You are on page 1of 304

University of Bradford eThesis

This thesis is hosted in Bradford Scholars – The University of Bradford Open Access
repository. Visit the repository for full metadata or to contact the repository team

© University of Bradford. This work is licenced for reuse under a Creative Commons
Licence.
AN IMMUNOHISTOPATHOLOGICAL AND FUNCTIONAL INVESTIGATION

OF β3 INTEGRIN ANTAGONISM AS A THERAPEUTIC STRATEGY IN

CANCER

F O F ALSHAMMARI

PhD

2013

I
AN IMMUNOHISTOPATHOLOGICAL AND FUNCTIONAL INVESTIGATION

OF β3 INTEGRIN ANTAGONISM AS A THERAPEUTIC STRATEGY IN

CANCER

Characterisation, development, and utilisation of preclinical cancer

models to investigate novel β3 integrin anatgonists

FATEMAH O F O ALSHAMMARI

Submitted for the degree of Doctor of Philosophy

Institute of Cancer Therapeutics

University of Bradford

2013

II
FATEMAH O F O ALSHAMMARI
Title: An immunohistopathological and functional investigation of β3
integrin antagonism as a therapeutic strategy in cancer

Key words: integrins, αIIbβ3, αVβ3, integrin antagonists, tumour cell migration,
histopathology of integrin expression, cancer therapy.

Abstract
Tumour cell dissemination is a major issue with the treatment of cancer, thus
new therapeutic strategies which can control this process are needed.
Antagonism of integrins highly expressed in tumours is one potential
strategy.
The integrins are transmembrane glycoprotein adhesive receptors. Two of
the integrins, αVβ3 and αIIbβ3, are highly expressed in a number of tumours
and induce bi-directional signalling through their interaction with extracellular
matrix proteins, and growth factor receptors. Through this signalling they play
an important role in a number of cellular processes that are involved in
tumour dissemination such as tumour growth, migration, invasion, metastasis
and angiogenesis. Dual αIIbβ3 and αVβ3 integrin antagonism will have a direct
effect on β3-expressing tumour cells that leads to the inhibition of cell
migration and dissemination. Furthermore, through targeting tumour cell
interaction with endothelial cells and platelets, this will also lead to inhibition
of angiogenesis and metastasis.
The aim of this project was to characterise the expression of αVβ3 and αIIbβ3
integrin in a panel of tumour cell lines and in human tumour xenograft
samples, and to develop and utilise cell-based models to investigate potential
novel β3 antagonists.
The expression of αV and β3 subunits was detected in xenograft tissue using
immunoblotting techniques. A panel of cell lines of different tumour types
including melanoma, prostate, breast, colon and non small cell lung
carcinoma was then characterised for αVβ3 and αIIbβ3 integrin expression
using immunoblotting and immunocytochemistry. Melanoma cell lines
demonstrated the strongest αVβ3 expression. No αIIbβ3 integrin expression
was seen in any of the cell lines evaluated. A selection of cell lines with
varying αVβ3 expression were then used to develop a functional test for cell
migration, the scratch wound healing assay. Migration of tumour cells that
expressed αVβ3 integrin was inhibited by the known β3 antagonists, cRGDfV
peptide and LM609 antibody. A panel of 12 potential novel β3 integrin
antagonists was screened for cytotoxicity and activity in the validated scratch
assay. ICT9055 was the most effective antagonist in inhibition of M14 cell
migration as determined by the scratch assay, with an IC50 of < 0.1 µM.
Therefore the work presented in this thesis has established models and tools
for evaluating potential novel β3 integrin antagonists, and identified a
promising molecule to progress for further preclinical evaluation.
i
Contents

Abstract ......................................................................................................... i

Contents ....................................................................................................... ii

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

List of Tables ............................................................................................... xi

Acknowledgement ..................................................................................... xii

Abbreviations ............................................................................................ xiii

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

1.1 Cancer background........................................................................... 1


1.1.1 Definition and aetiology of cancer ................................................ 1
1.1.2 Incidence of cancer ...................................................................... 2
1.1.3 Treatment of cancer ..................................................................... 3
1.1.3.1 Surgery ..................................................................................... 3
1.1.3.2 Radiotherapy ............................................................................ 3
1.1.3.3 Biological therapy ..................................................................... 4
1.1.1.1.1 Endocrine therapy .............................................................. 4
1.1.3.3.2 Gene therapy ..................................................................... 4
1.1.3.3.3 Antibodies .......................................................................... 5
1.1.3.4 Chemotherapy .......................................................................... 6
1.1.3.4.1 Conventional chemotherapy .............................................. 6
1.1.3.5 New strategies for cancer therapy ............................................ 7
1.1.4 The general pathobiology of cancer ............................................. 9

1.2 The Integrins.................................................................................... 15


1.2.1 Introduction ................................................................................ 15
1.2.2 Structure of integrins .................................................................. 15
1.2.3 Types of integrins ....................................................................... 19
1.2.4 Function of integrins ................................................................... 24
1.2.4.1 Function of αVβ3 integrin ......................................................... 27
1.2.4.1.1 Role of αvβ3 integrin in angiogenesis .............................. 28
1.2.4.2 Function of αIIbβ3 integrin ........................................................ 30
1.2.5 Role of αVβ3 and αIIbβ3 integrins in cancer development ............ 31
1.2.5.1 Role of αVβ3 integrin in cancer development .......................... 31
1.2.5.1.1 Role of αVβ3 integrin in breast cancer .............................. 33
1.2.5.1.2 Role of αVβ3 integrin in prostate cancer ........................... 35
1.2.5.1.3 Role of αVβ3 integrin in melanoma ................................... 36
1.2.5.1.4 αVβ3 integrin in glioma ...................................................... 38

ii
1.2.5.2 Role of αIIbβ3 integrin in cancer development ......................... 38
1.2.5.2.1 Role of αIIbβ3 integrin in melanoma .................................. 38
1.2.5.2.2 Role of αIIbβ3 integrin in prostate cancer .......................... 40
1.2.5.3 αVβ3 and αIIbβ3 integrin antagonism in cancer therapeutics .... 41
1.2.5.3.1 Anti-αVβ3 antibodies as cancer therapeutics .................... 41
1.2.5.3.2 Integrin binding peptides as cancer therapeutics ............. 42
1.2.5.3.3 Small molecule integrin antagonists as cancer therapeutics
………………………………………………………………….43
1.3.5.1.1 Dual αVβ3/αVβ5 integrin antagonism in cancer therapy ..... 45
1.2.5.3.4.1 Cilengitide, a dual αVβ3/αVβ5 integrin antagonist, in
cancer therapy………………………………………………………….47
1.2.5.3.5 Dual αVβ3/ αIIbβ3 integrin antagonism ............................... 49

1.3 Aims and objectives........................................................................ 57

2 Chapter 2: Characterisation of integrin αIIbβ3 and αVβ3 expression in


human xenograft tissue in mice using immunohistochemical and
immunoblotting techniques ...................................................................... 59

2.1 Introduction ..................................................................................... 60


2.1.1 Aims and objectives ................................................................... 63

2.2 Materials and methods ................................................................... 64


2.2.1 Materials .................................................................................... 64
2.2.2 Cell lines .................................................................................... 64
2.2.3 Methods ..................................................................................... 66
2.2.3.1 Collection of xenograft materials ............................................ 66
2.2.3.2 Slide coating ........................................................................... 66
2.2.3.3 Tissue fixation, processing, embedding and paraffin sectioning
………………………………………………………………………66
2.2.3.4 Haematoxylin and Eosin staining............................................ 68
2.2.3.5 Immunohistochemistry............................................................ 69
2.2.3.5.1 Immunodetection of αIIb and αVβ3 integrins expression in
FFPE human tumour xenograft sections ........................................... 69
2.2.3.5.1.1 Sample preparation ................................................... 69
2.2.3.5.1.2 Antigen retrieval......................................................... 69

A. Citrate buffer antigen retrieval ............................................................. 69

B. Pre-warmed pepsin digestion method antigen retrieval .................... 70

C. Proteinase K method antigen retrieval ................................................ 70


2.2.3.5.1.3 Antibody incubation ................................................... 70
2.2.3.5.2 Immunodetection of αIIbβ3 and αVβ3 integrins expression in
frozen human xenograft tumour ........................................................ 71
2.2.3.5.2.1 Sample preparation ................................................... 71

iii
2.2.3.5.2.2 Labelling procedure ................................................... 72
2.2.3.5.3 Immunodetection of β3 subunits and αVβ3 integrin
expression in FFPE and frozen human tumour xenograft sections by
using a mouse on mouse (M.O.M.) kit .............................................. 72
2.2.3.5.3.1 Sample preparation ................................................... 73
2.2.3.5.3.2 Preparation of M.O.M. kit working solutions .............. 73
2.2.3.5.3.3 Antibody incubations ................................................. 73
2.2.3.6 Immunoblotting (Western Blotting) ......................................... 74
2.2.3.6.1 Materials .......................................................................... 74
2.2.3.6.2 Tissue homogenisation .................................................... 74
2.2.3.6.2.1 Bradford assay .......................................................... 75
2.2.3.6.2.2 SDS-Polyacrylamide Gel Electrophoresis ................. 76
2.2.3.6.2.3 Western Blot for αV and β3 Integrins .......................... 77
2.2.3.6.2.4 Ponceau S Stain ........................................................ 77
2.2.3.6.2.5 Coomassie Brilliant Blue Staining .............................. 77
2.2.3.6.2.6 Immunoblotting of nitrocellulose membrane with anti-β3
and anti-αV integrin subunits .......................................................... 78

2.3 Results ............................................................................................. 80


2.3.1 Detection of the expression of αIIbβ3 and αVβ3 integrin in human
tumour xenograft tissue by IHC ................................................................ 80
2.3.1.1 Tumour xenograft ................................................................... 80
2.3.1.2 Studies on FFPE tissue samples ............................................ 81
2.3.1.3 IHC studies on Frozen Sections. ............................................ 84
2.3.2 Immunodetection of β3 and αVβ3 integrin subunits using an
M.O.M. kit. ………………………………………………………………………87
2.3.2.1 M.O.M. Immunodetection of β3 and αVβ3 integrins in FFPE
sections. ………………………………………………………………………87
2.3.2.2 M.O.M. Immunodetection of β3 and αVβ3 integrins in frozen
sections. ………………………………………………………………………92
2.3.3 Detection of the expression of αIIb, αV, and β3 subunits in
homogenised tumour xenograft mouse tissue by immunoblotting ............ 95
2.3.3.1 Expression of αV and β3 integrin in homogenised human
xenograft mouse tissue compared to negative control tissue and human
tumour cells .......................................................................................... 98

2.4 Discussion ..................................................................................... 100

2.5 Conclusion..................................................................................... 107

3 Chapter 3: Characterisation of integrin αIIbβ3 and αVβ3 expression in


a panel of human tumour cell lines ........................................................ 108

3.1 Introduction ................................................................................... 109


3.1.1 Aims and objectives ................................................................. 110

3.2 Materials and Methods.................................................................. 112


3.2.1 Materials .................................................................................. 112
iv
3.2.2 Methods ................................................................................... 112
3.2.2.1 Cell maintenance .................................................................. 112
3.2.2.2 Tumour cell line growth kinetics............................................ 115
3.2.2.3 Immunoblotting ..................................................................... 115
3.2.2.3.1 Cell Lysis........................................................................ 116
3.2.2.3.2 IMB of nitrocellulose membrane with different anti αV, αIIb
and β3 integrin antibodies ................................................................ 116
3.2.2.4 Immunocytochemistry (ICC) ................................................. 118
3.2.2.4.1 Detection of αV, αIIb and β3 subunits, and αVβ3 integrin
expression in different tumour cell lines by ICC .............................. 118
3.2.2.4.1.1 Materials .................................................................. 118
3.2.2.4.1.2 Cell preparation ....................................................... 118
3.2.2.4.1.3 Confocal microscopy ............................................... 119
3.2.2.4.1.4 ICC analysis ............................................................ 119

3.3 Results ........................................................................................... 121


3.3.1 Characterisation of cellular growth kinetics .............................. 121
3.3.2 Detection of αV, αIIb, and β3 integrins using IMB ....................... 123
3.3.2.1 Optimisation of IMB .............................................................. 123
3.3.2.2 Screening different tumour cell lines for αV and β3 integrin
expression by IMB technique using the optimised conditions for anti-αV,
Q20, and anti-β3, B7, antibodies ......................................................... 126
3.3.3 Detection of αV, αIIb, β3 and αVβ3 integrins using ICC ............... 129
3.3.3.1 Optimisation of ICC .............................................................. 129
3.3.3.2 Evaluation of the effect of method of cell harvesting on integrin
expression .......................................................................................... 132
3.3.3.3 Screening different cell lines for expression of α V, β3, αIIb and
αVβ3 integrins by ICC technique. ......................................................... 133
3.3.3.4 Analysis of the expression of αV, αIIb, β3 and αVβ3 integrin in
different human tumour cell lines detected by ICC. ............................ 136
3.3.3.5 Confirmation of sub-cellular immunolocalisation of αIIb, β3 and
αVβ3 integrin using confocal microscopy. ............................................ 137
3.3.4 Comparison of result of IMB and ICC....................................... 139

3.4 Discussion ..................................................................................... 140

3.5 Conclusion..................................................................................... 148

4 Chapter 4: Investigation of the effect of αVβ3 integrin inhibition on


tumour cell migration .............................................................................. 149

4.1 Introduction ................................................................................... 150


4.1.1 Aims and objectives ................................................................. 152

4.2 Materials and methods ................................................................. 154


4.2.1 Materials .................................................................................. 154
4.2.2 Methods ................................................................................... 154
4.2.2.1 Wound healing migration assay............................................ 154
v
4.2.2.1.1 Characterisation of the phenotype of the migrated cells in
terms of proliferation and integrin expression .................................. 156
4.2.2.2 Evaluation of the cytotoxicity of cRGDfV and LM609 using the
MTT assay .......................................................................................... 157
4.2.2.3 Validation of the scratch assay using the integrin antagonists
cRGDfV and LM609 ............................................................................ 158
4.2.2.4 Statistical analysis ................................................................ 160

4.3 Results ........................................................................................... 161


4.3.1 Development of the wound healing migration assay ................ 161
4.3.1.1 Seeding density .................................................................... 161
4.3.1.2 Healing time ......................................................................... 163
4.3.2 Evaluation of the cytotoxicity of cRGDfV and LM609 using the
MTT assay.............................................................................................. 167
4.3.3 The effect of cRGDfV on the migration of human tumour cell lines
………………………………………………………………………..168
4.3.4 The effect of LM609 on human tumour cell migration .............. 171

4.4 Discussion ..................................................................................... 173

4.5 Conclusion..................................................................................... 177

5 Chapter 5: Investigation of the cytotoxicity of potential novel β3


integrin antagonists and their effect on tumour cell migration ........... 178

5.1 Introduction ................................................................................... 179


5.1.1 Aims and objectives ................................................................. 180

5.2 Materials and methods ................................................................. 181


5.2.1 Materials .................................................................................. 181

Table 5.1 Compounds used .................................................................... 181


5.2.2 Methods ................................................................................... 182
5.2.2.1 Evaluation of the cytotoxicity of potential novel β3 integrin
antagonists using the MTT assay ....................................................... 182
5.2.2.2 Wound healing migration assay............................................ 182
5.2.2.3 Statistical analysis ................................................................ 182

5.3 Results ........................................................................................... 183


5.3.1 Evaluation of the cytotoxicity of potential novel β 3 integrin
antagonists ............................................................................................. 183
5.3.2 Characterisation of the anti-migratory effect of potential novel β3
integrin antagonists using the scratch assay .......................................... 185

5.4 Discussion ..................................................................................... 192

5.5 Conclusion..................................................................................... 194

vi
6 Chapter 6: Discussion and future work .......................................... 195

6.1 General discussion and future perspective ................................ 196

6.2 Conclusion..................................................................................... 206

7 Chapter 7: References and Appendixes ......................................... 207

7.1 References ..................................................................................... 208

7.2 Appendices .................................................................................... 280


7.2.1 Appendix 1 ............................................................................... 280
7.2.2 Appendix 2 ............................................................................... 281

vii
List of Figures
Figure 1.1 The basic steps of the cancer metastatic pathway (Bellahcene et
al. 2008) ....................................................................................................... 11
Figure 1.2 Role of integrins in cancer development ..................................... 14
Figure 1.3 The structure of αIIbβ3 integrin in its active and inactive state ..... 18
Figure 1.4 The integrin family....................................................................... 19
Figure 1.5 inside-out-signalling pathways for integrins (Askari et al. 2009) . 26
Figure 1.6 Interaction of active integrin with signalling molecules regulates
cellular function (Hynes 2002)...................................................................... 27
Figure 1.7 Interaction of αVβ3 integrin and VEGFR-2 (Somanath et al. 2009).
..................................................................................................................... 30
Figure 1.8 Active αIIbβ3 integrin in platelet activation and thrombus formation
(Millard, Odde & Neamati 2011)................................................................... 31
Figure 1.9 Anti-integrin antagonists ............................................................. 52
Figure 2.1 Representative calibration curve using Bradford assay. ............. 76
Figure 2.2 Confirmation of successful protein transfer ................................. 78
Figure 2.3 Haematoxylin and Eosin stain in M14 human melanoma xenograft
paraffin embedded tissues ........................................................................... 80
Figure 2.4 Optimisation of IHC to detect αIIbβ3 and αVβ3 integrin expression in
FFPE human xenografts, using different methods ....................................... 82
Figure 2.5 Optimisation of IHC to detect αV and β3 integrin expression in
FFPE M14 human melanoma xenografts using Q20 and B7 ....................... 83
Figure 2.6 Optimisation of the protocol used to characterise expression of
αIIbβ3 and αVβ3 integrin in frozen human xenograft mouse tissue by IHC ..... 85
Figure 2.7 IHC of αV integrin expression in human xenograft mouse frozen
tissue using Q20 .......................................................................................... 86
Figure 2.8 IHC detection of αVβ3 and β3 integrin in human FFPE tumours with
an M.O.M. kit................................................................................................ 89
Figure 2.9 IHC detection of β3 integrin expression using B7 with M.O.M. kit in
FFPE M14 melanoma human tumour xenografts in mouse ......................... 90
Figure 2.10 IHC detection of β3 integrin expression in FFPE human tumour
xenografts in mouse using B7 with M.O.M. kit ............................................. 91
Figure 2.11: IHC optimisation for αVβ3 and β3 integrin expression in frozen
sections using the M.O.M. detection kit. ...................................................... 93
Figure 2.12 Expression of β3 integrin in human tumour xenograft frozen
tissues immunolabelled with BV4 (anti-β3) using M.O.M. detection kit ......... 94

viii
Figure 2.13 Optimisation steps for extraction of protein from human xenograft
mouse tissue. ............................................................................................... 96
Figure 2.14 Expression of β3 subunit in M14 human xenograft mouse tissue
using B7 by IMB ........................................................................................... 97
Figure 2.15 Expression of αV and β3 integrin in homogenised tissue ........... 99
Figure 3.1 Growth parameters of human tumour cell lines ........................ 122
Figure 3.2 Optimisation of IMB protocol. .................................................... 124
Figure 3.3 Expression of αV and β3 integrin in a panel of human tumour cell
lines from a supernatant ............................................................................ 127
Figure 3.4 Expression of αV and β3 integrin protein in a panel of human
tumour cell lines from a pellet resuspended in lysis buffer ......................... 128
Figure 3.5 Detection of αV, αIIb, β3 and αVβ3 integrin in PC-3 using ICC. .... 130
Figure 3.6 Comparison of cell harvesting techniques ................................ 132
Figure 3.7 Screening of the first part of the cell line panel with anti-αV (Q20),
anti-αIIb (C20), anti-β3 (B7), and anti-αVβ3 (LM609) using ICC. ............... 134
Figure 3.8 Screening of the second part of the cell line panel with anti-αV
(Q20), anti-αIIb (C20), anti-β3 (B7), and anti-αVβ3 (LM609) using ICC. ....... 135
Figure 3.9 The expression of αIIb, β3, and αVβ3 integrin in human tumour cell
lines by confocal microscopy. .................................................................... 138
Figure 4.1 Wound healing assay steps (Hulkower 2011) ........................... 156
Figure 4.2 Scratch assay analysis ............................................................. 159
Figure 4.3 Formation of confluent monolayers for the cell line panel (cell
density per ml and time in hours) ............................................................... 162
Figure 4.4 Migration of different tumour cell lines as assessed by the scratch
assay ......................................................................................................... 164
Figure 4.5 Labelling M14 cells at initial wound perimeter (T=0) and after 24
hours of wound healing with LM609 and Ki-67 using ICC.......................... 166
Figure 4.6 Evaluation of cRGDfV and LM609 cytotoxicity against human
tumour cell lines M14 and HT-29 ............................................................... 167
Figure 4.7 Effects of cRGDfV on tumour cell migration in the cell panel using
the scratch assay ....................................................................................... 170
Figure 4.8 The effect of LM609 on tumour cell migration ........................... 172
Figure 5.1 Evaluation of the cytotoxicity of the potential novel β 3 integrin
antagonists in the tumour cell line panel .................................................... 184
Figure 5.2 Effect of potential β3 integrin antagonists on M14 cell migration 186
Figure 5.3 Effect of 10 µM β3 antagonists on the inhibition of M14 cell
migration by the scratch assay................................................................... 188

ix
Figure 5.4 Effect of potential β3 integrin antagonists in inhibition M14 cell. 190

x
List of Tables
Table 1.1 Drugs currently used for targeted therapy ...................................... 8
Table 1.2 Functions and ligands of different types of integrins .................... 20
Table 1.3 Further anti-integrin agents in clinical use and advanced
development ................................................................................................ 53
Table 2.1 Primary antibodies and related secondary antibodies used in
immunohistochemistry (IHC) and immunoblotting (IMB) .............................. 65
Table 3.1 Primary antibodies and related secondary antibodies used in
immunocytochemistry (ICC) and IMB techniques. ..................................... 113
Table 3.2 Cell lines .................................................................................... 115
Table 3.3 Conditions investigated while optimising detection of expression of
αV, αIIb, β3 and αVβ3 integrin in human tumour cell lines using ICC. ........... 120
Table 3.4 The optimised protocol for all antibodies used to detect the
expression of αV, αIIb, and β3 subunit and β-actin using IMB technique. .... 125
Table 3.5 Optimised protocols for all antibodies used to detect the expression
of αV, αIIb, β3 subunits, and αVβ3 integrin in cell membrane using ICC ....... 131
Table 3.6 Screening different tumour cell lines for the membranous
expression of αV, β3, αIIb and αVβ3 by ICC .................................................. 136
Table 3.7 Comparison of integrin expression in different tumour cell lines
using specific antibodies for αV and β3 integrin subunits using ICC and IMB.
................................................................................................................... 139
Table 4.1 Comparison of cell migration assays (Kramer et al. 2013) ......... 153
Table 5.1 Compounds used ....................................................................... 181
Table 5.2 IC50 values for the potential novel β3 integrin antagonists in the
tumour cell line panel ................................................................................. 185
Table 5.3 A summary of effect of the potential novel β3 integrin antagonists in
inhibition of M14 cell migration after 24 hours treatment with the scratch
assay ......................................................................................................... 191

xi
Acknowledgement

Firstly, I would like to express my profound gratitude to my supervisors Dr.

Steve Shnyder, Dr. Helen Sheldrake and Professor Laurence Patterson for

providing me with an exciting and challenging research project. I would like to

thank them for help and guidance and illustrious advice which they given me,

throughout my PhD studies.

I am extremely thankful to the staff of Institute of Cancer Therapeutics who

had helped in this research either by their support or encouragement. I would

like also to thank the staff in the lab: in the histology lab I would like to thank

Beryl Cronin and Patricia Cooper, in cell culture I would like to thank Dave

Healey and in the molecular lab I would like to thank Dr. Mark Sutherland and

Andrew Gordon (who provided PCR data shown in the appendix).

I would also like to thank the Public Authority for Applied Education and

Training (PAAET) for their funding.

My special thanks must also go to my dear parents (Khairyah Al Shammari

and Owayed Al Shammari), my husband Abdul Mohsen Al Shammari and for

my four daughters Sarah, Nourah, Noof and Shaikha.

xii
Abbreviations

ABC: Avidin biotin complex

ADMIDAS: Adjacent to metal-ion-dependent adhesion site

ADP: Adenosine diphosphate

Akt/PKB: Protein kinase B

Angpt13: Angiopoietin-like 3

APES: 3-aminopropyltriethoxysilane

ARMD: Age-related macular degeneration

ATCC: American Type Culture Collection

bFGF: basic fibroblast growth factor

BME: Basal membrane extract

BSA: Bovine serum albumin

BSP: Bone sialoprotein

CAMs: cell adhesion molecules

CD3: Cluster of differentiation 3

CMRIT: Combined radioimmunotherapy with cilengitide

cRGDfV : Arg-Gly-Asp-phe-Val

CTGF: Connective tissue growth factor

CTP: Cyclotetrapeptide

CYR61: Cysteine-rich angiogenic inducer 61

2D: Two dimensional

xiii
3D: Three dimensional

DAB: 3,3-diaminobenzidine tetrahydrochloride

DAPI: 4,6-Diamidino-2-phenylindole

Db: Double bands

dH2O: Distilled water

D/I: Dilution/incubation

DMSO: Dimethyl sulphoxide

DNA: Deoxyribonucleic acid

DPX: Distyrene/plasticiser/xylene

ECM: Extracellular matrix

EDTA: Ethylenediamine tetraacetic acid

EGF: Epidermal growth factor

EGFR: Epidermal Growth Factor Receptor

EMT: Epithelial mesenchymal transmission

EP: Endogenous peroxidase

ERK: Extracellular Signal-regulated Kinase

FAK: Focal adhesion kinase

FBS: Foetal bovine serum

FFPE: Formalin fixed paraffin embedded

FGFR: Fibroblast growth factor receptor

FITC: Fluorescein isothiocyanate

g: Gram
xiv
GAR: Goat anti rabbit

GIST: Gastrointestinal stromal tumour

GTPase: Guanosine triphosphate

H2O2: Hydrogen peroxide

HBSS: Hanks’s balanced salt solution

H&E: Haematoxylin and Eosin

HER2: Human Epidermal Growth Factor Receptor 2

HPV: Human papilloma virus

HRP: Horse-radish peroxidase

hrs: hours

HTS: High throughput screening

HUVECs: Human umbilical vein endothelial cells

IARC: International Agency for Research on Cancer

iC3b: Inactivated C3b

IC50: Inhibitory concentration

ICA: Ice pre-cooled acetone

ICAM: Interacellular Adhesion Molecule

ICC: Immunocytochemistry

ICM: Ice pre-cooled methanol

IF: Immunofluorescence

Ig: Immunoglobulin

IgG: Immunoglobulin g
xv
IHC: Immunohistochemistry

IMB: Immunoblotting

ISH: In-situ hybridisation

isoDGR: iso-Aspartic acid-Glycine-Arginine

kDa: Kilodalton

kg: Kilogram

LAP-TGF- β: Latency associated peptide-Transforming growth factor B

LIMBS: Ligand induced metal binding site

mA: Milliampere

MAdCAM-1: Mucosal vascular addressin cell adhesion molecule 1

MAPK: Mitogen-activated protein kinase

Mb: Multiple bands

MIDAS: Metal-Ion-Dependent Adhesion Site

ml: Milliliter

mM: Millimolar

MMAb: Monoclonal mouse antibody

MMPs: Matrix metalloproteinases

MMP-9: Matrix metalloproteinase-9

M.O.M.: Mouse on mouse

mTOR: mammalian Target Of Rapamycin

MTT: 3-(4,5-Dimethylthiazole-2-y1)-2,5-diphenyl tetrazolium bromide

NaCl: Sodium chloride


xvi
Nc: No clear band

NCI: National Cancer research

n.d.: Not done

NF-Kb: Nuclear factor kappa-light chain enhancer of activated B cells

NGS: Normal goat serum

NHS: Normal horse serum

nM: Nanomolar

NRS: Normal rabbit serum

NS: Normal serum

NSCLC: Non small cell lung cancer

NSP: Non specific binding

OPN: Osteopontin

P13k/Akt: Phosphotidylinositide 3-kinase/protein kinase B

PAb: Primary antibody

PBS: Phosphate buffered saline

PC: Polycarbonate

PCI: Percutaneous coronary intervention

PDGFR: Platelet Derived Growth Factor Receptor

PET: Polyethyleneterphthalate

PFA: Paraformaldehyde

PGAb: Polyclonal goat antibody

PGAR: Polyclonal goat anti rabbit


xvii
PKC: Protein kinase C

PKPD: Pharmacokinetic pharmacodynamic

PML: progressive multifocal leukoencephalopathy

PRAb: Polyclonal rabbit antibody

PRAG: Polyclonal rabbit anti goat

PRAM: Polyclonal rabbit anti mouse

PSI: Plexin-Semaphorin-integrin

RCF: Relative centrifugal force

RGD: Arginine-Glycine-Aspartic acid

RGP: Radial growth phase

RPM: Revolution per minute

RPMI: Roswell Park Memorial Institute

R.T.: Room temperature

RT-PCR: Reverse transcription-polymerase chain reaction

Sb: Single band

SD: Standard deviation

SDS: Sodium dodecyl sulphate

siRNA: small interfering RNA

SM: Skimmed milk

SU24: Untreated scratch after 24 hours

ST24: Treated scratch after 24 hours

TBS: Tris buffered saline


xviii
TEM: Transendothelial migration

TGF-β1: Tissue growth factor β1

THN: Tetramethylethylenediamine

TNF: Tumour necrosis factor

TRITC: Tetramethylrhodamin isothiocyanate

µg: Microgram

µl: Microliter

µM: Micromolar

VCAM-1: Vascular cell adhesion protein 1

VEGF: Vascular Endothelial Growth Factor

VEGFR-2: Vascular Endothelial Growth Factor receptor 2

VEGFR-A: Vascular Endothelial Growth Factor receptor A

VGP: Vertical growth phase

vWF: von Willebrand factor

xix
1 Chapter 1: Introduction

1.1 Cancer background

1.1.1 Definition and aetiology of cancer

Cancer is a complex family of diseases characterised by uncontrolled growth

of cells and spread from the primary tumour site to other sites (Varmus

2006). The formation of cancer is a multistep process that originates from a

single cell in which vital regulatory pathways have been disturbed. Cancer

has multiple causes, with a wide range of genetic, physiological and

histological features. However, genetic and environmental factors such as

chemical exposure, ionizing radiation exposure, and certain viruses (such as

human papilloma virus; HPV) are the main causes (Fearon 1997).

Clinically, tumours include two main types: benign and malignant. A benign

tumour grows locally, with a gradual increase in size, and causes local

pressure or obstruction within the surrounding tissue. In contrast, a malignant

tumour grows rapidly, metastasises via the lymph or blood vessels to distant

sites, and is dangerous due to its potential for invasion and destruction of

normal tissues (van Slooten et al. 1985).

1
1.1.2 Incidence of cancer

A wide variation is reported for the incidence and prevalence of cancer at

different anatomical sites, and for patients of different ages, sex and

geographical locations. The overall trend is for an increase in cancer

incidence and death annually. Statistics obtained from the International

Agency for Research on Cancer (IARC), showed that the incidence of cancer

in the world reached 12.7 million cases in 2008 with a mortality rate of 7.6

million. The incidence of cancer was estimated to increase to reach 13.2

million cases for both sexes in 2011 (Jemal et al. 2011). Europe estimated

1.3 million deaths from cancer in 2013 (Malvezzi et al. 2013). In the United

Kingdom, the cancer incidence reported in 2008 and cancer mortality in 2009

was 408,381 new cancer cases and 156,090 cancer deaths (CRUK 2012).

Moller indicated that the incidence rate of all cancers in the English

population will rise from 224,000 in 2001 to around 299,000 in 2020, or an

increase of 33 per cent (Moller et al. 2007). In the United States, cancer is

the second most common cause of death after heart disease and it is

estimated that one in four people will die from cancer. The American Cancer

Society estimates that in 2013 there will be 1.7 million new cancer cases

diagnosed and about 580,350 Americans are expected to die of cancer

(Siegel et al. 2013). These statistics support the need for continued research

into the causes of cancer and the development of effective preventive

methods and effective treatments.

2
1.1.3 Treatment of cancer

Treatment of cancer is a challenge. The choice of treatment depends on

tumour type, histological grade and stage of the tumour at the time of

diagnosis. The overall aim of the treatment is to remove all cancerous cells

without affecting normal tissue (Rosenberg 2001). The major types of

treatment for cancer are surgery, radiotherapy, biological therapy and

chemotherapy.

1.1.3.1 Surgery

In most cases, surgery is the first line of cancer treatment. However, before

diagnosis, the cancer may have already metastasised to another area of the

body; therefore, chemotherapy and/or radiotherapy must be used in

conjunction with surgical removal in order to slow tumour growth and prevent

further metastasis. In addition, some primary tumours, such as brain

tumours, are not easily removed by surgery. Therefore, neoadjuvant

chemotherapy (the use of chemotherapy prior to surgery or radiation) can

also play an important role. This treatment can reduce the tumour load

thereby making surgery or radiotherapy easier. The removal of residual

tumour after surgery can also be done by adjuvant treatment (Rosenberg

2001).

1.1.3.2 Radiotherapy

Radiotherapy is used to reduce the size of tumour before surgery

(Camporeale 2008), ensures that all cancer cells are destroyed following
3
surgery and slows down the progression of the disease. For example,

radiotherapy can be used in conjunction with surgery for treatment of

carcinomas of the pancreas (Alfieri et al. 2001), larynx, other head and neck

sites, cervix, breast, bladder and prostate. However, several studies have

shown that prostate cancer patients who undergo radiotherapy treatment can

have recurrence of the disease after a few months (Tefilli et al. 1998; Tefilli et

al. 1998; Tefilli et al. 1998). Furthermore, radiotherapy is not efficient at

curing metastasis.

1.1.3.3 Biological therapy

1.1.3.3.1 Endocrine therapy

The success of endocrine therapy depends on its ability to suppress further

growth of the tumour rather than its ability to kill cells. It can be used to treat

hormone dependent tumours such as breast and prostate cancer (Labrie

2004). The treatment depends on lowering the plasma concentration of a

particular hormone (e.g. by removing the producing organ such as by

orchidectomy (removal of the testes) (Suzuki et al. 2010) or by antagonising

its action at its receptor (e.g. tamoxifen, which binds to oestrogen receptors

on the mammary epithelium and thereby blocks the proliferative actions of

oestrogen) (Criscitiello et al. 2011).

1.1.3.3.2 Gene therapy

The principle of gene therapy involves changing the actual gene responsible

for the neoplastic processes. This therapy requires that the gene must
4
access to the cell via a vector. The vector may be a virus that is defective in

replication and into which the therapeutic gene has been inserted, along with

the promoter that will activate the gene once inside the cell. Gene therapy

may have one of the following aims: restoring the function of a defective

tumour-suppressor gene such as p53 to inhibit tumour growth; blocking the

action of a mutated or over-expressed oncogene or insertion of a gene that

activates a prodrug or produces other cell-killing effects thereby eliminating

cancer cells near the cell into which the gene has penetrated (Brown &

Lillicrap 2002; Edelstein et al. 2007). However, one type of gene therapy,

ganciclovir/Hstk, reached phase III clinical trials but did not show any

significant effect on malignant brain tumours due to low transduction of the

vector and immune response to vector-producing cells (Rainov 2000). Gene

therapy can cause damage to the immune system (Brown & Lillicrap 2002).

1.1.3.3.3 Antibodies

Monoclonal antibodies can disrupt cancer cells through a variety of

mechanisms (Strome et al. 2007; Trikha et al. 2002). Monoclonal antibodies

can trigger the immune system to attack cancer cells (e.g. Rituximab and

Alemtuzumab) (Murakami et al. 2011; Schweighofer & Wendtner 2010),

prevent the cancer cells from taking up proteins (e.g. Transtuzumab), block

angiogenesis through inhibition of VEGF-A (e.g. Bevacizumab) or carry

cancer drugs or radiation to cancer cells (e.g. Zevalin, Bexxar and Mylotarg).

However, a recent study reported that Bevacizumab administration led to

increased treatment-related mortality (Ranpura et al. 2011). Several

5
limitations also make antibodies unattractive for use as therapeutics,

including production costs, pharmacokinetics versus tissue penetration and

modes of action (Chames et al. 2009).

1.1.3.4 Chemotherapy

1.1.3.4.1 Conventional chemotherapy

Cancer chemotherapy is a systemic treatment and its selectivity depends on

the ability of the drugs to target the tumour. However, cancer chemotherapy

drugs also affect normal cells that replicate rapidly, such as hair follicles

(Botchkarev 2003), bone marrow, intestinal mucosa and gonads (Meirow &

Nugent 2001; Vijayalaxmi 2011). Chemotherapy works mainly by disrupting

cell proliferation, especially targeting DNA synthesis and cell division.

Traditional chemotherapy drugs include: anti metabolites such as 5-

fluorouracil (inhibits DNA synthesis), alkylating agents such as cisplatin

(blocks DNA replication), anti microtubule agents such as taxanes (blocks

microtubule depolymerisation) and anthracyclin antibiotics like doxorubicin

(interacts with topoisomerase II, intercalates between DNA nucleotides and

blocks DNA synthesis) (Kaye 1998).

Tumour cells may be resistant to cytotoxic drugs because of extracellular

circumstances that limit drug access to the tumour or due to defects in the

p53 gene that render the tumour cell resistant to the cytotoxic drug. Tumour

cell resistance to chemotherapy can also arise due to DNA damage,

decreased uptake of drug by the cell and increased drug efflux by a p-

6
glycoprotein that acts as a drug efflux pump and reduces the intracellular

concentrations of some cytotoxic agents. Another problem with

chemotherapy is that the drug may reach the target cells at insufficient

concentrations as a result of inactivation or removal of the drug from the

circulatory system (Moxley & McMeekin 2010).

1.1.3.5 New strategies for cancer therapy

While the increased incidence and mortality of cancer and the inadequacy of

the different kinds of therapy already mentioned, a clear need thus exists for

the development of treatments with greater effectiveness. The development

of targeted therapies has the aim of improving differentiation between normal

and neoplastic cells. The discoveries in molecular biology about the control of

cell cycle events highlighted that certain signalling pathways that control the

growth and differentiation were inappropriately activated in various tumours

(Hersey et al. 2009). Targeted agents can be used against cell adhesion

molecules, growth factors, intracellular signalling and angiogenesis (Gerber

2008; Korpanty et al. 2010; Lord & Ashworth 2010). Table 1.1 summarises

the targeted therapy drugs currently in clinical use (Collins & Workman 2006;

Malinowsky et al. 2010).

7
Table 1.1 Drugs currently used for targeted therapy
Drug Tumour type Target Ref
Trastuzumab Metastatic breast cancer, HER2 (Farolfi et
gastric cancer al. 2013)
Cetuximab Metastatic colorectal cancer EGFR (Stintzing
et al. 2013)
Imatinib Chronic myeloid leukkaemia Bcr/abl, c-kit, (Spiera et
Mesylate and Gastrointestinal stromal PDGFR, al. 2011)
(Gleevec) tumours with activated c-kit
receptor tyrosine kinase, other
sarcomas
Bevacizumab Colorectal cancer VEGF (Bennouna
et al. 2013)
Gefitinib Non-small-cell lung cancer mutant EGFR (Sugiura et
(Iressa) al. 2013)
Erlotinib Non-small-cell lung cancer mutant EGFR (Stevenson
(Tarveca) & El-Modir
2011)
Rapamycin Breast, prostate and renal mTOR (Armstrong
RAD001 cancer et al. 2010)
Sorafinib Melanoma RAF kinase (Panka et
BAY 43-9006 Renal carcinoma al. 2006;
Procopio et
al. 2013)
Vemurafenib Melanoma BRAF (Gonzalez
mutation et al. 2013)
Dasatinib Gastrointestinal stromal Kit, PDGFR (Caenepeel
BMS354825 tumour Dual Src/Abl et al. 2010;
Chronic myeloid leukemia kinase Quintas-
Cardama
et al. 2007)
Lapatinib Breast cancer EGFR, HER2 (Bachelot
et al. 2013)
Sunitinib Renal cell cancer VEGFR, (Blagoev et
PDGFR, cKit, al. 2013;
Flt-3 Huang et
al. 2010)
Pertuzumab Breast cancer HER2 (O'Sullivan
& Swain
2013)
Dasatinib Breast cancer Bcr/abl (Somlo et
al. 2013)
Key: HER2: Human Epidermal Growth Factor Receptor 2. EGFR: Epidermal Growth Factor
Receptor. PDGFR: Platelet Derived Growth Factor. VEGF: Vascular Epithelial Growth
Factor. mTOR: mammalian Target of Rapamycin GIST: Gastrointestinal stromal tumour.

8
1.1.4 The general pathobiology of cancer

Cell growth is usually well organised and controlled to meet the needs of the

body. In contrast, cancer cell division is uncontrolled. Growth factors such as

tumour necrosis factor (TNF) and extracellular matrix (ECM) components that

interact with cell surface receptors can lead to changes in cell division which

activate different protein pathways inside the cell (Jinka et al. 2012). With the

help of signal transducers inside the cell, these pathways will lead to changes

in gene expression and induce cell proliferation (Radeff-Huang et al. 2007).

Apoptosis is the process of programmed cell death. Apoptosis involves

biochemical events that lead to cell morphology changes and ultimately cell

death. These changes include blebbing, loss of cell membrane asymmetry

and attachment, cell shrinkage, nuclear fragmentation, chromatin

condensation and chromosomal DNA fragmentation. Tumour cells can

survive and proliferate if they can escape from undergoing apoptosis (Fulda

2009). Apoptosis can be mediated by both extrinsic and intrinsic pathways.

The extrinsic pathway is initiated by extracellular death ligand such as tumour

necrosis factor (TNF) and tumour necrosis factor-related apoptosis inducing

ligand (TRAIL), which leads to the recruitment of caspase-8 to form a death-

inducing signal complex. The extrinsic pathway can activate caspase-3 and

cleave BID to tBID which facilitates the release of the cytochrome c from the

mitochondria. The intrinsic pathway is mediated by BAX, BAK and BID

proapoptotic proteins which promote release of cytochrome c from the

9
mitochondria leading to activation of caspase-9 ultimately leading to

apoptosis.

The tumour suppressor gene p53 can bind to DNA and activates genes that

control the cell cycle and programmed cell death. If the cell has damaged

DNA, p53 can either repair the damage or induce apoptosis to prevent

proliferation of the defective cell and potential cancer development.

Therefore, inactivation of p53 can also play a role in the survival of tumour

cells. Cell adhesion molecules can also control cell apoptosis via anoikis

(apoptosis resulting from loss of interactions with the extracellular matrix),

direct induction of caspases and increased bcl-2 expression, p53 inactivation

and several other pathways related to survival, such as Extracellular Signal-

regulated Kinase (ERK) and protein kinase B/Akt (Guadamillas et al. 2011;

Stiewe 2007; Tan et al. 2004; Vachon 2011; Zhong & Rescorla 2012).

As a malignant tumour progresses it will metastasise (Bozzuto et al. 2010).

Metastasis is the process whereby the cancer cells migrate from the primary

tumour site to other parts of the body (Geho et al. 2005). In essence, it is the

fundamental difference between benign and malignant tumours. The ability of

cancer cells to metastasise depends on the interaction of their cell surface

molecules with the microenvironment including the ECM and neighbouring

cells. Several steps are involved in metastasis (Figure 1.1) including

migration, intravasation, transportation, extravasation and metastatic

colonisation (Hanahan & Weinberg 2000; Hanahan & Weinberg 2011;

Talmadge & Fidler 2010; Williams 1974).

10
Figure 1.1 The basic steps of the cancer metastatic pathway
(Bellahcene et al. 2008)
Normal cells are transformed into cancer cells (a). Cancer cells proliferate
(b), detach from each other, penetrate the basement membrane, migrate and
invade the surrounding tissues (c). Cells intravasate into blood vessels, travel
in the circulation to distant sites. They then extravasate through the vessel
wall (d) and migrate (e) to nearby tissue, where they may form distant
metastases (f).

The tools of cell migration are the cell adhesion molecules (CAMs) (Hood &

Cheresh 2002) and protease enzymes. Cell adhesion molecules such as

cadherins and integrins play a role in the interactions of similar and different

cells together. Cell adhesion molecules can also induce gene expression in

the nucleus by binding with transcription factors. Integrins can interact with

actin binding proteins and specific kinases such as focal adhesion kinase

(FAK) (Jockusch et al. 1995). FAK mediates cell motility through Src (Cary et

11
al. 1996) and activation of the RAS pathway (Figure 1.2) (Schlaepfer et al.

1994).

Further, integrins can promote the movement of metastatic cells by allowing

the cell to adhere to different ECM components. Cell migration starts with cell

polarization in the direction of movement and the formation of a protrusion.

Integrins serve as points of contact with the ECM and stabilise the protrusion

via connections to actin filaments. At the leading edge adhesion of the

protrusion to the substratum allows migration through enhancement of focal

adhesion formation (Gawecka et al. 2010; Wozniak et al. 2005). Finally, the

detachment of the cell from the previous attachment happens through

contraction of the cell from the edge toward the nucleus and the adhesion

receptor released from the cytoskeleton. This detachment is associated with

the release of cell vesicles and fragments (Lock et al. 2008; Sahai 2007).

The last factor that has a role in cell migration is the protease. Proteases

degrade a path through the ECM and stroma. The cancer cells in a tumour

can synthesise matrix metalloproteinases (MMPs); these MMPs also have an

association with integrins (Brooks et al. 1996; Jin et al. 2011), since integrins

induce the surrounding stromal cells to produce MMPs (Brooks et al. 1996).

Different integrin signalling pathways control MMP expression, thereby

controlling tumour cell invasion (Kubota et al. 1997; Westermarck & Kahari

1999). MMPs are involved in integrin processing and integrins can activate

MMPs in the cell. MMP activity can lead to proteolytic cleavage of the

12
extracellular domain of E-cadherin, which can lead to disruption of the ability

of E-cadherin to control cellular interactions (David & Rajasekaran 2012).

Migrating tumour cells will intravasate into the blood and lymphatic vessels.

During this process, tumour cells attach to the stromal face of the blood

vessel and degrade its basement membrane, passing between the

endothelial cells into the bloodstream. The tumour cells are then transported

with platelets in the direction of blood flow (Felding-Habermann et al. 2001;

Weis & Cheresh 2011). The role of integrins in this step will be discussed in

section 1.2.4.1.1

When a tumour cell reaches the metastatic site, it will escape from the blood

or lymph vessel (extravasation) by attaching to endothelial cells lining the

vessel and passing through them into the surrounding stroma. In this step,

another type of cell adhesion molecule can also play a role. Selectins, which

are expressed on endothelial cells, also help the attachment of cancer cells

to the endothelium (Barthel et al. 2013).

The last stage of metastasis is the colonisation of the tumour at a distant site.

With the help of angiogenesis (formation of new blood vessels), the tumour

can grow (Weis & Cheresh 2011). Vascular endothelial growth factor (VEGF)

and basic fibroblast growth factor (bFGF) induce and regulate angiogenesis

(Hanahan & Weinberg 2011; Weis & Cheresh 2011).

13
Figure 1.2 Role of integrins in cancer development

14
1.2 The Integrins

1.2.1 Introduction

Adhesion receptor molecules on the cell surface became attractive targets for

cancer therapy due to their important role in the control of tumour cell

dissemination (Okegawa et al. 2004; Simmons 2005). Many types of

adhesion cellular receptors (Shimaoka et al. 2002) are recognised including

cadherins, selectins, immunoglobulins and integrins (Mousa 2002). The

integrins are transmembrane glycoprotein adhesive receptors found on the

cell surface; they bind to the extracellular matrix components at the outer

membrane surface and interact with the cytoskeletal components at the inner

membrane surface. Integrins induce signals (Dedhar & Hannigan 1996;

Huveneers et al. 2007; Qin et al. 2004; Stupack 2005; Stupack & Cheresh

2002) through interaction with the extracellular matrix proteins (Brakebusch

et al. 2002) and growth factor receptors (Hynes 2002); these are important in

cell migration (Felding-Habermann 2003), cell cycle regulation (Fahraeus &

Lane 1999), gene expression and apoptosis (Jinka et al. 2012).

1.2.2 Structure of integrins

The integrins are heterodimers consisting of α and β chains that are non

covalently associated (Hynes 2002). The two chains each have a large

extracellular portion comprising multiple domains, a single transmembrane

domain and a short cytoplasmic tail (Figure 1.3) (Shimaoka et al. 2002;

Srichai 2010; Ulmer 2010).

15
The N-terminus of the α subunit consists of seven segments folded into a

single compact domain, forming a structure called the β-propeller. The α

subunit may also contain another domain that is inserted between blades 2

and 3 of the β propeller; this is known as the I domain, or von Willebrand

factor type A domain (Shimaoka et al. 2002). The I-domain is the key ligand-

binding domain in those integrins where it is present. The β subunit has a

highly conserved N-terminus region, which is similar to the I domain and

therefore named the β I-like domain; this domain is responsible for ligand

binding in integrins that do not contain an I domain (Figure 1.3) (Humphries

2000; Humphries 2004; Humphries et al. 2004; Takada et al. 2007; Takagi

2007).

The N-terminus of the integrin has a divalent cation binding site known as

MIDAS (Metal-Ion-Dependent Adhesion Site) located in each I and I-like

domain. Divalent cations bind here, as well as at other sites such as the

ligand induced metal binding site (LIMBS) and ADMIDAS, and serve a

central function in ligand recognition by the I domain and I like domain

(Humphries 2002; Lee et al. 1995; Shimaoka & Springer 2003).

The C-terminus of the α subunit includes three β sandwich domains as a

thigh, and two calf regions (calf 1, calf 2). In the β subunit, the C-terminus is

composed of the PSI (Plexin-Semaphorin-Integrin) domain and four

epidermal growth factor domains (EGF1, EGF2, EGF3 and EGF4). The

region where the integrin is bent in the resting state is termed the knee

region. In the β subunit, it forms a junction between the hybrid domain and

16
two EGF and PSI, while in the α subunit, it is found between the thigh and

the two calf regions (Figure 1.3). The terminal end of the C-terminus of both

subunits spans the plasma membrane to interact with the intracellular

cytoskeleton. During inside-out signalling (the regulation of integrin function

from within the cell) (Hynes & Lander 1992), the integrin undergoes a

structural change from the bent, closed or inactive conformation (Kalli et al.

2011), which has a low affinity for ligand binding, to an extended, open or

active form that has a high affinity for ligand binding (Chen et al. 2011; Hynes

2002). After activation, the signals are transmitted to the extracellular

domains of the integrin, which then alters the conformational of the ligand

binding site and hence the affinity for the ligand (Figure 1.3) (Hynes 2002;

Kim et al. 2011).

17
Figure 1.3 The structure of αIIbβ3 integrin in its active and inactive state
The resting inactive integrin (left), has a closed bent structure. The active
form (right) is open. The N-terminus includes the β-propeller and βA-domain
in αIIb and β3 subunits respectively. It has an activation-dependent ligand-
binding domain (indicated by the red arrow) for ECM proteins,
macromolecules or receptors on the surface of opposing cells. The C
terminus includes the thigh domain and two calf regions in αIIb and the hybrid
domain, PSI domain and β-tail domain in β3 region. The tails of both subunits
span the plasma membrane to receive signals from inside the cell (Shattil et
al. 2010).

18
1.2.3 Types of integrins

Humans have 24 integrins made up of a combination of 18 α and 8 β

subunits. As shown in Figure 1.4, the different types of integrins can be

classified according to specificity of ligand binding, and to the presence or

absence of the I domain. Each integrin has a different function (Table 1.2),

although a number of integrins can bind the same ECM proteins, providing

redundancy in cell signalling and adhesion pathways.

αIIb

α8 α5
β3
β5

β8 αV
α1
β6
α2
β1 α4 β7
α10

α9 αE
α11

αL αM

α3 α6 α7
β2

β4 αX αD
Figure 1.4 The integrin family
Different kinds of integrin subunits can bind together to form an integrin sub
family. There are 5 subfamilies: Red indicates the RGD receptors, yellow the
laminin the receptors, blue the collagen receptors and green the leukocyte
receptors. The black frame indicates an I-domain integrin.

19
Table 1.2 Functions and ligands of different types of integrins
Integrin Function Ligands Ref
(Bodary
&
McLean
Regulates cell adhesion, 1990;
migration and controls matrix Henry et
Laminin E1
accumulation. al. 2001;
α1β1 fragment,
Rossino
Collagen
Regulates epidermal growth et al.
factor receptors. 1991;
Tomaselli
et al.
1993)
Mediates the adhesion of
platelets to collagen. (Klein et
Collagen I-IV, al. 1991;
Used by fibroblasts for tissue Laminin, Schiro et
α2β1
collagen remodelling during Echovirus-1 al. 1991;
wound repair. Thrombospondin Staatz et
al. 1991)
Involved in tumour progression.
(Coopma
n et al.
Laminin5, 1996;
Regulates expression of MMP-9.
Epithelial Manohar
basement et al.
Mediates survival of
α3β1 membrane 2004;
keratinocytes.
protein,Epiligrin Pulido et
Thrombospondin al. 1991;
Participates in phagocytosis.
Wang et
al. 2004)

(Hsia et
al. 2005;
Maintains the structural integrity
VCAM-1, Moyano
of the placenta and heart during
Fibronectin, et al.
embryogenesis.
α4β1 Thrombospondin, 1997;
Osteopontin, Springer
Mediates adhesion of inflamed
Mad CAM-1 et al.
vascular endothelial cells to T
1994; Wu
cells.
et al.
1995)

20
Mediates lymphocyte adhesion to Mad CAM-1,
MAd CAM-1 on endothelial VCAM-1, (Erle et
α4β7
specific Peyer’s patches in the Fibronectin, al. 1994)
gut. Osteopontin
Mediates fibronectin assembly
into extracellular matrix,
fibronectin dependent cell
adhesion, spreading, migration
(Hynes
and signal transduction.
1992;
Fibronectin, Varner et
α5β1 Acts as a co-stimulatory
Osteopontin al. 1995;
molecule to the T cell receptor
Wu et al.
CD3 complex in T-cell activation
1993)
and enhanced phagocytosis of
opsonised particles by
monocytes.
Angiogenesis.
Platelet interaction with sub
endothelial basement membrane
(Leu et
laminin exposed on blood vessel
al. 2003;
damage.
α6β1 Laminin Sonnenb
erg et al.
Activation of T-cells.
1990)
Angiogenesis
Sonnenb
Adhesion to basement
α6β4 Laminin 5 erg et al.
membranes and matrix protein
1990
Involved in muscle development,
Laminin 1,2,4, E8 (Song et
α7β1 linkage between muscle fibre and
region al. 1992)
extracellular matrix.
(Bossy et
Osteopontin,
Kidney morphogenesis. al. 1991;
Fibronectin,
α8β1 Muller et
Vitronectin,
Expressed in neural tissues al. 1995)
Tenascin
(Desloge
s et al.
Involved in osteoclast formation
Osteopontin, 1998;
and function.
Tenascin-c Oommen
α9β1
VCAM-1 et al.
Cell migration and adhesion via
VEGF A,C,D 2010;
direct binding of VEGF.
Rao et al.
2006)
Expressed on chondrocytes, (Bengtss
Collagen II
α10β1 deficiency leads to on et al.
Laminin
chondrodysplasia. 2005)
21
Required on periodontal ligament (Carrace
fibroblasts for cell migration and do et al. ;
α11β1 collagen reorganisation to help Collagen I Popova
generate the forces needed for et al.
axial tooth movement. 2007)
Intestinal targeting of lymphocyte (Cepek et
αE β 7 E cadherin
subpopulations. al. 1994)
(Brockba
nk et al.
2005; Hu
et al.
Fibronectin, 1995;
Cell matrix and intracellular
αV β 1 Osteopontin Marshall
interaction.
LAP-TGF-β et al.
1995;
Vogel et
al. 1990)

Vitronectin, (Gladson
Vitronectin endocytosis and
αV β 5 Osteopontin, et al.
angiogenesis.
BSP 1997)
Response of epithelium to (Breuss
inflammatory stimuli and injury et al.
such as in asthma. 1995;
Fibronectin Erikson
αV β 6 Regulation of epithelial Osteopontin, et al.
proliferation in vitro and wound LAP-TGF-β 2009;
healing. Munger
et al.
Activation of LAP-TGF- β 1999)
(Chernou
sov &
Carey
2003;
Jackson
Neural function.
et al.
LAP-TGF-β
αV β 8 2004;
Receptor for foot and mouth Vitronectin
Nishimur
disease virus.
a et al.
1994;
Pozzi &
Zent
2011)

22
Involved in immune function, cell-
cell interactions during
(Springer
lymphocyte transit through
1990;
αLβ2 endothelium to sites of ICAM 1,2,3
Springer
inflammation
1990)
and during recirculation through
the lymph node.
Has a role in inflammation due to
its expression on many
leukocytes involved in immune
system.

Mediates adhesion to and


Complement
transmigration through vascular
fragment iC3b,
endothelium of monocytes and (Springer
αMβ2 ICAM 1,2,4,
granulocytes. 1990)
Fibrinogen,
Factor X
Participates in leucocyte
aggregation.

Involved in chemotaxis,
apoptosis and other phagocytic
activities.
(Keizer et
Complement
Cytotoxic T cell killing and al. 1987;
fragment iC3b,
αXβ2 monocyte and granulocyte Vorup-
Fibrinogen,
adhesion to endothelium. Jensen et
ICAM-1, Collagen
al. 2003)
(Grayson
Has role in phagocytosis by
αDβ2 ICAM-3, VCAM-1 et al.
macrophages.
1998)
(Buensuc
eso et al.
Fibrinogen, 2005;
Essential for normal platelet
Fibronectin, Muller et
adhesion to damaged vascular
αIIbβ3 Vitronectin, al. 1993;
endothelium and normal platelet
Thrombospondin, Podolniko
aggregation.
VWF va et al.
2003)

23
(Bader et
al. 1998;
Gavrilovs
kaya et
al. 1999;
Regulates angiogenesis, Humphrie
Endothelial cell adhesion to s et al.
ECM proteins. 2006;
Tumour progression and Vitronectin, Leavesle
invasion. Activation of MMPs. Fibronectin, y et al.
Fibrinogen, 1992;
αV β 3 Osteoclast mediated bone Thrombospondin, Nelsen-
resorption. Osteopontin, Salz et al.
BSP, 1999;
T cell activation. VWF Varner &
Cheresh
Act as a receptor for viruses like 1996;
adenovirus, Echo virus. Wadehra
et al.
2005;
Wickham
et al.
1993)

1.2.4 Function of integrins

As can be seen in Table 1.2, the integrins have multiple functions, including

the adhesion between the cell and the surrounding tissue (Hynes 1999;

Niland & Eble 2012). The integrins help the membrane to adhere to the

extracellular sites and create an attractive force that adheres the cells to their

surroundings (Schwartz 2010). Integrins are important in the regulation of cell

shape, migration (Schneider et al. 2011) and cell cycle regulation. They are

also involved in signalling pathways (Scales & Parsons 2011) that regulate

cell growth, proliferation, survival, division, differentiation and apoptosis (Guo

& Giancotti 2004).

24
The regulation of integrin function can occur internally within the cell through

signalling complexes pathways (Hynes 2002), where the activation of

integrins can result from binding to certain ligands or by inside-out-signalling

(Figure 1.5). Cell-matrix contact induces a wide range of signalling pathways

mediated by adaptor proteins and enzymes (Takada, Ye & Simon 2007). For

example, integrins can be activated intracellularly by signals from G-protein

coupled receptors that lead to phosphorylation of the cytoplasmic domain of

the β subunit. The cytoskeletal adaptor proteins such as talin, skelemin,

filamin, α-actinin, myosin and tensin can regulate integrin affinity, changing

the integrin into its active conformation. The adaptor protein head binds to

the β tail and induces conformational changes that are accompanied by

dissociation of α and β subunits and increased affinity for the ligand binding,

so that inside-out signals regulate the cell adhesion. When the integrins bind

to their ligands in the ECM, they cluster to form a focal adhesion, which

affects the organisation of the cytoskeleton underlying the plasma membrane

(Humphries 2000).

In the extracellular region, ligand binding transfers a signal to the interior of

the cell (outside-in signalling) and can induce conformational changes,

including separation of the α and β legs (Humphries 2000; Humphries et al.

2003; Legate et al. 2009). This separation allows the interaction of the

cytoplasmic tail with intracellular signalling molecules such as FAK/c-Src

(Huveneers et al. 2007), the small GTPases Ras & Rho and adaptors like

cas/crk and paxillin (Guo & Giancotti 2004; Takada, Ye & Simon 2007;

25
Yamada et al. 2003). These signals then regulate cell function (Figure 1.6)

(Millard et al. 2011).

This project will concentrate on the study of the αVβ3 and αIIbβ3 integrins (β3

subfamily). The subsequent sections will describe their role in cancer

development, survival, metastasis and invasion.

Figure 1.5 inside-out-signalling pathways for integrins (Askari et al.


2009)
Inside-out signals occur through binding of β integrin tail with cellular proteins
(B). The inside signals leads to integrin changing from inactive (A and B) to
active form, which is able to bind to ECM proteins (C). Binding ECM proteins
like fibrinogen, vitronectin, fibronectin, thrombospondin, von Willebrand factor
and osteopontin, and interaction with other proteins and receptors induce
various outside-in signals (D) which begin signalling cascades controlling
cellular processes (E).
26
Figure 1.6 Interaction of active integrin with signalling molecules
regulates cellular function (Hynes 2002).

1.2.4.1 Function of αVβ3 integrin

The αVβ3 integrin can recognise several ligands such as vitronectin,

fibronectin, fibrinogen, proteolysed collagen and other proteins. It does this

by recognising the tripeptide sequence Arginine-Glycine-Aspartic acid (RGD)

which is common to the above ligands. The αVβ3 integrin can also recognise

WGD, and isoDGR (iso-Aspartic acid-Glycine-Arginine) (Spitaleri et al. 2008)

and contains another non-RGD binding site for MMP-2 and MMP-9 (Brooks

et al. 1996).

αVβ3 is widely expressed on the endothelial cells (Takada et al. 2007),

epithelial cells, smooth muscle cells and monocytes. It is expressed on

activated endothelial cells and new vessels but it is absent in resting

endothelial cells and normal organ systems (Rusnati et al. 1997). It regulates

27
both angiogenesis in endothelial cells and cell adhesion to the extracellular

matrix (Soldi et al. 1999). It also has a role in osteoclast activity where it

mediates osteoclast adhesion and regulates the cytoskeletal organisation

required for cell migration (Duong et al. 2000; Nakamura et al. 1999; Srivatsa

et al. 1997; Tolar et al. 2004).

1.2.4.1.1 Role of αvβ3 integrin in angiogenesis

Angiogenesis is the physiological process involving the growth of new blood

vessels. Angiogenic blood vessels express αVβ3 at elevated level, whereas

αVβ3 expression is low in quiescent endothelial cells (Leu et al. 2002; Weis &

Cheresh 2011). Interaction between αVβ3 and the ECM can control

angiogenesis through pro-angiogenic and anti-angiogenic molecules that

originate from ECM (Robinson & Hodivala-Dilke 2011). The pro-angiogenic

function of αVβ3 is promoted through its co-operation with factors that

enhance angiogenesis such as VEGFR-2, vitronectin, fibronectin, Del1,

Angptl3, CYR61, bone sialoprotein (BSP) and thrombin. In contrast, the anti-

angiogenic effect occurs through its interaction with thrombospondin,

angiostatin and tumstatin (Hodivala-Dilke et al. 2003).

For example, adhesion of endothelial cells (expressing αVβ3) to vitronectin

leads to tyrosine phosphorylation of VEGFR-2, indicating an involvement of

αVβ3 in VEGFR-2 activation (Soldi et al. 1999). The αVβ3 integrin binds

directly to VEGFR-2, activating VEGFR-2 and leading to β3 subunit

phosphorylation. In turn, the phosphorylated αVβ3 will phosphorylate VEGFR-

2 in the presence of VEGF, indicating that the association of αVβ3 integrin


28
with VEGFR-2 promotes the activation of each receptor. Experiments using

mutant integrin have shown that the cytoplasmic tyrosine residues of β3

integrin (Y747 and Y759) are necessary for successful functional

communication between β3 integrin and VEGFR-2 (Figure 1.7) (Robinson &

Hodivala-Dilke 2011; Robinson et al. 2004). Studies using the same model

found that a mutant β3 (model had mutant β3 with two tyrosine residue Tyr747

and Tyr759 which were unable to induce signalling) integrin is expressed on

the cell surface but it is defective in signalling (Mahabeleshwar et al. 2006).

However, a study using a β3 knockout mouse model showed an induction of

angiogenesis and tumour growth, indicating role for the αV integrin as well as

in angiogenesis such as αVβ3 integrin (Reynolds et al. 2002).

The ability of αVβ3 integrin antagonists to block in vivo angiogenesis supports

a role for αVβ3 integrin in angiogenesis. LM609 anti αVβ3 integrin, a specific

antibody that targets αVβ3 integrin, has been used but LM609 cannot react

with mouse αVβ3 integrin. Thus, the effect of LM609 on angiogenesis was

studied by Brook et al., who used human breast carcinoma cells injected into

chimeric human/mouse model (this is a mouse model in which a piece of skin

is removed and replaced with a sutured piece of human skin). When MCF-

7PB cells were injected into the human skin, LM609 was able to block tumour

growth and angiogenesis in the human skin microenvironment (Brooks et al.

1995).

29
Figure 1.7 Interaction of αVβ3 integrin and VEGFR-2 (Somanath et al.
2009).

1.2.4.2 Function of αIIbβ3 integrin

The αIIbβ3 integrin (also known as GPIIb/IIIa) is expressed on the surface of

platelets, megakaryocytes, monocytes, granulocytes and lymphocytes. It is

inactive in resting platelets but is rapidly activated by thrombogenic stimuli

and promotes binding to fibrinogen (Huang et al. 1993; O'Toole et al. 1990),

von Willebrand factor and fibronectin. This integrin is necessary for platelet

aggregation (Bennett 2005).

Platelet aggregation occurs through the activation of platelets by

physiological molecules such as thrombin (Hodivala-Dilke et al. 1999),

collagen or ADP (Bhavaraju et al. 2011; Gay & Felding-Habermann 2011).

The activation of αIIbβ3 is mediated by a G-protein that acts through protein

kinase C (PKC) to cause a conformational change. Active αIIbβ3 binds to

fibrinogen, a multivalent ligand, through RGD and KQAGDV recognition

motifs, resulting in clustering of the integrin-ligand complex and formation of

adhesive patches on the surface of platelets, thereby allowing platelet cross-

30
linking and thrombus formation (Figure 1.8) (Geiger et al. 2009; Giuliano et

al. 2003; Quinn et al. 2003; Rocco et al. 1993; Ruoslahti 1996; Springer et al.

2008).

Figure 1.8 Active αIIbβ3 integrin in platelet activation and thrombus


formation (Millard, Odde & Neamati 2011)

1.2.5 Role of αVβ3 and αIIbβ3 integrins in cancer development

1.2.5.1 Role of αVβ3 integrin in cancer development

Alteration in the pattern of the αVβ3 integrin receptor is observed in tumour

cells (Arosio et al. 2009; Gasparini et al. 1998; Gladson & Cheresh 1991;

Landen et al. 2008; Max et al. 1997). The expression of αVβ3 integrin has

been detected in many cancer cell lines such as breast (Verbisck et al.

2009), ovarian (Landen et al. 2008) cervical carcinoma (Shen et al. 2006),

glioblastoma (Gladson & Cheresh 1991), melanoma (Petitclerc et al. 1999;

Seftor et al. 1992), prostate carcinoma (Chatterjee et al. 2001), kidney cancer

cells (Arosio et al. 2009) and lung cancer cells (Tsai et al. 2008). The β3 and

αVβ3 integrin receptor are also detected in clinical samples of melanoma

31
(Albelda et al. 1990; Neto et al. 2007) and ovarian carcinoma (Partheen et al.

2009). Breast cancer (Gasprini et al. 1998) and colon cancer show

expression of αVβ3 integrin (Reinmuth et al. 2003).

The αVβ3 integrin plays an important role in a number of cellular process that

lead to cancer development including malignant transformation, tumour

growth and progression, invasion, metastasis (Byzova et al. 2000), apoptosis

(Desgrosellier & Cheresh 2010; Jin & Varner 2004; Li et al. 2001; Lu et al.

2008; Mizejewski 1999; Mousa 2002) cell surface localisation of

metalloproteinases and angiogenesis (Jin & Varner 2004; Varner & Cheresh

1996). Consequently, it plays a key role in the regulation of tumour cell

survival and apoptosis. Signalling by αVβ3 integrin regulates both the

expression and activity of bcl-2 and suppresses the expression of the pro-

apoptotic protein Bax by down regulation of p53. αVβ3 increases the bcl-2:

BAX ratio, which leads to increased tumour cell survival by preventing the

mitochondrial apoptotic pathway that involves the release of cytochrome c

and caspase activation (Martin & Vuori 2004; Uhm et al. 1999). Crosstalk

between αVβ3 and fibroblast growth factor receptor (FGFR) also prevents

apoptosis through the mitogen-activated protein kinase (MAPK) pathway.

αVβ3 also enhances tumour progression by activation of the tyrosine kinase

SRC, which leads to activation of the FAK-independent survival pathway and

promotion of tumour growth (Desgrosellier & Cheresh 2010).

Increased expression of αVβ3 allows tumour cells to bind to extracellular

matrix proteins such as fibronectin, fibrinogen, vitronectin, von Willebrand

32
factor and osteopontin that are present in the tumour microenvironment.

These adhesive interactions provide survival signals for invading endothelial

cells (Desgrosellies & Cheresh 2010). MMP-2 binds to αVβ3 on the cell

surface and this interaction leads to activation of MMP-2 and localizes its

proteolytic activity to the invasive front of the cell (Varner & Cheresh 1996).

Breast, prostate, lung and thyroid tumour cell lines became more invasive

when αVβ3 forms a link with bone sialoprotein and MMP-2 (Karadag et al.

2004). The αVβ3-ERK1/2 pathway is essential for connective tissue growth

factor (CTGF) induced ERK1/2 activation and cellular migration in the MCF-7

breast carcinoma cell line (Chen et al. 2007).

1.2.5.1.1 Role of αVβ3 integrin in breast cancer

αVβ3 is expressed in human breast cancer tissue (Zhao et al. 2007), as well

as in breast cancer cell lines such as MDA-MB-231(Cai et al. 2006; Liu et al.

2009; Manzoni et al. 2009; Takayama et al. 2005). αVβ3 expression

correlates with the metastasis of MDA-MB-435 human breast cancer cell line,

which interacts with platelets to promote arrest of tumour cells during blood

flow and leads to haematogenous metastasis (Felding-Habermann 2003).

Expression of αVβ3 has been shown on cancer cells in the active

conformation and it activates platelets, leading to increased metastatic

activity to the lung. The activated state of αVβ3 in breast cancer cells may be

responsible for the tumour cell-platelet interaction that leads to arrest MDA-

MB-435 tumour cell during blood flow, resulting in increased binding of a

ligand mimetic antibody and increased cell migration toward vitronectin. The

33
metastasis of breast cancer cells was seen to be strongly inhibited by LM609

anti-αVβ3 antibody (Felding-Habermann et al. 2001).

Sloan et al. detected expression of αVβ3 on the cell surface of the 66c14

mammary cell line, which led to metastasis of breast tumour from the

mammary gland to the bone. The increase in metastasis resulted from the

ability of αVβ3 to adhere to vitronectin and invade the basement membrane

and the surrounding stroma, which depended on protease and MMP activity.

αVβ3 expression can induce haptotactic migration towards osteopontin (Sloan

et al. 2006).

Other studies have shown that αVβ3 is important for the development of bone

metastasis; expression of β3 was detected in human breast carcinoma with

bone metastasis and was stronger in tissue with bone metastasis than in

primary tissue. Tumour cells expressing αVβ3 integrin stimulate bone

destruction and osteoclast-mediated bone resorption once they are located in

the bone marrow (Zhao et al. 2007). Transfection of breast carcinoma cells

MDA-MB-231 with αVβ3 integrin or using cells from primary site of bone

metastasis led to increased number and area of osteolytic bone metastases

in vivo, due to expression of active αVβ3 integrin on the cell surface. Inhibition

of αVβ3 function by LM609 inhibited tumour cell invasion and adhesion to

cortical bone (Pecheur et al. 2002).

34
1.2.5.1.2 Role of αVβ3 integrin in prostate cancer

Prostate cancer cells express both αVβ3 and αV (Cooper et al. 2002; van der

Horst et al. 2011; Wang et al. 2005). The expression of αVβ3 may differ

between different prostate carcinoma cell lines and between labs (Saxena et

al. 2012; Zheng et al. 1999). A recent study that examined the expression of

αV and β3 integrin under different conditions reported the expression of αV

and β3 in the prostate carcinoma cell line, PC-3 and in human xenograft

mouse tissue. The expression differed in the in vitro and in vivo environments

(Taylor et al. 2011).

The expression of αVβ3 was correlated with the ability for adherence and

migration of the cells on vitronectin. The PC-3 cells expressing αV and αVβ3

integrin migrated on vitronectin and fibronectin whereas LNCaP cells, which

do not express αVβ3, migrated only on fibronectin. Transfection of LNCaP

cells with the β3 subunit resulted in adherence to vitronectin, which was

inhibited by LM609 (Zheng et al. 1999). In contrast, an immunocytochemistry

study using LM609 found higher expression of αVβ3 in LNCaP than PC-3

cells. After expression was characterised, the cells were treated with cRGDfV

(cyclic peptide: Arg-Gly-Asp-phe-Val) which was more effective in LNCaP

and had no effect on PC-3 cells. Blocking of αvβ3 integrin on LNCaP

membrane by cRGDfV treatment caused FAK cleavage, and reduced

Akt/PKB phosphorylation, which led to activation of caspase-9 and caspase-

3, indicating that cRGDfV induced programmed cell death in the LNCaP cells

(Chatterjee et al. 2001).

35
αVβ3 plays an important role in migration of prostate cancer to the bone;

expression of αVβ3 in PC-3 promoted cancer migration to the bone matrix

(McCabe et al. 2007). Barthel et al. found that prostate cancer extravasation

into bone marrow endothelium under physiological blood flow conditions

depended on the presence of E-selectin, αVβ3 integrin, β1, and Rac1/Rap1

GTPase activity (Barthel et al. 2012).

1.2.5.1.3 Role of αVβ3 integrin in melanoma

Melanoma cells and tissue express αVβ3, with the highest expression in

metastatic melanoma tissue (Hofmann et al. 2000). Using frozen melanoma

sections, Albelda et al. found that the expression of β3 integrin was more

common in invasive melanoma than in primary melanoma: vertical growth

phase (VGP) showed immunoreactivity with β3 integrin while radial growth

phase (RGP) failed to show any reactivity. The expression of β3 integrin was

detected only in metastatic melanoma cells and not in benign melanocytes.

These results suggested that the expression of β 3 integrin could be important

for the development of tumour invasiveness and could be useful as a marker

for melanoma cells entering the aggressive phase of the malignancy

process(Albelda et al. 1990).

Li et al. reported that high expression of αVβ3 integrin in vitro and in vivo can

increase metastasis of melanoma cells by mediating adhesion to vitronectin

(Li et al. 2001). The adhesion of αVβ3-expressing A375M melanoma cells to

vitronectin was inhibited by LM609, whereas the same antibody had no effect

on melanoma cell adhesion to fibronectin and laminin. However, an increase


36
in melanoma cells invasion through the basement membrane was not upon

exposure to either anti αVβ3 antibody or vitronectin. Exposure of melanoma

cells to vitronectin was associated with an increased production of type IV

collagenase as a result of αVβ3-initiated signalling indicated that αVβ3 integrin

had a role in melanoma cell invasion through modulation of protease levels

(Seftor et al. 1992).

Denatured collagen colocalised with αVβ3 integrin in melanoma cells and

attachment of M21 cells to vitronectin and denatured collagen led to an

increase in the bcl-2: BAX ratio. The αVβ3 antagonist LM609 could inhibit M21

cell growth in vivo. This antagonism inhibits tumour growth by inducing

apoptosis through the regulation of bcl-2 and BAX expression, indicating that

the role of αVβ3 integrin in tumour survival involves interaction with the ECM

(Petitclerc et al. 1999).

Voura et al. localised αVβ3 integrin expression on the cell surface of WM293

melanoma cells using immunofluorescence. The expression was localised in

cell-to-cell contact regions when the WM293 cells were co-cultured with

HUVECs and was diffuse before the cells started extravasation, increasing to

be strongest during extravasation. The study reported a 40% inhibition of

melanoma cell transmigration in response to the linear RGD peptide at

concentration 100 µM, whereas a cyclic RGD peptide (c-RGDfV) showed

50% inhibition of melanoma cell transmigration at concentration 5 µM and

had its greatest effect at 5 hours co-culture. Treatment with LM609 at

concentration 40 µg/ml led to 40-50% inhibition, indicating that this

37
melanoma extravasation is at least partially mediated by αVβ3 integrin (Voura

et al. 2001).

1.2.5.1.4 αVβ3 integrin in glioma

αVβ3 is overexpressed in glioblastoma tumour cells such as U-87MG (Cheng

et al. 2005), U-373MG and U-251MG (Mattern et al. 2005) and in endothelial

cells of tumour-associated proliferating microvessels. αVβ3 activation was

related to tumour cell invasion through an enhancement of angiogenesis that

prevents development of hypoxia in brain lesions (Lorger et al. 2009).

Glioblastoma cell line U-251MG also showed expression of αVβ3, which was

involved in cell adhesion to vitronectin (Gladson & Cheresh 1991).

αVβ3 is expressed on clinical glioma tumour samples and its expression

correlates with histological grade (Bello et al. 2001). Different glioblastoma

samples showed heterogenous expression of αVβ3 integrin and most of the

expression of αVβ3 was derived from glial tumour cells (Schnell et al. 2008). A

higher expression of αVβ3 was seen in grade III tumours than in grade I&II

tumours for glioblastoma.

1.2.5.2 Role of αIIbβ3 integrin in cancer development

1.2.5.2.1 Role of αIIbβ3 integrin in melanoma

The expression of αIIbβ3 integrin has been detected in tumour cells or tissues

(Chen et al. 1997; Trikha et al. 1998; Trikha et al. 1997). αIIb expression was

first seen in malignant melanoma by Puerschel et al. by

immunohistochemistry using the anti-αIIb monoclonal antibody sz22 (Santa


38
Cruz). In contrast, none of the tested tissue samples from patients with non-

metastatic melanoma showed αIIb immunoreactivity (Puerschel et al. 1996).

Chen et al. detected the expression of αIIbβ3 in melanoma cell line WM35 by

reverse transcription-polymerase chain reaction (RT-PCR) (Chen et al.

1997). Trikha et al. showed melanoma cells and tissue expressed high

affinity αIIbβ3 (Trikha et al. 1997).

The expression of αIIbβ3 was low in thin melanoma tissue (early stage

tumours) but increased in thick melanoma tissue (later stage disease),

whereas the expression of αVβ3 integrin was intense in thin melanoma. The

importance of αIIbβ3 integrin in human melanoma cell invasion, spreading and

migration was demonstrated using isogenic cell lines. The αIIbβ3 expressing

cells adhered to fibrinogen but couldnot adhere to vitronectin, whereas αIIbβ3

negative cells (expressing only αVβ3 integrin) adhered to vitronectin but

couldnot adhere to fibrinogen, suggesting that expression of αIIbβ3 integrin

suppresses the function of αVβ3 integrin. No difference was observed in the in

vitro growth between αIIbβ3 expressing cells and mock-transfected cells, but

αIIbβ3 expressing cells grew larger tumours in vivo due to a reduction in the in

vivo rate of apoptosis (Trikha et al. 2002). Dome et al. found that expression

of αIIbβ3 integrin in the vertical growth phase upregulate bFGF expression,

thereby providing an angiogenic growth factor for melanoma cells and

promoting tumour growth (Dome et al. 2005).

39
1.2.5.2.2 Role of αIIbβ3 integrin in prostate cancer

The expression of αIIb has been reported in human prostate tumour tissue

(Trikha et al. 1996), as well as in cell lines such as such as PC-3 and DU-145

(Chen et al. 1997).

Trikha et al. detected the expression of αIIb DNA/RNA in prostate carcinoma

tissue using an in situ hybridisation (ISH) technique using an antisense

riboprobe to αIIb mRNA. Prostate cancer tissues produced both αIIb and β3

integrin proteins. The presence of intact αIIbβ3 integrin was confirmed by flow

cytometry, dot blotting and cell adhesion assays. αIIbβ3 was stored

intracellularly and translocated to the cell surface in response to PKC

activation. Monoclonal antibodies directed to αIIbβ3 integrin blocked DU-145

cell invasion (Trikha et al. 1996).

Expression of αIIbβ3 differed between PC-3 and DU-145 cell lines. Although

both expressed αIIbβ3, the expression was intracellular in PC-3 whereas αIIbβ3

was localised on the cell surface of DU-145 cells and was involved in the

focal adhesion sites. DU-145 cells were also more invasive than PC-3. Mice

injected with PC-3 cells developed intraprostatic tumours whereas mice

injected with DU-145 developed tumours that infiltrated the surrounding

tissue and metastasised to the lymph nodes. The function blocking

antibodies, 10E5 and PAC-1 directed to αIIbβ3, inhibited lung colonization in a

tail vein injection model. These antibodies can bind to platelet αIIbβ3 to inhibit

platelet function and therefore platelet tumour interaction. Antibodies can also

block the interaction of DU-145 cells with host tissues by binding to the αIIbβ3
40
integrin expressed by tumour cells. Consequently, combined blockage of

αIIbβ3 integrins on multiple cell types can lead to a blockade of lung

colonisation by DU-145 cells (Trikha et al. 1998).

1.2.5.3 αVβ3 and αIIbβ3 integrin antagonism in cancer therapeutics

The role of integrins in cancer development, metastasis and angiogenesis

discussed in the previous sections suggests that the β3 subfamily may

represent attractive therapeutic targets (Cox et al. 2010; Desgrosellier &

Cheresh 2010; Eble & Haier 2006; Millard, Odde & Neamati 2011; Niu &

Chen 2011; Rust et al. 2002; Shimaoka & Springer 2003). Therapies based

on integrin antagonism that include antibodies, peptides, small molecules

and small interfering RNA (siRNA) are summarised in Figure 1.8 and Table

1.3 (Bisanz et al. 2005; Goodman & Picard 2012; Liu et al. 2008).

1.2.5.3.1 Anti-αVβ3 antibodies as cancer therapeutics

Abegrin (etaracizumab, previously known as vitaxin or MEDI-522) is an IgG1

humanized monoclonal antibody engineered from the murine monoclonal

antibody LM609. LM609 targets αVβ3 and has the ability to reduce tumour

growth in vivo and in vitro by inhibiting the tumour cells from interacting with

underlying extracellular matrix proteins such as fibrinogen. LM609 can block

human angiogenesis and reduce invasion; thus, it may be useful as an anti-

angiogenic approach for treatment of any cancer (Brooks et al. 1995).

Abegrin can inhibit the growth of ovarian cancer in vitro and in vivo (Landen

et al. 2008), although at high doses it can show impaired anti-tumour activity.

41
This is possibly because the high serum level might cause aggregation of the

antibody, limiting its availability (Mulgrew et al. 2006).

A phase I clinical trial conducted on advanced stage metastatic cancer found

Abegrin to have no significant toxicity (McNeel et al. 2005). Abegrin was then

used in a phase II clinical trial on stage IV melanoma, either as a single agent

or in combination with dacarbazine. However, Abegrin showed no tumour

response if used alone and the combination of Abegrin with dacarbazine had

only a low percentage of tumour response. Consequently, Abegrin is no

longer being investigated (Hersey et al. 2010).

1.2.5.3.2 Integrin binding peptides as cancer therapeutics

In addition to antibodies, peptides have been used as tool compounds in

cancer studies (Danhier et al. 2012; Millard, Odde & Neamati 2011; Temming

et al. 2005). Bello et al. examined the effects of the RGD peptide IS201 as an

inhibitor of αVβ3 integrin in vitro and in vivo on malignant glioma cells U87-

MG, 4373-MG, U118-MG and endothelial cells. The IS201 peptide inhibited

glioma growth in vivo, and its effects were associated with a decrease in

angiogenesis and cell proliferation and an increase in apoptosis (Bello et al.

2003).

Another peptide used by Chatterjee et al. who reported the effects of cyclic

cRGDfV and linear RGD peptides in glioma cells. They found that cRGDfV

blocked cell-to-cell adhesion and inhibited either survival or proliferation in

human glioma cells, whereas RGD had no inhibitory effect (Chatterjee et al.

42
2000). The same group also examined the effects of cRGDfV on prostate

cancer and found that it induced apoptosis in LNCaP prostate carcinoma

cells, whereas the linear RGD peptide caused only a partial reduction in the

number of prostate cancer cells. However, cRGDfV had a significant

cytotoxic effect on prostate cancer cells expressing αVβ3 integrin (Chatterjee

et al. 2001).

Another study by Allman et al. showed that cRGDfV inhibited melanoma cell

adhesion in vitro and prevented development of tumours in vivo. However,

the peptide was unable to induce apoptosis in malignant melanoma that

expressed high levels of bcl-2 (Allman et al. 2000). Furthermore, although

cRGDfV apparently induced melanoma cell detachment, the peptide did not

remain bound to the cell surface (Castel et al. 2001).

At present, the most advanced peptide antagonist undergoing clinical

development is Cilengitide which an N-methylated cRGDfV derivative

c(RGDf(NMe)V) that shows high affinity for αVβ3 and αVβ5 integrins. For

further details see section 1.2.5.3.4.1.

1.2.5.3.3 Small molecule integrin antagonists as cancer therapeutics

The small molecule antagonist IH0162 can induce anoikis in vitro through

inhibition of αVβ3 expressing M21 melanoma cells binding to vitronectin. It

was able to inhibit pulmonary metastasis of M21 cell in vivo. IH0162 activates

caspases 3, 8, and 9; both caspase 8 and 9 affect the intrinsic and extrinsic

43
apoptosis pathways. IH0162 can also decrease bcl-2 and survivin, extending

its effects further to the FAK pathway (Zhang et al. 2011).

The antagonism of αVβ3 can be exploited as a therapeutic target to prevent

skeletal metastasis. MDA-MB-435 breast carcinoma cells were transfected

with αVβ3 integrin and injected into tail vein of animal to determine the role of

αVβ3 integrin in development of bone metastasis in vivo. A lower bone

volume to tissue volume ratio, indicating higher bone destruction, was

observed when cells were injected directly into the tibial bone marrow cavity,

and a larger number of αVβ3 integrin expressing tumour cells were found

residing in the bone marrow; these stimulated osteoclast mediated bone

resorption. For example, long-term treatment with the αVβ3 integrin

antagonist PSK1404 resulted in multiple inhibitory effects on cancer cells,

endothelial cells, and osteoclasts and enhanced the anticancer efficiency.

while short term therapy inhibited tumour cell invasion in vivo (Zhao et al.

2007).

The effect of oral administration of XV454 (a specific αIIbβ3 antagonist) on

platelet aggregation, tumour induced thrombocytopenia and metastasis has

been investigated by Amirkhosravi et al. Oral administration of 5 mg/kg

XV545 led to complete inhibition of platelet aggregation within ten minutes.

Tumour cell induced thrombocytopenia was inhibited significantly by both oral

and intravenous administration of XV454 and it had anti-metastatic activity

with a reduced number of metastatic lung nodules seen (Amirkhosravi et al.

2003).

44
Bakewell et al. hypothesised that tumour cells require β3 integrin for

metastasis to bone and for induction of bone osteolysis. ML464 (a pro-drug

antagonist of αIIbβ3) protected mice from metastasis in vivo but it did not

disrupt platelet/tumour interactions in vitro because the pro-drug requires

activation in vivo (Bakewell et al. 2003).

Tirofiban, an αIIbβ3 antagonist, can block the interaction of αIIbβ3 integrin with

the col15 cell adhesion domain of collagen XVII, thereby blocking the

transmigration and invasion of the HSC-3 cell line (squamous cell carcinoma)

normally induced by col15. This transmigration is mediated by factors other

than αIIb. Since the blockade of α5 and αV reduced migration of HSC-3 in

response to col15 by 53%, whereas a 35% reduction in migration was

caused by tirofiban (Parikka et al. 2006).

1.2.5.3.4 Dual αVβ3/αVβ5 integrin antagonism in cancer therapy

Targeting more than one integrin with one compound is becoming an

attractive concept (Sheldrake & Patterson 2009). Dual antagonists can show

important antiangiogenic and antitumour effects, and may represent a

targeted approach for the inhibition of tumour angiogenesis and growth. One

pseudopeptide, ST1646, targets αVβ3/αVβ5 with antiangiogenic effects both in

vivo and in vitro, and has shown inhibitory effects on the growth of human

ovarian carcinoma A2780 cells injected into xenograft tissue in nude mice

(Belvisi et al. 2005).

45
S247 is a small molecule αVβ3/αVβ5 integrin antagonist that has been

investigated in both in vitro and in vivo models. Both continuous treatment

and short-term treatment with S247 were highly effective in reducing

metastasis, either by blocking early events such as stable attachment of

tumour cells to vasculature of distant organs or by blocking their

extravasation from vessels into nearby tissues evaluated by measurements

of gross lung weight, flow cytometric analysis, and histology. The effect of

S247 in inhibition of metastasis at the early stage (during stable attachment

of tumour cell to the vasculature of distant organ) was examined in another

tumour cell line because the 435/HAL cells did not colonise the lung after i.v.

injection. GFP-expressing B16-F10 murine melanoma cells were injected into

the tail vein of mice, following S247 injection just prior to tumour injection.

After first day of injection, 68% decrease in final tumour burden and an 86%

decrease in the tumour-associated gain in lung weight were observed.

Therefore, the study indicated that the use of αVβ3/αVβ5 integrin antagonists

shows promise for treatment of both early and late steps of metastasis

(Shannon et al. 2004).

Treatment with S247 prevented the development of colon cancer liver

metastasis in vivo but had no growth inhibitory effect on the primary tumour.

In vitro, S247 affected both endothelial cells and some types of tumour cell,

causing decreased cell proliferation, adhesion and migration and also

induced apoptosis (Reinmuth et al. 2003). A further study showed the effect

of S247 is increased if it is combined with radiotherapy (Abdollahi et al.

2005).
46
Kumar et al. found that a dual antagonist of αVβ3/αVβ5 may be useful in

blocking tumour induced angiogenesis by using the nonpeptide small

molecule SCH221153, which inhibits the binding of radiolabelled echistatin to

αVβ3/αVβ5 with an IC50 equal to 3.2 and 1.7 nM, respectively. SCH221153 has

the ability to block adhesion of foetal bovine aortic endothelial GM7373 cells

to vitronectin and FGF2. In addition, it inhibits HUVECs proliferation and the

mitogenic activity of FGF2 and VEGF. Testing in the human melanoma Lox

cell line (which expresses low levels of αVβ5) showed SCH221153 to be

ineffective in inhibiting cell proliferation in vitro, but it inhibited tumour growth

in vivo by decreasing the number of blood vessels surrounding a xenograft

tumour (Kumar et al. 2001).

1.2.5.3.4.1 Cilengitide, a dual αVβ3/αVβ5 integrin antagonist, in cancer

therapy

Cilengitide is another dual αVβ3/αVβ5 integrin antagonist. It is a cyclic

pentapeptide that binds to integrin αVβ3 and αVβ5 with high affinity (Alghisi et

al. 2009). Cilengitide has antiangiogenic and anti-proliferative effect in glioma

cells and xenografts (MacDonald et al. 2001; Mas-Moruno et al. 2010;

Mikkelsen et al. 2009; Tabatabai et al. 2010; Taga et al. 2002).

In phase I of a clinical trial, Cilengitide was well tolerated and showed limited

side effects, although the maximum tolerated dose in phase I was not

identified (MacDonald et al. 2008; Nabors et al. 2007). Hariharan et al.

carried out a clinical study using different doses of this peptide (600 or 1200

47
mg/ml) against different solid tumours, and concluded that Cilengitide has a

mild toxicity and can be used as antiangiogenic agent (Hariharan et al. 2007).

In a randomized phase II clinical trial by Reardon et al., patients were

assigned to receive either 500 mg or 2000 mg Cilengitide to determine

whether a higher dose would give a greater response. They recommended

Cilengitide as a safe and non-toxic agent and suggested that it could be used

in combination regimens for glioblastoma, because it had a high anti-tumour

activity (Reardon et al. 2008). Combination of Cilengitide with standard

chemotherapy such as temozolomide was safe and well tolerated (Stupp et

al. 2010).

A recent preclinical study by Mikkelsen et al. reported that treatment with

Cilengitide could lead to prolonged survival if it were given prior to

radiotherapy. Cilengitide increased the sensitivity of U-251MG cells and

HUVECs to radiation, leading to suppression of U-251MG growth and death

of the HUVECs in vitro. Cilengitide dramatically prolonged survival in vivo if it

was used with radiation rather than as a single agent. Further, Cilengitide

decreased the expression of p13k/pAkt, a gene in the NF-kB signalling

network with a recognised role in cell resistance to radiation in vivo

(Mikkelsen et al. 2009).

Cilengitide alone had no effect on breast tumour growth, whereas combined

radioimmunotherapy with Cilengitide (CMRIT) substantially increased the

cure rate (44% cure rate for CMRIT, 20% cure rate for RIT alone). CMRIT

48
increased apoptosis and decreased proliferation more than Cilengitide alone

(Burke et al. 2002).

Reynolds et al. found that nanomolar concentrations of Cilengitide enhanced

tumour growth and tumour vascularisation. When Cilengitide was

administered as a bolus at 200 mg/kg, its concentration in the plasma fell

rapidly from micromolar to nanomolar levels, which was then sustained for 16

to 24 hours after administration. This led to promotion of tumour growth by

direct stimulation of angiogenesis in vitro and in vivo. In β3/β5 deficient mice,

no increases in tumour growth or angiogenesis were observed. Cilengitide

therefore did not promote tumour growth directly but it acted in the cells that

expressed αVβ3/αVβ5 by enhancing cell migration in response to VEGF and

increased VEGFR-2/αVβ3 receptor recycling (Hodivala-Dilke 2008; Reynolds

et al. 2009).

1.2.5.3.5 Dual αVβ3/ αIIbβ3 integrin antagonism

Initial studies showed the antibody m7E3 (a monoclonal antibody created by

joining the Fab fragments of the mouse monoclonal antibody 7E3 to the FC

fragment of human Ig; it has high affinity and specificity for αIIbβ3/αVβ3)

reduced tumour growth and angiogenesis by blocking host αVβ3/αIIbβ3 in vivo

in β3 negative tumour xenografts in rat bone. m7E3 increased tumour-free

survival and decreased tumour volume and weight, but did not have any

effect on tumour metastasis (Engebraaten et al. 2009).

49
The c7E3 Fab fragment binds with equivalent affinity to αIIbβ3 and αVβ3, and

c7E3 can redistribute between αIIbβ3 and αVβ3 integrins. c7E3 completely

inhibited αVβ3-mediated cell adhesion of HUVECs and A375S2 cells to

fibrinogen and gelatin, and partially inhibited adhesion to fibrin and

vitronectin. c7E3 inhibited bFGF-mediated migration of HUVECs and A375S2

cells and also inhibited cell invasion through fibrin. c7E3 reduced proliferation

by inducing apoptosis of endothelial cells and completely inhibited

angiogenesis. Inhibition of both endothelial αVβ3 and the platelet αIIbβ3

receptor inhibited endothelial sprouting stimulated either by bFGF or by

platelets. c7E3 inhibited growth of human melanoma tumours in nude mice,

but blocking of expression of human melanoma cell-expressed αVβ3 integrin

without inhibition of mouse β3 integrin failed to completely inhibit tumour

growth in vivo. Taken together, these results suggested that a combined

blockade of both tumour cell expressed αVβ3 and host β3 integrin (endothelial

αVβ3 and platelets αIIbβ3) could enhance inhibition of tumour growth in vivo

(Trikha et al. 2002).

Another study investigated the contribution of αVβ3 and αIIbβ3 to MDA-MB-231

breast adenocarcinoma cells adhesion to endothelial cell ECM under flow

conditions. The αVβ3 antagonist (SB-273005) had no effect on tumour cell

adhesion to the ECM while αIIbβ3 antagonist lamifiban (Ro 44-9883)

decreased adhesion of tumour cell to ECM. However, the combination of two

antagonists (Ro 44-9883 and SB-273005) resulted in synergistic effect on

adhesion when compared to the effects of single antagonist. Further, the

previous finding of combining two antagonists was confirmed using c7E3


50
Fab, which targets both αVβ3 and αIIbβ3 integrins with the same affinity as

lamifiban Ro 44-9883 and SB-273005. The result was similar, indicating a co-

operation between αVβ3 and αIIbβ3 in the adhesion of MDA-MB-231 cells to

the ECM (Gomes et al. 2004).

Activation of platelets by thrombin leads to an increased capability for

migration by HeLa cells exposed to platelets. Migration and invasion of HeLa

cells decreased after inhibition of αIIbβ3 and αVβ3 integrin by eptifibatide and

RGDWE peptides respectively. HeLa cell migration depended on both

platelets and endothelial cells. Blocking αIIbβ3 on platelets and αVβ3 on HeLa

cells led to decreased adhesion of HeLa cells on monolayer HUVECs. The

study concluded that both αVβ3 and αIIbβ3 participated in the adhesion

interactions between cancer cells and endothelial cells (Liu et al. 2009).

51
Further anti-integrin agents in clinical use and advanced development

Single antagonist Dual antagonists Multiple antagonists

Anti-αVβ3/αVβ5
Anti-α2 Anti-α4 Anti-αIIbβ3 Anti-α5β1 αIIbβ3, αVβ3, αMβ2
Anti- α5β1,
Cilengitide SCH221153 S247 αVβ1, αVβ3,
22
E7820 Tysabri Abciximab αVβ5, αVβ6

22
Tirofiban 22
Eptifibatide
GLPG0187

JSM6427 Volcoximab PF-04605412

Figure 1.9 Anti-integrin antagonists

52
Table 1.3 Further anti-integrin agents in clinical use and advanced development
Agent Integrin Type of Disease indication Stage of Comments Ref
Target agent development
Abciximab αIIbβ3 Antibody Acute myocardial infarction Clinical use Adverse effect include bleeding, (Derer et al.
αV β 3 Unstable angina thrombocytopenia 2009; Jones
αMβ2 Percutaneous coronary Lead to double increase the need for red et al. 2008;
intervention (PCI) blood cells and platelets transfusion Kim et al.
2006;
Schwarz et
al. 2002)
Eptifibatide αIIbβ3 Peptide Percutaneous coronary Clinical use Adverse effect include bleeding, (Giugliano
intervention (PCI) thrombocytopenia and severe hypotension et al. 2009;
Unstable angina Pathak et
Myocardial infarction al. 2008)
Tirofiban αIIbβ3 Small Percutaneous coronary Clinical use Adverse effect includes intracranial (Valgimigli
Molecule intervention (PCI) retroperitoneal bleeding, pulmonary et al. 2010)
Unstable angina hemorrhage, spinal-epidural hematoma.
Myocardial infarction
Tysabri α4 Antibody Multiple sclerosis Suspended Inhibits immune cells from crossing blood (Gani et al.
,
Crohn s disease from clinical vessels to reach the affected organ. 2008; Ismail
use Several patients with MS in a long-term et al. 2009)
clinical trial of Tysabri recently died from
progressive multifocal leukoencephalopathy
(PML).
Treatment possibly associated with
melanoma development.
53
Cilengitide αVβ3/ Peptide Glioblastoma Phase III (Reynold
αV β 5 s et al.
2009)
CNTO-95 αV Antibody Solid tumours Phase I Safe, partial activity against angiosarcoma (Mullamit
ha et al.
2007;
Ning et
al. 2008)
Vitaxin αV β 3 Antibody Lymphoma, Leukemia, Phase II (Brooks
Rheumatoid arthritis, solid et al.
tumours 1995; Cai
et al.
2006)
Volociximab α5β1 Antibody Renal cell carcinoma Phase II Inhibits tumour angiogenesis by blocking (Bell-
Melanoma binding of activated endothelial cells to McGuinn
ARMD fibronectin present in the extracellular matrix, et al.
thereby inducing apoptosis and inhibiting 2011;
tumour growth. Ricart et
It has efficacy against kidney cancer but al. 2008)
showed poor clinical activity as a single
agent to treat melanoma.
PF-04605412 α5β1 Antibody Solid tumours Phase I Inhibits endothelial cell-cell interaction (Li et al.
It has anti-angiogenic and anti-metastatic 2010)
effects.

54
Ro 27-2441 α4β1, α4β7 Small Asthma Phase II (http://clin
molecule completed icaltrials.
gov/show
/NCT000
48022)
EMD525797 αV Antibody Colorectal and ovarian Phase I (Manfred
carcinoma with liver Wirth
metastasis 2013)
IMGN388 No target Antibody Solid tumours Phase I (Vater
revealed by conjugate 2008)
company
GLPG0187 αV β 1 Small Anti-angiogenic Phase I Inhibits osteoclast bone resorption and (van der
αV β 3 molecule Anti-metastasis of prostate angiogenesis in vitro and in vivo. Horst et
αV β 5 and breast cancer to the Inhibits the progression of bone metastasis al. 2010)
αV β 6 bone and the formation of new bone metastasis
α5β1 during the treatment.

JSM6427 α5β1 Small Proliferative Phase 1 Inhibits proliferation of retinal cells. (Zahn et
molecule vitreoretinopathy completed al. 2009)

55
ATN -161 α5β1 Peptide Lung cancer Phase II PHSCN derived from PHSRN of human (Donate
αV β 3 (linear) Renal carcinoma fibronectin in which a cysteine residue et al.
replaces the arginine in the original 2008)
sequence.
PHSRN of fibronectin interacts with α5β1
integrin and increases its avidity for RGD
PHSRN- increases tumour growth
ANT-161 inhibits angiogenesis and tumour
progression.
It inhibits tumour growth and metastasis.

Efalizumab αLβ2 Antibody Psoriasis Suspended Associated with PML fatal side effect through (Frampto
Raptiva from clinical its immune suppressive properties n &
use Plosker
2009)
E7820 α2 Small Solid tumours Phase II It can be used for treatment of advanced solid (Funahas
molecule tumour in combination with cetuximab hi et al.
2002;
Mita et al.
2011;
Semba et
al. 2004)

56
1.3 Aims and objectives

New drugs for the treatment of cancer that can control the tumour

dissemination process are required. As has been reviewed here; the β3

integrin subfamily (αIIbβ3 and αVβ3) plays an important role in this process.

The hypothesis underlying this work is that dual αIIbβ3 and αVβ3 integrin

antagonism will have an enhanced effect by having a direct effect on β3-

expressing tumour cells (compared to antagonism of αIIbβ3 and αVβ3

separately), inhibiting cell migration and dissemination. Furthermore, through

targeting tumour cell interaction with endothelial cells and platelets, this will

also lead to inhibition of angiogenesis and metastasis.

The main aims of the work described in this thesis are to assemble a panel of

human tumour cell lines characterised for their integrin expression which will

be utilised to evaluate potential novel β3 integrin antagonists.

These aims will be achieved through the following objectives:

 The characterisation of αIIbβ3 and αVβ3 integrin expression in human

tumour xenograft tissue.

 Evaluation of the expression of αIIbβ3 and αVβ3 integrins in a panel of

human tumour cell lines with different integrin expression to use in cell

based assays to investigate the effects of β3 integrin antagonists.

 Development and validation of cell based assays to investigate the

effect of β3 inhibition on tumour cell migration.


57
 Evaluation of the effect of potential novel β3 integrin antagonists in

inhibition of human tumour cell migration.

58
2 Chapter 2: Characterisation of integrin αIIbβ3 and αVβ3 expression in

human xenograft tissue in mice using immunohistochemical and

immunoblotting techniques

59
Chapter 2

2.1 Introduction

The development of effective targeted therapies for cancer relies on the

availability of in vivo models that have been characterised for expression of

the target (Teicher 2006). Preparation of an in vivo model can involve

subcutaneous implantation of a tumour cell line or orthotopic grafting at a site

where the tumour commonly metastasises; for example, into the lungs.

Subcutaneous transplantation models are considered effective for

investigating integrins. A study done by Gouon et al. used a subcutaneous

model to determine the difference in αVβ3 integrin expression in

subcutaneous vs. in vitro melanoma cells using HT-144 (weak expression of

αVβ3 integrin) and SK-MEL-2 (high expression of αVβ3 integrin) cell lines. The

HT-144 human melanoma cells showed up-regulation of β3 integrin, which

was correlated with MMP-9 expression, when the tumour was grown in nude

mice (Gouon et al. 1996). Trikha et al. found that subcutaneous implantation

of PC-3 and DU-145 cells, which express αIIbβ3 integrin, was tumorigenic but

not metastatic while an orthotopic in vivo model showed that DU-145 cells

were both tumorigenic and invasive (Trikha et al. 1998).

Tumour implants at a specific site in the model (orthotopic xenograft) produce

tumours with metastasis; these models permit study of compounds which

affect the tumour dissemination process in vivo (Bibby 2004). Such models

have been useful in identifying new therapeutics (Kerbel 2003; Popkov et al.

60
2006) and in anti-integrin drug evaluation. For example, Landen et al. used

an orthotopic model to evaluate the effects of the etaracizumab (anti-αVβ3)

antibody on ovarian carcinoma cell lines such as SKOV-3, A2780, and HeyA.

Etaracizumab was effective in inhibiting tumour growth and invasion (Landen

et al. 2008). Allman et al. used an in vivo model to determine the effect of

cRGDfV peptide on melanoma cells, with A375 administered subcutaneously

into nude mice. cRGDfV was able to decrease melanoma growth and

invasion (Allman et al. 2000). Furthermore, Zhao et al. used an in vivo model

to appraise the effect of the small molecule PSK1404 (anti-αVβ3) peptide on

breast and prostate carcinoma (Zhao et al. 2007). An orthotopic model was

also used to reveal the functional state of αIIbβ3 integrin and its role in tumour

metastasis in vivo. Injection of DU-145 cells had membranous expression of

αIIbβ3 integrin in mouse led to development of tumour which infiltrated in the

surrounding tissue, and metastasised to lymph node. Inhibition of αIIbβ3

integrin with 10E5 antibody (specific for αIIbβ3 integrin) led to block interaction

of DU-145 cells with host tissue and therefore led to blockade of colonisation

of DU-145 cells in the lung (Trikha et al. 1998).

Human prostate carcinoma cell lines with a high expression of αVβ3 integrin

are more tumorigenic in vivo; for example, PC-3 cells with a high expression

of αVβ3 integrin are more tumorigenic when xenografted, and grow more

rapidly when implanted in nude mice than do LNCaP cells which have a low

expression of αVβ3 integrin (Taylor et al. 2012). Trikha et al. reported that a

tumour cell line with expressed αIIbβ3 integrin can develop a large tumour in

61
vivo due to the decreased apoptotic rate in the cells expressing αIIbβ3 integrin

(Trikha et al. 2002).

Several studies have reported a lack of expression of αVβ3/αIIbβ3 integrin in

normal tissues. For example, using immunoblotting, Zheng et al. found no

expression of β3 integrin in normal human prostate tissue, whereas human

prostate adenocarcinoma tissue expressed this protein (Zheng et al. 1999).

Immunohistochemistry of FFPE tissue samples revealed no expression of β3

integrin in normal breast tissue, while breast carcinoma tissue clearly

expressed β3 integrin (Pontes-Junior et al. 2009; Wang et al. 2010). Havaki

et al. found no expression of β3 integrin in normal breast tissue by

immunoblotting, whereas breast carcinoma tissue expressed β3 integrin

(Havaki et al. 2007).

In the present study, immunohistochemistry and immunoblotting were used

to characterise the expression of αVβ3/αIIbβ3 integrin in human tumour

xenograft mouse tissue generated in house. Previous studies have detected

integrin expression using immunohistochemistry on either fresh frozen tissue

sections (Brooks et al. 1995; Li et al. 2001) or paraffin embedded tissue

sections (Bisanz et al. 2005; Brooks et al. 1995).

The mouse LM609 monoclonal antibody that targets the αVβ3 integrin can be

used to detect expression of αVβ3 in angiogenic vessels and in the tumour

cells of many human cancers such as colorectal carcinoma, pancreatic

carcinoma, breast and glioma (Vonlaufen et al. 2001). LM609 has been

reported to successfully detect integrins in breast xenograft mouse tissue,


62
formalin-fixed, paraffin embedded tissue (Brooks et al. 1995) as well as in

frozen tissue sections. LM609 was also used in an in vivo model to detect the

expression of αVβ3 integrin in ovarian carcinoma (Landen et al. 2008). Thus

this antibody was chosen for use in this part of the study to characterise the

expression of αVβ3 integrin in human xenograft mouse tissue.

2.1.1 Aims and objectives

The overall aim of the work described in this chapter is to evaluate αIIb and

αV β 3 integrin expression in human tumour xenografts, using

immunohistochemistry and immunoblotting. The information obtained will be

of great use in guiding the selection of cell lines to be characterised for

inclusion in the in vitro screening panel and for model selection, once novel

antagonists eventually progress to in vivo screening (the latter is beyond the

scope of this thesis).

The aims will be addressed by pursuing the following objectives:

1- Characterisation of the expression of αIIb and αVβ3 integrin in formalin

fixed paraffin embedded human tumour xenograft tissue using

immunohistochemistry.

2- Characterisation of the expression of αIIb and αVβ3 integrin in fresh

frozen human tumour xenograft tissue using immunohistochemistry.

3- Characterisation of the expression of αIIb, αV and β3 integrin in fresh

frozen human tumour xenograft tissue using immunoblotting.

63
2.2 Materials and methods

2.2.1 Materials

All general chemicals were obtained from Sigma-Aldrich (Poole, UK), unless

otherwise specified. Primary antibodies and related secondary antibodies

were used in this part of the study to detect expression of α V, αIIb, β3, αVβ3

and β actin as summarised in Table 2.1. The blocking reagents were bovine

serum albumin (BSA) (Sigma-Aldrich, Poole, UK), normal rabbit serum

(NRS), normal goat serum (NGS), and normal horse serum (NHS) obtained

from Vector Laboratories Ltd (Peterborough, UK). Vectastain Avidin Biotin

Complex (ABC) and the M.O.M. immunodetection basic kit were from Vector

Laboratories Ltd (Peterborough, UK). 3,3-diaminobenzidine

tetrahydrochloride (DAB) chromogen (Sigma-Aldrich, Poole, UK).

2.2.2 Cell lines

The cell lines used to generate the tumour xenograft material: PC-3 prostate

carcinoma, DLD-1 and HCT-116 colon adenocarcinoma, M14 and UACC-62

melanoma, SKOV-3 and A2780 ovarian carcinoma, A549 and H460 human

non small cell lung carcinoma, and PANC-1 pancreatic carcinoma. These

were all obtained from American Type Culture Collection (ATCC, Middlesex,

UK) except for M14 and UACC-62, which was from the US National Cancer

Institute (NCI, USA). One xenograft was analysed per cell line.

64
Table 2.1 Primary antibodies and related secondary antibodies used in immunohistochemistry (IHC) and immunoblotting
(IMB)
Primary Receptor Type Source Secondary Source Secondary antibody Source
antibody antibody (IHC) (IMB)
BV4 Anti-β3 MMAb BV4. ab 7167, biotinylated horse BA2000, Vector - -
Abcam (Cambridge, UK) anti-mouse Laboratories Ltd
(Peterborough, UK)
B7 Anti-β3 MMAb Clone B-7, SC-46655, biotinylated horse BA2000, Vector Polyclonal rabbit anti P0260,
Santa Cruz Biotechnology anti-mouse Laboratories Ltd mouse immunoglobulins Dako Cytomation,
(Santa Cruz, USA) (Peterborough, UK) horseradish peroxidase Glostrup, Denmark
C20 Anti-αIIb PGAb SC-6602, Santa Cruz biotinylated rabbit BA500, Vector - -
Biotechnology anti-goat Laboratories Ltd
(Santa Cruz, USA) (Peterborough, UK)
Q20 Anti-αV PRAb SC-6617-R, Santa Cruz biotinylated goat BA1000, Vector Polyclonal goat rabbit P0448,
Biotechnology anti-rabbit Laboratories Ltd immunoglobulins Dako Cytomation,
(Santa Cruz, USA) (Peterborough, UK) horseradish peroxidase. Glostrup, Denmark
LM609 Anti-αVβ3 MMAb Clone LM609, MAB 1976, biotinylated horse BA2000, Vector - -
Chemicon-Millipore, anti-mouse Laboratories Ltd
Watford, UK (Peterborough, UK)
A1978 β-actin MMAb Sigma-Aldrich, Poole, UK - - Polyclonal rabbit anti P0260,
mouse immunoglobulins DAKO Cytomation,
horseradish peroxidase Glostrup, Denmark
Key: MMAb: monoclonal mouse antibody, PGAb: polyclonal goat antibody, PRAb: polyclonal rabbit antibody.

65
2.2.3 Methods

2.2.3.1 Collection of xenograft materials

Creation of in vivo tumour models was carried out in-house under a project

licence issued by the UK Home Office and in accordance with the UK

National Cancer Research Institute Guidelines for the Welfare of Animals

(Workman et al. 2010). Human tumour cell lines were xenografted in

immunodeficient mice (Harlan, Loughborough, UK) aged six to eight weeks

under general inhalation anaesthesia with recovery. Animal welfare and

tumour growth was monitored frequently and when the tumours were

approximately 500 mm3 in volume, the mice were sacrificed and the tumours

removed and processed as indicated below.

2.2.3.2 Slide coating

Adherence of tissue sections to microscope slides during

immunohistochemical procedures was improved by coating all slides with 3-

aminopropyltriethoxysilane (APES). Slides were washed for two minutes with

acetone, then immersed for two minutes in a freshly prepared solution of 2%

APES in acetone. The slides were washed for two minutes in running tap

water and left to dry at room temperature and then stored in a dust free

environment.

2.2.3.3 Tissue fixation, processing, embedding and paraffin sectioning

Harvested tissue was placed for 24 to 48 hours in 10% neutral buffered

formalin (VWR International Ltd. Poole, UK) before transferring to 70%


66
ethanol. Then tissue was placed in plastic cassettes (Raymond A. Lamb Ltd,

UK) along with labels giving histology numbers. The cassettes were

processed using an automatic tissue processor (Leica TP 1020, Leica

Microsystems, Nussloch, Germany). The tissue processing steps were

achieved by incubating the tissues in 70% ethanol (Fisher Scientific, UK) for

one hour, 80% ethanol for one hour, 95% ethanol for one hour, 100% ethanol

for one hour (twice, in different tanks), 100% ethanol for two hours, Histoclear

for one hour (twice, in different tanks), Histoclear for two hours and paraffin

wax for two hours and thirty minutes (twice, in different tanks).

Finally, the tissue was placed in labelled plastic moulds (Polysciences,

Warrington, USA) which were filled with molten paraffin wax and left to set on

a refrigerated surface. Wax blocks were stored at room temperature until

required for sectioning.

The wax rigidity was increased before sectioning by placing the wax blocks

containing tumour tissues at -20 °C for at least 30 minutes. Then, 5 µm

sections were cut from each wax block using a Leitz rotary microtome (Leica

RM2155, Leica Microsystems, Nussloch, Germany). Sections were collected,

placed in a warm water bath and collected onto APES coated slides. Slides

were placed on a hot plate for thirty minutes to one hour to increase adhesion

of tissue to the slides and then stored at room temperature.

67
2.2.3.4 Haematoxylin and Eosin staining

All steps were carried out at room temperature. Sections were de-

paraffinised prior to staining by passing the sections through xylene and

different grades of ethanol. Sections were immersed in two different changes

of 100% xylene, 50% xylene/ethanol, two changes of 100% ethanol, 90%

ethanol and 70% ethanol for five minutes in each solution.

Deparaffinised sections were stained with Harris’s Haematoxylin solution for

ten minutes. Slides were washed with running tap water, and immersed in

acidic alcohol (0.25% hydrochloric acid in 70% ethanol) for five seconds to

differentiate staining. Differentiated sections were washed in tap water and

placed for two minutes in Scott’s Tap Water (3.5 g sodium bicarbonate and

20 g magnesium sulphate dissolved in 1000 ml H2O). Sections were

counterstained with 1% aqueous Eosin for one minute and washed briefly

with running tap water. Sections were then drained to allow the excess water

to run off.

Stained sections were dehydrated through different grades of ethanol and

xylene, sections were immersed sequentially for two minutes in absolute

ethanol, three minutes in 50% xylene/ethanol and three minutes in 100%

xylene (twice, in different tanks). Finally, sections were mounted with

Distyrene-plasticiser-xylene (DPX) medium (VWR International Ltd. Poole,

UK), a coverslip was applied, and the sections were left to dry.

68
2.2.3.5 Immunohistochemistry

2.2.3.5.1 Immunodetection of αIIb and αVβ3 integrins expression in FFPE

human tumour xenograft sections

2.2.3.5.1.1 Sample preparation

Five µm thick FFPE tissue sections were mounted on APES coated slides.

Sections were deparaffinised and rehydrated by sequential incubation in

100% xylene for ten minutes (twice, in different tanks), 50% xylene/ethanol

for five minutes, absolute ethanol for five minutes (twice, in different tanks),

90% ethanol for five minutes, 70% ethanol for five minutes and distilled water

for five minutes. Sections were incubated with 1% or 3% hydrogen peroxide

(H2O2) for thirty minutes to block the endogenous peroxidase activity.

Sections were washed in PBS, or 0.15 M TBS, pH 7.35 for 3 washes for 5

minutes each.

2.2.3.5.1.2 Antigen retrieval

Using antigen retrieval, attempts were made to expose antigens masked by

fixation. In this part of the study, three types of antigen retrieval methods

were evaluated (citrate buffer, pepsin digestion and proteinase K).

A. Citrate buffer antigen retrieval

Slides were placed in a plastic microwavable container filled with citrate

buffer (10 mM citrate buffer, pH 6). The container was covered with clingfilm

and heated in a microwave (NN-E201W, Panasonic) at 600 W for 3-6 × 5

69
minutes each time. The level of the citrate buffer was checked and topped up

if needed each time. Microwaved slides were allowed to cool at room

temperature for thirty minutes then washed in PBS for 3 washes of 5 minutes

each.

B. Pre-warmed pepsin digestion method antigen retrieval

Sections were incubated at 40 ˚C with pre-warmed pepsin (0.65 mg/ml) in

PBS at for five minutes then washed with PBS for 3 washes of 3 minutes

each.

C. Proteinase K method antigen retrieval

Sections were incubated with 0.1 µg/µl Proteinase K (MB2005, Melford Lab,

Ltd, Suffolk) in distilled water (dH2O) for five minutes at room temperature

then washed in 1X Tris buffered saline (TBS, 1.2 g Tris-HCl, 0.28 g Tris

base, 4.4 g NaCl dissolved in 450 ml dH2O, adjusted to pH 7.4 then volume

adjusted to 500 ml with dH2O), for five minutes.

2.2.3.5.1.3 Antibody incubation

Sections were incubated with normal blocking serum diluted with PBS or TBS

(1.5%) for twenty minutes at room temperature in order to avoid non-specific

binding of the antibody. The blocking serum was selected according to the

species in which the secondary antibody was raised. The excess blocking

serum was carefully removed by blotting with a tissue and primary antibodies

applied (see text for antibody dilution and incubation time). Sections were

then washed in PBS (3 washes for 3 minutes each). Secondary antibody;


70
horse anti mouse, goat anti rabbit, or rabbit anti goat (dilution, 1:200) was

applied for thirty minutes at room temperature, washed as described for the

primary antibody, incubated with peroxidase-labelled streptavidin for thirty

minutes at room temperature and then washed in 3 changes of PBS for 3

minutes in each change. The immunoreactivity was visualised by incubating

sections with DAB chromogen solution for two to five minutes. Sections were

then washed in dH2O for five minutes and counterstained with Harris’s

Haematoxylin solution for twenty seconds. Sections were washed well in

running tap water, and then incubated in Scott’s Tap Water substitute for two

minutes.

Sections were dehydrated and cleared by incubation in 70%, 90%, absolute

ethanol (twice, in different tanks), 50% xylene/ethanol for three minutes each,

xylene (two different tanks for five minutes each). Finally, coverslips were

mounted with DPX medium and left to dry at room temperature. The slides

were examined by light microscope (Leica DMRB, Leica Microsystems,

Wetzlar, Germany).

2.2.3.5.2 Immunodetection of αIIbβ3 and αVβ3 integrins expression in

frozen human xenograft tumour

2.2.3.5.2.1 Sample preparation

Fresh frozen human tumour xenografts (section 2.2.2) were sectioned (5 µm)

using a Cryostat (Leica CM1100, Leica Microsystems, Nussloch Germany)

71
and mounted on APES coated slides. Slides were air-dried at room

temperature then stored at -20 °C.

2.2.3.5.2.2 Labelling procedure

Slides were removed from the freezer and allowed to equilibrate at room

temperature for about twenty minutes. Sections were fixed with acetone,

methanol or 4% paraformaldehyde (PFA) for ten minutes, air-dried for ten

minutes then rehydrated in PBS for ten minutes. Sections were placed in a

humidified chamber and the non-specific binding of antibodies was blocked

by incubating with either normal serum (NS) diluted in PBS (1.5%) or 5%

BSA for twenty minutes at room temperature. The excess blocking solution

was removed and the primary antibody was applied (Table 2.1). Sections

were then washed in PBS (3 washes of 3 minutes each), incubated with

1:200 secondary antibody (Table 2.1) and washed as described for primary

antibody. Sections were incubated for thirty minutes with ABC peroxidase

labelled streptavidin at room temperature. The immunoreactivity was

visualised with DAB, counterstained with Harris’s Haematoxylin and mounted

with DPX as in section 2.2.3.5.1.3.

2.2.3.5.3 Immunodetection of β3 subunits and αVβ3 integrin expression

in FFPE and frozen human tumour xenograft sections by using

a mouse on mouse (M.O.M.) kit

The M.O.M. kit is specifically designed to localise mouse primary antibodies

on mouse tissues and should eliminate background staining in mouse tissue.

72
The M.O.M. kit was used during immunolocalisation for mouse primary

antibodies using FFPE and fresh frozen sections.

2.2.3.5.3.1 Sample preparation

FFPE tissue sections 5 µm-thick were de-waxed and rehydrated as stated in

section 2.2.3.5.1.1. Following rehydration and antigen retrieval sections were

treated with 3% hydrogen peroxide (H2O2) in order to block endogenous

peroxidase activity and then washed twice in PBS for two minutes.

Frozen sections were prepared as in section 2.2.3.5.2.1.

2.2.3.5.3.2 Preparation of M.O.M. kit working solutions

The mouse Ig blocking reagent was prepared by adding two drops of stock

solution to 2.5 ml of PBS. The diluent solution was prepared by diluting 600

µl of protein stock solution with 7.5 ml of PBS. Biotinylated anti-mouse IgG

reagent was prepared by mixing 10 µl of stock solution with 2.5 ml of diluent

solution prepared as above.

2.2.3.5.3.3 Antibody incubations

Blocking reagent was applied to the sections before the application of the

primary antibody in order to avoid non-specific binding, and incubated for one

to two hours followed by washing with PBS (2 washes for 3 minutes each).

Sections were incubated with diluent for five minutes, the excess diluent

removed, and the primary antibody directly applied. All incubations were

undertaken in a humid chamber at room temperature for all the

73
immunolabelling steps, except for primary antibody, which was either at room

temperature or at 4 °C depending on the optimised conditions for each

antibody. The negative control was incubated with M.O.M. mouse blocking

solution for the same period of time. Sections were then washed in PBS (2

washes for 3 minutes each). M.O.M. biotinylated anti-mouse IgG reagent was

applied for ten minutes then sections were washed in PBS (2 washes for 3

minutes each). Sections were incubated with the peroxidase-labelled

streptavidin for thirty minutes at room temperature and then washed in PBS

(2 washes for 3 minutes each). Sections were counterstained with Harris’s

haematoxylin, differentiated in Scott’s Tap Water, dehydrated and mounted

as described in section 2.2.3.5.1.3.

2.2.3.6 Immunoblotting (Western Blotting)

2.2.3.6.1 Materials

Primary and secondary antibodies used in this part of the study are

summarised in Table 2.1 and section 2.2.1.

2.2.3.6.2 Tissue homogenisation

Human xenograft mouse frozen tissue samples (-80°C) were weighed and

cut into approximately 2 mm3 pieces. Each piece was placed in a 1.5 ml

microfuge tube with lysis buffer (1:3 w/v compared to the weight of tissue to

be lysed). The lysis buffer was either PBS buffer alone, or was assembled as

follows: 2x lysis buffer [100 mM Tris pH 8.8, 5 mM EDTA, 300 mM NaCl, 2%

Triton X-100], protease inhibitor cocktail (Roche, Mannheim, Germany) and

74
water. The samples were incubated on ice for thirty minutes, and then

homogenised using an Ultra Turax homogeniser (IKA Labortechnik,

Germany), for 3 pulses of 5 seconds with cooling on ice between each

homogenisation to avoid overheating. The homogenate was centrifuged for

different speeds and times, and both the supernatant and pellet collected.

The samples were aliquoted and stored at -80 °C for later use.

2.2.3.6.2.1 Bradford assay

Protein concentration was measured using the Bradford Assay (Bradford

1976). Serial dilutions for six protein standards (0.0625 - 1.0 mg/ml) were

prepared using 2 mg/ml of BSA dissolved in dH2O. After preparing serial

dilutions, 50 µl from each BSA dilution was added to 1.5 ml of Bradford

reagent. All tubes were vortexed and left to stand at room temperature for ten

minutes to allow Coomassie Brilliant Blue dye to bind to protein. Dilutions of

tissue lysates were prepared in dH2O (50 µl in total) and added to 1.5 ml of

Bradford reagent. 1 ml from each BSA dilution (standard) and lysate were

vortexed and transferred to cuvettes. Absorbance readings were recorded at

595 nm using a spectrophotometer (Thermo Scientific Multiskan Spectrum,

1500, Thermo Fisher Scientific, Finland) and a standard curve plotted. If the

sample absorbance was outside of the range of the standard curve, then

appropriate dilutions were carried out to bring the reading within the range of

standard curve (Figure 2.1).

75
1
0.9
Absorbance at 595 nm 0.8
0.7
0.6
0.5
0.4
0.3
0.2
0.1
0
0 0.2 0.4 0.6 0.8 1

Protein concentration mg/ml

Figure 2.1 Representative calibration curve using Bradford assay.


Each data point represents the mean +/- SD of 3 readings at 595 nm. Linear
regression analysis gives a straight line with r2= 0.99.

2.2.3.6.2.2 SDS-Polyacrylamide Gel Electrophoresis

Proteins were separated using pre-cast 8% polyacrylamide SDS gels (Bio-

Rad, Hertfordshire, UK). The gel was immersed in running buffer (5X Tris-

glycine electrophoresis buffer: 25 mM Tris, 250 mM glycine pH 8.3, 0.1%

SDS). 20-75 µg of protein was diluted in 20 µl SDS loading buffer (50 mM

Tris Cl, pH 6.8, 100 mM dithiothreitol, 2% SDS, 0.1% bromophenol blue, 10%

glycerol) and heated at 70 °C for ten minutes followed by rapid incubation on

ice to denature proteins. 20 µl of sample was loaded into each well.

PageRulerTM Plus Prestained Protein Ladder marker (SM1811, Thermo

Fisher Scientific Inc. Rockford, USA) was loaded into first lane. The marker

76
had a range of size from 10 kDa to 250 kDa. The electrophoresis was run at

20 mA for 90 minutes to separate the proteins.

2.2.3.6.2.3 Western Blot for αV and β3 Integrins

Proteins were transferred from the SDS-polyacrylamide gel to nitrocellulose

membrane (GE Healthcare Life Sciences, Amersham, Buckinghamshire, UK)

using transfer buffer (2.9 g glycine, 5.8 g Tris base, 0.37 g SDS, 200 ml

methanol, 800 ml water). Protein transfer was run at 42 mA for 120 minutes.

After transfer, membranes were immersed in Ponceau S solution to verify

protein transfer, and the SDS-polyacrylamide gel was stained with

Coomassie Brilliant Blue to confirm that all proteins were transferred to the

membrane.

2.2.3.6.2.4 Ponceau S Stain

Protein blotted onto the nitrocellulose membrane was stained with Ponceau

S (1 g Ponceau S plus 50 ml acetic acid, made up to 1 l with distilled water)

for one to three minutes. The membrane was scanned (HP scan jet 2400)

then washed with distilled water three times in order to destain the

membrane. Figure 2.2A shows a representative Ponceau S membrane

where successful transfer has taken place.

2.2.3.6.2.5 Coomassie Brilliant Blue Staining

Complete protein transfer was confirmed by immersing SDS-polyacrylamide

gels in Coomassie Brilliant Blue solution (dissolved in methanol: H2O [1:1 v/v]

with 10% acetic acid) and fixing and staining them for one hour. The gels
77
were then destained in a solution of 40% methanol plus 10% acetic acid and

50% water. Gels were scanned with HP scan jet 2400. Figure 2.2B shows a

representative Coomassie Brilliant Blue stain gel where successful transfer of

most of the proteins has taken place.

A B

Figure 2.2 Confirmation of successful protein transfer


(A) Sample membrane stained with Ponceau S indicating that protein has
been transferred to the membrane from (B) SDS stained with Coomassie
Brilliant Blue and as can be seen most of the protein transferred to the gel.

2.2.3.6.2.6 Immunoblotting of nitrocellulose membrane with anti-β3 and

anti-αV integrin subunits

The nitrocellulose membranes were blocked with either 5% non-fat dried milk

in PBS with 0.05% Brij-35 solution (B4184) or 5% BSA in PBS- with 0.05%

Brij-35 solution for one hour at room temperature, followed by application of

primary antibody. The membrane was then washed with PBS with 0.05%

Brij-35 solution (3 washes for 10 minutes each) followed by application of

secondary antibody as specified in Table 2.1. The ECL plus Western blotting

detection system (NEL104, Perkin Elmer LAS) reagents A and B were mixed

78
in a 1:1 ratio and applied to the membrane for one minute after washing the

membrane with PBS with 0.05% Brij-35 solution (3 minutes for 5 minutes

each). The membrane was exposed to Kodak Scientific imaging film

(Amersham Biosciences) in the dark for five minutes. Films were developed

using IIford Rapid Multigrade developer (1/10 dilution) for three minutes,

rinsed with water and fixed in IIford rapid film and paper fixer solution (1/10

dilution) for two minutes. Expression of the integrin subunits was semi-

quantified by using Molecular Imager FX System (Bio-Rad Laboratories, Inc.,

UK) by dividing the signal intensity of the expression of αV or β3 protein by

the signal intensity of the expression of internal control β-actin.

79
2.3 Results

2.3.1 Detection of the expression of αIIbβ3 and αVβ3 integrin in human

tumour xenograft tissue by IHC

2.3.1.1 Tumour xenograft

Haematoxylin and Eosin (H & E) staining was carried out on M14 human

xenograft tissue sections. Microscopic examination revealed the tumours to

be poorly differentiated due to lack of organisational structure (Figure 2.3, A

and B).

A B

Figure 2.3 Haematoxylin and Eosin stain in M14 human melanoma


xenograft paraffin embedded tissues
Haematoxylin and Eosin stain shows histology of melanoma tissue. The
black arrows indicate tumour cells. Bar length = 200 µm (A) and 60 µm (B).

80
2.3.1.2 Studies on FFPE tissue samples

Detection of the expression of αIIbβ3 and αVβ3 integrin was attempted to

optimise by evaluating several antibodies in FFPE sections, and investigating

the effect of changing the antigen retrieval method, blocking procedure,

antibody incubation time and concentration.

Primary antibodies Q20 (anti-αV), C20 (anti-αIIb), BV4 (anti-β3), B7 (anti-β3)

and LM609 (anti-αVβ3) (Table 2.1) were used to characterise integrin

expression in FFPE tissue. However, assessment of all variables (as

summarised in Figure 2.4) revealed that all methods were unsuccessful for

immunodetection of αV, αIIb, β3 and αVβ3 (as shown in Figure 2.5). All

antibodies except Q20 (anti-αV) showed non-specific immunolabelling in both

sections with and without primary antibody. Q20 (anti-αV) had no

immunolabelling in the section without primary antibody whereas the section

with primary antibody had non-membranous immunolabelling.

81
Studies of FFPE tissue samples such as M14, UACC-62, PC-3, DLD-1 & HCT-116

1-Antigen retrieval

A-Proteinase K B-Pepsin digestion C-Citrate buffer

Damage tissue All primary antibodies: BV4 & B7 (anti-β3), Q20 (anti-αV), C20 (anti-αIIb) &
LM609 (anti-αVβ3) had non-specific immunolabelling in sections without
primary antibody. Q20 had non membranous immunolabelling.
Figure 2.5, A & B Figure 2.5, C & D
2- Blocking

A-Normal serum B-1-5 % BSA


Figure 2.5, E - H Figure 2.5, I - L

No change in the results with all antibodies

3- Primary antibody dilution :1:50 – 1:400

1 hour room temperature Overnight 4 C


Figure 2.5, M & N

4- Secondary antibody dilution 1:200-1:800. Figure 2.5, O & P

All attempts tried gave same result: BV4, B7, C20 &
LM609 gave non specific immunolabelling in
sections without primary antibody. Q20 had non
membranous immunolabelling.

Figure 2.4 Optimisation of IHC to detect αIIbβ3 and αVβ3 integrin


expression in FFPE human xenografts, using different methods

82
Proteinase K No primary antibody Pepsin No primary antibody
A B C D

B7 (anti-β3)

B7 (anti-β3)
Antigen retrieval
Citrate buffer No primary antibody
Citrate buffer No primary antibody
E F G H

Q20 (anti-αV)
B7 (anti-β3)
5% BSA No primary antibody 5% BSA No primary antibody
Blocking

I J K L

Q20 (anti-αV)
B7 (anti-β3)
Ab dilution / incubation

PAb Dilution No primary antibody PAb Dilution No primary antibody

M N O P

Q20 (anti-αV)
B7 (anti-β3)

Figure 2.5 Optimisation of IHC to detect αV and β3 integrin expression in


FFPE M14 human melanoma xenografts using Q20 and B7
M14 tissue (A-H & M-P), blocked with normal serum and immunolabelled
with primary antibodies: B7 (A-F) or Q20 (G&H) at dilution 1:50, overnight
incubation 4 °C. Proteinase K damaged M14 tissue immunolabelled with B7
(A) and without B7 (B). Pepsin digestion damaged M14 tissue with B7 (C)
and without B7 (D). A 30 minute citrate buffer antigen retrieval duration was
effective in sections immunolabelled with B7 (E) but non-specific
immunolabelling developed in sections without B7 (F). A 30 minute citrate
buffer antigen retrieval used in a M14 section immunolabelled with Q20 (G)
showed non-membranous immunolabelling whereas a section without Q20
(H) did not develop any non-specific binding. Under the same conditions with
blocking changed to 5% BSA, M14 immunolabelled with B7 (I) had
membranous immunolabelling but a section without B7 (J) developed non-
specific binding. Under the same conditions, non-membranous
immunolabelling developed in a section with Q20 (K) and a clean section
without Q20 (L). Dilution of B7 to 1:400 and incubation for 1 hour at room
temperature gave the same pattern of sections with (M) and without B7 (N).
Increasing the dilution of primary antibody to 1:400 and the secondary
antibody to 1:800 did not change the result in sections immunolabelled with
Q20 (O) and without Q20 (P). Sections shown are a representative example
of three independent experiments done using different primary antibodies on
different human tumour FFPE tissue. B7 in M14 tissue was chosen as a
representative example with the same results being seen for the other
primary antibodies used. Bar length = 60 µm.
83
2.3.1.3 IHC studies on Frozen Sections.

In order to avoid adverse effects of fixation and processing issues associated

with FFPE tissue, frozen sections were also evaluated. Frozen sections were

used with the same antibodies as used for the paraffin sections. Figure 2.6

summarises the steps of optimisation for different primary antibodies used to

characterise expression of αV, αIIb, β3 and αVβ3 integrins by IHC. The results

were similar to those obtained with FFPE tissues (Figure 2.7).

84
Studies of frozen tissue samples such as M14, UACC-62, SKOV-3, A2780, A549, H460, PANC-1, DLD-1, HCT-116 & PC-3

1- Differentfixatives tried

A-Acetone B-Methanol C-4% PFA

Damaged the tissue Q20 (anti-αV) had diffuse immunlabelling and clean section without primary
antibody .Other antibodies such as BV4 & B7 (anti-β3), C20 (anti-αIIb) & LM609
(anti-αVβ3) had immunoreactivity in both sections,with and withoutPAb.

2- Block endogenous peroxidase with 1%H2O2 for 1, 5 & 20 minutes

3- Block non specific binding of primary antibody

A-Normal serum B-5% BSA

4- Primary antibody at dilution 1:50 -1:400 incubated: 1 hour


at room temperature or at overnight 4 C.

5- Secondary antibody :1:200-1:800, 30 minutes at room


temperature then wash 3 5 minutes with PBS.

6-Avidin biotin complex (ABC) incubation


time decreased from 30 to 15 minutes

7- DAB time decreased from 5 to 2 minutes.

Figure 2.7

Figure 2.6 Optimisation of the protocol used to characterise expression


of αIIbβ3 and αVβ3 integrin in frozen human xenograft mouse tissue by
IHC

85
Acetone No primary antibody 1% H2O2 No primary antibody
A B G H

Block EP
Methanol No primary antibody 5% BSA No primary antibody
Different fixation

C D I J

Block NSB
4% PFA No primary antibody primary antibody No primary antibody
Primary antibody D/I
E F K L

Figure 2.7 IHC of αV integrin expression in human xenograft mouse


frozen tissue using Q20
IHC of M14 with anti αV antibody Q20: non-membranous immunolabelling
developed further to tissue damage in tissue fixed with acetone (A & B),
methanol (C & D) and diffuse immunolabelling in tissue fixed with 4% PFA,
blocked with NGS and incubated with 1:50 Q20 for overnight incubation at 4
°C (E & F). The immunolabelling of M14 tissue blocked with 1% H2O2 for
twenty minutes (G & H) did not change when blocking was changed to 5%
BSA (I & J) or when the Q20 incubation time was reduced to one hour at
room temperature and was diluted to 1:400 (K & L). EP: endogenous
peroxidise, NSB: non-specific binding, D/I: dilution/incubation. Bar length =
60 µm.

One reason why the use of antibodies for αV, αIIb, β3, and αVβ3 integrin was

unsuccessful for immunodetection in FFPE human tumour xenograft and

frozen tissues that is the use of mouse antibodies to detect the expression in

86
mouse tissue led to non-specific binding developing in sections without

primary antibody. Therefore, the next step was to address this issue using a

kit specifically designed to allow the use of mouse antibodies on mouse

tissues.

2.3.2 Immunodetection of β3 and αVβ3 integrin subunits using an

M.O.M. kit.

2.3.2.1 M.O.M. Immunodetection of β3 and αVβ3 integrins in FFPE

sections.

The kit was first used on FFPE sections, where M14 sections were incubated

with B7 (anti-β3), BV4 (anti-β3) or LM609 (anti-αVβ3), (changing variables as

shown in Figure 2.8). Different conditions were tested in order to optimise

final conditions of the above primary antibodies, such as dilution and

incubation: B7 gave concentration-dependent membranous immunoreactivity

at dilutions of 1:50 and 1:200. However, non-specific binding developed in

the blood vessels in the sections incubated with the blocking serum instead

of the B7 primary antibody, whereas sections without primary antibody and

without a secondary antibody did not develop any non-specific binding. This

result indicates that the non-specific binding that developed in the negative

control (without primary antibody) without using the M.O.M. kit was due to the

reaction of mouse secondary antibody on mouse tissue (Figure, 2.9 A - F).

The results of experiments on M14 xenograft sections immunolabelled with

B7 were confirmed by immunolabelling other human xenograft tissue that

87
supposedly have different expressions of β3 integrin; PC-3, DLD-1, HCT-116

and H460. As shown in Figure 2.10, low expression of β3 was found in PC-3

(A and B), weak expression in DLD-1 (C and D) and H460 (G and H), and no

expression was observed in HCT-116 (E and F).

88
Studies of FFPE tissue samples such as M14, using M.O.M kit

Citrate buffer antigen retrieval

3 5 minutes cycle 3 6 minutes cycle

Block endogenous peroxidase

1 % H2O2 3 % H2O2

Primary antibodies B7 and BV4 (anti-β3) & LM609 (anti-αVβ3) used at dilution 1:50

1 hour room temperature Overnight 4 C

Secondary antibody
1:250 for 10 minutes

BV4 and LM609 developed non-specific binding in sections without primary antibody.
B7 developed non-specific binding in section without primary antibody mainly in blood vessels.

B7 used only

1- Section had 30 minutes antigen 2- Same treatment as 3- Same treatment as section 1


retrieval, 3% H2O2 treatment, section 1 except using except using PBS instead of
immunolabelled with B7, 1:50, 1 hour PBS instead of primary primary antibody and secondary
at room temperature. Secondary antibody. antibody.
antibody 1:250 for 10 minutes.

M14 tissue had membranous M14 tissue had non-specific M14 tissue clean no reactivity
expression of β3 integrin. binding in blood vessels only. observed.

Figure 2.8 IHC detection of αVβ3 and β3 integrin in human FFPE tumours
with an M.O.M. kit

89
A B C

D E F

Figure 2.9 IHC detection of β3 integrin expression using B7 with M.O.M.


kit in FFPE M14 melanoma human tumour xenografts in mouse
Membranous expression was observed in M14 sections treated for 30
minutes with citrate buffer as a method of antigen retrieval, blocked with 3%
H2O2, immunolabelled with 1:50 B7 for one hour at room temperature (C) with
clean section without primary antibody (B) just the blood vessels that showed
non-specific binding. The non-specific binding disappeared when the same
tissue (used in B & C) had no primary and no secondary antibody (A). 1:50
dilution of B7 overnight incubation at 4 °C, gave a high background signal (E)
while a section without B7 had non-specific binding in blood vessels (D).
Dilution of B7 to 1:200 and incubation at room temperature (F) changed the
intensity of signal in comparison to Figure C. Bar length = 60 µm.

90
No primary antibody B7: primary antibody
A C E G

B D F H

Figure 2.10 IHC detection of β3 integrin expression in FFPE human


tumour xenografts in mouse using B7 with M.O.M. kit
All tumour tissues were treated with citrate buffer as a method of antigen
retrieval for 30 minutes, blocked with 3% H2O2 and immunolabelled with B7
1:50 for 1 hour at room temperature. PC-3 (A & B), DLD-1 (C & D), HCT-116
(E & F) and H460 (G & H) were used as comparison with the expression of
β3 seen in M14 tissue in Figure 2.9, C. Low expression of β3 was detected in
PC-3 (A), very weak in DLD-1 (C) and H460 (G) and no expression in HCT-
116 (E). Sections without B7 (B, D, F, H) developed non-specific binding in
blood vessels in a similar manner to M14 (Figure 2.9, B). Bar length = 60 µm.

91
2.3.2.2 M.O.M. Immunodetection of β3 and αVβ3 integrins in frozen

sections.

It was hoped that the use of the M.O.M. kit would enable the characterisation

of the β3 expression in FFPE xenograft tissue. However, sections without

primary antibody still showed non-specific binding. The same antibodies used

on FFPE sections were used to characterise the integrin expression in frozen

sections using the M.O.M. kit and optimisation of conditions described in

Figure 2.11. The expression of β3 was detected in PC-3 using BV4 (anti-β3)

(Figure 2.12). B7 (anti-β3) and LM609 (anti-αVβ3) showed non-specific

immunolabelling in both controls. BV4 (anti-β3) was further used to

investigate the expression of β3 in other human tumour xenograft mouse

tissues such as UACC-62, A549, H460, PANC-1, A2780 and SKOV-3, but

non-specific positive immunolabelling resulted. The same non-specific

immunolabelling was obtained with B7 (anti-β3) and LM609 (anti-αVβ3).

92
Studies of frozen tissue samples using M.O.M

Fixation

Ice cooled acetone 4% PFA

Block with 0.1% H2O2 Block with 1% H2O2

BV4 (anti-β3) B7 (anti-β3) LM609 (anti-αVβ3)

Primary antibody 1:50 -1:400, 1 hour room temperature.

Non-specific immunolabelling developed in both controls

Increase blocking time from 1 hour to 2 hours

BV4: high membraneous expression with clean B-7 & LM609: non-specific immunolabelling developed in
section without primary antibody in PC-3. both controls.

Screen other tumours such as A549, H460,


SKOV-3, A2780, PANC-1 & UACC-62.

Not all tumours screened developed the same


membranous expression found in PC-3, further
to non-specific binding developed in section
without primary antibody.

Figure 2.11: IHC optimisation for αVβ3 and β3 integrin expression in


frozen sections using the M.O.M. detection kit.

93
Prostate carcinoma PC-3
BV4(anti-β3) No primary antibody

BV4(anti-β3) No primary antibody BV4(anti-β3) No primary antibody BV4(anti-β3) No primary antibody


Pancreatic carcinoma

A2780 - Ovarian carcinoma - SKOV-3


H460 - Lung carcinoma - A 549
PANC
Melanoma
UACC-62

Figure 2.12 Expression of β3 integrin in human tumour xenograft frozen tissues immunolabelled with BV4 (anti-β3) using
M.O.M. detection kit
Tissue was fixed with 4% PFA, treated with 1% H2O2, blocked for 2 hours and immunolabelled with 1:50 BV4 for one hour at room
temperature, PC-3 had a membranous immunolabelling and no non-specific binding developed in section without primary antibody.
Screening a panel of human tumour cell line xenografts in mouse did not give similar results. Non-specific immunolabelling
developed in both sections with and without BV4. Bar length = 60 µm.
94
2.3.3 Detection of the expression of αIIb, αV, and β3 subunits in

homogenised tumour xenograft mouse tissue by immunoblotting

Integrin expression was also evaluated using immunoblotting (IMB) to

characterise the expression of αIIb, αV, and β3 integrin in human xenograft

mouse tissue. IMB was optimised for each antibody, taking into consideration

the type of lysis buffer, centrifugation speed and time, and protein

concentration loaded in the gel. The optimisation steps are summarised in

Figure 2.13. After the conditions for protein extraction had been optimised,

the optimised method was used for detecting expression of αV and β3 integrin.

No expression of αIIb was detected, probably because the cell lines tested

expressed αIIb at negligible levels (in-house data). Once again, non-specific

binding developed in protein from xenograft tissues with B7 (anti-β3) whereas

human tumour cell lines (Chapter 3) did not develop any non-specific binding

under the same conditions (Figure 2.14).

95
Optimise protocol for protein extraction from human xenograft mouse tissue

1- Buffer

PBS Lysis buffer

2- Centrifugation for different speed & time

9000 rcf, 20 minutes 20000 rcf, 20 minutes 100000 rcf, 1 hour

Supernatant collected Pellet resuspended either in lysis buffer or PBS

PBS excluded Lysis buffer selected at 9000 rcf, 20 minutes

Protein concentration

7.5 µg 25 µg 50 µg

It was selected as an optimum extraction of protein using lysis buffer and


centrifuged using 9000 rcf for 20 minutes .

B7 (anti-β3), mouse antibody had non-specific binding with mouse tissue. Confirmed by incubation of the
membrane without primary antibody (B7) and another membrane without primary antibody and without
secondary antibody in presence of human tumour cell line that did not develop any non -specific binding.

Figure 2.13 Optimisation steps for extraction of protein from human


xenograft mouse tissue.

96
B-7 No PAb No PAb, no 2nd Ab

6- M14 Tissue
2- M14 Tissue

4- M14 Tissue

7- M14 Cell
3- M14 Cell

5- M14 Cell
1-Marker
132

Non specific binding 95

72

Figure 2.14 Expression of β3 subunit in M14 human xenograft mouse


tissue using B7 by IMB
β3 expression was detected on the blot at 125 kDa. B7 mouse antibody had
non-specific binding with M14 human xenograft tissue (Lane 2). Non-specific
binding to mouse protein was confirmed by incubation the membrane without
B7 (Lane 4) and another membrane without B7 and without secondary
antibody (Lane 6). Protein extracted from M14 cells grown in vitro did not
demonstrate any non-specific binding (Lane 3, 5 and 7).

97
2.3.3.1 Expression of αV and β3 integrin in homogenised human

xenograft mouse tissue compared to negative control tissue

and human tumour cells

Once blotting conditions were optimised for each antibody, the expression

level of αV and β3 integrin subunits was evaluated in a panel of human

tumours. Figure 2.15 shows a representative blot and analysis using the

Molecular Imager FX System.

98
Cell Tissue Cell Tissue

Mouse liver

Mouse liver
UACC-62

UACC-62
HCT-116

HCT-116
Marker

DLD-1

DLD-1
HT-29

HT-29
PC-3

PC-3
M14

M14

M14

M14
αV
132
92 A B

132
β3
92
72 C D
β-actin 1

3
Expression of αv and β3 integrin

αV
αV
related to β-actin control

2.5 β3
β3
2
106

1.5

0.5

C: cell, T: tissue

Figure 2.15 Expression of αV and β3 integrin in homogenised tissue


In all membranes, 20 µg of M14 and HT-29 cell protein were loaded and 50 µg
of homogenised tissue. A: Expression of αV detected using Q20 antibody, B:
membrane treated same as A, but with PBS instead of Q20. C: Expression of
β3 detected using B7 antibody as bordered with red rectangle, D: membrane
treated with PBS instead of B7; anti-mouse secondary antibody shows non-
specific binding of mouse antibody on mouse tissue. Films were
semiquantitatively analysed by Bio-Rad, Molecular Imager FX.System. While
αV was similarly expressed in all human xenograft tissues, β3 showed a range
of expression with M14 and UACC-62 showed the highest expression followed
by PC-3 and weaker expression was seen in DLD-1 and HCT-116 cells.
Normal mouse liver tissue was negative for human integrins and used as a
control here since it had non-specific binding with B7. Human tumour cell lines
M14 and HT-29 used as a positive and negative control for β3 respectively.
Results in the graph calculated from three independent experiments.

99
2.4 Discussion

The overall aim of this part of the study was to characterise αIIbβ3 and αVβ3

integrin expression in human tumour xenograft tissue in mice. This is

because the ultimate goals of the drug discovery project of which this thesis

is a part require in vivo models that can be used to test the efficacy of novel

integrin antagonists as drug targets in cancer therapy, and also to guide

selection of cell lines for in vitro studies.

The expression of αIIb and αVβ3 integrin in tissue can be evaluated by

immunohistochemistry and immunoblotting (only αIIb subunit was investigated

because it hetrodimer with β3 subunit only so expression of αIIb subunit and

β3 indicated the expression of αIIbβ3). However, immunohistochemistry has

been the more widely used of the two techniques reported in the literature

because it gives information about specific location of integrin in tissue. In a

study on clinical human tumours, archival FFPE tissue was utilized and

immunohistochemistry was able to characterise the expression of αVβ3

integrin in glioma tissue, where it was associated with poor prognosis

(Schittenhelm et al. 2013). Bisanz et al. used immunohistochemistry to detect

the expression of αV subunit. The study treated PC-3 cells with anti-αV siRNA

inhibitor and indicated role of αV subunit mediated interaction of prostate

cancer cells with bone stroma when the tumour cells were implanted in

tumour as xenografts (Bisanz et al. 2005).

The expression of αIIbβ3 and αVβ3 integrin was initially investigated in FFPE

human xenograft tissues using immunohistochemistry. However,


100
immunohistochemistry was unsuccessful due to non-specific immunolabelling

that developed in the controls.

The problems experienced with immunohistochemistry may have been due

to masking antigenic epitopes; thus, different antigen retrieval methods were

tried. The use of citrate buffer as a method for antigen retrieval in human

xenograft tissue was unsuccessful. As shown in the results section, the use

of different primary antibodies did not reveal any that worked with the citrate

buffer microwave method of antigen retrieval. Therefore, pepsin and

proteinase K digestion were also tried.

Initially, LM609 was used to immunolabel different human xenograft mouse

tissue treated with pepsin, but pepsin digestion was unsuccessful with LM609

and also with other primary antibodies tried, such as B7 and BV4 (anti-β3),

C20 (anti-αIIb), and Q20 (anti-αv). Proteinase K antigen retrieval methods led

to tissue damage. All three different methods of antigen retrieval were

ineffective, due to the non-specific immunolabelling that developed in both

controls. The problem with antigen retrieval seen in this study has been

reported previously, where the embedding of tumour tissue in paraffin after

fixation can block integrin detection, requiring the use of an antigen retrieval

method to expose the integrin receptor to facilitate immunolabelling of the

integrin receptor with the primary antibody (Liapis & Hutton 1997). The failure

of the citrate buffer method in the present study was similar to that reported

in another study where the citrate buffer antigen retrieval method was

101
unsuccessful for integrin detection in formalin-fixed tissue (Cattoretti et al.

1993).

The non-specific binding observed in this study likely resulted from cross-

reactivity between the secondary antibody and endogenous mouse

immunoglobulins. Both sections (with primary antibody and without primary

antibody) developed immunoreactivity. These findings were confirmed by

using different fresh frozen human xenograft tumour tissue, which also

developed the same non-specific binding.

The detection of αVβ3 integrin in FFPE tissue remains a challenge. Goodman

et al. have summarised the problems with currently available antibodies, the

non specific staining developed in formalin fixed paraffin embedded tissue,

and have developed new rabbit monoclonal antibodies for characterisation of

integrin expression in archival FFPE human tumour tissue. Goodman’s

results reflected the problem faced in the present study with currently

available antibodies; LM609, BV4, B7 (mouse monoclonal antibodies) which

failed to detect specific expression of αVβ3 and β3 integrin in FFPE tissues

(Boger et al. 2013).

The use of the M.O.M. immunodetection kit has been proposed as a solution

for these issues (Jin et al. 2007). Using a mouse primary antibody in a mouse

xenograft tissue produced background labelling. The M.O.M. kit is specifically

designed to localize mouse primary antibodies in mouse tissues and to

eliminate background immunolabelling. The M.O.M. kit has been used

successfully with LM609 to detect the expression of α Vβ3 integrin in the


102
frozen tissue of human IGROV-1 ovarian carcinoma nodules disseminated in

the peritoneal cavity of a mouse (Garanger et al. 2005). Here, β3 expression

was detected using BV4 (anti-β3) in cryo-sections, but not in paraffin

sections, probably because the integrin epitopes are affected by fixation. BV4

(anti-β3) showed β3 subunit membranous immunolabelling, which was

observed in a PC-3 frozen section with a M.O.M. kit, but the resulting

expression was not reproducible. Furthermore, no expression was detected

in other fresh frozen human tumour xenografts using the same protocol,

although they were expected to express β3; the heavy background obscured

the results further to non-specific immunolabelling developed in sections

without BV4 (anti-β3) as a primary antibody.

The search for the β3 subunit integrin in tissue was continued using another

antibody B7 (anti-β3), aiming to result in membranous immunolabelling with a

clean negative control with a lack of non-specific binding. B7 gave a

membranous immunolabelling, but without a clean negative control. Although

a M.O.M. kit was used, the outcome was not changed: the same results were

obtained with the use of mouse monoclonal antibodies BV4, B7, and LM609.

This indicated that either non-specific immunolabelling developed in both

controls was due to the use of mouse antibody on mouse tissue, or it was

due to the inability of the primary antibodies used here to detect the

expression of αIIbβ3/αVβ3 in FFPE and frozen tissue using

immunohistochemistry.

103
The results obtained in the present study indicate an inability to detect the

expression of αIIbβ3 and αVβ3 integrin in xenograft tissue using

immunohistochemistry. Liapis and Hutton used a panel of primary antibodies

to detect the expression of α2β1, αVβ3, α3, α4, α5, α6, β1, and β5 integrin in

FFPE human ovarian carcinoma, and reported that not all antibodies work

with the microwave citrate buffer method of antigen retrieval, although they

used clinical tissues (Laurens et al. 2009; Liapis & Hutton 1997; Singh et al.

2000).

The second reason for the failure of antibodies used in this study may be due

to difficulties in characterising the expression of αIIbβ3 and αVβ3 integrin in

tumours grown in mouse tissue using a mouse antibody. The observation

that indirect immunohistochemical detection with a primary antibody

homologous with the test tissues usually results in non-specific background,

has been mentioned in the literature; for example, Lu et al. reported that

binding of a Fc fragment of secondary antibody to a mouse tissue component

was the cause of background immunolabelling (Lu & Partridge 1998).

Due to the issues of non-specificity seen in the immunohistochemistry

experiments, immunoblotting was also utilised to detect the expression of

human αVβ3 integrin, in the presence of normal mouse tissue as a negative

control. Expression of αV and β3 subunits was successfully detected using

the immunoblotting technique in a panel of human tumour xenograft tissue;

the xenograft tissues had varying levels of expression of β3 whereas αV was

expressed in all human tumour xenograft tissues tested. Expression of β 3

104
was highest in melanoma xenograft tissue (UACC-62 and M14), followed by

prostate adenocarcinoma (PC-3), which had moderate expression, with very

weak expression detected in colonic carcinoma (DLD-1 and HCT-116).

These results were confirmed by comparing them with normal mouse liver

tissue that has no expression of human αV and β3 integrin subunits (Taylor et

al. 2012).

The B7 antibody was able to reveal the expression of β3 but gave an

additional band just below the expected position of a band for β3 expression,

indicating non-specific binding. In order to confirm the non-specific binding

developed with B7, several variables were tried such as incubating the

membrane without B7 as a primary antibody and incubating another

membrane without a primary antibody and without a secondary antibody. It

was obvious that normal mouse liver tissue and human xenograft mouse

tissue developed non-specific binding in a membrane incubated with anti-

mouse secondary antibody only, due to the reaction of anti-mouse on mouse

tissue.

Further confirmation came from using human tumour cell lines M14 and HT-

29, which did not develop any non-specific binding in the blot membrane

without B7 as a primary antibody. The lack of non-specific binding in cell lines

indicates it is a problem with proteins present in mouse tissue, as shown in

Figure 2.15.

In addition, rabbit antibody Q20 (anti-αV) did not show any non-specific

binding in membrane without Q20 as a primary antibody, either in normal


105
mouse liver tissue or in human xenograft mouse tissue. This further confirms

the results obtained with B7 (anti-β3).

The results convincingly revealed that M14, UACC-62, PC-3, HCT-116 and

DLD-1 expressed αV. The expression of the αV integrin subunit has been

detected by immunoblotting in other types of human tumour cells however,

homogenised human tissue is rarely utilised to detect the expression of

integrin using immunoblotting. A study done by Schnell et al. detected the αV

and β3 integrin subunits in homogenised glioma tumour tissue using

immunoblotting (Schnell et al. 2008). The non-specific binding observed in

this study was also noted by Gouon et al. when immunoblotting was utilised

to detect the expression of β3 integrin in HT-144 melanoma homogenised

human xenograft mouse tissues (Gouon et al. 1996).

The expression of β3 varies among the different tissues, with the highest

levels seen in melanoma cells such as UACC-62 and M14. Studies found

strong β3 integrin expression was associated with the presence of tumour

necrosis, increased tumour thickness, tumour ulceration, vascular invasion,

tumour cell proliferation and high level of cytoplasmic p-cadherin. Significant

association of both αV and β3 integrin with tumour necrosis was also

associated with reduced overall survival in nodular melanoma (Bachmann et

al. 2008; Hieken et al. 1999; Si & Hersey 1994). The expression of β3 is

significant in malignancy, and αVβ3 integrin expression could be an important

step with in situ melanoma development and invasion.

106
The expression of αV, β3 subunits and αIIbβ3 integrin had previously been

detected by other groups in human melanoma tissue embedded in paraffin

(Trikha et al. 1997). Goodman has confirmed earlier work that found no β3

integrin expression in colon carcinoma tissue (Goodman et al. 2012). This

supports this study result where very weak expression of β3 integrin in colon

carcinoma xenograft tissue DLD-1 and HCT-116, was observed using

immunoblotting, whilst the αV subunit was detected in the same tissue

through immunohistochemistry labelling. In addition, this previous study

supports the finding of the present study where the αV subunit was detected

in all human xenograft tissues. Since no expression of β3 integrin was

observed, the αV subunit must be present as a heterodimer with another β

subunit such as β1, β5 and β6 (Agrez et al. 1996).

2.5 Conclusion

In conclusion, αIIb, αV, β3, and αVβ3 integrin expression could not be detected

in human xenograft tissue grown in mice using immunohistochemistry.

However, using the immunoblotting technique, αV and β3 integrin expression

could be successfully characterised in the panel of xenograft mouse tissues.

Differential β3 expression was seen over the panel of tumours, while αV levels

were similar across the panel whereas αIIb could not be detected due to in

ability of the primary antibody to work with IMB.

107
3 Chapter 3: Characterisation of integrin αIIbβ3 and αVβ3 expression in

a panel of human tumour cell lines

108
Chapter 3

3.1 Introduction

Evaluation of any novel targeted therapy requires careful selection of

appropriate models. In this thesis, the target is the β3 integrins. A panel of cell

lines with a range of expression levels of the different β3 integrins is therefore

required for the evaluation of novel integrin antagonists.

The expression of the β3 integrins is widely reported in the literature in

various human tumour cell lines and tissues, as reviewed in section 1.2.5.

Whilst the findings of the previous chapter may give some guidance for the

cell lines worthy of further investigation, the expression is not likely to be the

same in vitro as in vivo for all cell lines evaluated (Taylor et al. 2012). Thus,

this part of the study focused on characterisation of the expression in human

tumour cell lines grown in vitro.

Ideally, a panel for novel therapeutic evaluation should include: 1) cell lines

that express both αVβ3 and αIIbβ3 integrin; 2) cell lines that are negative for

both αVβ3 and αIIbβ3 integrin; 3) cell lines that express αVβ3 but are negative

for αIIbβ3; 4) cell lines that are negative for αVβ3 but express αIIbβ3 integrin.

The cell lines selected as models should ideally grow as a monolayer in cell

culture flasks whilst maintaining the specific integrin expression. They must

also have a reproducible expression of αIIbβ3 and αVβ3 integrins.

Optimisation of the protocols for the antibodies to be used in this part of the

study first requires a cell line known to express αVβ3 and αIIbβ3 integrin as a
109
positive control. A number of researchers have investigated the expression of

αVβ3 and αIIbβ3 in PC-3 cells, using different techniques for protein detection,

such as flow cytometry and immunoblotting (Cooper et al. 2002; Trikha et al.

1998; Wang et al. 2005). αVβ3 integrin plays an important role in prostate

cancer development and progression (Cooper et al. 2002; McCabe et al.

2007) and the expression of αVβ3 integrin has been detected in PC-3 cells

using the LM609 antibody (Arosio et al. 2009; Chatterjee et al. 2001). In

addition, Trikha et al. detected the expression of αIIb in PC-3 using the MAB

1990 antibody (Trikha et al. 1998).

3.1.1 Aims and objectives

The principle aim of this chapter will be to assemble a panel of cell lines that

can be used for screening novel antagonists. This will be done through

characterising integrin expression in human tumour cell lines.

The aims will be addressed using the following objectives:

1- Characterisation of the growth patterns of selected cell lines in order to

optimise seeding densities for further experiments.

2- Optimisation of the immunoblotting protocols for the antibodies used to

characterise the expression of αV, αIIb and β3 integrins.

3- Optimisation of the immunocytochemistry protocols for the antibodies

used to characterise the expression of αV, αIIb, β3, and αVβ3 integrins.

110
4- Evaluation of the expression of αV, αIIb, β3, integrin subunits in a panel

of cell lines using the immunoblotting and immunocytochemistry

protocols optimised in 2 and 3.

111
3.2 Materials and Methods

3.2.1 Materials

All general chemicals, media and media supplements were from Sigma-

Aldrich (Poole, UK) unless otherwise specified. Primary antibodies and

related secondary antibodies were used in this part of the study to detect

expression of αV, αIIb, β3 and αVβ3 as summarised in Table 3.1. The blocking

reagents (BSA, NRS, and NHS), and Vectashield hard-set mounting medium

with DAPI were all from Vector Laboratories Ltd (Peterborough, UK). All cell

lines used as summarised in Table 3.2 were stored in liquid nitrogen prior to

use.

3.2.2 Methods

3.2.2.1 Cell maintenance

The cell lines were selected to represent a range of tumour types (Table 3.2).

Cell culture procedures were performed in a sterile Micro-flow class II cabinet

(NUAIRE, biological safety cabinet, Plymouth, UK).

Cells were routinely maintained as monolayer cultures in 75 cm 2 cell culture

treated flasks (T75 flask) (Corning, Amsterdam, Netherlands) in 10 ml

complete Roswell Park Memorial Institute (RPMI) 1640 medium

supplemented with 10% foetal bovine serum (FBS) 200 mM L-glutamine and

100 mM sodium pyruvate.

112
Table 3.1 Primary antibodies and related secondary antibodies used in
immunocytochemistry (ICC) and IMB techniques.
PAb R Type Source 2nd Ab Source 2nd Ab Source
(IMB) (ICC)
Anti-β3 MMAb BV4 ab 7167, PRAM, P0260, Dako PRAM R0270,
Abcam, Ig HRP Cytomation, Igs, Dako
BV4

Cambridge, Glostrup, TRITC Cytomation,


UK Denmark Glostrup,
Denmark
Anti-β3 MMAb Clone B7, SC- PRAM, P0260, Dako PRAM R0270,
46655, Ig HRP Cytomation, Igs, Dako
Santa Cruz Glostrup, TRITC Cytomation
B7

Biotechnology Denmark Glostrup,


Santa Cruz, Denmark,
USA
Anti- PGAb SC-6602, Goat P0449, Dako PRAG F0250,
αIIb Santa Cruz Igs Cytomation, Igs, Dako
C20

Biotechnology Glostrup, FITC Cytomation,


Santa Cruz, Denmark Glostrup,
USA Denmark
Anti-αV MMAb SC-9969, PRAM, P0260, Dako PRAM R0270,
Biotechnology, Ig HRP Cytomation, Igs, Dako
P2W7

Santa Cruz Glostrup, TRITC Cytomation,


Biotechnology Denmark Glostrup,
Santa Cruz, Denmark
USA
Anti-αV PRAb SC-6617-R, PGAR, P0448, Dako GAR, A-11010,
Santa Cruz Ig HRP Cytomation, Alexa Invitrogen,
Q20

Biotechnology Glostrup, fluor California,


Santa Cruz, Denmark 546 CA, USA
USA
Anti- MMAb Clone LM609, - - PRAM R0270,
αV β 3
LM609

MAB 1976, Igs, Dako


Chemicon- TRITC Cytomation,
Millipore, Glostrup,
Watford, UK Denmark
Anti β- MMAb Sigma-Aldrich, PRAM, P0260, -
actin Poole, UK Ig HRP DAKO
A1978

Cytomation,
Glostrup,
Denmark
nd
Key: PAb: primary antibody, R: receptor, 2 Ab: secondary antibody, MMAb: monoclonal
mouse antibody, PGAb: polyclonal goat antibody, PRAb: polyclonal rabbit antibody, PRAM:
polyclonal rabbit anti mouse, Ig. Immunoglobulin, PGAR: polyclonal goat anti rabbit, PRAG:
polyclonal rabbit anti goat. TRITC: tetramethylrhodamine isothiocyanate, FITC: fluorescein
isothiocyanate. GAR: goat anti rabbit.

113
The cells were maintained at 37 °C in humidified conditions and a 5% CO2

atmosphere. When the cells attained 75 to 80% confluence, they were

passaged by discarding the medium and rinsing the contents of the flask

twice with 10 ml of Hank’s Balanced Salt Solution (HBSS), prior to

trypsinisation using 3 ml of a 0.25% trypsin-EDTA solution, and incubating at

37 ˚C for three to five minutes. When cells became detached from the flask,

10 ml of medium was added to deactivate the action of trypsin/EDTA. The

trypsinised cells were then centrifuged for five minutes at 1000 rcf at room

temperature (Heraeus Megafuge 1.0 centrifuge, DJB Labcare Ltd. UK). The

resulting supernatant was discarded and cell pellet was resuspended in 10

ml of fresh complete culture medium. Cells were then counted with 10 µl of

cell suspension placed in each chamber of a Neubauer Haemocytometer

(7201004, VWR International Ltd. Poole, UK). An inverted microscope

(Olympus, CK2, x-20 objective lens magnification) was used to count cells in

ten 1 mm2 areas of the haemocytometer and the mean count was calculated.

Cell counts were then expressed as (mean cell count) × 104 cells/ml medium,

and the required amount of cell suspension was seeded into new flasks

containing the appropriate amount of complete medium.

114
Table 3.2 Cell lines
Cell line Origin Identification code
A549 Human non small cell lung carcinoma ATCC CCL-185
(NSCLC)
DLD-1 Human colorectal adenocarcinoma ATCC CCL-221
DU-145 Prostate adenocarcinoma ATCC HTB-81
H460 Human NSCLC ATCC HTB-177
HCT-116 Human colorectal carcinoma ATCC CCL-247
HT-29 Human colorectal adenocarcinoma ATCC HTB-38
M14 Human melanoma NCI-0507466
MCF-7 Human breast adenocarcinoma cell line ATCC HTB-22
from metastatic site: pleural effusion
PC-3 Human prostate adenocarcinoma derived ATCC CRL-1435
from grade IV cancer metastasised to bone
SK-MEL-2 Human malignant melanoma ATCC HTB-68
UACC-62 Human malignant melanoma NCI-0507381
Key: ATCC: American Type Culture Collection. NCI: National Cancer Institute

3.2.2.2 Tumour cell line growth kinetics

The growth kinetics of all the tumour cell lines used in this study were

characterised to determine when cells would be in a log phase and hence be

functionally active. Cells were seeded at 1 × 104 cells/ml in 5 ml fresh

complete medium in T25 flasks, and were maintained at 37 °C, 5% CO2 as

described above. Cell counts were performed over 7 days and growth curves

plotted. Growth curves were carried out in this study using cells from different

passages. Readings are reported as the average (± SD) recorded from three

different flasks per day.

3.2.2.3 Immunoblotting

Immunoblotting (IMB) was carried out using whole cell lysates in order to

characterise the expression of αV, αIIb, and β3 integrin in a panel of human

tumour cell lines.


115
3.2.2.3.1 Cell Lysis

Once the cells lines reached 70 to 80 % confluence, cells were scraped from

the flask surface after a gentle wash with HBSS. Cells were centrifuged

(Eppendorf centrifuge, G417R, Natheler-Hizz, Hamburg, Germany) for five

minutes at 1000 rcf, the cell pellet was washed three times with PBS for four

minutes with centrifuging (4600 rcf) after each wash. Lysis buffer (2X lysis

buffer [100 mM Tris pH 8.8, 5 mM EDTA, 300 mM NaCl, 2% Triton X-100],

protease inhibitor (Complete mini EDTA-free (Roche, Mannheim, Germany)

and water) was applied for one hour on ice, after which sonication (Philip

Harries Scientific Sonicator, Scientific Laboratory) was carried out for two

pulses of five seconds each. Sonicated cells were centrifuged at 9000 rcf for

ten minutes at -20 °C, after which the cytosolic supernatant of the lysed cells

was collected and the cellular pellet resuspended in fresh lysis buffer. Since

functional integrin receptors span the cell membrane, but can also be

endocytosed or present in a storage pool within the cell both the plasma

membrane (cell pellet) and cytosolic content were collected here for further

confirmation of their integrin expression.

3.2.2.3.2 IMB of nitrocellulose membrane with different anti αV, αIIb and

β3 integrin antibodies

The Bradford assay, SDS-polyacrylamide gel electrophoresis and

immunoblotting were carried out using the same conditions as described in

Chapter 2. In this part of the study, the primary antibodies were optimised to

characterise expression of αIIb, αV and β3 integrin in human tumour cell lines.


116
With the initial optimisation protocol for each antibody, the nitrocellulose

membranes were blocked with either 5% non-fat dried milk in PBS-buffered

saline (with 0.05% Brij, 35 solution), or 5% BSA/PBS, for one hour at room

temperature. Then, the primary antibody as summarised in Table 3.1 was

applied at different dilutions ranging from 1:200 to 1:5000 for either one hour

at room temperature or overnight at 4 °C; each antibody had its own optimum

dilution and incubation condition (Table 3.4). The membrane was washed

with PBS (0.05% Brij) (3 washes of 10 minutes each). The recommended

secondary antibody (Table 3.1) was applied in the dilution range from 1:1000

to 1:5000 for one hour at room temperature followed by washing with PBS

(0.05% Brij) (3 washes for 5 minutes). The ECL Plus Western blotting

detection system (NEL104, Perkin Elmer LAS) reagents A and B were mixed

in 1:1 ratio and applied to membrane for one minute. The membrane was

exposed to Kodak Scientific Imaging film in the dark for five to thirty minutes.

Films were developed using IIford Rapid Multigrade developer (1/10 dilution)

for three minutes, rinsing with water and fixing in IIford Rapid Film and Paper

fixer solution (1/10 dilution) for two minutes. The immunoblotting was

repeated three times to confirm the reproducibility of the optimised protocol.

117
3.2.2.4 Immunocytochemistry (ICC)

3.2.2.4.1 Detection of αV, αIIb and β3 subunits, and αVβ3 integrin

expression in different tumour cell lines by ICC

3.2.2.4.1.1 Materials

All materials used here were the same as described in section 3.2.1 with the

addition to the primary antibody panel of LM609, anti-αVβ3 integrin.

3.2.2.4.1.2 Cell preparation

In order to detect the expression of αV, αIIb and β3 subunits and αVβ3 integrin,

1 × 104 to 1 × 105 cells/ml were seeded on autoclaved 22mm × 22mm cover

slips in a six-well plate and incubated at 5% CO2, 37 ˚C until cells were seen

adhering to the cover slip when viewed under the microscope no more than

12 hours. The medium was removed and the cells washed for two washes of

two minutes with 1 ml HBSS.

Cells were fixed with either ice pre-cooled methanol for twenty minutes at -20

°C, 1% PFA or 4% PFA for ten minutes at room temperature, or four to five

dips with ice pre-cooled acetone. The fixative was removed and the cells

allowed to air dry. Fixed cells were either used for immunolabelling directly or

stored at -20 °C. Following rehydration, blocking was followed by direct

application of primary antibody (Table 3.1) at different dilutions and

incubation conditions followed by washing for 3 washes for 5 minutes each

(Table 3.3). The secondary antibody (Table 3.1) was then applied at different

incubations times after labelling with secondary antibody, and cells washed
118
for 3 washes for 5 minutes each as shown in Table 3.3. Cells were mounted

with Vectashield with DAPI and kept at 4 °C until dry and examined by

fluorescence microscope (Leica DMRB, Leica Microsystems, Wetzlar,

Germany).

3.2.2.4.1.3 Confocal microscopy

Confocal microscopy was used in order to confirm the expression observed

by fluorescent microscope. Cells were prepared as in section 3.2.2.4.1.2 and

were analysed and images captured with a Zeiss LSM510 confocal system

attached to an Axiovert 200 M inverted microscope using LSM510 software

(all from Zwiss, Welwyn Garden City, UK).

3.2.2.4.1.4 ICC analysis

Semi-quantitative analysis of integrin expression in the tumour cells was

done by counting cells in 10 fields at 40 magnification, and giving a score to

the labelling of each cell dependent upon its intensity. The expression

intensity scoring scheme was confirmed by an independent observer. The

high expression was designated as +++, moderate ++, low + and weak ±.

The mean intensity was calculated for each sample.

119
Table 3.3 Conditions investigated while optimising detection of expression of αV, αIIb, β3 and αVβ3 integrin in human tumour
cell lines using ICC.
PAb Anti-β3 subunit, BV4 Anti-β3 subunit, B7 Anti-αV subunit, Q20 Anti-αIIb subunit, C20 Anti- αVβ3 integrin, LM609
Rehydration PBS 5 minutes, R.T. PBS 5 minutes, R.T. PBS 5 minutes, R.T. PBS 5 minutes, R.T. PBS 5 minutes, R.T. or
PBS + 0.1% Triton 5
minutes, R.T. or 4 °C
Blocking 2 & 15% NRS/PBS, 2% NRS/PBS, 10 2% NGS/PBS, 10 2% NRS/PBS, 10 2% NRS/PBS, 10 minutes,
10 minutes, R.T. minutes, R.T., or 5% minutes, R.T. or 5% minutes, R.T. or 1- 5% R.T. 5% BSA/PBS, 1 hour
BSA/PBS, 1 hour. R.T. BSA/PBS, 1 hour. BSA/PBS, 1 hour R.T. R.T.
R.T.
Primary 1:50 - 1:800 1:20 - 1:400 1:25 - 1:400 1:20 - 600 1:50 – 1:400
antibody dilution
Primary 30 minutes & 1hour at 30 minutes & 1hour at 30 minutes at R.T., or 30 minutes & 1 hour at 30 minutes & 1 hour at R.T.,
antibody incubation R.T., or 1 hour at 37° R.T., or 1 hour at 37 1 hour at 37° C, or R.T., or 1 hour at 37 or 1 hour at 37 °C, or
C, or overnight at 4 °C °C, or overnight at 4 °C overnight at 4 °C °C, or overnight at 4 °C overnight at 4 °C
Secondary antibody PRAM, TRITC PRAM, TRITC GAR IgG, Alexa fluor PRAG, FITC, 1:50 in PRAM, TRITC
dilution 1:50/PBS 1:50/PBS 546, 1:50 in PBS, or PBS, or1:50 in 5% 1:50 in PBS, or 1:50 in 5%
or 1:50/5%BSA/PBS
1:50 in 5% BSA/PBS BSA/PBS, or 1:50 in BSA/PBS
1% BSA/PBS
Secondary antibody 30 minutes to 1 hour in 30 minutes to 1 hour in 30 minutes to 1 hour 30 minutes to 1 hour in 30 minutes to 1 hour in dark
Incubation dark dark in dark dark
Washing PBS (3 × 5 minutes) PBS or 5% BSA/PBS PBS or 5% BSA/PBS PBS or 5% BSA/PBS PBS or 5% BSA/PBS
after primary and (3 × 5 minutes) (3 × 5 minutes) or 1% BSA/PBS (3 × 5 minutes)
secondary antibody (3 × 5 minutes)
Key: PAb: Primary antibody, PBS: Phosphate buffered saline, R.T: room temperature, BSA: bovine serum albumin, NGS: normal goat serum. NRS: normal
rabbit serum, PRAM: polyclonal rabbit anti mouse, GAR: goat anti rabbit, PRAG: polyclonal rabbit anti goat.
120
3.3 Results

3.3.1 Characterisation of cellular growth kinetics

Growth rates varied between tumour cell lines as demonstrated by

differences in their growth curves and the duration of lag, log and plateau

phases.

Most of the cell lines had a lag phase, except DU-145 and DLD-1, which

were started in log phase and showed exponential growth from the first day.

Cell lines were in plateau phase once the cells became confluent except

DLD-1 and PC-3 which continued in log phase until day 7. Figure 3.1 shows

the growth curves obtained for the cell panel.

121
104/ml

Number of cell × 104/ml


Number of cell × 104/ ml
H460 HT-29

Number of cell × 104/ml


100.00 A549 100.00 100.00 MCF-7 100.00

10.00 10.00 10.00 10.00

Number of cell
1.00 1.00 1.00 1.00

0.10 0.10 0.10 0.10


0 2 4 6 0 2 4 6 0 2 4 6 0 2 4 6
Time (Days) Time (Days) Time (Days) Time (Days)

PC-3
Number of cell 104/ ml

104/ml
DLD-1

Number of cell × 104/ml


Number of cell × 104/ml
100.00 100.00 HCT-116 100.00 100.00 DU-145

10.00 10.00

Number of cell
10.00 10.00

1.00 1.00 1.00 1.00

0.10 0.10 0.10 0.10


0 2 4 6 0 2 4 6 0 2 4 6 0 2 4 6
Time (Days) Time (Days) Time (Days) Time (Days)

M14

Number of cell × 104/ml


100.00 UACC-62 100.00 SK-MEL-2
Number of cell × 104/ml
Number of cell × 104/ ml

100.00

10.00 10.00 10.00

1.00 1.00 1.00

0.10 0.10 0.10


0 2 4 6 0 2 4 6 0 2 4 6
Time (Days) Time (Days) Time (Days)

Figure 3.1 Growth parameters of human tumour cell lines


Growth parameters were obtained as described in section 3.3.1.
Values correspond to the mean ± SD of 3 independent experiments.

122
3.3.2 Detection of αV, αIIb, and β3 integrins using IMB

3.3.2.1 Optimisation of IMB

In order to optimise the IMB protocols for each antibody used here, different

variables were tried. These variables as shown in the flowchart in Figure 3.2

included: protein concentrations, blocking the non-specific binding of primary

antibody, primary antibody dilution and incubation time, and film exposure

time. All primary antibodies detected expression of integrin at a protein

concentration of 20 µg except C20 (anti-αIIb). C20 did not detect expression

even when using a protein concentration of 100 µg. B7 (anti-β3) and Q20

(anti-αV) were able to detect expression in membrane blocked with 5%

BSA/PBS whereas the rest of the antibodies worked only with 5% non-fat

milk. The different dilutions and incubations tried for primary antibodies

clearly showed that 1:2000 dilution, overnight incubation at 4 °C was

optimum for B7 (anti-β3) and Q20 (anti-αV) whereas 1:500 at room

temperature for two hours was optimum for the rest of the antibodies. All

antibodies detected expression after five minutes exposure time except C20

(anti-αIIb) which did not detect clear expression even if exposed for thirty

minutes. Thus no further work was carried out for this antibody and hence

attempts at detecting αIIb. Figure 3.2 shows an example of a representative

blot (three independent experiments were done for each antibody using

optimum conditions for each antibody as summarised in Table 3.4). From the

above results B7 (anti-β3) and Q20 (anti-αV) were identified as specific

antibodies that can be used to screen the expression of α V and β3 integrin in

123
a panel of different tumour cell lines using the optimised protocol as

summarised in Table 3.4.

IMB optimisation of primary antibodies used for detection αV, αIIb, and β3

1- Loading PC-3 different protein concentrations: 5, 10, 15, 20, 75 & 100 µg

Q20, P2W7, B7 & BV4 cannot detect expression < 20 µg C20 did not detect expression even at 100 µg

2- Blocked with 5% non fat milk/PBS or 5% BSA/PBS1 hour R.T

BV4, P2W75%
and non fat with
C20 work milk/PBS
5% non fat milk/PBS B7 and Q20 work with 5% BSA/PBS

3- primary antibody dilution / incubation

1:500 1:1000 1:2000 1:3000

2 hours R.T. Overnight 4 C

BV4, P2W7 & C20 at 1:500. B7 & Q20 at 1:2000


.

4- Secondary antibody (2nd Ab) dilution, 1:2000 to 1:5000 1 hour R.T

5- Film exposing time: 5, 10 15 & 30 minutes

BV4: double band at 2nd P2W7: multiple band, 2nd Ab C20: no clear band, 2nd Ab B7 & Q20: single band, 2nd
Ab1:5000 , 5 minutes exposing 1:5000, 5 minutes exposing 1:3000, 30 minutes exposing Ab,1:3000, 5 minutes exposing
αIIb αV β3

C20 P2W7 Q20 BV4 B7

132

95

72

No No Yes No Yes

Figure 3.2 Optimisation of IMB protocol.


Expression of αV, αIIb and β3 were detected in PC-3 cells using different
antibodies. Q20 and P2W7 were anti-αV, C20 anti- αIIb, and BV4 and B7 were
anti-β3. Protein concentrations, blocking conditions, PAb dilutions and
incubations and film exposing time were optimised for each antibody.
Key: Nc: no clear band. Mb: multiple bands. Db: double bands. Sb: single band.

124
Table 3.4 The optimised protocol for all antibodies used to detect the expression of αV, αIIb, and β3 subunit and β-actin
using IMB technique.
Immunolabelling BV4 anti-β3 B7 anti-β3 P2W7 anti-αV Q20 anti-αV C20 anti-αIIb β-Actin
conditions
Protein 20 µg 20 µg 20 µg 20 µg 100 µg 20 µg
concentration
Blocking 5% non-fat milk, 5% BSA/PBS, 5% non-fat milk, 5% BSA/PBS , 5% non-fat milk, 5% BSA/PBS ,
1 hour R.T. 1 hour R.T. 1 hour R.T. 1 hour R.T. 1hour R.T. 1 hour R.T.
Primary antibody 1:500 in 5% non- 1:2000 in 1:500 in 5% non- 1:2000 1:500 in 5% 1:5000
Dilution/Incubation fat milk, 5%BSA/PBS, fat milk, in 5% BSA/PBS non-fat milk, in 5% BSA/PBS
2 hour R.T. overnight 4 °C 2 hour R.T. overnight 4 °C 2 hour R.T. overnight 4 °C
Washing PBS PBS (0.05 % Brij), PBS (0.05 % Brij) PBS (0.05 % Brij) PBS (0.05 % Brij) PBS (0.05 % Brij)
(0.05 % Brij), (3 × 10 minutes) (3 × 5 minutes) (3 × 10 minutes) (3 × 5 minutes) (3 × 10 minutes)
(3 × 5 minutes)
Secondary PRAM 1:5000 in PRAM 1:3000 in PRAM 1:5000 in PGAR 1:3000 Goat Igs 1:3000 PRAM 1:5000
antibody non-fat milk, non-fat milk, non-fat milk, in non-fat milk, in non-fat milk, in non-fat milk,
Dilution/Incubation 1 hour R.T. 1 hour R.T. 1 hour R.T. 1 hour R.T. 1 hour R.T. 1 hour R.T.
Washing PBS (0.05% Brij), PBS (0.05% Brij), PBS (0.05% Brij) PBS (0.05% Brij) PBS (0.05% Brij) PBS (0.05% Brij)
(3 × 5 minutes) (3 × 5 minutes) (3 × 5 minutes) (3 × 5 minutes) (3 × 5 minutes) (3 × 5 minutes)
Exposing 5 minutes 5 minutes 5 minutes 5 minutes 30 minutes 2 minutes
Result Double bands Single band size Multiple bands Single band size No detectable Single band size
125 KDa 132 KDa band 42 KDa

Key: PGAb: polyclonal goat antibody. PRAM: polyclonal rabbit anti mouse. R.T: room temperature.

125
3.3.2.2 Screening different tumour cell lines for αV and β3 integrin

expression by IMB technique using the optimised conditions

for anti-αV, Q20, and anti-β3, B7, antibodies

In the preceding section, two antibodies were identified as giving reliable and

specific detection of integrin subunits, Q20, anti-αV and B7, anti-β3. These

were therefore used to screen the extended cell line panel. The expression of

αV and β3 was detected in the supernatant (Figure 3.3), as well as in the

pellet resuspended in lysis buffer (Figure 3.4) indicating expression of the

integrin subunits in both the cell membrane and cytosol. αV was expressed in

all cell lines with HCT-116 demonstrating a notably lower expression,

whereas β3 expression levels showed more variation between cell lines. The

melanoma cell lines UACC-62, M14 and SK-MEL-2 had the highest

expression of β3 subunit. Moderate expression of β3 was seen for the

prostate lines PC-3 and DU-145, whilst weak expression was seen in the

NSCLC lines, A549 and H460 followed by the colon lines DLD-1 and HCT-

116 and very weak in breast line MCF-7. As would be expected from earlier

studies (section 2.3.3.1), no expression was seen in the HT-29 colon line

which used here as a negative control.

126
A

SK-MEL-2

UACC-62
HCT-116
DU1-45

DLD-1
MCF-7

HT-29
H460
A549

PC-3

M14
M
M
132

92 αV αV

132

92
β3 β3
72

β-actin

B 3
β-6
to10
to β actin control

2.5
V and β3
αv and β3 xof10α6 related

2
control

1.5 αv
αV
Expression
related

1 β3
β3
actin

0.5
Expression of

Figure 3.3 Expression of αV and β3 integrin in a panel of human tumour


cell lines from a supernatant
A. Expression of αV and β3 integrin subunits in a panel of human tumour cell
lines. All cell lines expressed the αV integrin subunit. Expression of β3 integrin
was highest in M14 followed by UACC-62 and SK-MEL-2. Moderate
expression of β3 integrin was seen in the PC-3 and DU-145. A549, H460,
DLD-1 and HCT-116 and MCF-7 showed weak expression of β3 integrin. HT-
29 was negative with no clear band observed. αV integrin was expressed at
molecular weight 132 kDa and β3 at 125 kDa. Data shown are representative
of at least three independent experiments. Lane 1 is Page Ruler Plus
Prestained Protein Ladder marker range from 10 to 250 kDa but because of
a short exposure time of five minutes it is not clearly seen on the film. B.
Results are presented as mean ± SD of three independent experiments.
Films analysed using Bio Rad Molecular Imager FX. The expression of α V
and β3 band density and β-actin band density were subtracted from the film
background density. αV and β3 signal intensity were divided by β-actin signal
intensity to obtain relative expression levels.
127
A

SK-MEL-2
UACC-62
HCT-116
DU-145
MCF-7

DLD-1

HT-29
H460
A549

PC-3

M14
M

M
132

92 αV αV

132

92
β3 β3
72

β-actin

B 3
Expression of αV and β3 106

2.5
related to β actin control

1.5 αv
αV

1 β3
β3
0.5

Figure 3.4 Expression of αV and β3 integrin protein in a panel of human


tumour cell lines from a pellet resuspended in lysis buffer
A. Expression of αV and β3 integrin subunit in the panel of human tumour cell
lines pellets resuspended in lysis buffer. Expression in the cell pellet showed
that αV and β3 subunit levels correlated with the expression found in cell
supernatant (Figure 3.3) except UACC-62 and PC-3 cell lines where the
expression was higher in their supernatant. B. Results are presented as
mean ± SD of three independent experiments. Films were analysed as
described in Figure 3.3.

128
3.3.3 Detection of αV, αIIb, β3 and αVβ3 integrins using ICC

3.3.3.1 Optimisation of ICC

Whilst IMB identifies the expression of a specific integrin subunit within a cell

line, it does not give any information as to the localisation of the integrin

expression within the cell and further cannot detect intact heterodimers.

Therefore, the ICC technique was also used to evaluate the cell panel. In

order to optimise the detection of expression of αV, αIIb, β3 and αVβ3 by ICC,

several variables were evaluated as shown in Figure 3.5. The optimised

protocol for each antibody is summarised in Table 3.5.

129
Optimise protocol for PAbs to characterise expression of αVβ3 and αIIbβ3 in human tumour cell lines by

Different fixatives tried

ICM ICA 4% PFA 1% PFA

Block non-specific binding of PAb

15% NS 2 % NS 5 % BSA 1 % BSA

All PAbs had non- BV4: +VE All other LM609 +VE Q20 weak All other C20 weak All other PAbs
specific binding in with all PAbs -VE with 4% PFA with 4% PFA PAbs -VE with 1% PFA -VE
section without PAb fixatives

PAbs dilution 1:50 , incubated 1 hour R.T, 1 hour. 37 C and 4 C overnight

BV4: +VE at R.T & 37 C Q20, B7, P2W7 & LM609: +VE 4 C only with 4%PFA C20 +VE at 4 C with all fixatives

Dilution of 1:50 -1:800

BV4: can be detected B7 & C20: can be detected Q20, P2W7 & LM609 can
untill 1:600 at 1:50 & 1:100 only be detected untill 1:400

Q20 (anti-αV) P2W7 (anti-αV) C20 (anti-αIIb)

BV4 (anti-β3) B7 (anti-β3) LM609 (anti-αVβ3)

Figure 3.5 Detection of αV, αIIb, β3 and αVβ3 integrin in PC-3 using ICC.
Different antibodies: Q20 and P2W7 anti-αV, BV-4, and B7 anti-β3, C20, anti-
αIIb, and LM609, anti-αVβ3 integrin were used to immunolabel PC-3 cells
using the optimum conditions for each antibody as summarised in Table 3.5.
Bar length = 60 µm.
KEY: ICM: ice-cooled methanol, ICA: ice-cooled acetone, PFA: paraformaldehyde.
130
Table 3.5 Optimised protocols for all antibodies used to detect the expression of αV, αIIb, β3 subunits, and αVβ3 integrin in
cell membrane using ICC
Labelling P2W7 (anti-αV) Q20 (anti-αV) C20 (anti-αIIb) BV4 (anti-β3) B7 (anti-β3) LM609 (anti-αVβ3)
Conditions
Fixation 4% PFA, 4% PFA, 1 or 4% PFA, ICM, 20 minutes 4% PFA, 4% PFA,
10 minutes R.T. 10 minutes R.T. 10 minutes R.T. or at -20 °C 10 minutes at R.T. 10 minutes at R.T.
ICM 20 minutes
at -20 °C

Blocking 5% BSA/PBS, 5% BSA/PBS, 1% BSA/PBS, 2% NRS/PBS, 5% BSA/PBS, 5% BSA/PBS,


1 hour R.T. 1 hour R.T. 1 hour R.T. 10 minutes R.T. 1 hour R.T. 1 hour R.T.
PAb D/I 1:50 in 5% 1:50 in 5% 1:50 1:50 in 2% 1:50 in 5% 1:50 in 5%
BSA/PBS, BSA/PBS, in 1% BSA/PBS, NRS/PBS, BSA/PBS, BSA/PBS,
overnight 4 °C overnight 4 °C overnight 4 °C 30 minutes R.T. overnight 4 °C overnight 4 °C
2nd Ab D/I TRITC, TRITC, 1:50 TRITC, 1:50 TRITC,1:50 TRITC, 1:50 TRITC, 1:50 in 5%
1:50 in PBS, in 5% BSA/PBS, in 1% BSA/PBS, in 5%BSA/PBS, in 5%BSA/PBS, BSA/PBS,
30 minutes in dark 1 hour in dark 1 hour in dark 1 hour in dark 1 hour in dark 1 hour in dark
Washing PBS 5% BSA/PBS 1% BSA/PBS 5% BSA/PBS 5% BSA/PBS 5% BSA/PBS
(3 × 5 minutes) (3 × 5 minutes) (3 × 5 minutes) (3 × 5 minutes) (3 × 5 minutes) (3 × 5 minutes)
nd
Key: ICM: ice pre-cooled methanol, PAb: primary antibody, 2 Ab: secondary antibody, D/I: dilution/ incubation.

131
3.3.3.2 Evaluation of the effect of method of cell harvesting on integrin

expression

Trypsinisation of cells can affect integrin expression (Brown et al. 2007).

Since cells were harvested by scraping for IMB and trypsinisation for ICC,

any effect on the integrin expression was investigated to ensure correlation

between expression of integrins by both techniques. Both scraped and

trypsinised PC-3 cells were immunolabelled with LM609 (anti-αVβ3) following

the optimum ICC protocol (Table 3.5). As shown in Figure 3.6, there was no

obvious difference in αVβ3 integrin expression between scraped and

trypsinised PC-3 cells using ICC.

A B

Figure 3.6 Comparison of cell harvesting techniques


No difference was seen in αVβ3 integrin expression on the membrane of PC-3
cells which had been scraped (A) and trypsinised (B). Cells were
immunolabelled with LM609 using ICC. Bar length = 60 µm.

132
3.3.3.3 Screening different cell lines for expression of αV, β3, αIIb and

αVβ3 integrins by ICC technique.

A panel of tumour cell lines was tested for the expression of αV, β3, αIIb

subunits and αVβ3 integrin immunoreactivity according to the optimised

protocols (Table 3.5). Following optimisation of the antibodies for ICC, the

panel of cell lines was screened for expression of αV, β3, αIIb subunits and

αVβ3 integrin (Figure 3.7 and 3.8).

133
Q20, anti-αV C20, anti-αIIb B7, anti-β3 LM609, anti-αVβ3

A549
H460
DU-145
PC-3
M14
SK-MEL-2

Figure 3.7 Screening of the first part of the cell line panel with anti-αV
(Q20), anti-αIIb (C20), anti-β3 (B7), and anti-αVβ3 (LM609) using ICC.
The antibodies were used for immunodetection of α V, αIIb, β3 subunits and
αVβ3 integrin in the panel of human tumour cell lines (A549, H460, DU-145,
PC-3, M14 and SK-MEL-2) according to protocols optimised above. Apart
from C20, all samples were counterlabelled with DAPI (blue) to visualize cell
nuclei. Bar length = 60 µm
134
Q20, anti-αV C20, anti-αIIb B7, anti-β3 LM609, anti-αVβ3

UACC-62
MCF-7
DLD-1
HCT-116
HT-29

Figure 3.8 Screening of the second part of the cell line panel with anti-
αV (Q20), anti-αIIb (C20), anti-β3 (B7), and anti-αVβ3 (LM609) using ICC.
The antibodies were used for immunodetection of α V, αIIb, β3 subunits and
αVβ3 integrin in the panel of human tumour cell lines (UACC-62, MCF-7,
DLD-1, HCT-116 and HT-29) according to protocols optimised above. Apart
from C20, all samples were counterlabelled with DAPI (blue) to visualize cell
nuclei. Bar length = 60 µm.

135
3.3.3.4 Analysis of the expression of αV, αIIb, β3 and αVβ3 integrin in

different human tumour cell lines detected by ICC.

After screening a panel of human tumour cell lines for the expression of α V,

αIIb, β3 and αVβ3 integrin using ICC technique, the expression was as either:

high expression, moderate expression, low expression, weak expression or

no expression. Table 3.6 summarises the expression seen in all cell lines

based on three independent experiments for each cell line.

Table 3.6 Screening different tumour cell lines for the membranous
expression of αV, β3, αIIb and αVβ3 by ICC
Cell lines BV4 B7 Q20 LM609 C20
anti-β3 anti-β3 anti-αV anti-αVβ3 anti-αIIb
Lung cancer cell lines
A549 + ± +++ ± ±
H460 ± ± + ± ±
Colon cancer cell lines
DLD-1 ++ ± +++ ++ +
HCT-116 ++ ± + ± +
HT-29 ± - ++ - ++
Prostate adenocarcinoma
cell lines
DU-145 ++ ++ ++ ++ ++
PC-3 +++ ++ ++ +++ +
Melanoma cell lines
M14 + +++ ++ ++ ±
SK-MEL-2 +++ ++ +++ ++ ±
UACC-62 ++ +++ +++ ++ +
Breast carcinoma cell line
MCF-7 + ± +++ ± +
Key: +++: High cell membrane expression, ++: Moderate cell membrane expression, +: Low
cell membrane expression, ±: Weak cell membrane expression.

136
3.3.3.5 Confirmation of sub-cellular immunolocalisation of αIIb, β3 and

αVβ3 integrin using confocal microscopy.

The sub-cellular localisation of expression of αIIb, β3 and αVβ3 integrins were

confirmed using ICC by observing the expression using confocal microscopy.

As shown in Figure 3.9, membranous expression of αVβ3 integrin and the β3

subunit were observed in human tumour cells with high, moderate, and weak

expression of β3, αVβ3, and αIIb integrin according to IMB and ICC result for

M14, PC-3, and MCF-7.

137
No primary antibody B7 No primary antibody LM609 No primary antibody C20

M14
PC-3
MCF-7

Figure 3.9 The expression of αIIb, β3, and αVβ3 integrin in human tumour cell lines by confocal microscopy.
The human melanoma tumour cell line, M14, the human prostate carcinoma cell line, PC-3, and the human breast carcinoma, MCF-
7, were selected as examples of high, moderate, and weak expression of β3 and αVβ3 respectively, and were examined using
confocal microscopy. β3 and αVβ3 integrin immunoreactivity was detected in the cell membrane using B7 and LM609, respectively.
Weak expression of αIIb was detected in M14, and low expression was detected in PC-3 and MCF-7 cell membrane using C20
under the optimum conditions summarised in Table 3.5. The white arrow indicates the different expression of β 3 and αVβ3 integrin in
human tumour cell line and weak and low expression of αIIb. Bar length for C20 images = 30 µm and for B7 and LM609 = 150 µm.
138
3.3.4 Comparison of result of IMB and ICC

The inability to detect αIIb by IMB and the possibility that expression obtained

by ICC might represent false positive expression (since no expression was

detected by IMB), meant that this was excluded from comparison of integrin

expression between the two detection techniques, as shown in Table 3.7.

The expression of αV and β3 integrin was similar in all the human tumour cell

lines included the panel.

Table 3.7 Comparison of integrin expression in different tumour cell


lines using specific antibodies for αV and β3 integrin subunits using ICC
and IMB.
Human tumour cell lines Q20 (anti-αV) B7 (anti-β3)
IMB ICC IMB ICC
A549 +++ +++ ± ±
H460 ++ + ± ±
DLD-1 ++ +++ ± ±
HCT-116 + + ± ±
HT-29 ++ ++ - -
DU-145 ++ ++ + ++
PC-3 ++ ++ + ++
M14 ++ ++ +++ +++
SK-MEL-2 +++ +++ ++ ++
UACC-62 ++ +++ +++ +++
MCF-7 +++ +++ ± ±
Key for IMB and ICC: +++: High expression, ++: Moderate expression, +: Low expression,
±: Weak expression, -: no expression.
The expression of αV and β3 integrin subunits was quantified visually by giving the highest
cell expression +++ and correlated to the rest of the cell lines expression.

139
3.4 Discussion

The main aim of this chapter was to characterise αIIb and αVβ3 integrin

expression in a panel of human tumour cell lines, in order to develop a set of

models to be use in screening novel β3 integrin antagonists.

Initially, growth curves were recorded to identify the log phase where cells

functionally active in the days where cells reach 70 to 80% confluency. Cells

were collected in this period of growth to detect expression of αIIbβ3 and αVβ3

integrin because, according to previous reports, integrin expression is

reduced when cells are highly confluent (Haywood-Reid et al. 1997). In the

present study the expression was detected only in the log phase of the

tumour cells, to know how the expression of αIIbβ3 and αVβ3 integrin changed

during different growth phase, further study needs to be done.

The expression of αIIbβ3 and αVβ3 integrin was investigated by

immunoblotting, thereby confirming integrin protein expression in human

tumour cell lines and the specificity of the primary antibodies used.

Immunocytochemistry was then used to localise the site of expression as

either membranous or intracellular, using the same antibodies.

The expression of αV and β3 integrin subunits in tumour cells lines was

successfully detected using the immunoblotting technique but αIIb expression

was not. Different antibodies were used comprising anti-β3 BV4 and B7 and

anti-αV P2W7 and Q20 and anti-αIIb C20. Looking at this range of antibodies,

some seemed to be non-specific; for example, BV4 (anti-β3) and P2W7 (anti-

140
αV) showed double and multiple bands in immunoblotting. A specific antibody

should give a single band on the exposed film and any non-specificity that

develops is due to binding of the primary antibody to other non-target

proteins. C20 (anti-αIIb) did not reveal any obvious expression, which is most

likely due to the presence of the target protein at negligible levels in the cell

lines investigated. Further investigations involving this antibody should use

known expressors (e.g. human platelet lysates) (Lau et al. 2004) or inducible

samples (PMA treatment of K562) (Sevinsky et al. 2004) as a positive

controls to optimise the antibody reaction. For these reasons, P2W7, C20

and BV4 were excluded from further study. Q20 and B7 successfully

detected the expression of integrin subunits in tumour cell lines showing a

single band in the immunoblotting indicating the specificity in detecting the

expression. B7 binds to amino acids 635-730, mapping near the C-terminus

of the β3 integrin and Q20 recognises a peptide mapping to the C-terminus of

integrin αV indicating the chance of αV and β3 subunits to form αVβ3 integrin

heterodimer. Q20 and B7 were then used to screen a panel of cell lines for

the expression of αV and β3 integrin using immunoblotting. Q20 had been

used previously for immunoblotting in human 1321N1 astrocytoma (Liao et

al. 2007).

After optimising the protocol for each antibody, specific antibodies were

utilised to screen a panel of human tumour cell lines. All the human tumour

cell lines expressed αV integrin, whereas the expression of β3 integrin differed

between different tumour cell lines. High expression of β3 integrin was

141
observed in melanoma human tumour cell lines which agrees with the

literature (Li et al. 2001; Voura et al. 2001).

In order to support the expression seen by immunoblotting,

immunocytochemistry was utilised using the same antibodies in addition to

an anti-αVβ3 integrin antibody LM609. This antibody binds to a conformational

epitope resulting from the post-translational association of the αV and β3

subunits and detects the αVβ3 complex, so it could not be used with the

immunoblotting technique because cell lysis causes dissociation of the dimer.

LM609 has been used in several studies to detect the expression of α Vβ3

integrin in different cell membranes (Arosio et al. 2009; Hofmann et al. 2000;

Seftor et al. 1992). The protocol of Arosio et al. (Arosio et al. 2009) was

followed for immunodetection of αVβ3 by immunocytochemistry using LM609.

Optimised protocols for other antibodies were developed by altering variables

until optimisation was reached, indicating that each antibody had different

protocol requirements.

Moderate expression of αV and β3 and αVβ3 integrin was observed in PC-3

using Q20, B7, and LM609, respectively, by immunocytochemistry and

immunoblotting. The human prostate carcinoma cell line PC-3 was selected

to optimise the methodologies because earlier studies have documented high

expression of αV, αIIb, β3 and αVβ3 in this prostate adenocarcinoma cell line

(Arosio et al. 2009; Trikha et al. 1998; Trikha et al. 1996; Zheng et al. 1999).

Zheng et al. detected high expression of αVβ3 integrin in a PC-3 cell line

using flow cytometry analysis and immunoblotting techniques, and αV was

142
shown to complex with β3 using an immunoprecipitation technique (Zheng et

al. 1999). Trikha et al. detected intracellular expression of αIIbβ3 in PC-3 and

DU-145 cells, and showed that αIIbβ3 can translocate to the cell surface

following the activation of PKC (Trikha et al. 1996). They had previously

detected membranous expression of αIIbβ3 in DU-145, using

immunocytochemistry using with the anti-αIIbβ3 integrin antibody, MAB 1990

(Trikha et al. 1998). However, very little expression was reported by some

others; for example, Arosio et al. found little expression of αVβ3 integrin in

PC-3 using LM609 by immunofluorescence technique (Arosio et al. 2009).

Chatterjee et al. also reported little expression of αVβ3 integrin in PC-3 in

comparison to LNCaP (Chatterjee et al. 2001), and Goodman et al. found no

expression of αVβ3 and αIIbβ3 integrin in PC-3 (Goodman, Grote & Wilm

2012). This variation in αVβ3 expression in the PC-3 cell line may be due to

different culture conditions, as proposed by Nemeth et al. who suggested that

expression of αVβ3 integrin in PC-3 can be lost after a long period of culturing

(Nemeth et al. 2003). Here in the present study, the cells were not used after

passage 10 to avoid loss of the expression which can occur if late-passage

cells are used. Variation in the expression reported between laboratories may

be due to different culture conditions, and the use of fetal bovine serum from

different batches (Haywood-Reid, Zipf & Springer 1997).

After successful detection of integrin expression in PC-3 cells, a panel of

tumour cell lines was screened for the expression of αV, αIIb, β3 and αVβ3

integrins. To address the objectives of the immunodetection of αIIb, αV and β3

subunits and αVβ3 integrins in melanoma, three cell lines were used; M14,
143
UACC-62 and SK-MEL-2. The αV, β3 subunits and αVβ3 integrin complex

were expressed in all cell line membranes whereas αIIb subunit expression

was weak. Melanoma cell lines had the highest expression of α V, β3 subunits

and αVβ3 integrin. This is the first study to investigate the expression of αV, β3

and αVβ3 integrin in the UACC-62 melanoma cell line using immunoblotting

and immunocytochemistry techniques.

Expression of αIIbβ3 and αVβ3 and their respective subunits have been

reported in melanoma cell lines. For example, Seftor et al. found that αVβ3

integrin was expressed in the melanoma cell line A375M using LM609 with

immunofluorescence (Seftor et al. 1992; Trikha et al. 1997). Petitclerc et al.

found the expression of αVβ3 integrin in the M21 melanoma cell line and used

it as a model to explore αVβ3 integrin expression on melanoma growth in vivo

(Petitclerc et al. 1999). Del Bufalo et al. found that M14 expresses αV and β3

integrin subunits but did not show any expression of αIIb indicating that M14

expressed αVβ3 integrin as the only β3 containing receptor complex (Del

Bufalo et al. 1998). Seoane et al. reported the expression of αV and β3

integrin and no expression of αIIb in another melanoma cell line (SK-MEL-28)

using PCR and flow cytometry techniques (Seoane et al. 2010). Trikha et al.

reported that the expression of αIIbβ3 integrin was not limited to only one

single cell line; all melanoma cell lines expressed αIIbβ3 integrin, furthermore,

translocation of αIIbβ3 integrin from the intracellular pool to the cell surface

can be mediated under the effect of an agonist (Trikha et al. 1997).

144
Both positive and negative control cell lines needed to be investigated for

each integrin subunit. The presence of positive and negative controls in the

study confirms the expression detected in other cell lines is true expression

rather than a false negative/positive.

In this part of the study, MCF-7 was utilised and as indicated by

immunoblotting very weak expression of β3 integrin. Previous studies have

detected the expression of αV, αIIb, β3 subunits and αVβ3 integrin in breast

carcinoma cell lines, such as MCF-7, using different techniques such as

immunoblotting and immunocytochemistry (Beauvais et al. 2004). The weak

β3 subunit expression in MCF-7 was also seen by Wang et al. who found that

MCF-7 had a lower expression than another breast cell line, MDA-MB-231

(Wang et al. 20100.

The A549 human NSCLC cell line was used as a possible negative control

for use in the functional assay for αVβ3. Goodman reported no expression of

αVβ3 integrin in A549 (Goodman, Grote & Wilm 2012) however, in the present

study α Vβ 3 expression was detected, by immunoblotting and

immunocytochemistry, in human NSCLC cell lines such as A549 and H460,

with both cell lines showing weak expression of the β 3 subunit and αVβ3. This

difference may be due to different culture conditions between labs.

The expression of αV and β3 subunits and αVβ3 integrin was also detected in

the colon carcinoma cell lines, DLD-1 and HCT-116, by immunoblotting and

immunocytochemistry, but no expression of β3 was noted in HT-29. The

immunodetection of β3 integrin by immunocytochemistry indicated that colon


145
carcinoma cell lines DLD-1 and HCT-116 had weak expression of the β3

subunit; the expression of the αVβ3 integrin was lower than the β3 subunit.

Few reports describe the expression of αV, β3 and αIIb subunit and αVβ3

integrin in colon cancer. Most of the previous reports concentrate on β1

integrins rather than β3 (Gong et al. 1997; O'Brien et al. 1996).

Immunocytochemistry and immunoprecipitation detected different integrins

expression in HT-29, i.e. α2, α3, α6, β1 and β5 but weak expression of αV and

no expression of β3 integrin (Haier et al. 1999). Burbridge et al. detected the

expression of αVβ3 integrin in HCT-116 and no expression in HT-29 by

immunoblotting and immunocytochemistry (Burbridge et al. 2003). This result

agrees with Trikha et al. who used LM609 and reported that HT-29 was

negative for αVβ3 integrin (Trikha et al. 2002), and other studies which also

found no expression of αVβ3 integrin in HT-29 cell (Caltabiano et al. 1999)

using the immunofluorescence technique (Liu et al. 2011).

αV was expressed at a high level in all screened cell lines, which is to be

expected as αV does not bind exclusively to β3 but can also bind to β1, β5, β6,

and β8. The expression obtained from immunoblotting and

immunocytochemistry in this chapter was similar to previous reports (Trikha

et al. 1998; Trikha et al. 2002; Zheng et al. 1999; Wang et al. 2005). β3

integrin expression differed between cell lines; β3 expression was highest in

the melanoma cell lines M14, SK-MEL-2 and UACC-62, followed by the

prostate carcinoma cell lines: PC-3 and DU-145. Lung carcinoma cell lines

A549 and H460, colon carcinoma cell line DLD-1 and HCT-116 cells and

146
breast carcinoma cell line MCF-7 showed weak expression of β3 integrin. The

HT-29 colon carcinoma cell line did not express β3 integrin.

A weak expression of the αIIb subunit was noted using immunocytochemistry

technique. However, although there was evidence among a panel of tumour

cell lines not all of them had an expression, it was not convincing. The result

obtained by immunocytochemistry did not agree with immunoblotting

technique which suggests the expression of αIIb observed by

immunocytochemistry is may be false positive result or the conditions in

western blot did not optimised properly. In addition the serum added to the

media may also play an important role in the expression because the growth

factors and the serum can be differes between labs

αVβ3 is believed to be present over the entire surface of the cell (Wang et al.

2005). Confocal microscopy was used to localise expression as intracellular

or on the cell surface. αVβ3 integrin was found to be membranous in all cell

lines tested. Immunolabelling was done on cells adhering to a coverslip, so

reducing the chance for cell to cell contact resulting in increasing

membranous expression, since the expression of αVβ3 was known to be

more prominent when cells start to adhere and migrate (Carreiras et al.

1999).

147
3.5 Conclusion

Immunoblotting and immunocytochemistry were used to characterise the

expression of αV and β3 integrins in a panel of cell lines, whose growth

characteristics have been determined. Expression of αVβ3 was successfully

detected and found to be highest in M14, UACC-62 and SK-MEL-2,

moderate in PC-3 and DU-145, weak in A549, H460, DLD-1, HCT-116 and

MCF-7, negative in HT-29. The αIIb subunit was expressed in the human

tumour cell lines by ICC but not with IMB. The αV subunit was expressed in

all human tumour cell lines while expression levels of the β3 subunit were

more variable.

148
4 Chapter 4: Investigation of the effect of αVβ3 integrin inhibition on

tumour cell migration

149
Chapter 4

4.1 Introduction

There is strong evidence that αVβ3 integrin plays a role in tumour

dissemination, and therefore developing a therapeutic strategy to antagonise

this integrin should be of value in reducing dissemination.

One of the key stages of the tumour dissemination process is cell migration.

Tumour cell migration is initiated by cell polarization and the formation of

membrane protrusions at the leading edge of the cell (Wang et al. 2005;

Brakebusch et al. 2002). There are two types of tumour cell migration: single

cell migration and collective cell migration. When a single cell migrates, it

needs to polarize and protrude at the leading edge, then attach the leading

edge to the substratum over which it migrates, followed by proteolytic

degradation of the tissue component, actomyosin contraction, and finally,

forward sliding of the cell rear. Collective cell migration occurs when cells

maintain cell-cell junctions at the leading edge and lateral regions inside the

moving cell group via cadherins (Vicente-Manzanares et al. 2005).

As previously discussed, research supports the view that the β3 integrins,

particularly αVβ3, play an important role in various stages of tumour

metastasis including migration (Seftor et al. 1992; McCabe et al. 2007;

Takayama et al. 2005). Voura et al. indicated that the strongest expression of

the αVβ3 integrin was seen in the protrusions and pseudopods of migrating

cells, and also in cells spreading on matrigel. This indicates a role for αVβ3

150
integrin in extravasation and in cell-extracellular matrix interactions (Voura et

al. 2001). Blocking β3 integrin can inhibit interaction of tumour cells with the

ECM and block tumour cell migration.

Several experimental assays (summarised in Table 4.1) are available for

measuring tumour cell migration in vitro, such as the Boyden chamber assay,

ring assay and scratch assay (Kramer et al. 2013). The scratch assay

involves scratching a confluent monolayer of the cells and then evaluating

the extent of tumour cell migration into the scratch. This assay has several

advantages that make it an attractive assay; it is simple, inexpensive and can

record the cell movement in real time (Liang et al. 2007). Furthermore, the

scratch assay can be used to study cell migration by cell-cell interaction and

cell interaction with ECM when cells are seeded on coated plate as a

monolayer, rather than held in suspension, which disrupts cell-to-cell and

cell-to-ECM interactions (Goetsch & Niesler 2011; Liang, Park & Guan 2007).

The scratch assay has been used to investigate the effect of anti-integrin

antagonists in inhibition of tumour cell migration in vitro and to study the

functional activity of tumour cells. For example, it has been used to evaluate

the anticancer effects of the disintegrin contortrostatin on the PC-3 prostate

cancer cell line (Lin et al. 2010), and to investigate the role of receptor

glycosylation on αVβ3 function (Kremser et al. 2008).

In order to utilise this assay in the evaluation of novel β 3 antagonists, the

assay must be initially validated using known anti-αVβ3 integrin antagonists.

Cyclic RGD peptides such as (cRGDfV) (section 1.2.5.3.2) (Manzoni et al.


151
2009) and the LM609 antibody are known αVβ3 integrin antagonists (section

1.2.5.3.1). Whilst LM609 is specific to the αVβ3 integrin (Trikha et al. 2002;

Voura et al. 2001; Zheng et al. 1999), cRGDfV may also antagonise the αVβ5

integrin (Manzoni et al. 2009).

4.1.1 Aims and objectives

The overall aim of the work described in this chapter is to develop and

validate a functional assay of cell migration, the scratch assay, for use in the

evaluation of β3 integrin antagonists.

This aim was achieved by addressing the following objectives:

1- Setting up the scratch assay using the cell line models characterised

in Chapter 3.

2- Determination of a safe dose of the known αVβ3 integrin antagonists,

cRGDfV and LM609.

3- Validation of the assay using cRGDfV and LM609.

152
Table 4.1 Comparison of cell migration assays (Kramer et al. 2013)
Trans-well Wound Cell Fence assay Micro- Spheroid Capillary Capillary tube Leukocyte Colloidal Time-lapse
migration healing assay exclusion carrier bead migration chamber assays migration particle cell tracking
assay (Scratch zone assay assay assay assays agarose assay
assay) technique
assay
Dimensionality 2D 2D 2D 2D 2D (3D) 2D (3D) 2D 2D 2D 2D 2D
Chemotaxis + - - - - - + + + - Assay
dependent
Stable - - - - - - + + + - Assay
chemokine dependent
gradient
Measurement Cell count Migration area Migration area Migration area Migration Migration Cell count Migration Migration Cell Cell
fluorescence area area migration distance distance migration migration
area area path path
Live imaging - + + + + + + + + + +
IF, IHC - + + + + + + - n.d. - +
Substrate PC, PET Plastic, glass, Plastic, glass, Plastic, glass, Plastic, Plastic, Glass, Glass Plastic, Plastic, Plastic,
membrane or coated or coated or coated or coated or coated or coated glass, glass, glass,
or coated or coated or coated or coated
Direction of Horizontal Horizontal Horizontal Horizontal Vertical then Mixed then Horizontal Horizontal Horizontal Horizontal Horizontal
movement then vertical horizontal horizontal
HTS + + + ± ± ± - - - + +
Type of analysis End point Kinetic kinetic Kinetic kinetic kinetic kinetic Endpoint Endpoint kinetic kinetic
(kinetic) (kinetic)
Recapitulated in Single cell Collective Collective Collective Attachment Migration Single cell Leukocyte Leukocyte Single cell Assay
vivo migration migration migration of migration of migration of and from migration, migration, migration migration dependent
mode chemotaxis epithelial epithelial epithelial migration cell cluster, Chemotaxis single cell
sheets, EMT sheets, EMT sheets, EMT established migration
cell-cell
interaction
Key: 3D: Three dimensional, - : Not suitable, +: Suitable, PC: Polycarbonate, PET: Polyethylene terphthalate, BME: Basal membrane extract, IF:
Immunofluorescence, IHC: Immunohistochemistry, HTS: High throughput screening, n.d.: Not done. EMT: Epithelial mesenchymal transmission

153
4.2 Materials and methods

4.2.1 Materials

Human tumour cell lines (Table 3.2) were maintained as described in section

3.2.2.1. All general chemicals, media and media supplements were

purchased from Sigma-Aldrich (Poole, UK), unless otherwise specified.

cRGDfV (ENZO Life Sciences, Farmingdale, NY) 4.3 Mm was dissolved in

PBS, aliquoted and stored at -20 °C, and LM609 (see Table 2.1) was diluted

in RPMI medium and used fresh. Ki-67 rabbit polyclonal primary antibody

(AB9260, Chemicon Millipore Watford, UK), LM609 (see Table 2.1) and the

Alexa fluor 546, anti rabbit, IgG (A11010, Invitrogen, USA) were used for

immunocytochemical studies.

4.2.2 Methods

4.2.2.1 Wound healing migration assay

Cells were seeded into six-well plates (culture area: 9.6 cm2) at different

concentrations in 2 ml of RPMI 1640 medium and then incubated at 37 C in

5% CO2 humidified atmosphere for three different durations (24, 48 and 72

hours) to determine the concentration and time at which a confluent

monolayer was formed. Once a confluent monolayer of cells had formed, the

cell monolayer was scratched in a straight line with a sterile P200 pipette tip

to create a gap (approximately 2 cm in length and 650 µm in width) in the

centre of the well. After scratching, the medium was slowly aspirated and

discarded, and the cells were washed with 1 ml of HBSS to remove the
154
debris and to smooth the edge of the scratch. After washing, fresh medium

was added to the cells, and the plate was incubated for 3, 6, 12, 24, 48 or 72

hours in order to investigate the time of wound healing for each cell line

(Figure 4.1). During incubation, the plates were periodically monitored using

phase-contrast microscopy (Olympus, Model: CK2). The real-time tracking of

the scratch perimeter was done by measuring the average between the

leading cells at five points along the scratch field using an eyepiece graticule.

After determining the time of wound healing for each cell line, the cells were

washed twice with HBSS and fixed with ice pre-cooled methanol for thirty

minutes at -20 °C. After fixation, the cells were hydrated by two washes in

PBS and counterstained with Harris’s Haematoxylin solution for two minutes.

The cells were then washed in tap water for one minute and left to dry at

room temperature.

The resulting plates were observed using an inverted microscope and

images were taken (Nikon camera, model: C-D 55230, Japan) at ten

positions throughout the scratch area. Scratch width was calculated using the

grid and percentage migration was calculated as follows:

Migration at time x (%) = (average scratch width at Tx /average scratch width

at T0 ) × 100.

The data were expressed as means ± SD of three independent experiments.

155
Figure 4.1 Wound healing assay steps (Hulkower 2011)
Cells were seeded as a confluent monolayer in six-well plates (A). The
monolayer was scratched using a sterile P200 pipette tip (B). The initial
perimeter was washed twice with HBSS and incubated with 1 ml of RPMI
media for 24 hours, the initial scratch measured at five points as indicated by
blue arrows (C). Cells that migrated toward the wound area were indicated by
narrowing of the scratch area after cell migration as indicated by blue arrows
(D).

4.2.2.1.1 Characterisation of the phenotype of the migrated cells in

terms of proliferation and integrin expression

ICC was carried out with LM609 to confirm that cells migrating into the wound

area expressed αVβ3 integrin, and with Ki-67, a marker for proliferation

(Gasparini et al. 1998), to confirm that wound healing was due to cell

migration rather than proliferation. M14 cells were seeded on autoclaved

coverslips in six-well plates using the optimum seeding concentration and the

required time for each cell line to grow to give a confluent monolayer. The

confluent monolayer was scratched and left to heal for 24 hours. Both healed

and initial perimeter scratches were immunolabelled by ICC technique as

previously described in Chapter 3. For Ki-67 immunolabelling, M14 cells were

fixed with 4% PFA, and blocked with 5% BSA/PBS for one hour at room

temperature. Cells were immunolabelled with Ki-67, 1:50 at 4 °C overnight

156
then the secondary antibody (Alexa fluor 546, anti rabbit) was applied, 1:50

for one hour in the dark at room temperature (for optimisation see Figure

3.5).

4.2.2.2 Evaluation of the cytotoxicity of cRGDfV and LM609 using the

MTT assay

The cytotoxicity of cRGDfV and LM609 was measured in M14 and HT-29

human tumour cell lines using the MTT assay (Mosmann 1983). The assay is

based on the ability of viable cells to reduce a yellow water-soluble

tetrazolium salt (MTT: 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyl tetrazolium

bromide) to a purple water-insoluble formazan product by mitochondrial

succinate dehydrogenase, with the amount of formazan produced being

proportional to the number of viable cells present. The formazan products are

dissolved in DMSO and the absorbance of the resulting solution is

determined spectrophotometrically. In the first lane (8 wells per lane) of a 96-

well plate, 200 µl of RPMI medium was added to serve as a blank. To

subsequent lanes, 1×104 /ml tumour cells were plated in a final volume of 180

µl of medium and incubated for 24 hours. 20 µl of the test compounds: 0.01,

0.05, 0.5, 5.0 and 50 µM cRGDfV and 2.5, 5.0, 7.5 and 10 µg/ml LM609 were

then added to all wells except the RPMI blank control and untreated control

for 96 hours of continuous drug exposure at 37 °C in a humidified 5% CO 2

atmosphere.

Following incubation, the medium was removed and 200 µl of MTT solution

(5.0 mg/ml) was added per well. The plate was incubated for four hours in the
157
dark at 37 °C, 5% CO2 humidified atmosphere. The plate was centrifuged at

1000 rcf for five minutes, the supernatant was removed and 150 µl/well

DMSO was added with vigorous mixing in the pipettor to dissolve the

formazan crystals. The absorbance of the resulting solution was determined

at 550 nm using a Lab system Multiscan Plus spectrophotometer (Lab-

systems Group, UK). The adjusted mean absorbance for each compound

concentration was calculated by subtracting the mean background

absorbance (average of the blank well) from the mean absorbance of the test

wells. A dose response curve of adjusted mean absorbance against

concentration was constructed, and the IC50 values for each compound

against each cell line were calculated using least squares method in

Microsoft Excel.

4.2.2.3 Validation of the scratch assay using the integrin antagonists

cRGDfV and LM609

The scratch assay was carried out as described in section 4.2.2.1. cRGDfV

used on A549, DLD-1, DU-145, M14, PC-3 and UACC-62. LM609 used on

A549, DLD-1, M14 and PC-3. cRGDfV (0.05, 0.5, 5.0 and 50 µM) and LM609

(0.5 and 2.5 µg/ml) were diluted in a final volume of 1 ml RPMI 1640

medium, added to the scratched monolayer after the washing step, and

incubated for 24 hours. In each six-well plate, two wells were used as

controls, one for the initial perimeter, and the other for 24 hours of wound

healing. The specific positions were photographed as described in section

4.2.2.1 and analysed using a superimposed grid (Figure 4.2, A & B).

158
Migration compared to untreated control (%) was calculated as % migration =

100 × (ST0-ST24)/ (SU0-SU24) × 100.

Where ST0 is scratch at initial perimeter, SU24 is untreated scratch after 24

hours and ST24 is treated scratch after 24 hours. The data were expressed as

the means ± SD of three independent experiments.

Scratch at time 0 (ST0) Untreated scratch after 24 Treated scratch after 24


hours (SU24) hours (ST24)
Figure 4.2 Scratch assay analysis
Figure A shows a black line drawn at the back of each well to separate it into
two to ensure the images were captured at the same position each time.
After image capture, the original image was analysed by superimposing a
grid over the image, as shown in B.

159
4.2.2.4 Statistical analysis

The student’s t-test and Kruskall wallis test were used for statistical analysis

of the scratch assays, with results considered statistically significant and

highly significant for p < 0.05 and p < 0.01 respectively. Data are presented

as mean ± standard deviation, with each experiment repeated at least three

times.

160
4.3 Results

4.3.1 Development of the wound healing migration assay

A scratch wound healing assay was performed using cell lines that showed

high, low, weak or no expression of αVβ3 integrin. Developing this assay

required: optimisation of seeding density required for each cell line to form a

confluent monolayer, working on the scratching technique to produce a

consistent scratch width, determination of the wound healing times for the

different cell lines, and confirmation that cells occupying the wound area had

migrated there and were not just the result of proliferation by cells at the

wound edges.

4.3.1.1 Seeding density

As the scratch assay method is dependent on the cell density per well

leading to a confluent monolayer before scratching, the different tumour cell

lines were seeded into six-well plates at different concentrations for 24, 48

and 72 hours in order to determine the optimum seeding concentration for

each cell line.

Initially different concentrations of cells were seeded into six-well plates,

ranging from 1 × 103 to 1 × 106 cells /ml and incubated at 37 °C, 5% CO2 for

24 hours. After 24 hours, the appearance of the monolayer was analysed

visually, and if a confluent monolayer was reproducibly formed, then the

concentration was deemed suitable for seeding. Cell lines that did not form a

confluent monolayer after 24 hours were incubated for longer times. Different

161
cell lines required different seeding concentrations and incubation times to

form a confluent monolayer (Figure 4.3).

A549 (2 105, 48 hrs) H460 (2 105, 48 hrs) MCF-7 (1 106, 48 hrs)

DLD-1 (2 105, 48 hrs) HT-29 (5 105, 72 hrs) HCT-116 (1 106, 24 hrs)

M14 (8 105, 48 hrs) UACC-62 (2 105, 48 hrs) SK-MEL-2 (1 106, 48 hrs)

PC-3 (2 105 , 48 hrs) DU-145 (3 105, 24 hrs)

Figure 4.3 Formation of confluent monolayers for the cell line panel
(cell density per ml and time in hours)
Bar length = 200 µm.

162
4.3.1.2 Healing time

The time required for wound closure in each cell line was evaluated. After 24

hours, only M14 cells showed a high percentage (85%) of wound closure.

PC-3, DLD-1, UACC-62, DU-145 and A549 showed a moderate wound

closure in comparison to the initial perimeter after 24 hours; PC-3, DLD-1 and

UACC-62 monolayers were respectively 94%, 90%, and 78% healed after 48

hours and DU-145 100% healed after 72 hours. H460, HT-29, SK-MEL-2,

MCF-7 and HCT-116 had slow wound closure and scratch was not healed

after 72 hours (Figure 4.4).

163
A
T0 T 24 hours T 48 hours T 72 hours

A549

H460

MCF-7

DLD-1

HT-29

HCT-116

M14

UACC-62

SK-MEL-2

PC-3

DU-145

B 100
** ** **
**
** **
** ** **
**
80

** **
** **
60 **
** **
T 24 hours
40 ** T 48 hours.
Migration (%)

T 72 hours

* * * * * *
20 * * *
* * * * *
*

Figure 4.4 Migration of different tumour cell lines as assessed by the


scratch assay
A: A confluent monolayer was scratched and incubated with complete
medium and wound closure was measured using the grid. Images are
representative of three independent experiments. Bar length = 200 µm.
B: Results were analysed and the mean percentage of wound closure shown
±SD based on three independent experiments. ** indicated p < 0.01 and *
indicated p <0.05.

164
As a part of assay development, the phenotype of the cells seen in the

wound area was characterised using ICC. The αVβ3 integrin expression of

M14 cells migrating into the wound area was evaluated by labelling cells with

LM609. As shown in Figure 4.5, all cells found in the wound area expressed

αVβ3 integrin (Figure 4.5 A).

M14 cells were immunolabelled with Ki-67 to determine whether cells moving

into the wound area had migrated and not proliferated. Nuclear labelling was

obtained in cells which had moved toward the wound area, however there did

not seem to be a higher rate of proliferation in the cells found in the wound

area (Figure 4.5 B).

165
A TRITC DAPI Overlay X10 Overlay X40

Initial perimeter
24 hours wound healing

B
TRITC DAPI Overlay x40 Overlay x10
24 hours wound healing

Figure 4.5 Labelling M14 cells at initial wound perimeter (T=0) and after
24 hours of wound healing with LM609 and Ki-67 using ICC
A. Expression of αVβ3 integrin at the initial wound perimeter and after 24
hours of wound healing using LM609, anti-αVβ3 integrin antibody. All M14
cells were labelled with LM609 at the initial wound perimeter and after 24
hours. Cells migrating into the wound area showed αVβ3 integrin expression
as indicated by arrow. B. The immunolabelling with Ki-67 confirmed that M14
cells moving into the wound area had migrated. Examining M14 cells under
higher magnification showed that Ki-67 is a nuclear immunolabelling and it
did not seem that M14 had a high rate of proliferation. Bar length (A) = 60 µm
and (B) = 200 µm.

166
4.3.2 Evaluation of the cytotoxicity of cRGDfV and LM609 using the

MTT assay

The MTT assay was used to assess the cytotoxicity of cRGDfV against M14

and HT-29 cells and LM609 against M14 cells over a range of different

concentrations for 96 continuous hours of drug exposure. As shown in Figure

4.6, neither antagonist demonstrated cytotoxicity in the cells at the

concentration tested, and was it not possible to obtain an IC50 for either

compound. Thus, both antagonists are non-toxic within this concentration

range for these cell lines and could be used in the scratch assay.

100 * * * *
* 100
* *
80 80
Cell survival (%)
Cell survival (%)

60 60

40 40
M14
20 Ht29
HT-29 20 M14

0 0
0.01 0.05 0.5 5 50 2.5 5 7.5 10
cRGDfV (µM) LM609 (µg)

Figure 4.6 Evaluation of cRGDfV and LM609 cytotoxicity against human


tumour cell lines M14 and HT-29
Cells were exposed to the antagonists for 96 hours. There was a negligible
effect on cell survival by either compound. The results was statistically
significant p< 0.05.

167
4.3.3 The effect of cRGDfV on the migration of human tumour cell lines

Cell lines with varying αVβ3 integrin expression that demonstrated definite

wound healing above were used to validate the wound healing assay

because HT-29 (no αVβ3 integrin expression) can not be used as a model in

scratch assay due to slow wound healing. Initially the M14 cell line was used

due to its high expression of αVβ3 integrin in addition to its good growth and

rapid wound healing. All the concentrations of cRGDfV tested led to inhibition

of M14 migration. UACC-62 also showed high expression of αVβ3 integrin,

and again all cRGDfV concentrations led to inhibition of UACC-62 migration,

although with less of an effect than on M14 except at 0.05 µM, which showed

the same percentage of inhibition in both cell lines with other concentrations

tried. 50 µM cRGDfV was not toxic to M14 and UACC-62 cells. The effect of

all cRGDfV concentrations was statistically significant p < 0.01 expect with

DLD-1 the significant reached at 50 and 5 µM only.

50 µM cRGDfV was toxic to PC-3 and A549 cells (Figure 4.7). Thus, the

effect of cRGDfV on the migration of PC-3 and A549 cells was tested at 5.0,

0.5 and 0.05 µM and it was statistically significant p < 0.01 except for A549 at

0.05 µM, p < 0.05 (Figure 4.7).

DLD-1, used as tumour model of weak expression of αVβ3 integrin, did not

show a dose dependent response to cRGDfV. The inhibition of DLD-1

migration seen with high concentrations of cRGDfV may be due to inhibition

of αVβ5 integrin in DLD-1 (Appendix 1). The effect of 50 and 5 µM was

168
statistically significant with p <0.01. There was no significant effect of

cRGDfV at 0.5 and 0.05 µM on DLD-1 cell migration (Figure 4.7).

cRGDfV was effective in inhibition of all tumour cell migration at 5.0, 0.5, and

0.05 µM except DLD-1 which had an effect at 50 and 5.0 µM only. It was vital

to know why cell lines with moderate and weak expression of αVβ3 integrin

were affected by cRGDfV. These cell lines may also express αVβ5 integrin

(Appendix 1) which can also be antagonised by cRGDfV. To investigate this

possibility, LM609, which is reported to be specific to αVβ3 integrin, was also

used in the scratch assay.

169
A T0 Untreated 0.05 µM 0.5 µM 5.0 µM 50 µM

DLD-1
UACC-62
M14
PC-3
A549
DU-145

B PC-3 A549

100 *
** **
** ** **
**
80
** **
**
** **
**
Untreated
60 **
0.05 µM
Migration (%)

**
** ** 0.5 µM
** **
40
5.0
5 µMµM
50 µM
20 **

T T
0
DLD-1 UACC-62
UACC62 M14 PC-3 A549 DU-145

Figure 4.7 Effects of cRGDfV on tumour cell migration in the cell panel
using the scratch assay
A: Migration of different human tumour cell lines toward wound area was
inhibited using different concentrations of cRGDfV. Images shown as are
representative of three independent experiments. Bar length = 200 µm.
B: Toxicity was observed with PC-3 and A549 at 50 µM as indicated by (T) in
the chart. ** indicated p < 0.01 and * indicated p <0.05.

170
4.3.4 The effect of LM609 on human tumour cell migration

LM609 was used first with M14 cell line at two different concentrations, 0.5

and 2.5 µg/ml. LM609 at both of concentrations inhibited M14 cell migration,

but had little effect on migration of the PC-3 and DLD-1 cells and a negligible

effect on the migration of A549 cells, which showed cytotoxicity with cRGDfV

(Figure 4.8). The effect of LM609 on tumour cells migration was statistically

significant with p < 0.01. M14, PC-3, DLD-1 and A549 (as one cell line from

each type of tumour) were selected as a model of high, moderate and weak

expression to validate the scratch assay.

171
A T0 Untreated 0.5 µg/ml 2.5 µg/ml

M14
PC-3
A549
DLD-1

100
**

**
80
**

**
**
60
Untreated
cell migration

**
(%)

0.5 µg/ml LM609


40 2.5 µg/ml LM609
% TumourMigration

20

0
M14 PC-3 A549 DLD-1

Figure 4.8 The effect of LM609 on tumour cell migration


A: Migration of tumour cells toward the scratch area was inhibited by LM609.
Bar length = 200 µm. B: Images were analysed and aveage result shown as
representative of three independent experiments. ** indicated P < 0.01.

172
4.4 Discussion

The main aim of the work described in this part of the study was to develop a

scratch assay that can be used to measure one of functional aspects of

antagonising integrin expression, migration, for assessing novel integrin

antagonists.

The scratch assay has some key advantages that led to its use in this study.

The assay is easy to set up and does not require any specific equipment, and

all the required materials are available in any cell culture laboratory (Liang,

Park & Guan 2007). Nevertheless, the in vitro scratch assay presented some

technical challenges: the first was the difficulty of creating scratched areas of

equal sizes. This variation in wound size between wells of the same plate

was resolved by practicing to achieve scratches of the same width (Appendix

2). Furthermore, there was difficulty achieving a wound without any cells.

This problem was overcome by tilting the plate and washing the wound from

the corner of the plate, which resulted in the removal of all unwanted cells

from the wound area.

The intention was to use cell lines with different levels of αVβ3 integrin

expression to investigate the effect of αVβ3 antagonism. Thus, tumour cell

lines with different αVβ3 integrin expression which demonstrated clear wound

healing were identified for use. The screening of a panel of tumour cell lines

for wound healing (Figure 4.4) offered a chance to select the best model for

testing β3 antagonists. Not all cell lines showed rapid wound healing due to

the differing properties of the various cells in growing cultures. Some cells
173
grow in clumps rather than in flat spreading way. Cells growing in clumps are

associated with difficulty in defining the starting point of cell migration which

complicates image analysis.

Melanoma cell lines that exhibited high expression of αVβ3 integrin were

screened for wound healing in order to select the best model. The only cell

line that showed rapid wound healing (scratch nearly closed after 24 hours)

was M14, followed by UACC-62, which exhibited moderate wound healing

whereas SK-MEL-2 showed slow wound healing that did not close even after

72 hours. SK-MEL-2 has previously been shown to migrate very slowly, and

to lack a response to TGF-β1, which caused a change in phenotype and

signalling promoting cell migration in other melanoma cell lines (Janji et al.

1999).

PC-3 and DU-145 cell lines exhibited moderate wound healing after 24

hours. PC-3 has been reported to migrate rapidly, with a scratch being

completely healed after 12 hours (Lin et al. 2010). The slower wound healing

of PC-3 seen here may be due to different culture conditions or due to the

original study seeding PC-3 cells on matrigel coated plates which promote

integrin-mediated adhesion, whereas in the present study the PC-3 cells

were seeded on uncoated plates which may have led to a slower migration of

PC-3. Wang et al. found that wound healing was faster in PC-3 than in DU-

145 (Wang et al. 2005), which supported the present study result where both

cell types showed moderate wound healing after 24 hours, but PC-3 had

wound closure after 48 hours, whereas DU-145 did not.

174
Colon carcinoma cell lines were screened for wound healing to be used as a

αVβ3 weak or negative control models. However, only DLD-1 had moderate

wound healing after 24 hours whereas HCT-116 and HT-29 did not show

wound closure even after 72 hours. Thus, both HCT-116 and HT-29 were

excluded from the rest of the scratch assay study, and DLD-1 was selected

for further scratch assays as a model of weak expression of αVβ3 integrin.

Other cell lines with low αVβ3 integrin expression were also investigated.

MCF-7 (weak αVβ3) did not achieve wound closure after 72 hours. Lung

carcinoma cell lines had weak αVβ3 integrin expression; moderate wound

healing was observed for A549 while H460 did not have wound closure even

after 72 hours. Cell lines that showed very slow wound healing (HCT-116,

HT-29, H460 and MCF-7) in the scratch migration assay were excluded from

the rest of the study.

The scratch assay provides valuable results regarding the expression of the

αVβ3 integrin in tumour cell migration. Immunolabelling with LM609 of M14

cells in the scratch assay has never been done before, although this

technique has been demonstrated by Carreiras et al. who showed that

IGROV-1 ovarian carcinoma cells expressing αVβ3 integrin migrated toward a

scratch area by immunolabelling migrating cells with LM609 (Carreiras et al.

1999).

The assay was validated before utilising it to evaluate novel anti-β3 integrin

antagonists by using well-characterised commercially available anti-αVβ3

integrin antagonists as positive controls. Before using the antagonists on


175
tumour cells, the cytotoxicity of cRGDfV and LM609 was tested using the

MTT assay, which showed that cRGDfV had no toxic effects on the M14 and

HT-29 human tumour cell lines. LM609 also did not show any toxicity to M14

cells. The only toxicity observed to other cell lines (did not tested by MTT

assay) was with 50 µM cRGDfV to PC-3 and A549 only.

An anti-migratory effect was obtained with cRGDfV on cells with high,

moderate and low αVβ3 integrin expression (Figure 4.7). PC-3 cells showed

moderate expression of the αVβ3 integrin with high response to cRGDfV at

5.0, 0.5 and 0.05 µM. This may be due to expression of αVβ5, which is also

antagonised by cRGDfV. Bisanz et al. have previously shown that the

expression of β3 was lower than for αVβ1 and αVβ5 in PC-3 cells (Bisanz et al.

2005).

The LM609 antibody has been used to detect the expression of the αVβ3

integrin and as an inhibitor (Leavesley et al. 1992; Seftor et al. 1992; Trikha

et al. 2002). A previous study used 20 µg/ml LM609 to study endothelial cell

migration (Li et al. 2006). However, use of 20 µg/ml on M14 cells led to loss

of M14 adhesion to the plate. Thus, lower concentrations (2.5 and 0.5 µg/ml)

were used to study the effect on migration.

LM609 is specific for αVβ3, and does not antagonise related integrins such as

αVβ5. The observed inhibition of cell migration suggests that αVβ3 plays a

significant role in the migration of M14, and PC-3 cells and may also be

involved in migration of DLD-1. αVβ3 integrin has been shown to play an

important role in migration of other cell lines; for example, a study done by
176
Leavesley et al. reported that LM609 inhibited migration of pancreatic FG

cells transfected with αVβ3 integrin in the Boyden chamber migration assay

(Leavesley et al. 1992).

4.5 Conclusion

This work has developed and validated an assay for evaluating integrin-

mediated cell migration, which can be used to evaluate novel αVβ3

antagonists in the following chapter.

177
5 Chapter 5: Investigation of the cytotoxicity of potential novel β3

integrin antagonists and their effect on tumour cell migration

178
Chapter 5

5.1 Introduction

The previous chapter described the development and validation of the

scratch assay to evaluate novel β3 integrin antagonists. M14 cells were

chosen as the optimal cell line to use in the scratch assay due to their high

αVβ3 integrin expression and rapid wound healing. αVβ3 integrin is required

for melanoma cell motility and it has been reported that blocking αVβ3 integrin

leads to blocking of melanoma cell migration (Li et al. 2001).

In this chapter, the optimised scratch assay was utilised to evaluate potential

novel β3 integrin antagonists synthesized at the Institute of Cancer

Therapeutics, using M14 as the model for testing for inhibition of tumour cell

migration due to its high αVβ3 integrin expression.

In the present study the cytotoxic effect of potential novel β3 integrin

antagonists will be tested on tumour cells before they are evaluated in the

functional scratch assay, in order to know that the effect of the potential novel

β3 integrin antagonists was due to inhibition β 3 integrin function in mediating

tumour cell migration rather than cytotoxicity. Dayam et al. identified highly

selective αVβ3 integrin antagonists through screening a panel of potential

αVβ3 integrin antagonists they had synthesized. Six compounds were tested

in breast tumour cells, MDA-MB-435 and MCF-7 as high and low αVβ3

models respectively. They found one of anti αVβ3 small molecules was toxic

whereas the others showed no toxicity. This study indicated the importance

179
of testing the toxicity of small molecules before evaluating their effects on

cancer cell migration and attachment (Dayam et al. 2006).

5.1.1 Aims and objectives

The overall aim of this chapter was to evaluate potential novel β3 integrin

antagonists in terms of their cytotoxicity and effect on tumour cell migration.

This aim was achieved through the following objectives:

1- Evaluation of the cytotoxicity of potential novel β3 integrin antagonists

using the MTT assay for cytotoxicity.

2- Determination of the effects of potential novel β3 integrin antagonists

on M14 cell migration using the scratch assay.

180
5.2 Materials and methods

5.2.1 Materials

Human tumour cell lines M14, MCF-7, and HT-29 (see Table 3.2 for details)

were used in this chapter and maintained as described in section 3.2.2.1. All

general chemicals, media and media supplements were purchased from

Sigma-Aldrich (Poole, UK), unless otherwise specified. cRGDfV, was

sourced as described in section 4.2.1. A series of potential novel β3 integrin

antagonists was synthesized at the Institute of Cancer Therapeutics (see

Table 5.1 for details). All antagonists dissolved readily in 0.1% DMSO and a

stock of 100 mM was prepared. For compound structures see attached file.

LM609 details are summarised in Table 2.1.

Table 5.1 Compounds used


No Compound Molecular weight
1 ICT9003 320
2 ICT9018 531
3 ICT9019 503
4 ICT9020 517
5 ICT9053 538
6 ICT9055 542
7 ICT9057 500
8 ICT9062 542
9 ICT9064 528
10 ICT9068 421
11 ICT9072 474
12 ICT9073 516

181
5.2.2 Methods

5.2.2.1 Evaluation of the cytotoxicity of potential novel β3 integrin

antagonists using the MTT assay

The cytotoxicity of the potential novel β3 integrin antagonists was evaluated

in M14 (high expression of αVβ3 integrin), MCF-7 (weak expression of αVβ3

integrin), and HT-29 (no expression of αVβ3 integrin) human tumour cell lines

using the MTT assay as described in section 4.2.2. The β3 integrin

antagonists were used at the following concentrations 0.01, 0.1, 1.0, 10 and

100 µM.

5.2.2.2 Wound healing migration assay

The anti-migratory effect of the potential novel β3 integrin antagonists on M14

cell migration was evaluated using the scratch assay as described in section

4.2.3. Briefly, wounded cultures of cells were exposed to 0.01, 0.1, 1.0, 10

and 100 µM of the potential novel β3 integrin antagonists for 24 hours, and

then the cultures were processed as described in Chapter 4. cRGDfV at 0.5

and 5.0 µM was included as a positive control.

5.2.2.3 Statistical analysis

The student’s t-test (parametric) and kruskall wallis (non parametric) test

were used for statistical analysis of the scratch assays, with results

considered statistically significant and highly significant for p < 0.05 and p <

0.01 respectively. Data are presented as mean ± standard deviation, with

each experiment repeated at least three times.


182
5.3 Results

5.3.1 Evaluation of the cytotoxicity of potential novel β3 integrin

antagonists

The MTT assay was used to assess the cytotoxicity of potential novel β3

integrin antagonists against M14, HT-29 and MCF-7 cells over a range of

different concentrations for 96 continuous hours of drug exposure. As shown

in Figure 5.1 and Table 5.2, the majority of compounds demonstrated

minimal toxicity, with it not being possible to obtain IC50 at the highest dose

assayed, 100 µM.

As can be seen from Table 5.2, ICT9062, ICT9072, and ICT9073 were toxic

to all cell lines with IC50 calculated from three independent experiments.

183
100 100 100 100

80 80 80 80

Cell survival (%)

Cell survival (%)

Cell survival (%)


Cell survival (%)

60 60 60 60

40 40 40 40
M14 M14 M14 M14
20 MCF-7 20 MCF-7 20 MCF-7 20 MCF-7
HT-29 HT-29 HT-29 HT-29
0 0 0 0
0.01 0.1 1 10 100 0.01 0.1 1 10 100 0.01 0.1 1 10 100 0.01 0.1 1 10 100
ICT9003 (µM) ICT9018 (µM) ICT9019 (µM) ICT9020 (µM)

100 100 100 100

80 80 80 80

Cell survival (%)

Cell survival (%)


Cell survival (%)

Cell survival (%)


60 60 60 60

40 40 40 40
M14 M14 M14 M14
MCF-7 MCF-7 MCF-7 MCF-7
20 20 20 20
HT-29 HT-29 HT-29
HT-29
0 0 0 0
0.01 0.1 1 10 100 0.01 0.1 1 10 100 0.01 0.1 1 10 100 0.01 0.1 1 10 100
ICT9053 (µM) ICT9055 (µM) ICT9057 (µM) ICT9062 (µM)
100
100 100 100
80
80 80 80

Cell survival (%)


Cell survival (%)
Cell survival (%)

Cell survival (%)


60
60 60 60

40 40 40 40
M14 M14 M14 M14
MCF-7 MCF-7 20 MCF-7 MCF-7
20 20 20
HT-29 HT-29 HT-29 HT-29
0 0 0 0
0.01 0.1 1 10 100 0.01 0.1 1 10 100 0.01 0.1 1 10 100 0.01 0.1 1 10 100
ICT9064 (µM) ICT9068 (µM) ICT9072 (µM) ICT9073 (µM)

Figure 5.1 Evaluation of the cytotoxicity of the potential novel β3 integrin antagonists in the tumour cell line panel
184
Table 5.2 IC50 values for the potential novel β3 integrin antagonists in
the tumour cell line panel
Compound M14 MCF-7 HT-29
IC50 ± SD (µM) IC50 ± SD (µM) IC50 ± SD (µM)
ICT9003 > 100 >100 >100
ICT9018 > 100 >100 >100
ICT9019 > 100 >100 >100
ICT9020 36 ± 18.6 >100 80 ± 5.8
ICT9053 72.7 ± 9.8 >100 >100
ICT9055 > 100 >100 25 ± 5.8
ICT9057 > 100 >100 >100
ICT9062 3.3 ± 0.8 1.4 ± 0.5 21.2 ± 30
ICT9064 > 100 >100 >100
ICT9068 > 100 9.5 ± 9 >100
ICT9072 50 ± 47 14 ± 8.5 0.8 ± 0.8
ICT9073 61 ± 32.9 6.3 ± 6.9 36.7 ± 42.2

5.3.2 Characterisation of the anti-migratory effect of potential novel β3

integrin antagonists using the scratch assay

The potential novel β3 integrin antagonists were tested in two batches; with

information obtained from the first batch informing the concentrations used in

testing the second batch. In the first batch, ICT9003, ICT9055, ICT9057 and

ICT9064 were screened at four concentrations 0.1, 1.0, 10 and 100 µM. At

the lowest concentration, ICT9055 showed the highest effect of inhibiting

M14 cell migration; whilst the other potential novel β3 integrin antagonists had

lower effects at the same concentrations (Figure 5.2). The results were seen

to be highly statistically significant p < 0.01 for all novel β3 integrin

antagonists. IC50 values were calculated as 4.8 ± 0.2 µM for ICT9003, 1.0 ±

0.09 µM for ICT9057 and 0.2 ± 0.06 µM for ICT9064 (Table 5.3).

185
A T0 T24 hours 0.5 µM cRGDfV 5.0 µM cRGDfV

0.1 µM 1.0 µM 10 µM 100 µM


ICT9003

CB57
ICT9055
ICT9057
ICT9064

B
100

80
** ** **
0.1 µM
** **
60 ** 1.0
1 µMµM
Migration (%)

**
** ** ** 10 µM
** 100 µM
40 **
**
** 0.5 µM
**
20 **
55.0
µMµM
** **

0
ICT9003 ICT9055 ICT9057 ICT9064 cRGDfV

Figure 5.2 Effect of potential β3 integrin antagonists on M14 cell


migration
A: M14 cells were wounded and then treated with different concentrations of
different potential novel β3 integrin antagonists for 24 hours. Bar length = 200
µm. B: The migrated cells were counted and related to 100% untreated
cells. Results were statistically significant for all antagonists with p <0.01.

186
A concentration of 10 µM was selected as an initial screening concentration

for the second batch of compounds, which were ICT9018, ICT9019,

ICT9020, ICT9053, ICT9062, ICT9068, ICT9072 and ICT9073. As seen in

the cytotoxicity assay, some of the antagonists (ICT9062, ICT9072 and

ICT9073) were observed to be toxic. In the MTT assay cells were treated for

96 hours, whereas in the scratch assay the M14 cells were treated for only

24 hours, thus as the exposure time was shorter it was assumed that these

concentrations would not have a cytotoxic effect, however this was not the

case and the toxicity was still observed (Figure 5.3). Therefore, the β3

integrin anatgonists were tried at lower concentrations which were 1.0, 0.1

and 0.01 µM.

187
T0 T24 hours 0.5 µM cRGDfV 5.0 µM cRGDfV ICT9018 ICT9019

ICT9020 ICT9053 ICT9062 ICT9068 ICT9072 ICT9073


Figure 5.3 Effect of 10 µM β3 antagonists on the inhibition of M14 cell migration by the scratch assay.
A confluent monolayer of M14 cells was scratched, and then treated with 10 µM of the second batch of novel anti-β3 integrin
antagonists for 24 hours. Bar length = 200 µm.

188
Further screening was carried out for the β3 integrin antagonists that showed

an inhibitory effect on M14 cell migration using a more extensive

concentration range, from 0.01 µM up to 10 µM (Figure 5.4). The compounds

that were toxic at 10 µM were also evaluated, and these demonstrated a high

percentage of inhibition of M14 cell migration at lower concentrations, with

ICT9073 having the highest percentage of inhibition at 0.01 µM. IC50s were

9.5 ± 0.9 µM for ICT9019, 0.15 ± 0.03 µM for ICT9062 and 0.15 ± 0.04 µM

for ICT9072 (Table 5.3).

189
A 0.01 µM 0.1 µM 1.0 µM 10 µM

ICT9019
ICT9062
ICT9068
ICT9072
ICT9073

100
**
** **
**
**
80 **
** ** **
**
**

**
60 **
** 0.01 µM
**
Migration (%)

** 0.1 µM
40 1 µM
**
10 µM
**
20 0.5 µM
**
5 µM
T T T
0
ICT 9019 ICT 9062 ICT 9068 ICT 9072 ICT 9073 cRGDfV

Figure 5.4 Effect of potential β3 integrin antagonists in inhibition M14


cell
migration by scratch assay
A: M14 cells were wounded then treated with different concentrations of
different potential novel β3 integrin antagonists for 24 hours. Bar length = 200
µm. B: The migrated cells were counted and related to 100% untreated
cells. Result was statistically significant p <0.01. ICT9062, ICT9072 and
ICT9073 were toxic at 10 µM as indicated by T.

190
Table 5.3 A summary of effect of the potential novel β3 integrin
antagonists in inhibition of M14 cell migration after 24 hours treatment
with the scratch assay
Compound IC50
ICT9003 4.8 ± 0.2 µM
ICT9018 -
ICT9019 9.5 ± 0.9 µM
ICT9020 -
ICT9053 -
ICT9055 < 0.1 µM
ICT9057 1.0 ± 0.09 µM
ICT9062 0.15 ± 0.03 µM
ICT9064 0.2 ± 0.06 µM
ICT9068 > 50 µM
ICT9072 0.15 ± 0.04 µM
ICT9073 < 0.01 µM

191
5.4 Discussion

In the previous chapters characterised cell lines were selected for their

integrin expression patterns and then used to set up and validate a functional

assay of integrin antagonism, the scratch assay. In this chapter the validated

assay was used to evaluate novel antagonists in inhibition of migration of a

cell line expressing β3 integrin, M14.

Initially β3 integrin antagonists were evaluated for their relative cytotoxicity.

As shown in Table 5.2, different anti β3 integrin antagonists had different

cytotoxic effects. ICT9003, ICT9018, ICT9019 and ICT9057 were not toxic to

M14, MCF-7 and HT-29 cells. ICT9053 had a cytotoxic effect on M14 only,

and ICT9055 was cytotoxic to HT-29 only. ICT9064, the free acid of ICT9055

showed no obvious cytotoxicity with all tested cell lines. ICT9062 showed a

cytotoxic effect with all tested cell lines, which is surprising since it differs

from ICT9055 only in the stereochemical arrangement of bonds around the

central linker. ICT9068 showed cytotoxic effect with MCF-7 only, and

ICT9020 was cytotoxic with both M14 and HT-29.

The cytotoxic effect was evaluated using the MTT assay, to ensure that the

results observed were due to blockade of integrin function (reduction in cell

migration in the scratch assay), rather than general toxicity.

There was no obvious pattern in cytotoxicity across compounds and cell

lines. Since compounds that were shown to be active in the scratch assay

(and other work in-house) such as ICT9055, are not toxic in the high β 3

192
expressing M14 cells, the toxicities observed are unlikely to be β 3-mediated.

However, cell line specific cytotoxicity may be due to effects mediated by

other integrins: for example, αVβ6 is known to be important in colon cancer

survival, so the toxicity observed with HT-29 treated with ICT9055 may result

from antagonism of β6 (Peng et al. 2009). Future work on these compounds

should involve assessment of their selectivity for similar integrins.

ICT9072 and ICT9073 had a cytotoxic effect with all tested cell lines, which

may be due to the presence of the amino pyrimidine group at the start of the

carbon chain, which has previously been demonstrated to cause toxicity

(Gorneva 2005). Cilengitide induced cytotoxicity in glioma cells U-251MG

and U-87MG (Lomonaco et al. 2011) whereas cRGDfV did not show any

cytotoxic effect to same cells (Chatterjee et al. 2000).

The effect of the potential novel β3 integrin antagonists was investigated by

the scratch assay. The results indicated that different antagonists had

different effects on inhibition of M14 cell migration. ICT9003 had the lowest

effect, which may be due to the absence of both tetrahydronaphthyridine

(THN) at the beginning of the carbon chain and the NHSO 2Mes exosite

binding group at the end of the carbon chain.

As shown in Figure 5.3, ICT9018, ICT9019, ICT9020 and ICT9003

(pyrimidine) (Figure 5.4) at 10 µM were not effective in inhibiting M14 cell

migration. In contrast, ICT9055, ICT9057 and ICT9064 which all contain the

THN group, were all effective inhibitors of M14 cell migration. This consistent

effect of ICT9057, ICT9062 and ICT9064 indicated that the presence of the
193
exosite binding NHSO2Mes group is also required for activity. The ICT9062

had the same structure as ICT9055, but with different stereochemistry which

led to it being cytotoxic to all tested cell lines suggesting that stereochemistry

is a major consideration in antagonist design. ICT9055 was thus the most

effective functional antagonist with no cytotoxic effect on M14 cells, among

the compounds screened.

5.5 Conclusion

A total of 12 potential β3 integrin antagonists were screened in the scratch

assay, with some of the compounds demonstrating significant antagonistic

effects. ICT9055 was the most effective of the compounds screened and

would warrant further investigation.

194
6 Chapter 6: Discussion and future work

195
6.1 General discussion and future perspective

The current strategy for cancer therapy is still inadequate; chemotherapy

continues to be associated with long-term toxicities and drug resistance,

while surgery and radiotherapy are not suitable for treating the whole body

after cancer metastasis. Thus, research has begun to focus on new

strategies for cancer therapy based on targeted therapy. The development of

targeted therapies improves differentiation between normal and neoplastic

cells.

Integrins have become an attractive receptor target for cancer therapy

(Goodman & Picard 2012; Sheldrake & Patterson 2009). However, among

the panel of integrin antagonists summarised in Table 1.3, none of them

specifically targets tumour cell integrins and their interaction with platelet and

endothelial cells to inhibit angiogenesis and tumour metastasis. Furthermore,

a large number of the available antagonists available are either antibodies or

peptides and small molecules are preferred as antagonists as they are less

costly to manufacture than antibodies, have better stability and

pharmacokinetic/pharmacodynamic (PKPD) properties, and can be

administered orally (Alig et al. 1992; Millard, Odde & Neamati 2011).

Blockade of αIIbβ3 and αVβ3 integrins has a critical role in preventing cancer

metastasis and angiogenesis (Liu et al. 2009). Blockade of both tumour cell

αVβ3 and host cell β3 integrins (endothelial αVβ3 and platelets αIIbβ3) can

enhance inhibition of tumour growth in vivo (Trikha et al. 2002). At the

Institute of Cancer Therapeutics we hypothesise that dual αIIbβ3 and αVβ3


196
antagonism will have a direct effect on β3-expressing tumour cells and it will

also through targeting tumour cell interaction with endothelial cells and

platelets lead to inhibition of angiogenesis and metastasis.

In this thesis, the aim was to evaluate the effect of potential novel β3 integrin

antagonists in inhibition of human tumour cell migration using cell based

assays. The main objectives of the work were to detect αIIbβ3 and αVβ3

integrin expression in human tumour cell lines and human xenograft tissue to

develop models that can be used for both in vitro and in vivo evaluation of

novel β3 integrin antagonists.

After characterisation of αIIbβ3/αVβ3 integrin expression in a panel of human

tumour cell lines, M14 was selected as a suitable cell line to be used as a

model in the scratch assay. This cell line allows the study of migration

mediated by αVβ3 integrin due to negligible expression of αIIbβ3 integrin in the

cell line. Using this cell line the potential novel β3 integrin antagonists

ICT9055 and ICT9073 were identified as providing blockade of cell migration

at non-toxic concentrations, with IC50 < 0.1 and < 0.01 µM respectively.

However, the other antagonists that had cytotoxic effect need to be

confirmed that it non toxic to control cells in other functional assay.

Initially, the expression of αIIbβ3 and αVβ3 was investigated in human

xenograft mouse tissue using immunohistochemistry in order to obtain

information that would guide the selection of cell lines to be used when the

potential novel β3 integrin antagonists progress to in vivo screening.

Detection of integrin expression in xenograft mouse tissue was really


197
important in preparing an in vivo model since it is known that integrin

expression varies in response to extracellular environment (Zhao et al. 2007).

After different methodologies were followed, all the primary antibodies

evaluated in these studies gave non-specific immunolabelling in sections

both with and without primary antibodies, except B7 (anti-β3) using the

M.O.M. kit which gave a clean negative control with non-specific

immunolabelling of blood vessels only. Furthermore, non-membranous

immunolabelling developed with Q20 (anti-αV). These results were confirmed

by immunoblotting which showed that mouse antibody (B7) on mouse tissue

developed non-specific binding while rabbit antibody Q20 did not develop any

non-specific binding.

According to tissue homogenisation and protein extraction in this study, it can

be said that the human xenograft tissues used in this study expressed αV and

β3 integrin subunits. The problem faced with immunohistochemistry was due

to the inability of the mouse primary antibodies used to selectively detect the

expression of human integrin subunits in the presence of mouse tissue. In

normal mouse tissue there was non-specific binding whereas there was no

non-specific binding in samples from human tumour cell lines grown in vitro

when the membrane was incubated without primary antibody. In order to

investigate integrin expression in an in vivo model using either

immunohistochemistry or immunoblotting it seems to be vital to avoid using

mouse antibodies on mouse tissue. For example anti-αV (Q20) did not

198
develop any non-specific binding with immunohistochemistry or with

immunoblotting.

The results from immunoblotting encouraged evaluation of the expression of

αVβ3 and αIIbβ3 integrin in other human tumour cell lines. After optimisation of

a panel of primary antibodies, the expression of αV, β3 and αVβ3 integrin was

detected on the surface of a panel of tumour cell lines using

immunocytochemistry, and also the expression of αV and β3 integrin subunits

was detected in different cell lines using immunoblotting. However, the

overall aim of this study can be best investigated in vitro using a model

expressing both αIIbβ3 and αVβ3 integrin and I did not detect expression of αIIb

integrin in this study. Thus, to achieve a model of α IIbβ3/αVβ3 integrin dual

expression, it will be vital to transfect the cells chosen for the study with the

αIIb integrin subunit (O'Toole et al. 1989). The transfection of αIIbβ3 integrin

has been achieved previously; for example, αIIbβ3 integrin was transfected

into the melanoma cell line WM 983B which lead to increased tumour cell

adhesion, spreading and invasion in vitro and decreased apoptotic rate in

vivo (Trikha et al. 2002).

Here, αV protein was expressed in all tumour cell lines evaluated, and was

observed to be localised to the cell surface using immunocytochemistry. This

result was expected since αV can bind to other β subunits besides β3.

Goodman et al. have also shown that αV is expressed in all human tumour

cell lines (Goodman, Grote & Wilm 2012). αV has been shown to play an

important role in prostate cancer angiogenesis and bone metastasis through

199
validation of αV as a target by knockdown, and also pharmacological

inhibition by GLPG0187 (Table 1.3), using in vivo and in vitro models.

GLPG0187 was effective in inhibiting angiogenesis and bone metastasis

through its action on prostate cancer stem cells (van der Host et al. 2011). In

addition, αV blocking with GLPG0187 in PC-3 cells prevented the growth of

the tumour in vivo (van der Host et al. 2010). The anti-αV antibody CNTO95

(Table 1.3) inhibits HUVECs and melanoma cell (A375.S2) adhesion,

migration, and invasion in vitro. CNTO95 also leads to inhibition of tumour

proliferation and angiogenesis in vivo (Trikha et al. 2004). Bisanz et al.

treated PC-3 cells with αV si-RNA which lead to decreased growth of PC-3

xenografts in bone, suggesting that αV mediated communication between

tumour cell and bone microenvironment is required for bone metastasis

development (Bisanz et al. 2005). The above indicate that αV has an

important role in cancer. In the present study, it was not possible to use a

model with no αVβ3 integrin because the only line explored that had no αVβ3

integrin expression, HT-29, could not be used in the scratch assay due to its

low rate of migration/wound closure.

The present study developed a semi-quantitative method for the detection of

αVβ3 integrin expression in human tumour cell lines and xenografts. A more

quantitative measure would be to use flow cytometry, it was the advantage

over the immunocytochemistry is being able to detect even weak expression

(Goodman, Grote & Wilm 2012). LM609 has been used previously to

characterise the expression of αVβ3 integrin in human tumour cell lines using

flow cytometry (Landen et al. 2008). However, since flow cytometry does not
200
assess morphological expression, it must still be run in parallel with

immunohistochemistry or immunocytochemistry.

The overall aims of this study were to characterise αVβ3/αIIbβ3 integrin

expression in tumour cell lines and tissues, and to evaluate the

pharmacological effect of novel small molecule, anti αVβ3 and/or αIIbβ3

antagonists. Using different tumour cell lines with different α Vβ3 integrin

expression, a range of wound-healing responses was seen in the scratch

assay for migration. LM609 and cRGDfV were used as positive controls to

demonstrate that M14 migration was mediated by αVβ3 integrin. Albelda et al.

showed increased expression of αVβ3 integrin correlates with increased

melanoma cell migration (Albelda et al. 1990). Although both inhibitors were

effective in inhibition of M14 cell migration, cRGDfV (target αVβ3/αVβ5 integrin)

and LM609 (target αVβ3 integrin) this indicated that αVβ5 integrin may also had

a role in M14 cell migration.

After optimising the scratch assay conditions, the potential novel β3 integrin

antagonists were screened. The effect of potential novel β3 integrin

antagonists in inhibition of M14 cell migration was evaluated. From screening

a panel of 12 novel β3 integrin antagonists, ICT9055 had the highest effect in

inhibition of M14 cell migration. This small molecule did not exert any

cytotoxic effect even at high concentrations on M14 cells but was toxic to HT-

29. ICT9053 was toxic to M14 cells only, ICT9020 was toxic to M14 and HT-

29 but not to MCF-7 and ICT9068 was toxic to MCF-7 not to M14 and HT-29.

Different human tumour cell lines may have different survival pathways and

201
this can be explained by the variation in cytotoxic effect of some potential

novel β3 integrin antagonists on some tumour cells and not others. For

example, while cRGDfV was non-toxic to LNCaP prostate carcinoma cells

(Chatterjee et al. 2001), and U-87MG and U-373MG glioma cells (Chatterjee

et al. 2000), Cilengitide was toxic to glioma cells U-251MG and U-87MG

plated onto vitronectin coated dishes. It has been shown Cilengitide can

induce autophagy leading to cell death (Lomonaco et al. 2011) whilst

cRGDfV can induce apoptosis through blocking FAK which mediates the

signal transduction pathway necessary for cell survival (Chatterjee et al.

2001).

ICT9055 was toxic to HT-29 and this cytotoxicity may be due to expression of

β6 integrin (in house data, Andrew Gordon) which has been shown to have a

role in colon cancer cell survival through ERK signalling inducing tumour cell

proliferation (Peng et al. 2009). 10D5 (IgG2a, monoclonal antibody target

αVβ6) induced apoptosis of HT-29 cells through increase in caspase-3 and

caspase-9 expression and release of cytochrome c from the mitochondria to

the cytoplasm (Zhao-Yang et al. 2008). The above result suggests that the

effects of the novel β3 antagonists tested in the present study on HT-29

maybe were via antagonism of αVβ6 integrin, and further work needs to be

done to prove this by using anti αVβ6 integrin inhibitor.

As shown in the scratch assay done here, not all cell lines were adherent or

had rapid cell migration toward wound area, therefore another assay should

be evaluated that may work with a broader range of cell type. Another assay

202
that could be used to investigate integrin antagonism is the Boyden chamber

transwell migration assay. The transwell migration assay has been used to

test the effect of compounds on the inhibition of cell migration in vitro, by

using a transwell migration assay chamber coated with ligands such as

laminin, vitronectin or fibronectin (Trusolino et al. 2000). Using the transwell

migration assay coated with substrates (Byzova et al. 2000) can give an idea

about the effect of potential novel β3 integrin antagonists in inhibition of

tumour cell migration in vivo. Further, the use of the transwell assay will give

a chance to detect the effect of integrin antagonists on adherent and non

adherent cells. The Boyden chamber assay permits migration of cells toward

the ECM protein. Cell contact allows cells to exchange information that can

be used to control the cell migration process (Carreiras et al. 1999).

Here, the wound healing assay was done using uncoated plates. Coating

plates with substrate would provide a more physiologically relevant model to

help to identify integrin antagonists. The choice of extracellular matrix ligand

can be used to determine the selectivity of the antagonists or the relative

contribution of related integrin to cell migration. PC-3 cells which have no

expression of αVβ3 integrin can adhere and migrate on vitronectin mediated

by αVβ1 and αVβ5 integrin (Bisanz et al. 2005). PC-3 cell expressing αVβ5 and

αVβ1 and α5β1 integrin have been shown to migrate on fibronectin and

vitronectin, since these cells had little expression of αVβ3 a selective αVβ3

antagonist did not have an effect on PC-3 plated on vitronectin (Lin et al.

2010). Thus to fully understand our scratch assay results, the expression of

other integrins must be detected; for example, other studies have reported
203
expression of α5β1 integrin in melanoma cell lines (Leavesley et al. 1992;

Seftor et al. 1992). Eventually, targeting multiple integrins in tumour cells and

the microenvironment may be investigated as a therapeutic strategy (Mark

Sutherland 2012).

It will be vital to test the effects of novel integrin antagonists on the ability of

tumour cell lines to adhere to ECM protein such as vitronectin or fibronectin.

The adhesion assay can be used to identify novel integrin antagonists and to

characterise their selectivity, using antibody controls and cell lines with fully

characterised integrin expression (Caltabiano et al. 1999; Del Bufalo et al.

1998). Gentilucci, et al. used an adhesion assay to test the selectivity of

cyclotetrapeptides as αVβ3 and/or α5β1 antagonists. Selectivity was

determined by studying the effects of the peptides on the inhibition of

adhesion of K562 (human erythroleukemic cells) expressing α5β1 integrin, or

SK-MEL-24 (human melanoma cells) expressing αVβ3 integrin to fibronectin,

and HUVECs expressing αVβ3 integrin to vitronectin (Gentilucci et al. 2010).

Wang et al. investigated the role of β3 integrins in transendothelial migration

in vitro by evaluating the migratory capacity of PC-3 cells through a

monolayer of HUVECs. The membranous expression of β3 integrin in PC-3

was associated with active motility from its location at the edges of

pseudopodial extensions, and the study concluded that PC-3 played an

important role in TEM through its interaction with the matrix underneath the

endothelium. Blocking β3 integrin led to inhibition of the interaction of PC-3

cell with the ECM (Wang et al. 2005). Voura et al. found that cRGDfV (5 µM)

204
was able to inhibit transmigration of melanoma cell WM 239 through

HUVECs by 70% and LM609 (40 µg/ml) led to 40-50% inhibition indicating

that transmigration was partly mediated by αVβ5. These assays will be useful

in identifying compounds that would be effective against early stages of

metastasis (Voura et al. 2001).

The effect of potential novel β3 integrin antagonists on downstream cell

signalling needs to be tested since as has been described earlier, integrins

function through inside-out and out-sidein signalling. The

RAS/RAF/MEK/ERK signalling pathway is commonly activated in melanoma

(Meier et al. 2005). Ligation of αVβ3 integrin with ECM has been shown to

lead to focal contact formation and autophosphorylation of FAK at Y397

which provides a recognition site for c-src SH2 domain, and recruitment of

Src that lead to phosphorylation of FAK at Y925 and provides an additional

site to SH2 (adaptor protein) of Grb-2 that recruits SOS to activate Ras then

Raf then MEK then ERK (Chen et al. 2007). For example, cRGDfV blocking

FAK activation led to cleavage of FAK and reduced expression of Akt/PKB

further to activation of caspase 3 and caspase 9 which induced programmed

cell death in LNCaP, prostate carcinoma cells (Chatterjee et al. 2001).

Cilengitide has been used with HUVECs and glioma cells where it induced

cell detachment and apoptosis in cells grown on uncoated plates. The effect

of Cilengitide was associated with reduction in FAK and Src phosphorylation

in both endothelial and glioma cells. Akt phosphorylation was reduced in

glioma cells, but there was no effect on ERK in HUVECs (Oliveira-Ferrer et

al. 2008).
205
After successfully identifying potential novel β3 integrin antagonists that

inhibit tumour cell adhesion, migration and invasion using the functional

assays in vitro, the effect of novel antagonists can be evaluated in term of

inhibition of tumour cell angiogenesis, migration and invasion in vivo.

The results presented in this thesis indicated a very good start for the

development of potential novel β3 integrin antagonists. However, much work

still needs to be done to evaluate the selectivity of novel integrin antagonists.

This will be a big challenge since the cell line model prepared to use with the

scratch assay may not work with the other assays such as adhesion assay

and invasion assays. Therefore, other models will need to be investigated.

6.2 Conclusion

In conclusion, tumour dissemination is a major issue with the treatment of

cancer, and there is a pressing need for therapies which can control this

process. As has been discussed here, the integrins represent a good

potential target for therapeutic intervention to control tumour dissemination.

Using characterised cell lines in a validated functional assay for migration,

promising potential novel β3 integrin antagonists have been identified. Further

work is required to confirm their activity as anti-cancer agents.

206
7 Chapter 7: References and Appendixes

207
7.1 References

Abdollahi, A., D. W. Griggs, H. Zieher, A. Roth, K. E. Lipson, R. Saffrich, H. J.

Grone, D. E. Hallahan, R. A. Reisfeld, J. Debus, A. G. Niethammer & P. E.

Huber 2005. Inhibition of alpha(v)beta3 integrin survival signaling enhances

antiangiogenic and antitumor effects of radiotherapy. Clin Cancer Res 11:

6270-9.

Agrez, M. V., R. C. Bates, D. Mitchell, N. Wilson, N. Ferguson, P. Anseline &

D. Sheppard 1996. Multiplicity of fibronectin-binding alpha V integrin

receptors in colorectal cancer. Br J Cancer 73: 887-92.

Albelda, S. M., S. A. Mette, D. E. Elder, R. Stewart, L. Damjanovich, M.

Herlyn & C. A. Buck 1990. Integrin distribution in malignant melanoma:

association of the beta 3 subunit with tumor progression. Cancer Res 50:

6757-64.

Alfieri, S., A. G. Morganti, A. Di Giorgio, V. Valentini, M. Bossola, L. Trodella,

N. Cellini & G. B. Doglietto 2001. Improved survival and local control after

intraoperative radiation therapy and postoperative radiotherapy: a

multivariate analysis of 46 patients undergoing surgery for pancreatic head

cancer. Arch Surg 136: 343-7.

208
Alghisi, G. C., L. Ponsonnet & C. Ruegg 2009. The integrin antagonist

cilengitide activates alphaVbeta3, disrupts VE-cadherin localization at cell

junctions and enhances permeability in endothelial cells. PLoS ONE 4:

e4449.

Alig, L., A. Edenhofer, P. Hadvary, M. Hurzeler, D. Knopp, M. Muller, B.

Steiner, A. Trzeciak & T. Weller 1992. Low molecular weight, non-peptide

fibrinogen receptor antagonists. J Med Chem 35: 4393-407.

Allman, R., P. Cowburn & M. Mason 2000. In vitro and in vivo effects of a

cyclic peptide with affinity for the alpha(nu)beta3 integrin in human melanoma

cells. Eur J Cancer 36: 410-22.

Amirkhosravi, A., S. A. Mousa, M. Amaya, S. Blaydes, H. Desai, T. Meyer &

J. L. Francis 2003. Inhibition of tumor cell-induced platelet aggregation and

lung metastasis by the oral GpIIb/IIIa antagonist XV454. Thromb Haemost

90: 549-54.

Armstrong, A. J., G. J. Netto, M. A. Rudek, S. Halabi, D. P. Wood, P. A.

Creel, K. Mundy, S. L. Davis, T. Wang, R. Albadine, L. Schultz, A. W. Partin,

A. Jimeno, H. Fedor, P. G. Febbo, D. J. George, R. Gurganus, A. M. De

Marzo & M. A. Carducci 2010. A pharmacodynamic study of rapamycin in

men with intermediate- to high-risk localized prostate cancer. Clin Cancer

Res 16: 3057-66.

209
Arosio, D., L. Manzoni, E. M. Araldi, A. Caprini, E. Monferini & C. Scolastico

2009. Functionalized cyclic RGD peptidomimetics: conjugable ligands for

alphavbeta3 receptor imaging. Bioconjug Chem 20: 1611-7.

Askari, J. A., P. A. Buckley, A. P. Mould & M. J. Humphries 2009. Linking

integrin conformation to function. J Cell Sci 122: 165-70.

Bachelot, T., G. Romieu, M. Campone, V. Dieras, C. Cropet, F. Dalenc, M.

Jimenez, E. Le Rhun, J. Y. Pierga, A. Goncalves, M. Leheurteur, J. Domont,

M. Gutierrez, H. Cure, J. M. Ferrero & C. Labbe-Devilliers 2013. Lapatinib

plus capecitabine in patients with previously untreated brain metastases from

HER2-positive metastatic breast cancer (LANDSCAPE): a single-group

phase 2 study. Lancet Oncol 14: 64-71.

Bachmann, I. M., R. G. Ladstein, O. Straume, G. N. Naumov & L. A. Akslen

2008. Tumor necrosis is associated with increased alphavbeta3 integrin

expression and poor prognosis in nodular cutaneous melanomas. BMC

Cancer 8: 362.

Bader, B. L., H. Rayburn, D. Crowley & R. O. Hynes 1998. Extensive

vasculogenesis, angiogenesis, and organogenesis precede lethality in mice

lacking all alpha v integrins. Cell 95: 507-19.

210
Bakewell, S. J., P. Nestor, S. Prasad, M. H. Tomasson, N. Dowland, M.

Mehrotra, R. Scarborough, J. Kanter, K. Abe, D. Phillips & K. N. Weilbaecher

2003. Platelet and osteoclast beta3 integrins are critical for bone metastasis.

Proc Natl Acad Sci U S A 100: 14205-10.

Barthel, S. R., D. Hays, E. M. Yazawa, M. J. Opperman, K. C. Walley, L.

Nimrichter, M. M. Burdick, B. M. Gillard, M. T. Moser, K. Pantel, B. A. Foster,

K. J. Pienta & C. J. Dimitroff 2012. Definition of molecular determinants of

prostate cancer cell bone extravasation. Cancer Res

Barthel, S. R., D. L. Hays, E. M. Yazawa, M. Opperman, K. C. Walley, L.

Nimrichter, M. M. Burdick, B. M. Gillard, M. T. Moser, K. Pantel, B. A. Foster,

K. J. Pienta & C. J. Dimitroff 2013. Definition of molecular determinants of

prostate cancer cell bone extravasation. Cancer Res 73: 942-52.

Beauvais, D. M., B. J. Burbach & A. C. Rapraeger 2004. The syndecan-1

ectodomain regulates alphavbeta3 integrin activity in human mammary

carcinoma cells. J Cell Biol 167: 171-81.

Bell-McGuinn, K. M., C. M. Matthews, S. N. Ho, M. Barve, L. Gilbert, R. T.

Penson, E. Lengyel, R. Palaparthy, K. Gilder, A. Vassos, W. McAuliffe, S.

Weymer, J. Barton & R. J. Schilder 2011. A phase II, single-arm study of the

anti-alpha5beta1 integrin antibody volociximab as monotherapy in patients

211
with platinum-resistant advanced epithelial ovarian or primary peritoneal

cancer. Gynecol Oncol 121: 273-9.

Bellahcene, A., V. Castronovo, K. U. Ogbureke, L. W. Fisher & N. S. Fedarko

2008. Small integrin-binding ligand N-linked glycoproteins (SIBLINGs):

multifunctional proteins in cancer. Nat Rev Cancer 8: 212-26.

Bello, L., M. Francolini, P. Marthyn, J. Zhang, R. S. Carroll, D. C. Nikas, J. F.

Strasser, R. Villani, D. A. Cheresh & P. M. Black 2001. Alpha(v)beta3 and

alpha(v)beta5 integrin expression in glioma periphery. Neurosurgery 49: 380-

9; discussion 390.

Bello, L., V. Lucini, C. Giussani, G. Carrabba, M. Pluderi, F. Scaglione, G.

Tomei, R. Villani, P. M. Black, A. Bikfalvi & R. S. Carroll 2003. IS20I, a

specific alphavbeta3 integrin inhibitor, reduces glioma growth in vivo.

Neurosurgery 52: 177-85; discussion 185-6.

Belvisi, L., T. Riccioni, M. Marcellini, L. Vesci, I. Chiarucci, D. Efrati, D.

Potenza, C. Scolastico, L. Manzoni, K. Lombardo, M. A. Stasi, A. Orlandi, A.

Ciucci, B. Nico, D. Ribatti, G. Giannini, M. Presta, P. Carminati & C. Pisano

2005. Biological and molecular properties of a new

alpha(v)beta3/alpha(v)beta5 integrin antagonist. Mol Cancer Ther 4: 1670-

80.

212
Bengtsson, T., A. Aszodi, C. Nicolae, E. B. Hunziker, E. Lundgren-Akerlund

& R. Fassler 2005. Loss of alpha10beta1 integrin expression leads to

moderate dysfunction of growth plate chondrocytes. J Cell Sci 118: 929-36.

Bennett, J. S. 2005. Structure and function of the platelet integrin

alphaIIbbeta3. J Clin Invest 115: 3363-9.

Bennouna, J., J. Sastre, D. Arnold, P. Osterlund, R. Greil, E. Van Cutsem, R.

von Moos, J. M. Vieitez, O. Bouche, C. Borg, C. C. Steffens, V. Alonso-

Orduna, C. Schlichting, I. Reyes-Rivera, B. Bendahmane, T. Andre, S.

Kubicka & M. L. S. Investigators 2013. Continuation of bevacizumab after

first progression in metastatic colorectal cancer (ML18147): a randomised

phase 3 trial. Lancet Oncol 14: 29-37.

Bhavaraju, K., P. R. Lakhani, R. T. Dorsam, J. Jin, I. S. Hitchcock, A. Sanjay

& S. P. Kunapuli 2011. G(12/13) signaling pathways substitute for integrin

alphaIIbbeta3-signaling for thromboxane generation in platelets. PLoS ONE

6: e16586.

Bibby, M. C. 2004. Orthotopic models of cancer for preclinical drug

evaluation: advantages and disadvantages. Eur J Cancer 40: 852-7.

Bisanz, K., J. Yu, M. Edlund, B. Spohn, M. C. Hung, L. W. Chung & C. L.

Hsieh 2005. Targeting ECM-integrin interaction with liposome-encapsulated

213
small interfering RNAs inhibits the growth of human prostate cancer in a

bone xenograft imaging model. Mol Ther 12: 634-43.

Blagoev, K. B., J. Wilkerson, W. D. Stein, R. J. Motzer, S. E. Bates & A. T.

Fojo 2013. Sunitinib does not accelerate tumor growth in patients with

metastatic renal cell carcinoma. Cell Rep 3: 277-81.

Bodary, S. C. & J. W. McLean 1990. The integrin beta 1 subunit associates

with the vitronectin receptor alpha v subunit to form a novel vitronectin

receptor in a human embryonic kidney cell line. J Biol Chem 265: 5938-41.

Boger, C., H. Kalthoff, S. L. Goodman & C. Rocken 2013. Validation and

Comparison of Anti-alphavbeta3 and Anti-alphavbeta5 Rabbit Monoclonal

Versus Murine Monoclonal Antibodies in Four Different Tumor Entities. Appl

Immunohistochem Mol Morphol

Bossy, B., E. Bossy-Wetzel & L. F. Reichardt 1991. Characterization of the

integrin alpha 8 subunit: a new integrin beta 1-associated subunit, which is

prominently expressed on axons and on cells in contact with basal laminae in

chick embryos. EMBO J 10: 2375-85.

Botchkarev, V. A. 2003. Molecular mechanisms of chemotherapy-induced

hair loss. J Investig Dermatol Symp Proc 8: 72-5.

214
Bozzuto, G., P. Ruggieri & A. Molinari 2010. Molecular aspects of tumor cell

migration and invasion. Ann Ist Super Sanita 46: 66-80.

Bradford, M. M. 1976. A rapid and sensitive method for the quantitation of

microgram quantities of protein utilizing the principle of protein-dye binding.

Anal Biochem 72: 248-54.

Brakebusch, C., D. Bouvard, F. Stanchi, T. Sakai & R. Fassler 2002.

Integrins in invasive growth. J Clin Invest 109: 999-1006.

Breuss, J. M., J. Gallo, H. M. DeLisser, I. V. Klimanskaya, H. G. Folkesson,

J. F. Pittet, S. L. Nishimura, K. Aldape, D. V. Landers, W. Carpenter & et al.

1995. Expression of the beta 6 integrin subunit in development, neoplasia

and tissue repair suggests a role in epithelial remodeling. J Cell Sci 108 ( Pt

6): 2241-51.

Brockbank, E. C., J. Bridges, C. J. Marshall & E. Sahai 2005. Integrin beta1

is required for the invasive behaviour but not proliferation of squamous cell

carcinoma cells in vivo. Br J Cancer 92: 102-12.

Brooks, P. C., S. Stromblad, R. Klemke, D. Visscher, F. H. Sarkar & D. A.

Cheresh 1995. Antiintegrin alpha v beta 3 blocks human breast cancer

growth and angiogenesis in human skin. J Clin Invest 96: 1815-22.

215
Brooks, P. C., S. Stromblad, L. C. Sanders, T. L. von Schalscha, R. T.

Aimes, W. G. Stetler-Stevenson, J. P. Quigley & D. A. Cheresh 1996.

Localization of matrix metalloproteinase MMP-2 to the surface of invasive

cells by interaction with integrin alpha v beta 3. Cell 85: 683-93.

Brooks, P. C., S. Strömblad, L. C. Sanders, T. L. von Schalscha, R. T.

Aimes, W. G. Stetler-Stevenson, J. P. Quigley & D. A. Cheresh 1996.

Localization of Matrix Metalloproteinase MMP-2 to the Surface of Invasive

Cells by Interaction with Integrin αvβ3. Cell 85: 683-693.

Brown, B. D. & D. Lillicrap 2002. Dangerous liaisons: the role of "danger"

signals in the immune response to gene therapy. Blood 100: 1133-40.

Brown, M. A., C. S. Wallace, C. C. Anamelechi, E. Clermont, W. M. Reichert

& G. A. Truskey 2007. The use of mild trypsinization conditions in the

detachment of endothelial cells to promote subsequent endothelialization on

synthetic surfaces. Biomaterials 28: 3928-35.

Buensuceso, C. S., A. Obergfell, A. Soriani, K. Eto, W. B. Kiosses, E. G.

Arias-Salgado, T. Kawakami & S. J. Shattil 2005. Regulation of outside-in

signaling in platelets by integrin-associated protein kinase C beta. J Biol

Chem 280: 644-53.

216
Burbridge, M. F., V. Venot, P. J. Casara, F. Perron-Sierra, J. A. Hickman &

G. C. Tucker 2003. Decrease in survival threshold of quiescent colon

carcinoma cells in the presence of a small molecule integrin antagonist. Mol

Pharmacol 63: 1281-8.

Burke, P. A., S. J. DeNardo, L. A. Miers, K. R. Lamborn, S. Matzku & G. L.

DeNardo 2002. Cilengitide targeting of alpha(v)beta(3) integrin receptor

synergizes with radioimmunotherapy to increase efficacy and apoptosis in

breast cancer xenografts. Cancer Res 62: 4263-72.

Byzova, T. V., W. Kim, R. J. Midura & E. F. Plow 2000. Activation of integrin

alpha(V)beta(3) regulates cell adhesion and migration to bone sialoprotein.

Exp Cell Res 254: 299-308.

Caenepeel, S., L. Renshaw-Gegg, A. Baher, T. L. Bush, W. Baron, T. Juan,

R. Manoukian, A. S. Tasker, A. Polverino & P. E. Hughes 2010. Motesanib

inhibits Kit mutations associated with gastrointestinal stromal tumors. J Exp

Clin Cancer Res 29: 96.

Cai, W., Y. Wu, K. Chen, Q. Cao, D. A. Tice & X. Chen 2006. In vitro and in

vivo characterization of 64Cu-labeled Abegrin, a humanized monoclonal

antibody against integrin alpha v beta 3. Cancer Res 66: 9673-81.

217
Caltabiano, S., W. T. Hum, G. J. Attwell, D. N. Gralnick, L. J. Budman, A. M.

Cannistraci & F. J. Bex 1999. The integrin specificity of human recombinant

osteopontin. Biochem Pharmacol 58: 1567-78.

Camporeale, J. 2008. Basics of radiation treatment. Clin J Oncol Nurs 12:

193-5.

Carracedo, S., N. Lu, S. N. Popova, R. Jonsson, B. Eckes & D. Gullberg The

fibroblast integrin {alpha}11{beta}1 is induced in a mechanosensitive manner

involving activin A and regulates myofibroblast differentiation. J Biol Chem

Carreiras, F., V. Rigot, S. Cruet, F. Andre, P. Gauduchon & J. Marvaldi 1999.

Migration properties of the human ovarian adenocarcinoma cell line IGROV1:

importance of alpha(v)beta3 integrins and vitronectin. Int J Cancer 80: 285-

94.

Cary, L. A., J. F. Chang & J. L. Guan 1996. Stimulation of cell migration by

overexpression of focal adhesion kinase and its association with Src and

Fyn. J Cell Sci 109 ( Pt 7): 1787-94.

Castel, S., R. Pagan, F. Mitjans, J. Piulats, S. Goodman, A. Jonczyk, F.

Huber, S. Vilaro & M. Reina 2001. RGD peptides and monoclonal antibodies,

antagonists of alpha(v)-integrin, enter the cells by independent endocytic

pathways. Lab Invest 81: 1615-26.

218
Cattoretti, G., S. Pileri, C. Parravicini, M. H. Becker, S. Poggi, C. Bifulco, G.

Key, L. D'Amato, E. Sabattini, E. Feudale & et al. 1993. Antigen unmasking

on formalin-fixed, paraffin-embedded tissue sections. J Pathol 171: 83-98.

Cepek, K. L., S. K. Shaw, C. M. Parker, G. J. Russell, J. S. Morrow, D. L.

Rimm & M. B. Brenner 1994. Adhesion between epithelial cells and T

lymphocytes mediated by E-cadherin and the alpha E beta 7 integrin. Nature

372: 190-3.

Chames, P., M. Van Regenmortel, E. Weiss & D. Baty 2009. Therapeutic

antibodies: successes, limitations and hopes for the future. Br J Pharmacol

157: 220-33.

Chatterjee, S., K. H. Brite & A. Matsumura 2001. Induction of apoptosis of

integrin-expressing human prostate cancer cells by cyclic Arg-Gly-Asp

peptides. Clin Cancer Res 7: 3006-11.

Chatterjee, S., A. Matsumura, J. Schradermeier & G. Y. Gillespie 2000.

Human malignant glioma therapy using anti-alpha(v)beta3 integrin agents. J

Neurooncol 46: 135-44.

Chen, P. S., C. C. Wang, C. D. Bortner, G. S. Peng, X. Wu, H. Pang, R. B.

Lu, P. W. Gean, D. M. Chuang & J. S. Hong 2007. Valproic acid and other

histone deacetylase inhibitors induce microglial apoptosis and attenuate

219
lipopolysaccharide-induced dopaminergic neurotoxicity. Neuroscience 149:

203-212.

Chen, P. S., M. Y. Wang, S. N. Wu, J. L. Su, C. C. Hong, S. E. Chuang, M.

W. Chen, K. T. Hua, Y. L. Wu, S. T. Cha, M. S. Babu, C. N. Chen, P. H. Lee,

K. J. Chang & M. L. Kuo 2007. CTGF enhances the motility of breast cancer

cells via an integrin-alphavbeta3-ERK1/2-dependent S100A4-upregulated

pathway. J Cell Sci 120: 2053-65.

Chen, W., J. Lou, J. Hsin, K. Schulten, S. C. Harvey & C. Zhu 2011.

Molecular Dynamics Simulations of Forced Unbending of Integrin

alpha(V)beta(3). PLoS Comput Biol 7: e1001086.

Chen, Y. Q., M. Trikha, X. Gao, R. Bazaz, A. T. Porter, J. Timar & K. V. Honn

1997. Ectopic expression of platelet integrin alphaIIb beta3 in tumor cells

from various species and histological origin. Int J Cancer 72: 642-8.

Cheng, Z., Y. Wu, Z. Xiong, S. S. Gambhir & X. Chen 2005. Near-infrared

fluorescent RGD peptides for optical imaging of integrin alphavbeta3

expression in living mice. Bioconjug Chem 16: 1433-41.

Chernousov, M. A. & D. J. Carey 2003. alphaVbeta8 integrin is a Schwann

cell receptor for fibrin. Exp Cell Res 291: 514-24.

220
Collins, I. & P. Workman 2006. New approaches to molecular cancer

therapeutics. Nat Chem Biol 2: 689-700.

Cooper, C. R., C. H. Chay & K. J. Pienta 2002. The role of alpha(v)beta(3) in

prostate cancer progression. Neoplasia 4: 191-4.

Coopman, P. J., D. M. Thomas, K. R. Gehlsen & S. C. Mueller 1996. Integrin

alpha 3 beta 1 participates in the phagocytosis of extracellular matrix

molecules by human breast cancer cells. Mol Biol Cell 7: 1789-804.

Cox, D., M. Brennan & N. Moran 2010. Integrins as therapeutic targets:

lessons and opportunities. Nat Rev Drug Discov 9: 804-20.

Criscitiello, C., D. Fumagalli, K. S. Saini & S. Loi 2011. Tamoxifen in early-

stage estrogen receptor-positive breast cancer: overview of clinical use and

molecular biomarkers for patient selection. Onco Targets Ther 4: 1-11.

Danhier, F., A. Le Breton & V. Preat 2012. RGD-based strategies to target

alpha(v) beta(3) integrin in cancer therapy and diagnosis. Mol Pharm 9:

2961-73.

David, J. M. & A. K. Rajasekaran 2012. Dishonorable discharge: the

oncogenic roles of cleaved E-cadherin fragments. Cancer Res 72: 2917-23.

221
Dayam, R., F. Aiello, J. Deng, Y. Wu, A. Garofalo, X. Chen & N. Neamati

2006. Discovery of small molecule integrin alphavbeta3 antagonists as novel

anticancer agents. J Med Chem 49: 4526-34.

Dedhar, S. & G. E. Hannigan 1996. Integrin cytoplasmic interactions and

bidirectional transmembrane signalling. Curr Opin Cell Biol 8: 657-69.

Del Bufalo, D., C. Leonetti, B. Bucci, C. Amedeo, R. Falcioni, A. Biroccio & G.

Zupi 1998. N-methylformamide induces changes on adhesive properties and

lung-colonizing potential of M14 melanoma cells. Br J Cancer 77: 210-5.

Derer, W., E. S. Barnathan, E. Safak, P. Agarwal, H. Heidecke, M. Mockel,

M. Gross, C. Oezcelik, R. Dietz & R. Dechend 2009. Vitronectin

concentrations predict risk in patients undergoing coronary stenting. Circ

Cardiovasc Interv 2: 14-9.

Desgrosellier, J. S. & D. A. Cheresh 2010. Integrins in cancer: biological

implications and therapeutic opportunities. Nat Rev Cancer 10: 9-22.

Desloges, N., N. Basora, N. Perreault, Y. Bouatrouss, D. Sheppard & J. F.

Beaulieu 1998. Regulated expression of the integrin alpha9beta1 in the

epithelium of the developing human gut and in intestinal cell lines: relation

with cell proliferation. J Cell Biochem 71: 536-45.

222
Dome, B., E. Raso, J. Dobos, L. Meszaros, N. Varga, L. G. Puskas, L. Z.

Feher, T. Lorincz, A. Ladanyi, M. Trikha, K. V. Honn & J. Timar 2005. Parallel

expression of alphaIIbbeta3 and alphavbeta3 integrins in human melanoma

cells upregulates bFGF expression and promotes their angiogenic

phenotype. Int J Cancer 116: 27-35.

Donate, F., G. C. Parry, Y. Shaked, H. Hensley, X. Guan, I. Beck, Z. Tel-

Tsur, M. L. Plunkett, M. Manuia, D. E. Shaw, R. S. Kerbel & A. P. Mazar

2008. Pharmacology of the novel antiangiogenic peptide ATN-161 (Ac-

PHSCN-NH2): observation of a U-shaped dose-response curve in several

preclinical models of angiogenesis and tumor growth. Clin Cancer Res 14:

2137-44.

Duong, L. T., P. Lakkakorpi, I. Nakamura & G. A. Rodan 2000. Integrins and

signaling in osteoclast function. Matrix Biol 19: 97-105.

Eble, J. A. & J. Haier 2006. Integrins in cancer treatment. Curr Cancer Drug

Targets 6: 89-105.

Edelstein, M. L., M. R. Abedi & J. Wixon 2007. Gene therapy clinical trials

worldwide to 2007--an update. J Gene Med 9: 833-42.

223
Engebraaten, O., M. Trikha, S. Juell, S. Garman-Vik & O. Fodstad 2009.

Inhibition of in vivo tumour growth by the blocking of host alpha(v)beta3 and

alphaII(b)beta3 integrins. Anticancer Res 29: 131-7.

Erikson, D. W., R. C. Burghardt, K. J. Bayless & G. A. Johnson 2009.

Secreted phosphoprotein 1 (SPP1, osteopontin) binds to integrin alpha v

beta 6 on porcine trophectoderm cells and integrin alpha v beta 3 on uterine

luminal epithelial cells, and promotes trophectoderm cell adhesion and

migration. Biol Reprod 81: 814-25.

Erle, D. J., M. J. Briskin, E. C. Butcher, A. Garcia-Pardo, A. I. Lazarovits & M.

Tidswell 1994. Expression and function of the MAdCAM-1 receptor, integrin

alpha 4 beta 7, on human leukocytes. The Journal of Immunology 153: 517-

28.

Fahraeus, R. & D. P. Lane 1999. The p16(INK4a) tumour suppressor protein

inhibits alphavbeta3 integrin-mediated cell spreading on vitronectin by

blocking PKC-dependent localization of alphavbeta3 to focal contacts. EMBO

J 18: 2106-18.

Farolfi, A., E. Melegari, M. Aquilina, E. Scarpi, T. Ibrahim, R. Maltoni, S. Sarti,

L. Cecconetto, E. Pietri, C. Ferrario, A. Fedeli, M. Faedi, O. Nanni, G. L.

Frassineti, D. Amadori & A. Rocca 2013. Trastuzumab-induced cardiotoxicity

224
in early breast cancer patients: a retrospective study of possible risk and

protective factors. Heart

Fearon, E. R. 1997. Human cancer syndromes: clues to the origin and nature

of cancer. Science 278: 1043-50.

Felding-Habermann, B. 2003. Integrin adhesion receptors in tumor

metastasis. Clin Exp Metastasis 20: 203-13.

Felding-Habermann, B. 2003. Targeting tumor cell-platelet interaction in

breast cancer metastasis. Pathophysiol Haemost Thromb 33 Suppl 1: 56-8.

Felding-Habermann, B., T. E. O'Toole, J. W. Smith, E. Fransvea, Z. M.

Ruggeri, M. H. Ginsberg, P. E. Hughes, N. Pampori, S. J. Shattil, A. Saven &

B. M. Mueller 2001. Integrin activation controls metastasis in human breast

cancer. Proc Natl Acad Sci U S A 98: 1853-8.

Frampton, J. E. & G. L. Plosker 2009. Efalizumab: a review of its use in the

management of chronic moderate-to-severe plaque psoriasis. Am J Clin

Dermatol 10: 51-72.

Fulda, S. 2009. Tumor resistance to apoptosis. Int J Cancer 124: 511-5.

225
Funahashi, Y., N. H. Sugi, T. Semba, Y. Yamamoto, S. Hamaoka, N.

Tsukahara-Tamai, Y. Ozawa, A. Tsuruoka, K. Nara, K. Takahashi, T. Okabe,

J. Kamata, T. Owa, N. Ueda, T. Haneda, M. Yonaga, K. Yoshimatsu & T.

Wakabayashi 2002. Sulfonamide derivative, E7820, is a unique angiogenesis

inhibitor suppressing an expression of integrin alpha2 subunit on

endothelium. Cancer Res 62: 6116-23.

Gani, R., G. Giovannoni, D. Bates, B. Kemball, S. Hughes & J. Kerrigan

2008. Cost-effectiveness analyses of natalizumab (Tysabri) compared with

other disease-modifying therapies for people with highly active relapsing-

remitting multiple sclerosis in the UK. Pharmacoeconomics 26: 617-27.

Garanger, E., D. Boturyn, Z. Jin, P. Dumy, M. C. Favrot & J. L. Coll 2005.

New multifunctional molecular conjugate vector for targeting, imaging, and

therapy of tumors. Mol Ther 12: 1168-75.

Gasparini, G., P. C. Brooks, E. Biganzoli, P. B. Vermeulen, E. Bonoldi, L. Y.

Dirix, G. Ranieri, R. Miceli & D. A. Cheresh 1998. Vascular integrin

alpha(v)beta3: a new prognostic indicator in breast cancer. Clin Cancer Res

4: 2625-34.

Gavrilovskaya, I. N., E. J. Brown, M. H. Ginsberg & E. R. Mackow 1999.

Cellular entry of hantaviruses which cause hemorrhagic fever with renal

syndrome is mediated by beta3 integrins. J Virol 73: 3951-9.

226
Gawecka, J. E., G. S. Griffiths, B. Ek-Rylander, J. W. Ramos & M. L. Matter

2010. R-Ras regulates migration through an interaction with filamin A in

melanoma cells. PLoS One 5: e11269.

Gay, L. J. & B. Felding-Habermann 2011. Contribution of platelets to tumour

metastasis. Nat Rev Cancer 11: 123-34.

Geho, D. H., R. W. Bandle, T. Clair & L. A. Liotta 2005. Physiological

mechanisms of tumor-cell invasion and migration. Physiology (Bethesda) 20:

194-200.

Geiger, B., J. P. Spatz & A. D. Bershadsky 2009. Environmental sensing

through focal adhesions. Nat Rev Mol Cell Biol 10: 21-33.

Gentilucci, L., G. Cardillo, S. Spampinato, A. Tolomelli, F. Squassabia, R. De

Marco, A. Bedini, M. Baiula, L. Belvisi & M. Civera 2010. Antiangiogenic

effect of dual/selective alpha(5)beta(1)/alpha(v)beta(3) integrin antagonists

designed on partially modified retro-inverso cyclotetrapeptide mimetics. J

Med Chem 53: 106-18.

Gerber, D. E. 2008. Targeted therapies: a new generation of cancer

treatments. Am Fam Physician 77: 311-9.

227
Giugliano, R. P., J. A. White, C. Bode, P. W. Armstrong, G. Montalescot, B.

S. Lewis, A. van 't Hof, L. G. Berdan, K. L. Lee, J. T. Strony, S. Hildemann,

E. Veltri, F. Van de Werf, E. Braunwald, R. A. Harrington, R. M. Califf & L. K.

Newby 2009. Early versus delayed, provisional eptifibatide in acute coronary

syndromes. N Engl J Med 360: 2176-90.

Giuliano, S., W. S. Nesbitt, M. Rooney & S. P. Jackson 2003. Bidirectional

integrin alphaIIbbeta3 signalling regulating platelet adhesion under flow:

contribution of protein kinase C. Biochem J 372: 163-72.

Gladson, C. L. & D. A. Cheresh 1991. Glioblastoma expression of vitronectin

and the alpha v beta 3 integrin. Adhesion mechanism for transformed glial

cells. J Clin Invest 88: 1924-32.

Gladson, C. L., C. Dennis, T. C. Rotolo, D. R. Kelly & J. R. Grammer 1997.

Vitronectin expression in differentiating neuroblastic tumors: integrin alpha v

beta 5 mediates vitronectin-dependent adhesion of retinoic-acid-differentiated

neuroblastoma cells. Am J Pathol 150: 1631-46.

Goetsch, K. P. & C. U. Niesler 2011. Optimization of the scratch assay for in

vitro skeletal muscle wound healing analysis. Anal Biochem 411: 158-60.

Gomes, N., J. Vassy, C. Lebos, B. Arbeille, C. Legrand & F. Fauvel-Lafeve

2004. Breast adenocarcinoma cell adhesion to the vascular subendothelium

228
in whole blood and under flow conditions: effects of alphavbeta3 and

alphaIIbbeta3 antagonists. Clin Exp Metastasis 21: 553-61.

Gong, J., D. Wang, L. Sun, E. Zborowska, J. K. Willson & M. G. Brattain

1997. Role of alpha 5 beta 1 integrin in determining malignant properties of

colon carcinoma cells. Cell Growth Differ 8: 83-90.

Gonzalez, D., L. Fearfield, P. Nathan, P. Taniere, A. Wallace, E. Brown, C.

Harwood, J. Marsden & S. Whittaker 2013. BRAF mutation testing algorithm

for vemurafenib treatment in melanoma: recommendations from an expert

panel. Br J Dermatol 168: 700-707.

Goodman, S. L., H. J. Grote & C. Wilm 2012. Matched rabbit monoclonal

antibodies against alphav-series integrins reveal a novel alphavbeta3-LIBS

epitope, and permit routine staining of archival paraffin samples of human

tumors. Biol Open 1: 329-40.

Goodman, S. L. & M. Picard 2012. Integrins as therapeutic targets. Trends

Pharmacol Sci 33: 405-12.

Gouon, V., G. C. Tucker, L. Kraus-Berthier, G. Atassi & N. Kieffer 1996. Up-

regulated expression of the beta3 integrin and the 92-kDa gelatinase in

human HT-144 melanoma cell tumors grown in nude mice. Int J Cancer 68:

650-62.

229
Grayson, M. H., M. Van der Vieren, S. A. Sterbinsky, W. Michael Gallatin, P.

A. Hoffman, D. E. Staunton & B. S. Bochner 1998. alphadbeta2 integrin is

expressed on human eosinophils and functions as an alternative ligand for

vascular cell adhesion molecule 1 (VCAM-1). J Exp Med 188: 2187-91.

Guadamillas, M. C., A. Cerezo & M. A. Del Pozo 2011. Overcoming anoikis--

pathways to anchorage-independent growth in cancer. J Cell Sci 124: 3189-

97.

Guo, W. & F. G. Giancotti 2004. Integrin signalling during tumour

progression. Nat Rev Mol Cell Biol 5: 816-26.

Haier, J., M. Nasralla & G. L. Nicolson 1999. Different adhesion properties of

highly and poorly metastatic HT-29 colon carcinoma cells with extracellular

matrix components: role of integrin expression and cytoskeletal components.

Br J Cancer 80: 1867-74.

Hanahan, D. & R. A. Weinberg 2000. The hallmarks of cancer. Cell 100: 57-

70.

Hanahan, D. & R. A. Weinberg 2011. Hallmarks of cancer: the next

generation. Cell 144: 646-74.

230
Hariharan, S., D. Gustafson, S. Holden, D. McConkey, D. Davis, M. Morrow,

M. Basche, L. Gore, C. Zang, C. L. O'Bryant, A. Baron, D. Gallemann, D.

Colevas & S. G. Eckhardt 2007. Assessment of the biological and

pharmacological effects of the alpha nu beta3 and alpha nu beta5 integrin

receptor antagonist, cilengitide (EMD 121974), in patients with advanced

solid tumors. Ann Oncol 18: 1400-7.

Havaki, S., M. Kouloukoussa, K. Amawi, Y. Drosos, L. D. Arvanitis, N.

Goutas, D. Vlachodimitropoulos, S. D. Vassilaros, E. Z. Katsantoni, I.

Voloudakis-Baltatzis, V. Aleporou-Marinou, C. Kittas & E. Marinos 2007.

Altered expression pattern of integrin alphavbeta3 correlates with actin

cytoskeleton in primary cultures of human breast cancer. Cancer Cell Int 7:

16.

Haywood-Reid, P. L., D. R. Zipf & W. R. Springer 1997. Quantification of

integrin subunits on human prostatic cell lines--comparison of nontumorigenic

and tumorigenic lines. Prostate 31: 1-8.

Henry, M. D., J. S. Satz, C. Brakebusch, M. Costell, E. Gustafsson, R.

Fassler & K. P. Campbell 2001. Distinct roles for dystroglycan, beta1 integrin

and perlecan in cell surface laminin organization. J Cell Sci 114: 1137-44.

Hersey, P., L. Bastholt, V. Chiarion-Sileni, G. Cinat, R. Dummer, A. M.

Eggermont, E. Espinosa, A. Hauschild, I. Quirt, C. Robert & D. Schadendorf

231
2009. Small molecules and targeted therapies in distant metastatic disease.

Ann Oncol 20 Suppl 6: vi35-40.

Hersey, P., J. Sosman, S. O'Day, J. Richards, A. Bedikian, R. Gonzalez, W.

Sharfman, R. Weber, T. Logan, M. Buzoianu, L. Hammershaimb, J. M.

Kirkwood & G. Etaracizumab Melanoma Study 2010. A randomized phase 2

study of etaracizumab, a monoclonal antibody against integrin

alpha(v)beta(3), + or - dacarbazine in patients with stage IV metastatic

melanoma. Cancer 116: 1526-34.

Hieken, T. J., S. G. Ronan, M. Farolan, A. L. Shilkaitis & T. K. Das Gupta

1999. Molecular prognostic markers in intermediate-thickness cutaneous

malignant melanoma. Cancer 85: 375-82.

Hodivala-Dilke, K. 2008. alphavbeta3 integrin and angiogenesis: a moody

integrin in a changing environment. Curr Opin Cell Biol 20: 514-9.

Hodivala-Dilke, K. M., K. P. McHugh, D. A. Tsakiris, H. Rayburn, D. Crowley,

M. Ullman-Cullere, F. P. Ross, B. S. Coller, S. Teitelbaum & R. O. Hynes

1999. Beta3-integrin-deficient mice are a model for Glanzmann

thrombasthenia showing placental defects and reduced survival. J Clin Invest

103: 229-38.

232
Hodivala-Dilke, K. M., A. R. Reynolds & L. E. Reynolds 2003. Integrins in

angiogenesis: multitalented molecules in a balancing act. Cell Tissue Res

314: 131-44.

Hofmann, U. B., J. R. Westphal, E. T. Waas, J. C. Becker, D. J. Ruiter & G.

N. van Muijen 2000. Coexpression of integrin alpha(v)beta3 and matrix

metalloproteinase-2 (MMP-2) coincides with MMP-2 activation: correlation

with melanoma progression. J Invest Dermatol 115: 625-32.

Hood, J. D. & D. A. Cheresh 2002. Role of integrins in cell invasion and

migration. Nat Rev Cancer 2: 91-100.

Hsia, D. A., S. T. Lim, J. A. Bernard-Trifilo, S. K. Mitra, S. Tanaka, J. den

Hertog, D. N. Streblow, D. Ilic, M. H. Ginsberg & D. D. Schlaepfer 2005.

Integrin alpha4beta1 promotes focal adhesion kinase-independent cell

motility via alpha4 cytoplasmic domain-specific activation of c-Src. Mol Cell

Biol 25: 9700-12.

Hu, D. D., E. C. Lin, N. L. Kovach, J. R. Hoyer & J. W. Smith 1995. A

biochemical characterization of the binding of osteopontin to integrins alpha v

beta 1 and alpha v beta 5. J Biol Chem 270: 26232-8.

Huang, D., Y. Ding, Y. Li, W. M. Luo, Z. F. Zhang, J. Snider, K. Vandenbeldt,

C. N. Qian & B. T. Teh 2010. Sunitinib acts primarily on tumor endothelium

233
rather than tumor cells to inhibit the growth of renal cell carcinoma. Cancer

Res 70: 1053-62.

Huang, M. M., L. Lipfert, M. Cunningham, J. S. Brugge, M. H. Ginsberg & S.

J. Shattil 1993. Adhesive ligand binding to integrin alpha IIb beta 3 stimulates

tyrosine phosphorylation of novel protein substrates before phosphorylation

of pp125FAK. J Cell Biol 122: 473-83.

Humphries, J. D., A. Byron & M. J. Humphries 2006. Integrin ligands at a

glance. J Cell Sci 119: 3901-3.

Humphries, M. J. 2000. Integrin structure. Biochem Soc Trans 28: 311-39.

Humphries, M. J. 2002. Insights into integrin-ligand binding and activation

from the first crystal structure. Arthritis Res 4 Suppl 3: S69-78.

Humphries, M. J. 2004. Monoclonal antibodies as probes of integrin priming

and activation. Biochem Soc Trans 32: 407-11.

Humphries, M. J., P. A. McEwan, S. J. Barton, P. A. Buckley, J. Bella & A. P.

Mould 2003. Integrin structure: heady advances in ligand binding, but

activation still makes the knees wobble. Trends Biochem Sci 28: 313-20.

234
Humphries, M. J., M. A. Travis, K. Clark & A. P. Mould 2004. Mechanisms of

integration of cells and extracellular matrices by integrins. Biochem Soc

Trans 32: 822-5.

Huveneers, S., H. Truong & H. J. Danen 2007. Integrins: signaling, disease,

and therapy. Int J Radiat Biol 83: 743-51.

Huveneers, S., I. van den Bout, P. Sonneveld, A. Sancho, A. Sonnenberg &

E. H. Danen 2007. Integrin alpha v beta 3 controls activity and oncogenic

potential of primed c-Src. Cancer Res 67: 2693-700.

Hynes, R. O. 1992. Integrins: versatility, modulation, and signaling in cell

adhesion. Cell 69: 11-25.

Hynes, R. O. 1999. Cell adhesion: old and new questions. Trends Cell Biol 9:

M33-7.

Hynes, R. O. 2002. Integrins: bidirectional, allosteric signaling machines. Cell

110: 673-87.

Hynes, R. O. & A. D. Lander 1992. Contact and adhesive specificities in the

associations, migrations, and targeting of cells and axons. Cell 68: 303-22.

235
Ismail, A., J. Kemp & B. Sharrack 2009. Melanoma complicating treatment

with natalizumab (tysabri) for multiple sclerosis. J Neurol 256: 1771-2.

Jackson, T., S. Clark, S. Berryman, A. Burman, S. Cambier, D. Mu, S.

Nishimura & A. M. King 2004. Integrin alphavbeta8 functions as a receptor

for foot-and-mouth disease virus: role of the beta-chain cytodomain in

integrin-mediated infection. J Virol 78: 4533-40.

Janji, B., C. Melchior, V. Gouon, L. Vallar & N. Kieffer 1999. Autocrine TGF-

beta-regulated expression of adhesion receptors and integrin-linked kinase in

HT-144 melanoma cells correlates with their metastatic phenotype. Int J

Cancer 83: 255-62.

Jemal, A., F. Bray, M. M. Center, J. Ferlay, E. Ward & D. Forman 2011.

Global cancer statistics. CA Cancer J Clin 61: 69-90.

Jin, H. & J. Varner 2004. Integrins: roles in cancer development and as

treatment targets. Br J Cancer 90: 561-5.

Jin, Y. J., I. Park, I. K. Hong, H. J. Byun, J. Choi, Y. M. Kim & H. Lee 2011.

Fibronectin and vitronectin induce AP-1-mediated matrix metalloproteinase-9

expression through integrin alpha(5)beta(1)/alpha(v)beta(3)-dependent Akt,

ERK and JNK signaling pathways in human umbilical vein endothelial cells.

Cell Signal 23: 125-34.

236
Jin, Z. H., V. Josserand, S. Foillard, D. Boturyn, P. Dumy, M. C. Favrot & J.

L. Coll 2007. In vivo optical imaging of integrin alphaV-beta3 in mice using

multivalent or monovalent cRGD targeting vectors. Mol Cancer 6: 41.

Jinka, R., R. Kapoor, P. G. Sistla, T. A. Raj & G. Pande 2012. Alterations in

Cell-Extracellular Matrix Interactions during Progression of Cancers. Int J Cell

Biol 2012: 219196.

Jockusch, B. M., P. Bubeck, K. Giehl, M. Kroemker, J. Moschner, M.

Rothkegel, M. Rudiger, K. Schluter, G. Stanke & J. Winkler 1995. The

molecular architecture of focal adhesions. Annu Rev Cell Dev Biol 11: 379-

416.

Jones, R. G., I. Davagnanam, S. Colley, R. J. West & D. A. Yates 2008.

Abciximab for treatment of thromboembolic complications during

endovascular coiling of intracranial aneurysms. AJNR Am J Neuroradiol 29:

1925-9.

Kalli, A. C., I. D. Campbell & M. S. Sansom 2011. Multiscale simulations

suggest a mechanism for integrin inside-out activation. Proc Natl Acad Sci U

S A 108: 11890-5.

237
Karadag, A., K. U. Ogbureke, N. S. Fedarko & L. W. Fisher 2004. Bone

sialoprotein, matrix metalloproteinase 2, and alpha(v)beta3 integrin in

osteotropic cancer cell invasion. J Natl Cancer Inst 96: 956-65.

Kaye, S. B. 1998. New antimetabolites in cancer chemotherapy and their

clinical impact. Br J Cancer 78 Suppl 3: 1-7.

Keizer, G. D., A. A. Te Velde, R. Schwarting, C. G. Figdor & J. E. De Vries

1987. Role of p150,95 in adhesion, migration, chemotaxis and phagocytosis

of human monocytes. Eur J Immunol 17: 1317-22.

Kerbel, R. S. 2003. Human tumor xenografts as predictive preclinical models

for anticancer drug activity in humans: better than commonly perceived-but

they can be improved. Cancer Biol Ther 2: S134-9.

Kim, C., F. Ye & M. H. Ginsberg 2011. Regulation of Integrin Activation. Annu

Rev Cell Dev Biol

Kim, W., M. H. Jeong, K. H. Kim, I. S. Sohn, Y. J. Hong, H. W. Park, J. H.

Kim, Y. K. Ahn, J. G. Cho, J. C. Park, D. L. Cho & J. C. Kang 2006. The

clinical results of a platelet glycoprotein IIb/IIIa receptor blocker (abciximab:

ReoPro)-coated stent in acute myocardial infarction. J Am Coll Cardiol 47:

933-8.

238
Klein, C. E., D. Dressel, T. Steinmayer, C. Mauch, B. Eckes, T. Krieg, R. B.

Bankert & L. Weber 1991. Integrin alpha 2 beta 1 is upregulated in fibroblasts

and highly aggressive melanoma cells in three-dimensional collagen lattices

and mediates the reorganization of collagen I fibrils. J Cell Biol 115: 1427-36.

Korpanty, G., E. Smyth, L. A. Sullivan, R. A. Brekken & D. N. Carney 2010.

Antiangiogenic therapy in lung cancer: focus on vascular endothelial growth

factor pathway. Exp Biol Med (Maywood) 235: 3-9.

Kramer, N., A. Walzl, C. Unger, M. Rosner, G. Krupitza, M. Hengstschlager &

H. Dolznig 2013. In vitro cell migration and invasion assays. Mutat Res 752:

10-24.

Kremser, M. E., M. Przybylo, D. Hoja-Lukowicz, E. Pochec, A. Amoresano,

A. Carpentieri, M. Bubka & A. Litynska 2008. Characterisation of alpha3beta1

and alpha(v)beta3 integrin N-oligosaccharides in metastatic melanoma WM9

and WM239 cell lines. Biochim Biophys Acta 1780: 1421-31.

Kubota, S., H. Ito, Y. Ishibashi & Y. Seyama 1997. Anti-alpha3 integrin

antibody induces the activated form of matrix metalloprotease-2 (MMP-2)

with concomitant stimulation of invasion through matrigel by human

rhabdomyosarcoma cells. Int J Cancer 70: 106-11.

239
Kumar, C. C., M. Malkowski, Z. Yin, E. Tanghetti, B. Yaremko, T. Nechuta, J.

Varner, M. Liu, E. M. Smith, B. Neustadt, M. Presta & L. Armstrong 2001.

Inhibition of angiogenesis and tumor growth by SCH221153, a dual

alpha(v)beta3 and alpha(v)beta5 integrin receptor antagonist. Cancer Res

61: 2232-8.

Labrie, F. 2004. Current status of endocrine therapy in localized prostate

cancer: cure has become a strong possibility. Int Braz J Urol 30: 3-11.

Landen, C. N., T. J. Kim, Y. G. Lin, W. M. Merritt, A. A. Kamat, L. Y. Han, W.

A. Spannuth, A. M. Nick, N. B. Jennnings, M. S. Kinch, D. Tice & A. K. Sood

2008. Tumor-selective response to antibody-mediated targeting of

alphavbeta3 integrin in ovarian cancer. Neoplasia 10: 1259-67.

Lau, L. M., J. L. Wee, M. D. Wright, G. W. Moseley, P. M. Hogarth, L. K.

Ashman & D. E. Jackson 2004. The tetraspanin superfamily member CD151

regulates outside-in integrin alphaIIbbeta3 signaling and platelet function.

Blood 104: 2368-75.

Laurens, N., M. A. Engelse, C. Jungerius, C. W. Lowik, V. W. van Hinsbergh

& P. Koolwijk 2009. Single and combined effects of alphavbeta3- and

alpha5beta1-integrins on capillary tube formation in a human fibrinous matrix.

Angiogenesis 12: 275-85.

240
Leavesley, D. I., G. D. Ferguson, E. A. Wayner & D. A. Cheresh 1992.

Requirement of the integrin beta 3 subunit for carcinoma cell spreading or

migration on vitronectin and fibrinogen. J Cell Biol 117: 1101-7.

Lee, J. O., L. A. Bankston, M. A. Arnaout & R. C. Liddington 1995. Two

conformations of the integrin A-domain (I-domain): a pathway for activation?

Structure 3: 1333-40.

Legate, K. R., S. A. Wickstrom & R. Fassler 2009. Genetic and cell biological

analysis of integrin outside-in signaling. Genes Dev 23: 397-418.

Leu, S. J., S. C. Lam & L. F. Lau 2002. Pro-angiogenic activities of CYR61

(CCN1) mediated through integrins alphavbeta3 and alpha6beta1 in human

umbilical vein endothelial cells. J Biol Chem 277: 46248-55.

Leu, S. J., Y. Liu, N. Chen, C. C. Chen, S. C. Lam & L. F. Lau 2003.

Identification of a novel integrin alpha 6 beta 1 binding site in the angiogenic

inducer CCN1 (CYR61). J Biol Chem 278: 33801-8.

Li, G., L. Zhang, E. Chen, J. Wang, X. Jiang, J. H. Chen, G. Wickman, K.

Amundson, S. Bergqvist, J. Zobel, D. Buckman, S. M. Baxi, S. L. Bender, G.

F. Casperson & D. D. Hu-Lowe 2010. Dual functional monoclonal antibody

PF-04605412 targets integrin alpha5beta1 and elicits potent antibody-

dependent cellular cytotoxicity. Cancer Res 70: 10243-54.

241
Li, J., L. Zhou, H. T. Tran, Y. Chen, N. E. Nguyen, M. A. Karasek & M. P.

Marinkovich 2006. Overexpression of laminin-8 in human dermal

microvascular endothelial cells promotes angiogenesis-related functions. J

Invest Dermatol 126: 432-40.

Li, X., J. Regezi, F. P. Ross, S. Blystone, D. Ilic, S. P. Leong & D. M. Ramos

2001. Integrin alphavbeta3 mediates K1735 murine melanoma cell motility in

vivo and in vitro. J Cell Sci 114: 2665-72.

Liang, C. C., A. Y. Park & J. L. Guan 2007. In vitro scratch assay: a

convenient and inexpensive method for analysis of cell migration in vitro. Nat

Protoc 2: 329-33.

Liao, Z., C. I. Seye, G. A. Weisman & L. Erb 2007. The P2Y2 nucleotide

receptor requires interaction with alpha v integrins to access and activate

G12. J Cell Sci 120: 1654-62.

Liapis, H. & K. Hutton 1997. Detection of integrins in formalin-fixed, paraffin-

embedded tissues. J Histochem Cytochem 45: 737-41.

Lin, E., Q. Wang, S. Swenson, H. Jadvar, S. Groshen, W. Ye, F. S. Markland

& J. Pinski 2010. The disintegrin contortrostatin in combination with docetaxel

is a potent inhibitor of prostate cancer in vitro and in vivo. Prostate 70: 1359-

70.

242
Liu, Y., F. Zhao, W. Gu, H. Yang, Q. Meng, Y. Zhang & Q. Duan 2009. The

roles of platelet GPIIb/IIIa and alphavbeta3 integrins during HeLa cells

adhesion, migration, and invasion to monolayer endothelium under static and

dynamic shear flow. J Biomed Biotechnol 2009: 829243.

Liu, Z., B. Jia, H. Zhao, X. Chen & F. Wang 2011. Specific targeting of

human integrin alpha(v)beta (3) with (111)In-labeled Abegrin in nude mouse

models. Mol Imaging Biol 13: 112-20.

Liu, Z., F. Wang & X. Chen 2008. Integrin alpha(v)beta(3)-Targeted Cancer

Therapy. Drug Dev Res 69: 329-339.

Lock, J. G., B. Wehrle-Haller & S. Stromblad 2008. Cell-matrix adhesion

complexes: master control machinery of cell migration. Semin Cancer Biol

18: 65-76.

Lomonaco, S. L., S. Finniss, C. Xiang, H. K. Lee, W. Jiang, N. Lemke, S. A.

Rempel, T. Mikkelsen & C. Brodie 2011. Cilengitide induces autophagy-

mediated cell death in glioma cells. Neuro Oncol 13: 857-65.

Lord, C. J. & A. Ashworth 2010. Biology-driven cancer drug development:

back to the future. BMC Biol 8: 38.

243
Lorger, M., J. S. Krueger, M. O'Neal, K. Staflin & B. Felding-Habermann

2009. Activation of tumor cell integrin alphavbeta3 controls angiogenesis and

metastatic growth in the brain. Proc Natl Acad Sci U S A 106: 10666-71.

Lu, Q. L. & T. A. Partridge 1998. A new blocking method for application of

murine monoclonal antibody to mouse tissue sections. J Histochem

Cytochem 46: 977-84.

Lu, X., D. Lu, M. Scully & V. Kakkar 2008. The role of integrins in cancer and

the development of anti-integrin therapeutic agents for cancer therapy.

Perspect Medicin Chem 2: 57-73.

MacDonald, T. J., C. F. Stewart, M. Kocak, S. Goldman, R. G. Ellenbogen, P.

Phillips, D. Lafond, T. Y. Poussaint, M. W. Kieran, J. M. Boyett & L. E. Kun

2008. Phase I clinical trial of cilengitide in children with refractory brain

tumors: Pediatric Brain Tumor Consortium Study PBTC-012. J Clin Oncol 26:

919-24.

MacDonald, T. J., T. Taga, H. Shimada, P. Tabrizi, B. V. Zlokovic, D. A.

Cheresh & W. E. Laug 2001. Preferential susceptibility of brain tumors to the

antiangiogenic effects of an alpha(v) integrin antagonist. Neurosurgery 48:

151-7.

244
Mahabeleshwar, G. H., W. Feng, D. R. Phillips & T. V. Byzova 2006. Integrin

signaling is critical for pathological angiogenesis. J Exp Med 203: 2495-507.

Malinowsky, K., C. Wolff, S. Gundisch, D. Berg & K. Becker 2010. Targeted

therapies in cancer - challenges and chances offered by newly developed

techniques for protein analysis in clinical tissues. J Cancer 2: 26-35.

Malvezzi, M., P. Bertuccio, F. Levi, C. La Vecchia & E. Negri 2013. European

cancer mortality predictions for the year 2013. Ann Oncol 24: 792-800.

Manohar, A., S. G. Shome, J. Lamar, L. Stirling, V. Iyer, K. Pumiglia & C. M.

DiPersio 2004. Alpha 3 beta 1 integrin promotes keratinocyte cell survival

through activation of a MEK/ERK signaling pathway. J Cell Sci 117: 4043-54.

Manzoni, L., L. Belvisi, D. Arosio, M. Civera, M. Pilkington-Miksa, D. Potenza,

A. Caprini, E. M. Araldi, E. Monferini, M. Mancino, F. Podesta & C. Scolastico

2009. Cyclic RGD-containing functionalized azabicycloalkane peptides as

potent integrin antagonists for tumor targeting. ChemMedChem 4: 615-32.

Marshall, J. F., D. C. Rutherford, A. C. McCartney, F. Mitjans, S. L. Goodman

& I. R. Hart 1995. Alpha v beta 1 is a receptor for vitronectin and fibrinogen,

and acts with alpha 5 beta 1 to mediate spreading on fibronectin. J Cell Sci

108 ( Pt 3): 1227-38.

245
Martin, S. S. & K. Vuori 2004. Regulation of Bcl-2 proteins during anoikis and

amorphosis. Biochim Biophys Acta 1692: 145-57.

Mas-Moruno, C., F. Rechenmacher & H. Kessler 2010. Cilengitide: the first

anti-angiogenic small molecule drug candidate design, synthesis and clinical

evaluation. Anticancer Agents Med Chem 10: 753-68.

Mattern, R. H., S. B. Read, M. D. Pierschbacher, C. I. Sze, B. P. Eliceiri & C.

A. Kruse 2005. Glioma cell integrin expression and their interactions with

integrin antagonists: Research Article. Cancer Ther 3A: 325-340.

Max, R., R. R. Gerritsen, P. T. Nooijen, S. L. Goodman, A. Sutter, U.

Keilholz, D. J. Ruiter & R. M. De Waal 1997. Immunohistochemical analysis

of integrin alpha vbeta3 expression on tumor-associated vessels of human

carcinomas. Int J Cancer 71: 320-4.

McCabe, N. P., S. De, A. Vasanji, J. Brainard & T. V. Byzova 2007. Prostate

cancer specific integrin alphavbeta3 modulates bone metastatic growth and

tissue remodeling. Oncogene 26: 6238-43.

McNeel, D. G., J. Eickhoff, F. T. Lee, D. M. King, D. Alberti, J. P. Thomas, A.

Friedl, J. Kolesar, R. Marnocha, J. Volkman, J. Zhang, L. Hammershaimb, J.

A. Zwiebel & G. Wilding 2005. Phase I trial of a monoclonal antibody specific

for alphavbeta3 integrin (MEDI-522) in patients with advanced malignancies,

246
including an assessment of effect on tumor perfusion. Clin Cancer Res 11:

7851-60.

Meier, F., B. Schittek, S. Busch, C. Garbe, K. Smalley, K. Satyamoorthy, G.

Li & M. Herlyn 2005. The RAS/RAF/MEK/ERK and PI3K/AKT signaling

pathways present molecular targets for the effective treatment of advanced

melanoma. Front Biosci 10: 2986-3001.

Meirow, D. & D. Nugent 2001. The effects of radiotherapy and chemotherapy

on female reproduction. Hum Reprod Update 7: 535-43.

Mikkelsen, T., C. Brodie, S. Finniss, M. E. Berens, J. L. Rennert, K. Nelson,

N. Lemke, S. L. Brown, D. Hahn, B. Neuteboom & S. L. Goodman 2009.

Radiation sensitization of glioblastoma by cilengitide has unanticipated

schedule-dependency. Int J Cancer 124: 2719-27.

Millard, M., S. Odde & N. Neamati 2011. Integrin targeted therapeutics.

Theranostics 1: 154-88.

Mita, M., K. R. Kelly, A. Mita, A. D. Ricart, O. Romero, A. Tolcher, L. Hook, C.

Okereke, I. Krivelevich, D. P. Rossignol, F. J. Giles, E. K. Rowinsky & C.

Takimoto 2011. Phase I study of E7820, an oral inhibitor of integrin {alpha}-2

expression with antiangiogenic properties, in patients with advanced

malignancies. Clin Cancer Res 17: 193-200.

247
Mizejewski, G. J. 1999. Role of integrins in cancer: survey of expression

patterns. Proc Soc Exp Biol Med 222: 124-38.

Moller, H., L. Fairley, V. Coupland, C. Okello, M. Green, D. Forman, B. Moller

& F. Bray 2007. The future burden of cancer in England: incidence and

numbers of new patients in 2020. Br J Cancer 96: 1484-8.

Mosmann, T. 1983. Rapid colorimetric assay for cellular growth and survival:

application to proliferation and cytotoxicity assays. J Immunol Methods 65:

55-63.

Mousa, S. A. 2002. Anti-integrin as novel drug-discovery targets: potential

therapeutic and diagnostic implications. Curr Opin Chem Biol 6: 534-41.

Moxley, K. M. & D. S. McMeekin 2010. Endometrial carcinoma: a review of

chemotherapy, drug resistance, and the search for new agents. Oncologist

15: 1026-33.

Moyano, J. V., B. Carnemolla, C. Dominguez-Jimenez, M. Garcia-Gila, J. P.

Albar, P. Sanchez-Aparicio, A. Leprini, G. Querze, L. Zardi & A. Garcia-Pardo

1997. Fibronectin type III5 repeat contains a novel cell adhesion sequence,

KLDAPT, which binds activated alpha4beta1 and alpha4beta7 integrins. J

Biol Chem 272: 24832-6.

248
Mulgrew, K., K. Kinneer, X. T. Yao, B. K. Ward, M. M. Damschroder, B.

Walsh, S. Y. Mao, C. Gao, P. A. Kiener, S. Coats, M. S. Kinch & D. A. Tice

2006. Direct targeting of alphavbeta3 integrin on tumor cells with a

monoclonal antibody, Abegrin. Mol Cancer Ther 5: 3122-9.

Mullamitha, S. A., N. C. Ton, G. J. Parker, A. Jackson, P. J. Julyan, C.

Roberts, G. A. Buonaccorsi, Y. Watson, K. Davies, S. Cheung, L. Hope, J.

W. Valle, J. A. Radford, J. Lawrance, M. P. Saunders, M. C. Munteanu, M. T.

Nakada, J. A. Nemeth, H. M. Davis, Q. Jiao, U. Prabhakar, Z. Lang, R. E.

Corringham, R. A. Beckman & G. C. Jayson 2007. Phase I evaluation of a

fully human anti-alphav integrin monoclonal antibody (CNTO 95) in patients

with advanced solid tumors. Clin Cancer Res 13: 2128-35.

Muller, B., H. G. Zerwes, K. Tangemann, J. Peter & J. Engel 1993. Two-step

binding mechanism of fibrinogen to alpha IIb beta 3 integrin reconstituted into

planar lipid bilayers. J Biol Chem 268: 6800-8.

Muller, U., B. Bossy, K. Venstrom & L. F. Reichardt 1995. Integrin alpha 8

beta 1 promotes attachment, cell spreading, and neurite outgrowth on

fibronectin. Mol Biol Cell 6: 433-48.

Munger, J. S., X. Huang, H. Kawakatsu, M. J. Griffiths, S. L. Dalton, J. Wu, J.

F. Pittet, N. Kaminski, C. Garat, M. A. Matthay, D. B. Rifkin & D. Sheppard

1999. The integrin alpha v beta 6 binds and activates latent TGF beta 1: a

249
mechanism for regulating pulmonary inflammation and fibrosis. Cell 96: 319-

28.

Murakami, M., T. Fujimaki, S. Asano, H. Nakaguchi, S. M. Yamada, K. Hoya,

K. Yamazaki, Y. Ishida & A. Matsuno 2011. Combination therapy with

rituximab and temozolomide for recurrent and refractory primary central

nervous system lymphoma. Yonsei Med J 52: 1031-4.

Nabors, L. B., T. Mikkelsen, S. S. Rosenfeld, F. Hochberg, N. S. Akella, J. D.

Fisher, G. A. Cloud, Y. Zhang, K. Carson, S. M. Wittemer, A. D. Colevas & S.

A. Grossman 2007. Phase I and correlative biology study of cilengitide in

patients with recurrent malignant glioma. J Clin Oncol 25: 1651-7.

Nakamura, I., M. F. Pilkington, P. T. Lakkakorpi, L. Lipfert, S. M. Sims, S. J.

Dixon, G. A. Rodan & L. T. Duong 1999. Role of alpha(v)beta(3) integrin in

osteoclast migration and formation of the sealing zone. J Cell Sci 112 ( Pt

22): 3985-93.

Nelsen-Salz, B., H. J. Eggers & H. Zimmermann 1999. Integrin alpha(v)beta3

(vitronectin receptor) is a candidate receptor for the virulent echovirus 9

strain Barty. J Gen Virol 80 ( Pt 9): 2311-3.

Nemeth, J. A., M. L. Cher, Z. Zhou, C. Mullins, S. Bhagat & M. Trikha 2003.

Inhibition of alpha(v)beta3 integrin reduces angiogenesis, bone turnover, and

250
tumor cell proliferation in experimental prostate cancer bone metastases. Clin

Exp Metastasis 20: 413-20.

Neto, D. S., L. Pantaleao, B. C. de Sa & G. Landman 2007. Alpha-v-beta3

integrin expression in melanocytic nevi and cutaneous melanoma. J Cutan

Pathol 34: 851-6.

Niland, S. & J. A. Eble 2012. Integrin-mediated cell-matrix interaction in

physiological and pathological blood vessel formation. J Oncol 2012: 125278.

Ning, S., J. A. Nemeth, R. L. Hanson, K. Forsythe & S. J. Knox 2008. Anti-

integrin monoclonal antibody CNTO 95 enhances the therapeutic efficacy of

fractionated radiation therapy in vivo. Mol Cancer Ther 7: 1569-78.

Nishimura, S. L., D. Sheppard & R. Pytela 1994. Integrin alpha v beta 8.

Interaction with vitronectin and functional divergence of the beta 8

cytoplasmic domain. J Biol Chem 269: 28708-15.

Niu, G. & X. Chen 2011. Why integrin as a primary target for imaging and

therapy. Theranostics 1: 30-47.

O'Brien, V., S. M. Frisch & R. L. Juliano 1996. Expression of the integrin

alpha 5 subunit in HT29 colon carcinoma cells suppresses apoptosis

triggered by serum deprivation. Exp Cell Res 224: 208-13.

251
O'Sullivan, C. C. & S. M. Swain 2013. Pertuzumab : evolving therapeutic

strategies in the management of HER2-overexpressing breast cancer. Expert

Opin Biol Ther 13: 779-90.

O'Toole, T. E., J. C. Loftus, X. P. Du, A. A. Glass, Z. M. Ruggeri, S. J. Shattil,

E. F. Plow & M. H. Ginsberg 1990. Affinity modulation of the alpha IIb beta 3

integrin (platelet GPIIb-IIIa) is an intrinsic property of the receptor. Cell Regul

1: 883-93.

O'Toole, T. E., J. C. Loftus, E. F. Plow, A. A. Glass, J. R. Harper & M. H.

Ginsberg 1989. Efficient surface expression of platelet GPIIb-IIIa requires

both subunits. Blood 74: 14-8.

Okegawa, T., R. C. Pong, Y. Li & J. T. Hsieh 2004. The role of cell adhesion

molecule in cancer progression and its application in cancer therapy. Acta

Biochim Pol 51: 445-57.

Oliveira-Ferrer, L., J. Hauschild, W. Fiedler, C. Bokemeyer, J. Nippgen, I.

Celik & G. Schuch 2008. Cilengitide induces cellular detachment and

apoptosis in endothelial and glioma cells mediated by inhibition of

FAK/src/AKT pathway. J Exp Clin Cancer Res 27: 86.

252
Oommen, S., S. K. Gupta & N. E. Vlahakis 2010. Vascular endothelial growth

factor - A (VEGF-A) induces endothelial and cancer cell migration through

direct binding to integrin {alpha}9{beta}1: identification of a specific

{alpha}9{beta}1 binding site. J Biol Chem

Panka, D. J., W. Wang, M. B. Atkins & J. W. Mier 2006. The Raf inhibitor

BAY 43-9006 (Sorafenib) induces caspase-independent apoptosis in

melanoma cells. Cancer Res 66: 1611-9.

Parikka, M., L. Nissinen, T. Kainulainen, L. Bruckner-Tuderman, T. Salo, J.

Heino & K. Tasanen 2006. Collagen XVII promotes integrin-mediated

squamous cell carcinoma transmigration--a novel role for alphaIIb integrin

and tirofiban. Exp Cell Res 312: 1431-8.

Partheen, K., K. Levan, L. Osterberg, I. Claesson, K. Sundfeldt & G. Horvath

2009. External validation suggests Integrin beta 3 as prognostic biomarker in

serous ovarian adenocarcinomas. BMC Cancer 9: 336.

Pathak, A., R. Zhao, J. Huang & G. A. Stouffer 2008. Eptifibatide and

abciximab inhibit insulin-induced focal adhesion formation and proliferative

responses in human aortic smooth muscle cells. Cardiovasc Diabetol 7: 36.

Pecheur, I., O. Peyruchaud, C. M. Serre, J. Guglielmi, C. Voland, F. Bourre,

C. Margue, M. Cohen-Solal, A. Buffet, N. Kieffer & P. Clezardin 2002.

253
Integrin alpha(v)beta3 expression confers on tumor cells a greater propensity

to metastasize to bone. FASEB J 16: 1266-8.

Peng, C., X. Liu, E. Liu, K. Xu, W. Niu, R. Chen, J. Wang, Z. Zhang, P. Lin,

M. Agrez & J. Niu 2009. Norcantharidin induces HT-29 colon cancer cell

apoptosis through the alphavbeta6-extracellular signal-related kinase

signaling pathway. Cancer Sci 100: 2302-8.

Petitclerc, E., S. Stromblad, T. L. von Schalscha, F. Mitjans, J. Piulats, A. M.

Montgomery, D. A. Cheresh & P. C. Brooks 1999. Integrin alpha(v)beta3

promotes M21 melanoma growth in human skin by regulating tumor cell

survival. Cancer Res 59: 2724-30.

Podolnikova, N. P., V. P. Yakubenko, G. L. Volkov, E. F. Plow & T. P.

Ugarova 2003. Identification of a novel binding site for platelet integrins alpha

IIb beta 3 (GPIIbIIIa) and alpha 5 beta 1 in the gamma C-domain of

fibrinogen. J Biol Chem 278: 32251-8.

Pontes-Junior, J., S. T. Reis, M. Dall'Oglio, L. C. Neves de Oliveira, J. Cury,

P. A. Carvalho, L. A. Ribeiro-Filho, K. R. Moreira Leite & M. Srougi 2009.

Evaluation of the expression of integrins and cell adhesion molecules through

tissue microarray in lymph node metastases of prostate cancer. J Carcinog 8:

3.

254
Popkov, M., C. Rader, B. Gonzalez, S. C. Sinha & C. F. Barbas, 3rd 2006.

Small molecule drug activity in melanoma models may be dramatically

enhanced with an antibody effector. Int J Cancer 119: 1194-207.

Popova, S. N., M. Barczyk, C. F. Tiger, W. Beertsen, P. Zigrino, A. Aszodi, N.

Miosge, E. Forsberg & D. Gullberg 2007. Alpha11 beta1 integrin-dependent

regulation of periodontal ligament function in the erupting mouse incisor. Mol

Cell Biol 27: 4306-16.

Pozzi, A. & R. Zent 2011. TGF-beta sequestration by mesangial cell integrin

alphavbeta8: A novel mechanism of glomerular endothelial cell regulation.

Am J Pathol 178: 485-9.

Procopio, G., J. Bellmunt, J. Dutcher, S. Bracarda, J. Knox, A. Brueckner, I.

Molnar, B. Escudier & T. E. Hutson 2013. Sorafenib tolerability in elderly

patients with advanced renal cell carcinoma: results from a large pooled

analysis. Br J Cancer 108: 311-8.

Puerschel, W. C., M. Gawaz, W. I. Worret & J. Ring 1996. Immunoreactivity

of glycoprotein IIb is present in metastasized but not in non-metastasized

primary malignant melanoma. Br J Dermatol 135: 883-7.

Pulido, R., M. J. Elices, M. R. Campanero, L. Osborn, S. Schiffer, A. Garcia-

Pardo, R. Lobb, M. E. Hemler & F. Sanchez-Madrid 1991. Functional

255
evidence for three distinct and independently inhibitable adhesion activities

mediated by the human integrin VLA-4. Correlation with distinct alpha 4

epitopes. J Biol Chem 266: 10241-5.

Qin, J., O. Vinogradova & E. F. Plow 2004. Integrin bidirectional signaling: a

molecular view. PLoS Biol 2: e169.

Quinn, M. J., T. V. Byzova, J. Qin, E. J. Topol & E. F. Plow 2003. Integrin

alphaIIbbeta3 and its antagonism. Arterioscler Thromb Vasc Biol 23: 945-52.

Quintas-Cardama, A., H. Kantarjian, D. Jones, C. Nicaise, S. O'Brien, F.

Giles, M. Talpaz & J. Cortes 2007. Dasatinib (BMS-354825) is active in

Philadelphia chromosome-positive chronic myelogenous leukemia after

imatinib and nilotinib (AMN107) therapy failure. Blood 109: 497-9.

Radeff-Huang, J., T. M. Seasholtz, J. W. Chang, J. M. Smith, C. T. Walsh &

J. H. Brown 2007. Tumor necrosis factor-alpha-stimulated cell proliferation is

mediated through sphingosine kinase-dependent Akt activation and cyclin D

expression. J Biol Chem 282: 863-70.

Rainov, N. G. 2000. A phase III clinical evaluation of herpes simplex virus

type 1 thymidine kinase and ganciclovir gene therapy as an adjuvant to

surgical resection and radiation in adults with previously untreated

glioblastoma multiforme. Hum Gene Ther 11: 2389-401.

256
Ranpura, V., S. Hapani & S. Wu 2011. Treatment-related mortality with

bevacizumab in cancer patients: a meta-analysis. JAMA 305: 487-94.

Rao, H., G. Lu, H. Kajiya, V. Garcia-Palacios, N. Kurihara, J. Anderson, K.

Patrene, D. Sheppard, H. C. Blair, J. J. Windle, S. J. Choi & G. D. Roodman

2006. Alpha9beta1: a novel osteoclast integrin that regulates osteoclast

formation and function. J Bone Miner Res 21: 1657-65.

Reardon, D. A., K. L. Fink, T. Mikkelsen, T. F. Cloughesy, A. O'Neill, S.

Plotkin, M. Glantz, P. Ravin, J. J. Raizer, K. M. Rich, D. Schiff, W. R.

Shapiro, S. Burdette-Radoux, E. J. Dropcho, S. M. Wittemer, J. Nippgen, M.

Picard & L. B. Nabors 2008. Randomized phase II study of cilengitide, an

integrin-targeting arginine-glycine-aspartic acid peptide, in recurrent

glioblastoma multiforme. J Clin Oncol 26: 5610-7.

Reinmuth, N., W. Liu, S. A. Ahmad, F. Fan, O. Stoeltzing, A. A. Parikh, C. D.

Bucana, G. E. Gallick, M. A. Nickols, W. F. Westlin & L. M. Ellis 2003.

Alphavbeta3 integrin antagonist S247 decreases colon cancer metastasis

and angiogenesis and improves survival in mice. Cancer Res 63: 2079-87.

Reynolds, A. R., I. R. Hart, A. R. Watson, J. C. Welti, R. G. Silva, S. D.

Robinson, G. Da Violante, M. Gourlaouen, M. Salih, M. C. Jones, D. T.

Jones, G. Saunders, V. Kostourou, F. Perron-Sierra, J. C. Norman, G. C.

257
Tucker & K. M. Hodivala-Dilke 2009. Stimulation of tumor growth and

angiogenesis by low concentrations of RGD-mimetic integrin inhibitors. Nat

Med 15: 392-400.

Reynolds, L. E., L. Wyder, J. C. Lively, D. Taverna, S. D. Robinson, X.

Huang, D. Sheppard, R. O. Hynes & K. M. Hodivala-Dilke 2002. Enhanced

pathological angiogenesis in mice lacking beta3 integrin or beta3 and beta5

integrins. Nat Med 8: 27-34.

Ricart, A. D., A. W. Tolcher, G. Liu, K. Holen, G. Schwartz, M. Albertini, G.

Weiss, S. Yazji, C. Ng & G. Wilding 2008. Volociximab, a chimeric

monoclonal antibody that specifically binds alpha5beta1 integrin: a phase I,

pharmacokinetic, and biological correlative study. Clin Cancer Res 14: 7924-

9.

Robinson, S. D. & K. M. Hodivala-Dilke 2011. The role of beta3-integrins in

tumor angiogenesis: context is everything. Curr Opin Cell Biol 23: 630-7.

Robinson, S. D., L. E. Reynolds, L. Wyder, D. J. Hicklin & K. M. Hodivala-

Dilke 2004. Beta3-integrin regulates vascular endothelial growth factor-A-

dependent permeability. Arterioscler Thromb Vasc Biol 24: 2108-14.

Rocco, M., B. Spotorno & R. R. Hantgan 1993. Modeling the alpha IIb beta 3

integrin solution conformation. Protein Sci 2: 2154-66.

258
Rossino, P., P. Defilippi, L. Silengo & G. Tarone 1991. Up-regulation of the

integrin alpha 1/beta 1 in human neuroblastoma cells differentiated by

retinoic acid: correlation with increased neurite outgrowth response to

laminin. Cell Regul 2: 1021-33.

Ruoslahti, E. 1996. RGD and other recognition sequences for integrins. Annu

Rev Cell Dev Biol 12: 697-715.

Rusnati, M., E. Tanghetti, P. Dell'Era, A. Gualandris & M. Presta 1997.

alphavbeta3 integrin mediates the cell-adhesive capacity and biological

activity of basic fibroblast growth factor (FGF-2) in cultured endothelial cells.

Mol Biol Cell 8: 2449-61.

Rust, W. L., S. W. Carper & G. E. Plopper 2002. The Promise of Integrins as

Effective Targets for Anticancer Agents. J Biomed Biotechnol 2: 124-130.

Sahai, E. 2007. Illuminating the metastatic process. Nat Rev Cancer 7: 737-

49.

Saxena, P., M. Trerotola, T. Wang, J. Li, A. Sayeed, J. Vanoudenhove, D. S.

Adams, T. J. Fitzgerald, D. C. Altieri & L. R. Languino 2012. PSA regulates

androgen receptor expression in prostate cancer cells. Prostate 72: 769-76.

259
Scales, T. M. & M. Parsons 2011. Spatial and temporal regulation of integrin

signalling during cell migration. Curr Opin Cell Biol

Schiro, J. A., B. M. Chan, W. T. Roswit, P. D. Kassner, A. P. Pentland, M. E.

Hemler, A. Z. Eisen & T. S. Kupper 1991. Integrin alpha 2 beta 1 (VLA-2)

mediates reorganization and contraction of collagen matrices by human cells.

Cell 67: 403-10.

Schittenhelm, J., E. I. Schwab, J. Sperveslage, M. Tatagiba, R. Meyermann,

F. Fend, S. L. Goodman & B. Sipos 2013. Longitudinal Expression Analysis

of alphav Integrins in Human Gliomas Reveals Upregulation of Integrin

alphavbeta3 as a Negative Prognostic Factor. J Neuropathol Exp Neurol 72:

194-210.

Schlaepfer, D. D., S. K. Hanks, T. Hunter & P. van der Geer 1994. Integrin-

mediated signal transduction linked to Ras pathway by GRB2 binding to focal

adhesion kinase. Nature 372: 786-91.

Schneider, J. G., S. R. Amend & K. N. Weilbaecher 2011. Integrins and bone

metastasis: integrating tumor cell and stromal cell interactions. Bone 48: 54-

65.

Schnell, O., B. Krebs, E. Wagner, A. Romagna, A. J. Beer, S. J. Grau, N.

Thon, C. Goetz, H. A. Kretzschmar, J. C. Tonn & R. H. Goldbrunner 2008.

260
Expression of integrin alphavbeta3 in gliomas correlates with tumor grade

and is not restricted to tumor vasculature. Brain Pathol 18: 378-86.

Schwartz, M. A. 2010. Integrins and extracellular matrix in

mechanotransduction. Cold Spring Harb Perspect Biol 2: a005066.

Schwarz, M., T. Nordt, C. Bode & K. Peter 2002. The GP IIb/IIIa inhibitor

abciximab (c7E3) inhibits the binding of various ligands to the leukocyte

integrin Mac-1 (CD11b/CD18, alphaMbeta2). Thromb Res 107: 121-8.

Schweighofer, C. D. & C. M. Wendtner 2010. First-line treatment of chronic

lymphocytic leukemia: role of alemtuzumab. Onco Targets Ther 3: 53-67.

Seftor, R. E., E. A. Seftor, K. R. Gehlsen, W. G. Stetler-Stevenson, P. D.

Brown, E. Ruoslahti & M. J. Hendrix 1992. Role of the alpha v beta 3 integrin

in human melanoma cell invasion. Proc Natl Acad Sci U S A 89: 1557-61.

Semba, T., Y. Funahashi, N. Ono, Y. Yamamoto, N. H. Sugi, M. Asada, K.

Yoshimatsu & T. Wakabayashi 2004. An angiogenesis inhibitor E7820 shows

broad-spectrum tumor growth inhibition in a xenograft model: possible value

of integrin alpha2 on platelets as a biological marker. Clin Cancer Res 10:

1430-8.

261
Seoane, A. I., V. L. Tran, E. E. Sanchez, S. A. White, J. L. Choi, B. Gaytan,

N. Chavez, S. R. Reyes, C. J. Ramos, L. H. Tran, S. E. Lucena, M. Sugarek,

J. C. Perez, S. A. Mandal, S. Ghorab, A. Rodriguez-Acosta, B. K. Fung & J.

G. Soto 2010. The mojastin mutant Moj-DM induces apoptosis of the human

melanoma SK-Mel-28, but not the mutant Moj-NN nor the non-mutated

recombinant Moj-WN. Toxicon 56: 391-401.

Sevinsky, J. R., A. M. Whalen & N. G. Ahn 2004. Extracellular signal-

regulated kinase induces the megakaryocyte GPIIb/CD41 gene through

MafB/Kreisler. Mol Cell Biol 24: 4534-45.

Shannon, K. E., J. L. Keene, S. L. Settle, T. D. Duffin, M. A. Nickols, M.

Westlin, S. Schroeter, P. G. Ruminski & D. W. Griggs 2004. Anti-metastatic

properties of RGD-peptidomimetic agents S137 and S247. Clin Exp

Metastasis 21: 129-38.

Shattil, S. J., C. Kim & M. H. Ginsberg 2010. The final steps of integrin

activation: the end game. Nat Rev Mol Cell Biol 11: 288-300.

Sheldrake, H. M. & L. H. Patterson 2009. Function and antagonism of beta3

integrins in the development of cancer therapy. Curr Cancer Drug Targets 9:

519-40.

262
Shen, M. R., Y. M. Hsu, K. F. Hsu, Y. F. Chen, M. J. Tang & C. Y. Chou

2006. Insulin-like growth factor 1 is a potent stimulator of cervical cancer cell

invasiveness and proliferation that is modulated by alphavbeta3 integrin

signaling. Carcinogenesis 27: 962-71.

Shimaoka, M. & T. A. Springer 2003. Therapeutic antagonists and

conformational regulation of integrin function. Nat Rev Drug Discov 2: 703-

16.

Shimaoka, M., J. Takagi & T. A. Springer 2002. Conformational regulation of

integrin structure and function. Annu Rev Biophys Biomol Struct 31: 485-516.

Si, Z. & P. Hersey 1994. Immunohistological examination of the relationship

between metastatic potential and expression of adhesion molecules and

'selectins' on melanoma cells. Pathology 26: 6-15.

Siegel, R., D. Naishadham & A. Jemal 2013. Cancer statistics, 2013. CA

Cancer J Clin 63: 11-30.

Simmons, D. L. 2005. Anti-adhesion therapies. Curr Opin Pharmacol 5: 398-

404.

263
Singh, B., C. Fu & J. Bhattacharya 2000. Vascular expression of the

alpha(v)beta(3)-integrin in lung and other organs. Am J Physiol Lung Cell Mol

Physiol 278: L217-26.

Sloan, E. K., N. Pouliot, K. L. Stanley, J. Chia, J. M. Moseley, D. K. Hards &

R. L. Anderson 2006. Tumor-specific expression of alphavbeta3 integrin

promotes spontaneous metastasis of breast cancer to bone. Breast Cancer

Res 8: R20.

Soldi, R., S. Mitola, M. Strasly, P. Defilippi, G. Tarone & F. Bussolino 1999.

Role of alphavbeta3 integrin in the activation of vascular endothelial growth

factor receptor-2. EMBO J 18: 882-92.

Somanath, P. R., N. L. Malinin & T. V. Byzova 2009. Cooperation between

integrin alphavbeta3 and VEGFR2 in angiogenesis. Angiogenesis 12: 177-

85.

Somlo, G., F. Atzori, L. C. Strauss, W. J. Geese, J. M. Specht, W. J.

Gradishar, A. Rybicki, O. Sy, L. T. Vahdat & J. Cortes 2013. Dasatinib plus

Capecitabine for Advanced Breast Cancer: Safety and Efficacy in Phase I

Study CA180004. Clin Cancer Res 19: 1884-93.

264
Song, W. K., W. Wang, R. F. Foster, D. A. Bielser & S. J. Kaufman 1992.

H36-alpha 7 is a novel integrin alpha chain that is developmentally regulated

during skeletal myogenesis. J Cell Biol 117: 643-57.

Sonnenberg, A., C. J. Linders, J. H. Daams & S. J. Kennel 1990. The alpha 6

beta 1 (VLA-6) and alpha 6 beta 4 protein complexes: tissue distribution and

biochemical properties. J Cell Sci 96 ( Pt 2): 207-17.

Spiera, R. F., J. K. Gordon, J. N. Mersten, C. M. Magro, M. Mehta, H. F.

Wildman, S. Kloiber, K. A. Kirou, S. Lyman & M. K. Crow 2011. Imatinib

mesylate (Gleevec) in the treatment of diffuse cutaneous systemic sclerosis:

results of a 1-year, phase IIa, single-arm, open-label clinical trial. Ann Rheum

Dis 70: 1003-9.

Spitaleri, A., S. Mari, F. Curnis, C. Traversari, R. Longhi, C. Bordignon, A.

Corti, G. P. Rizzardi & G. Musco 2008. Structural basis for the interaction of

isoDGR with the RGD-binding site of alphavbeta3 integrin. J Biol Chem 283:

19757-68.

Springer, B. A., M. W. Pantoliano, F. A. Barbera, P. L. Gunyuzlu, L. D.

Thompson, W. F. Herblin, S. A. Rosenfeld & G. W. Book 1994. Identification

and concerted function of two receptor binding surfaces on basic fibroblast

growth factor required for mitogenesis. J Biol Chem 269: 26879-84.

265
Springer, T. A. 1990. Adhesion receptors of the immune system. Nature 346:

425-34.

Springer, T. A. 1990. Leucocyte adhesion to cells. Scand J Immunol 32: 211-

6.

Springer, T. A., J. Zhu & T. Xiao 2008. Structural basis for distinctive

recognition of fibrinogen gammaC peptide by the platelet integrin

alphaIIbbeta3. J Cell Biol 182: 791-800.

Srivatsa, S. S., L. A. Fitzpatrick, P. W. Tsao, T. M. Reilly, D. R. Holmes, Jr.,

R. S. Schwartz & S. A. Mousa 1997. Selective alpha v beta 3 integrin

blockade potently limits neointimal hyperplasia and lumen stenosis following

deep coronary arterial stent injury: evidence for the functional importance of

integrin alpha v beta 3 and osteopontin expression during neointima

formation. Cardiovasc Res 36: 408-28.

Staatz, W. D., K. F. Fok, M. M. Zutter, S. P. Adams, B. A. Rodriguez & S. A.

Santoro 1991. Identification of a tetrapeptide recognition sequence for the

alpha 2 beta 1 integrin in collagen. J Biol Chem 266: 7363-7.

Stevenson, R. & A. El-Modir 2011. Unilateral onycholysis in a patient taking

erlotinib (Tarceva). BMJ Case Rep 2011:

266
Stiewe, T. 2007. The p53 family in differentiation and tumorigenesis. Nat Rev

Cancer 7: 165-8.

Stintzing, S., C. Kapaun, R. P. Laubender, A. Jung, J. Neumann, D. P.

Modest, C. Giessen, N. Moosmann, A. Wollenberg, T. Kirchner & V.

Heinemann 2013. Prognostic value of cetuximab-related skin toxicity in

metastatic colorectal cancer patients and its correlation with parameters of

the epidermal growth factor receptor signal transduction pathway: results

from a randomized trial of the GERMAN AIO CRC Study Group. Int J Cancer

132: 236-45.

Strome, S. E., E. A. Sausville & D. Mann 2007. A mechanistic perspective of

monoclonal antibodies in cancer therapy beyond target-related effects.

Oncologist 12: 1084-95.

Stupack, D. G. 2005. Integrins as a distinct subtype of dependence

receptors. Cell Death Differ 12: 1021-30.

Stupack, D. G. & D. A. Cheresh 2002. Get a ligand, get a life: integrins,

signaling and cell survival. J Cell Sci 115: 3729-38.

Stupp, R., M. E. Hegi, B. Neyns, R. Goldbrunner, U. Schlegel, P. M. Clement,

G. G. Grabenbauer, A. F. Ochsenbein, M. Simon, P. Y. Dietrich, T. Pietsch,

C. Hicking, J. C. Tonn, A. C. Diserens, A. Pica, M. Hermisson, S. Krueger, M.

267
Picard & M. Weller 2010. Phase I/IIa study of cilengitide and temozolomide

with concomitant radiotherapy followed by cilengitide and temozolomide

maintenance therapy in patients with newly diagnosed glioblastoma. J Clin

Oncol 28: 2712-8.

Sugiura, Y., E. Nemoto, O. Kawai, Y. Ohkubo, H. Fusegawa & S. Kaseda

2013. Skin rash by gefitinib is a sign of favorable outcomes for patients of

advanced lung adenocarcinoma in Japanese patients. Springerplus 2: 22.

Suzuki, H., S. Hinotsu, H. Akaza, Y. Fujii, S. Kawakami, K. Kihara, K.

Akakura, M. Suzuki, T. Kitamura, Y. Homma & A. Mizokami 2010. Hormonal

therapy for prostate cancer: current topics and future perspectives. Int J Urol

17: 302-13.

Tabatabai, G., M. Weller, B. Nabors, M. Picard, D. Reardon, T. Mikkelsen, C.

Ruegg & R. Stupp 2010. Targeting integrins in malignant glioma. Target

Oncol 5: 175-81.

Taga, T., A. Suzuki, I. Gonzalez-Gomez, F. H. Gilles, M. Stins, H. Shimada,

L. Barsky, K. I. Weinberg & W. E. Laug 2002. alpha v-Integrin antagonist

EMD 121974 induces apoptosis in brain tumor cells growing on vitronectin

and tenascin. Int J Cancer 98: 690-7.

Takada, Y., X. Ye & S. Simon 2007. The integrins. Genome Biol 8: 215.

268
Takagi, J. 2007. Structural basis for ligand recognition by integrins. Curr Opin

Cell Biol 19: 557-64.

Takayama, S., S. Ishii, T. Ikeda, S. Masamura, M. Doi & M. Kitajima 2005.

The relationship between bone metastasis from human breast cancer and

integrin alpha(v)beta3 expression. Anticancer Res 25: 79-83.

Talmadge, J. E. & I. J. Fidler 2010. AACR centennial series: the biology of

cancer metastasis: historical perspective. Cancer Res 70: 5649-69.

Tan, C., S. Cruet-Hennequart, A. Troussard, L. Fazli, P. Costello, K. Sutton,

J. Wheeler, M. Gleave, J. Sanghera & S. Dedhar 2004. Regulation of tumor

angiogenesis by integrin-linked kinase (ILK). Cancer Cell 5: 79-90.

Taylor, R. M., V. Severns, D. C. Brown, M. Bisoffi & L. O. Sillerud 2011.

Prostate cancer targeting motifs: Expression of alpha(nu) beta(3) ,

neurotensin receptor 1, prostate specific membrane antigen, and prostate

stem cell antigen in human prostate cancer cell lines and xenografts.

Prostate

Taylor, R. M., V. Severns, D. C. Brown, M. Bisoffi & L. O. Sillerud 2012.

Prostate cancer targeting motifs: Expression of alpha(nu) beta(3) ,

neurotensin receptor 1, prostate specific membrane antigen, and prostate

269
stem cell antigen in human prostate cancer cell lines and xenografts.

Prostate 72: 523-32.

Taylor, R. M., V. Severns, D. C. Brown, M. Bisoffi & L. O. Sillerud 2012.

Prostate cancer targeting motifs: expression of alphanu beta3, neurotensin

receptor 1, prostate specific membrane antigen, and prostate stem cell

antigen in human prostate cancer cell lines and xenografts. Prostate 72: 523-

32.

Tefilli, M. V., E. L. Gheiler, B. Shekarriz, J. G. de Oliveira, R. Tiguert, D.

Grignon & J. E. Pontes 1998. Primary adenocarcinoma of the urethra with

metastasis to the glans penis: successful treatment with chemotherapy and

radiation therapy. Urology 52: 517-9.

Tefilli, M. V., E. L. Gheiler, R. Tiguert, M. Banerjee, J. Forman, J. E. Pontes &

D. P. Wood, Jr. 1998. Salvage surgery or salvage radiotherapy for locally

recurrent prostate cancer. Urology 52: 224-9.

Tefilli, M. V., E. L. Gheiler, R. Tiguert, U. Barroso, Jr., C. D. Barton, D. P.

Wood, Jr. & J. E. Pontes 1998. Quality of life in patients undergoing salvage

procedures for locally recurrent prostate cancer. J Surg Oncol 69: 156-61.

Teicher, B. A. 2006. Tumor models for efficacy determination. Mol Cancer

Ther 5: 2435-43.

270
Temming, K., R. M. Schiffelers, G. Molema & R. J. Kok 2005. RGD-based

strategies for selective delivery of therapeutics and imaging agents to the

tumour vasculature. Drug Resist Updat 8: 381-402.

Tolar, J., S. L. Teitelbaum & P. J. Orchard 2004. Osteopetrosis. N Engl J

Med 351: 2839-49.

Tomaselli, K. J., P. Doherty, C. J. Emmett, C. H. Damsky, F. S. Walsh & L. F.

Reichardt 1993. Expression of beta 1 integrins in sensory neurons of the

dorsal root ganglion and their functions in neurite outgrowth on two laminin

isoforms. J Neurosci 13: 4880-8.

Trikha, M., E. Raso, Y. Cai, Z. Fazakas, S. Paku, A. T. Porter, J. Timar & K.

V. Honn 1998. Role of alphaII(b)beta3 integrin in prostate cancer metastasis.

Prostate 35: 185-92.

Trikha, M., J. Timar, S. K. Lundy, K. Szekeres, Y. Cai, A. T. Porter & K. V.

Honn 1997. The high affinity alphaIIb beta3 integrin is involved in invasion of

human melanoma cells. Cancer Res 57: 2522-8.

Trikha, M., J. Timar, S. K. Lundy, K. Szekeres, K. Tang, D. Grignon, A. T.

Porter & K. V. Honn 1996. Human prostate carcinoma cells express

functional alphaIIb(beta)3 integrin. Cancer Res 56: 5071-8.

271
Trikha, M., J. Timar, A. Zacharek, J. A. Nemeth, Y. Cai, B. Dome, B. Somlai,

E. Raso, A. Ladanyi & K. V. Honn 2002. Role for beta3 integrins in human

melanoma growth and survival. Int J Cancer 101: 156-67.

Trikha, M., L. Yan & M. T. Nakada 2002. Monoclonal antibodies as

therapeutics in oncology. Curr Opin Biotechnol 13: 609-14.

Trikha, M., Z. Zhou, J. A. Nemeth, Q. Chen, C. Sharp, E. Emmell, J. Giles-

Komar & M. T. Nakada 2004. CNTO 95, a fully human monoclonal antibody

that inhibits alphav integrins, has antitumor and antiangiogenic activity in

vivo. Int J Cancer 110: 326-35.

Trikha, M., Z. Zhou, J. Timar, E. Raso, M. Kennel, E. Emmell & M. T. Nakada

2002. Multiple roles for platelet GPIIb/IIIa and alphavbeta3 integrins in tumor

growth, angiogenesis, and metastasis. Cancer Res 62: 2824-33.

Trusolino, L., S. Cavassa, P. Angelini, M. Ando, A. Bertotti, P. M. Comoglio &

C. Boccaccio 2000. HGF/scatter factor selectively promotes cell invasion by

increasing integrin avidity. FASEB J 14: 1629-40.

Tsai, W. H., C. W. Chang, Y. S. Lin, W. J. Chuang, J. J. Wu, C. C. Liu, P. J.

Tsai & M. T. Lin 2008. Streptococcal pyrogenic exotoxin B-induced apoptosis

in A549 cells is mediated through alpha(v)beta(3) integrin and Fas. Infect

Immun 76: 1349-57.

272
Uhm, J. H., N. P. Dooley, A. P. Kyritsis, J. S. Rao & C. L. Gladson 1999.

Vitronectin, a glioma-derived extracellular matrix protein, protects tumor cells

from apoptotic death. Clin Cancer Res 5: 1587-94.

Vachon, P. H. 2011. Integrin signaling, cell survival, and anoikis: distinctions,

differences, and differentiation. J Signal Transduct 2011: 738137.

Valgimigli, M., G. Biondi-Zoccai, M. Tebaldi, A. W. van't Hof, G. Campo, C.

Hamm, J. ten Berg, L. Bolognese, F. Saia, G. B. Danzi, C. Briguori, E.

Okmen, S. B. King, D. J. Moliterno & E. J. Topol 2010. Tirofiban as

adjunctive therapy for acute coronary syndromes and percutaneous coronary

intervention: a meta-analysis of randomized trials. Eur Heart J 31: 35-49.

van der Horst, G., C. van den Hoogen, J. T. Buijs, H. Cheung, H. Bloys, R. C.

Pelger, G. Lorenzon, B. Heckmann, J. Feyen, P. Pujuguet, R. Blanque, P.

Clement-Lacroix & G. van der Pluijm 2011. Targeting of alpha(v)-integrins in

stem/progenitor cells and supportive microenvironment impairs bone

metastasis in human prostate cancer. Neoplasia 13: 516-25.

van der Horst, G., C. van den Hoogen, J. T. Buijs, H. Cheung, J. H. M.

Feyen, P. Pujuguet, B. Heckmann, R. C. M. Pelger, P. Clement-Lacroix & G.

van der Pluijm 2010. Targeting of alpha(v) Integrins, a potential marker for

tumor-initiating cells, in Prostate Cancer and Bone Stroma Inhibits Bone

Metastasis Formation. Bone 47: S320-S320.

273
van Slooten, H., A. Schaberg, D. Smeenk & A. J. Moolenaar 1985.

Morphologic characteristics of benign and malignant adrenocortical tumors.

Cancer 55: 766-73.

Varmus, H. 2006. The new era in cancer research. Science 312: 1162-5.

Varner, J. A. & D. A. Cheresh 1996. Integrins and cancer. Curr Opin Cell Biol

8: 724-30.

Varner, J. A. & D. A. Cheresh 1996. Tumor angiogenesis and the role of

vascular cell integrin alphavbeta3. Important Adv Oncol 69-87.

Varner, J. A., D. A. Emerson & R. L. Juliano 1995. Integrin alpha 5 beta 1

expression negatively regulates cell growth: reversal by attachment to

fibronectin. Mol Biol Cell 6: 725-40.

Verbisck, N. V., E. T. Costa, F. F. Costa, F. P. Cavalher, M. D. Costa, A.

Muras, V. A. Paixao, R. Moura, M. F. Granato, D. F. Ierardi, T. Machado, F.

Melo, K. B. Ribeiro, I. W. Cunha, V. C. Lima, S. Maciel Mdo, A. L. Carvalho,

F. F. Soares, S. Zanata, M. C. Sogayar, R. Chammas & A. A. Camargo

2009. ADAM23 negatively modulates alpha(v)beta(3) integrin activation

during metastasis. Cancer Res 69: 5546-52.

274
Vicente-Manzanares, M., D. J. Webb & A. R. Horwitz 2005. Cell migration at

a glance. J Cell Sci 118: 4917-9.

Vogel, B. E., G. Tarone, F. G. Giancotti, J. Gailit & E. Ruoslahti 1990. A novel

fibronectin receptor with an unexpected subunit composition (alpha v beta 1).

J Biol Chem 265: 5934-7.

Vonlaufen, A., G. Wiedle, B. Borisch, S. Birrer, P. Luder & B. A. Imhof 2001.

Integrin alpha(v)beta(3) expression in colon carcinoma correlates with

survival. Mod Pathol 14: 1126-32.

Vorup-Jensen, T., C. Ostermeier, M. Shimaoka, U. Hommel & T. A. Springer

2003. Structure and allosteric regulation of the alpha X beta 2 integrin I

domain. Proc Natl Acad Sci U S A 100: 1873-8.

Voura, E. B., R. A. Ramjeesingh, A. M. Montgomery & C. H. Siu 2001.

Involvement of integrin alpha(v)beta(3) and cell adhesion molecule L1 in

transendothelial migration of melanoma cells. Mol Biol Cell 12: 2699-710.

Wadehra, M., A. Forbes, N. Pushkarna, L. Goodglick, L. K. Gordon, C. J.

Williams & J. Braun 2005. Epithelial membrane protein-2 regulates surface

expression of alphavbeta3 integrin in the endometrium. Dev Biol 287: 336-45.

275
Wang, H., W. Fu, J. H. Im, Z. Zhou, S. A. Santoro, V. Iyer, C. M. DiPersio, Q.

C. Yu, V. Quaranta, A. Al-Mehdi & R. J. Muschel 2004. Tumor cell

alpha3beta1 integrin and vascular laminin-5 mediate pulmonary arrest and

metastasis. J Cell Biol 164: 935-41.

Wang, R., Z. Q. Li, X. Han, B. L. Li, X. Y. Mi, L. M. Sun, M. Song, Y. C. Han,

Y. Zhao & E. H. Wang 2010. Integrin beta3 and its ligand regulate the

expression of uPA through p38 MAPK in breast cancer. APMIS 118: 909-17.

Wang, X., A. M. Ferreira, Q. Shao, D. W. Laird & M. Sandig 2005. Beta3

integrins facilitate matrix interactions during transendothelial migration of PC3

prostate tumor cells. Prostate 63: 65-80.

Weis, S. M. & D. A. Cheresh 2011. alphav Integrins in Angiogenesis and

Cancer. Cold Spring Harb Perspect Med 1: a006478.

Weis, S. M. & D. A. Cheresh 2011. Tumor angiogenesis: molecular pathways

and therapeutic targets. Nat Med 17: 1359-70.

Westermarck, J. & V. M. Kahari 1999. Regulation of matrix metalloproteinase

expression in tumor invasion. FASEB J 13: 781-92.

276
Wickham, T. J., P. Mathias, D. A. Cheresh & G. R. Nemerow 1993. Integrins

alpha v beta 3 and alpha v beta 5 promote adenovirus internalization but not

virus attachment. Cell 73: 309-19.

Williams, D. C. 1974. Factors in tumour dissemination. Proc R Soc Med 67:

847-9.

Workman, P., E. O. Aboagye, F. Balkwill, A. Balmain, G. Bruder, D. J.

Chaplin, J. A. Double, J. Everitt, D. A. Farningham, M. J. Glennie, L. R.

Kelland, V. Robinson, I. J. Stratford, G. M. Tozer, S. Watson, S. R. Wedge,

S. A. Eccles & I. Committee of the National Cancer Research 2010.

Guidelines for the welfare and use of animals in cancer research. Br J

Cancer 102: 1555-77.

Wozniak, M. A., L. Kwong, D. Chodniewicz, R. L. Klemke & P. J. Keely 2005.

R-Ras controls membrane protrusion and cell migration through the spatial

regulation of Rac and Rho. Mol Biol Cell 16: 84-96.

Wu, C., J. S. Bauer, R. L. Juliano & J. A. McDonald 1993. The alpha 5 beta 1

integrin fibronectin receptor, but not the alpha 5 cytoplasmic domain,

functions in an early and essential step in fibronectin matrix assembly. J Biol

Chem 268: 21883-8.

277
Wu, C., A. J. Fields, B. A. Kapteijn & J. A. McDonald 1995. The role of alpha

4 beta 1 integrin in cell motility and fibronectin matrix assembly. J Cell Sci

108 ( Pt 2): 821-9.

Yamada, K. M., R. Pankov & E. Cukierman 2003. Dimensions and dynamics

in integrin function. Braz J Med Biol Res 36: 959-66.

Zahn, G., K. Volk, G. P. Lewis, D. Vossmeyer, R. Stragies, J. S. Heier, P. E.

Daniel, Jr., A. P. Adamis, E. A. Chapin, S. K. Fisher, F. G. Holz, K. U. Loffler

& J. Knolle 2009. Assessment of the integrin alpha5beta1 antagonist

JSM6427 in proliferative vitreoretinopathy using in vitro assays and a rabbit

model of retinal detachment. Invest Ophthalmol Vis Sci 51: 1028-35.

Zhang, Y., M. Yang, Q. Ji, D. Fan, H. Peng, C. Yang, D. Xiong & Y. Zhou

2011. Anoikis induction and metastasis suppression by a new integrin

alphavbeta3 inhibitor in human melanoma cell line M21. Invest New Drugs

29: 666-73.

Zhao-Yang, Z., X. Ke-Sen, H. Qing-Si, N. Wei-Bo, W. Jia-Yong, M. Yue-

Tang, W. Jin-Shen, W. Guo-Qiang, Y. Guang-Yun & N. Jun 2008. Signaling

and regulatory mechanisms of integrin alphavbeta6 on the apoptosis of colon

cancer cells. Cancer Lett 266: 209-15.

278
Zhao, Y., R. Bachelier, I. Treilleux, P. Pujuguet, O. Peyruchaud, R. Baron, P.

Clement-Lacroix & P. Clezardin 2007. Tumor alphavbeta3 integrin is a

therapeutic target for breast cancer bone metastases. Cancer Res 67: 5821-

30.

Zheng, D. Q., A. S. Woodard, M. Fornaro, G. Tallini & L. R. Languino 1999.

Prostatic carcinoma cell migration via alpha(v)beta3 integrin is modulated by

a focal adhesion kinase pathway. Cancer Res 59: 1655-64.

Zhong, X. & F. J. Rescorla 2012. Cell surface adhesion molecules and

adhesion-initiated signaling: Understanding of anoikis resistance

mechanisms and therapeutic opportunities. Cell Signal 24: 393-401.

279
7.2 Appendices

7.2.1 Appendix 1

The expression of integrins subunits in tumour cell lines: PC-3, DLD-1, DU-

145 and HT-29 by PCR.

1.5 PC3 DLD-1


1.5
Relative Intensity

Relative Intensity
1 1

0.5 0.5

0 0
GAPDH αIIb α5 αv β1 β3 β5 β6 GAPDH αIIb α5 αv β1 β3 β5 β6

1.5 DU-145 HT-29


0.3
Relative Intensity

Relative Intensity

0.25
1 0.2
0.15
0.5 0.1
0.05
0 0
a2 a2b a5 aV b1 b3 b5 aV b6 b8

280
7.2.2 Appendix 2

Analysis of scratch width for each cell line in the panel.

A549 H460 MCF-7 DLD-1 HCT-116 HT-29

PC-3 DU-145 SK-MEL-2 UACC62 M14

180
160
140
Width of initial scratch

120
100
80 Width of
60 wound at To
40
20
0

Tumour cell lines

281
282

You might also like