227-37.

TAO, W., SOUTH, V. J., ZHANG, Y., DAVIDE, J. P., FARRELL, L., KOHL, N. E., SEPP-LORENZINO, L. & LOBELL, R. B. 2005. Induction of apoptosis by an inhibitor of the mitotic kinesin KSP requires both activation of the spindle assembly checkpoint and mitotic slippage. Cancer Cell, 8, 49-59.

TATSUKA, M., KATAYAMA, H., OTA, T., TANAKA, T., ODASHIMA, S., SUZUKI, F. & TERADA, Y. 1998. Multinuclearity and increased ploidy caused by overexpression of the aurora- and Ipl1-like midbody-associated protein mitotic kinase in human cancer cells. Cancer Res, 58, 4811-6.

Page 179: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

162

TEWEY, K. M., ROWE, T. C., YANG, L., HALLIGAN, B. D. & LIU, L. F. 1984. Adriamycin-induced DNA damage mediated by mammalian DNA topoisomerase II. Science, 226, 466-8.

THIKE, A. A., CHEOK, P. Y., JARA-LAZARO, A. R., TAN, B., TAN, P. & TAN, P. H. 2010. Triple-negative breast cancer: clinicopathological characteristics and relationship with basal-like breast cancer. Mod Pathol, 23, 123-33.

THOMAS, S., SHARMA, N., GOLDEN, E. B., CHO, H., AGARWAL, P., GAFFNEY, K. J., PETASIS, N. A., CHEN, T. C., HOFMAN, F. M., LOUIE, S. G. & SCHONTHAL, A. H. 2012. Preferential killing of triple-negative breast cancer cells in vitro and in vivo when pharmacological aggravators of endoplasmic reticulum stress are combined with autophagy inhibitors. Cancer Lett, 325, 63-71.

THOMPSON, L. J. & FIELDS, A. P. 1996. betaII protein kinase C is required for the G2/M phase transition of cell cycle. J Biol Chem, 271, 15045-53.

TISCHKOWITZ, M., BRUNET, J. S., BEGIN, L. R., HUNTSMAN, D. G., CHEANG, M. C., AKSLEN, L. A., NIELSEN, T. O. & FOULKES, W. D. 2007. Use of immunohistochemical markers can refine prognosis in triple negative breast cancer. BMC Cancer, 7, 134.

TURNER, N., LAMBROS, M. B., HORLINGS, H. M., PEARSON, A., SHARPE, R., NATRAJAN, R., GEYER, F. C., VAN KOUWENHOVE, M., KREIKE, B., MACKAY, A., ASHWORTH, A., VAN DE VIJVER, M. J. & REIS-FILHO, J. S. 2010. Integrative molecular profiling of triple negative breast cancers identifies amplicon drivers and potential therapeutic targets. Oncogene, 29, 2013-23.

TURNER, N., TUTT, A. & ASHWORTH, A. 2004. Hallmarks of 'BRCAness' in sporadic cancers. Nat Rev Cancer, 4, 814-9.

TUTT, A. N., LORD, C. J., MCCABE, N., FARMER, H., TURNER, N., MARTIN, N. M., JACKSON, S. P., SMITH, G. C. & ASHWORTH, A. 2005. Exploiting the DNA repair defect in BRCA mutant cells in the design of new therapeutic strategies for cancer. Cold Spring Harb Symp Quant Biol, 70, 139-48.

UCKUN, F. M., ZHENG, Y., CETKOVIC-CVRLJE, M., VASSILEV, A., LISOWSKI, E., WAURZYNIAK, B., CHEN, H., CARPENTER, R. & CHEN, C. L. 2002. In vivo pharmacokinetic features, toxicity profile, and chemosensitizing activity of alpha-cyano-beta-hydroxy-beta- methyl-N-(2,5-dibromophenyl)propenamide (LFM-A13), a novel antileukemic agent targeting Bruton's tyrosine kinase. Clin Cancer Res, 8, 1224-33.

UEDA, T., BRENNER, S., MALECH, H. L., LANGEMEIJER, S. M., PERL, S., KIRBY, M., PHANG, O. A., KROUSE, A. E., DONAHUE, R. E., KANG, E. M. & TISDALE, J. F. 2005. Cloning and functional analysis of the rhesus macaque ABCG2 gene. Forced expression confers an SP phenotype among hematopoietic stem cell progeny in vivo. J Biol Chem, 280, 991-8.

VAN BEERS, E. H., VAN WELSEM, T., WESSELS, L. F., LI, Y., OLDENBURG, R. A., DEVILEE, P., CORNELISSE, C. J., VERHOEF, S., HOGERVORST, F. B., VAN'T VEER, L. J. & NEDERLOF, P. M. 2005. Comparative genomic hybridization profiles in human BRCA1 and BRCA2 breast tumors highlight differential sets of genomic aberrations. Cancer Res, 65, 822-7.

VARNA, M., LEHMANN-CHE, J., TURPIN, E., MARANGONI, E., EL-BOUCHTAOUI, M., JEANNE, M., GRIGORIU, C., RATAJCZAK, P., LEBOEUF, C., PLASSA, L. F., FERREIRA, I., POUPON, M. F., JANIN, A., DE THE, H. & BERTHEAU, P. 2009. p53 dependent cell-cycle arrest triggered by chemotherapy in xenografted breast tumors. Int J Cancer, 124, 991-7.

VERGOTE, I. B., CHEKEROV, R., AMANT, F., HARTER, P., CASADO, A., EMERICH, J., BAUKNECHT, T., MANSOURI, K., MYRAND, S. P., NGUYEN, T. S., SHI, P. &

Page 180: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

163

SEHOULI, J. 2013. Randomized, phase II, placebo-controlled, double-blind study with and without enzastaurin in combination with paclitaxel and carboplatin as first-line treatment followed by maintenance treatment in advanced ovarian cancer. J Clin Oncol, 31, 3127-32.

VITALE, I., GALLUZZI, L., CASTEDO, M. & KROEMER, G. 2011. Mitotic catastrophe: a mechanism for avoiding genomic instability. Nat Rev Mol Cell Biol, 12, 385-92.

WAHL, A. F., DONALDSON, K. L., FAIRCHILD, C., LEE, F. Y., FOSTER, S. A., DEMERS, G. W. & GALLOWAY, D. A. 1996. Loss of normal p53 function confers sensitization to Taxol by increasing G2/M arrest and apoptosis. Nat Med, 2, 72-9.

WALDMAN, T., LENGAUER, C., KINZLER, K. W. & VOGELSTEIN, B. 1996. Uncoupling of S phase and mitosis induced by anticancer agents in cells lacking p21. Nature, 381, 713-6.

WALLACE, J. A., PITARRESI, J. R., SHARMA, N., PALETTAS, M., CUITINO, M. C., SIZEMORE, S. T., YU, L., SANDERLIN, A., ROSOL, T. J., MEHTA, K. D., SIZEMORE, G. M. & OSTROWSKI, M. C. 2014. Protein kinase C Beta in the tumor microenvironment promotes mammary tumorigenesis. Front Oncol, 4, 87.

WANG, T. H., CHAN, Y. H., CHEN, C. W., KUNG, W. H., LEE, Y. S., WANG, S. T., CHANG, T. C. & WANG, H. S. 2006. Paclitaxel (Taxol) upregulates expression of functional interleukin-6 in human ovarian cancer cells through multiple signaling pathways. Oncogene, 25, 4857-66.

WANG, T. H., POPP, D. M., WANG, H. S., SAITOH, M., MURAL, J. G., HENLEY, D. C., ICHIJO, H. & WIMALASENA, J. 1999. Microtubule dysfunction induced by paclitaxel initiates apoptosis through both c-Jun N-terminal kinase (JNK)-dependent and -independent pathways in ovarian cancer cells. J Biol Chem, 274, 8208-16.

WARNER, S. L., BEARSS, D. J., HAN, H. & VON HOFF, D. D. 2003. Targeting Aurora-2 kinase in cancer. Mol Cancer Ther, 2, 589-95.

WATERS, J. C., CHEN, R. H., MURRAY, A. W. & SALMON, E. D. 1998. Localization of Mad2 to kinetochores depends on microtubule attachment, not tension. J Cell Biol, 141, 1181-91.

WEIGELT, B., BAEHNER, F. L. & REIS-FILHO, J. S. 2010. The contribution of gene expression profiling to breast cancer classification, prognostication and prediction: a retrospective of the last decade. J Pathol, 220, 263-80.

WEISS, R. B. 1992. The anthracyclines: will we ever find a better doxorubicin? Semin Oncol, 19, 670-86.

WERTZ, I. E., KUSAM, S., LAM, C., OKAMOTO, T., SANDOVAL, W., ANDERSON, D. J., HELGASON, E., ERNST, J. A., EBY, M., LIU, J., BELMONT, L. D., KAMINKER, J. S., O'ROURKE, K. M., PUJARA, K., KOHLI, P. B., JOHNSON, A. R., CHIU, M. L., LILL, J. R., JACKSON, P. K., FAIRBROTHER, W. J., SESHAGIRI, S., LUDLAM, M. J., LEONG, K. G., DUEBER, E. C., MAECKER, H., HUANG, D. C. & DIXIT, V. M. 2011. Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7. Nature, 471, 110-4.

WILSON, T. R., LONGLEY, D. B. & JOHNSTON, P. G. 2006. Chemoresistance in solid tumours. Ann Oncol, 17 Suppl 10, x315-24.

WOOD, K. W., CHUA, P., SUTTON, D. & JACKSON, J. R. 2008. Centromere-associated protein E: a motor that puts the brakes on the mitotic checkpoint. Clin Cancer Res, 14, 7588-92.

Page 181: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

164

WU, B., ZHOU, H., HU, L., MU, Y. & WU, Y. 2013. Involvement of PKCalpha activation in TF/VIIa/PAR2-induced proliferation, migration, and survival of colon cancer cell SW620. Tumour Biol, 34, 837-46.

YADAV, B. S., SHARMA, S. C., CHANANA, P. & JHAMB, S. 2014. Systemic treatment strategies for triple-negative breast cancer. World J Clin Oncol, 5, 125-33.

YAMAUCHI, C., FUJII, S., KIMURA, T., KUWATA, T., WADA, N., MUKAI, H., MATSUMOTO, N., FUKAYAMA, M. & OCHIAI, A. 2011. E-cadherin expression on human carcinoma cell affects trastuzumab-mediated antibody-dependent cellular cytotoxicity through killer cell lectin-like receptor G1 on natural killer cells. Int J Cancer, 128, 2125-37.

YANG, H., OU, C. C., FELDMAN, R. I., NICOSIA, S. V., KRUK, P. A. & CHENG, J. Q. 2004. Aurora-A kinase regulates telomerase activity through c-Myc in human ovarian and breast epithelial cells. Cancer Res, 64, 463-7.

YANG, J. M., SULLIVAN, G. F. & HAIT, W. N. 1997. Regulation of the function of P-glycoprotein by epidermal growth factor through phospholipase C. Biochem Pharmacol, 53, 1597-604.

YANG, Z. F., HO, D. W., NG, M. N., LAU, C. K., YU, W. C., NGAI, P., CHU, P. W., LAM, C. T., POON, R. T. & FAN, S. T. 2008. Significance of CD90+ cancer stem cells in human liver cancer. Cancer Cell, 13, 153-66.

YARDEN, Y. & SLIWKOWSKI, M. X. 2001. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol, 2, 127-37.

YIP, N. C., FOMBON, I. S., LIU, P., BROWN, S., KANNAPPAN, V., ARMESILLA, A. L., XU, B., CASSIDY, J., DARLING, J. L. & WANG, W. 2011. Disulfiram modulated ROS-MAPK and NFkappaB pathways and targeted breast cancer cells with cancer stem cell-like properties. Br J Cancer, 104, 1564-74.

YOULE, R. J. & STRASSER, A. 2008. The BCL-2 protein family: opposing activities that mediate cell death. Nat Rev Mol Cell Biol, 9, 47-59.

YU, F. X. & GUAN, K. L. 2013. The Hippo pathway: regulators and regulations. Genes Dev, 27, 355-71.

YU, Y., XIAO, C. H., TAN, L. D., WANG, Q. S., LI, X. Q. & FENG, Y. M. 2014. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of breast cancer cells through paracrine TGF-beta signalling. Br J Cancer, 110, 724-32.

ZHANG, J., ANASTASIADIS, P. Z., LIU, Y., THOMPSON, E. A. & FIELDS, A. P. 2004. Protein kinase C (PKC) betaII induces cell invasion through a Ras/Mek-, PKC iota/Rac 1-dependent signaling pathway. J Biol Chem, 279, 22118-23.

ZHANG, L. L., CAO, F. F., WANG, Y., MENG, F. L., ZHANG, Y., ZHONG, D. S. & ZHOU, Q. H. 2015. The protein kinase C (PKC) inhibitors combined with chemotherapy in the treatment of advanced non-small cell lung cancer: meta-analysis of randomized controlled trials. Clin Transl Oncol, 17, 371-7.

ZHAO, C., DAHLMAN-WRIGHT, K. & GUSTAFSSON, J. A. 2008. Estrogen receptor beta: an overview and update. Nucl Recept Signal, 6, e003.

ZHAO, H., YANG, M., ZHAO, J., WANG, J., ZHANG, Y. & ZHANG, Q. 2013. High expression of LC3B is associated with progression and poor outcome in triple-negative breast cancer. Med Oncol, 30, 475.

ZHONG, Q., GAO, W., DU, F. & WANG, X. 2005. Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis. Cell, 121, 1085-95.

ZHOU, M., LIU, Z., ZHAO, Y., DING, Y., LIU, H., XI, Y., XIONG, W., LI, G., LU, J., FODSTAD, O., RIKER, A. I. & TAN, M. 2010. MicroRNA-125b confers the resistance of breast cancer cells to paclitaxel through suppression of pro-

Page 182: IDENTIFICATION OF NOVEL ONCOGENIC PATHWAYS IN ...Figure 3.3.2 Top hit of the kinase inhibitor screen Figure 3.4.1 Validation of the top 9 hits identified by the kinase screen Figure

165

apoptotic Bcl-2 antagonist killer 1 (Bak1) expression. J Biol Chem, 285, 21496-507.