validation and mechanism studies of novel therapeutic compounds modulating angiogenesis ·...

162
VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS By Jennifer Tat A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Physiology University of Toronto Supervisor: Dr. Xiao-Yan Wen Co-supervisor: Dr. Mingyao Liu © Copyright by Jennifer Tat 2013

Upload: others

Post on 01-Aug-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC

COMPOUNDS MODULATING ANGIOGENESIS

By Jennifer Tat

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

Department of Physiology

University of Toronto

Supervisor: Dr. Xiao-Yan Wen

Co-supervisor: Dr. Mingyao Liu

© Copyright by Jennifer Tat 2013

Page 2: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

ii

ABSTRACT

Validation and mechanism studies of novel therapeutic compounds modulating angiogenesis

Jennifer Tat

Master of Science 2013 Department of Physiology

University of Toronto

Discovering novel compounds that stimulate or abrogate angiogenesis can lead to development of new therapeutic agents that may effectively treat diseases with pathological angiogenesis. The zebrafish model allows for a whole-organism approach to drug discovery. Advantages over other animal models include small embryo size, fecundity, rapid embryonic development, optical clarity and easy accessibility of the embryos. My goal is to validate the therapeutic efficacy and identify the molecular mechanisms of action of three compounds identified from our previous chemical genetic screens. Fenretinide promoted angiogenesis in zebrafish embryos but inhibited the angiogenesis-dependent process of fin regeneration. The pro-angiogenic effects of fenretinide appear secondary to the stimulation of somitogenesis. I3M potently inhibited angiogenesis and fin regeneration, and may act partially through the notch pathway. Lastly, I validated the anti-angiogenic effect of a novel compound DHM. Comprehensively, my studies support the utility of zebrafish as a versatile tool for anti-angiogenic drug discovery.

Page 3: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

iii

ACKNOWLEDGEMENTS

I was forewarned about the isolative nature of research prior to my entry into graduate school. This is partly true, as I can account for dozens of hours of solitary data collection and analysis. However, I can also affirm that I have received a wealth of advice, support and sense of community in the course of this program, which has made the past two years fruitful as well as enjoyable.

I am grateful to my supervisor Dr. Xiao-Yan Wen for accepting me into his laboratory, for providing essential guidance during my training and for giving me opportunities to present my work and to act as a mentor to others. Additionally, I would like to express gratitude to my co-supervisor Dr. Mingyao Liu for his kind advice and critical feedback throughout my research training. I would also like to thank my committee members, Drs. Philip Marsden and Haibo Zhang, for their valued input, offered resources and words of encouragement.

I would like to recognize past and present Wen lab members - Wing, Dave, Suzan, Youdong, Dingyan, Rei, Antonio and Filip – and 5th floor LKS researchers for their technical assistance, advice and reagents. Thanks to Dave and Pamela for their critical reading of abstracts and manuscripts. Thanks to Wing, Pamela and Dingyan for their help with cell and molecular work. Thanks to Suzan for enduring RNA synthesis with me, and Youdong for his expertise and patience with the innumerable amount of microinjections we did together. There are a lot of beautiful zebrafish images throughout my thesis that I cannot entirely take credit for, as I received microscope training and general help from Youdong, Judy and Antonio. I would also like to express appreciation for the camaraderie, the laughs and the counsel of my fellow occupants in grad room 512 (A.K.A the fish bowl).

The external research community has been very open and supportive. Within the University of Toronto, I would like to thank Dr. Jason Fish for his vital tips on ISH and notch, Dr. Ian Scott for providing the Tg(flk1:EGFP) and Tg(GATA1:dsRED) strains, James (Medin lab) for transfecting our cancer cell lines, and Keisha (Gerlai lab) and Angela (Scott lab) for their tips on improving the survival of larval zebrafish. Additional thanks to Dr. Sarah Childs and Christian Lawrence (Zon lab) for providing the Tg(fli1:nEGFP)y7 and Tg(fli1:EGFP;casper) strains respectively, and Dr. Konstatin Stoletov for the MDA435 cell line and advice with xenograft fish models.

Lastly, I would like to thank my family and friends for putting up with me through my master’s degree and beyond. I am greatly indebted to their abundant support during the high and low points of the past two years.

I must also recognize the sacrifice of thousands of zebrafish for their essential contributions to my project and the field of biomedical research as a whole.

I am not certain what the future holds but aspire to continue contributing to research knowledge and its applications to improve outcomes in clinical practice.

Page 4: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

iv

TABLE OF CONTENTS

ABSTRACT ................................................................................................................................... ii

ACKNOWLEDGEMENTS ........................................................................................................ iii

TABLE OF CONTENTS ............................................................................................................ iv

LIST OF ABBREVIATIONS .................................................................................................... vii

LIST OF FIGURES .................................................................................................................... xii

LIST OF APPENDICES ........................................................................................................... xiii

CHAPTER 1 – GENERAL INTRODUCTION ......................................................................... 1

CHAPTER 2 – RATIONALE and HYPOTHESES ................................................................ 33 2.1 Rationale .......................................................................................................................................... 33 2.2 Objective and General Hypothesis ................................................................................................ 38 2.3 Specific Hypotheses ......................................................................................................................... 39

CHAPTER 3: ROLES OF 4-HYDROXYPHENYL RETINAMIDE IN ANGIOGENESIS AND ITS POTENTIAL CELLULAR AND MOLECULAR MECHANISMS .................... 40

3.1 Introduction ..................................................................................................................................... 40 3.1.1 Retinoids: Physiology and pharmaceutical development ....................................................... 40 3.1.2 Fenretinide as an anti-cancer agent ......................................................................................... 42 3.1.3 Additional applications of fenretinide .................................................................................... 44 3.1.4 Rationale ................................................................................................................................. 45

3.2 Materials and Methods ................................................................................................................... 46 3.2.1 Preparation of drugs and reagents ........................................................................................... 46 3.2.2 Zebrafish strains and husbandry ............................................................................................. 46 3.2.3 Zebrafish developmental angiogenesis assay ......................................................................... 47 3.2.4 In vivo quantification of EC number ...................................................................................... 48 3.2.5 Fin regeneration assay ............................................................................................................. 49 3.2.6 Whole-mount immunohistochemistry (IHC) .......................................................................... 50 3.2.7 Whole-mount RNA in situ hybridization (ISH) ...................................................................... 51

3.3 Results .............................................................................................................................................. 56 3.3.2 Fenretinide alters the number of endothelial cells in blood vessels ....................................... 57 3.3.3 Fenretinide inhibits fin regeneration ...................................................................................... 58 3.3.5 Fenretinide increases RAR expression and may decrease FGFR expression ........................ 62

3.4 Discussion ......................................................................................................................................... 64 3.4.1 Fenretinide promotes developmental angiogenesis but inhibits EC migration ...................... 64 3.4.2 Fenretinide inhibits the process of fin regeneration ................................................................ 65 3.4.3 Fenretinide promotes angiogenesis through somitogenesis .................................................... 66 3.4.4 Fenretinide’s effect on somitogenesis ..................................................................................... 67 3.4.5 The teratology of retinoids and fenretinide ............................................................................. 69 3.4.6 Conclusions and future directions ........................................................................................... 71

3.5 Appendix .......................................................................................................................................... 73

Page 5: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

v

CHAPTER 4: ANTI-ANGIOGENIC ACTIVITY AND POTENTIAL MOLECULAR MECHANISMS OF INDIRUBIN-3-MONOXIME ................................................................. 75

4.1 Introduction ..................................................................................................................................... 75 4.1.1 Traditional Chinese medicine and the discovery of indirubin ................................................ 75 4.1.2 Applications and properties of indirubin ................................................................................ 76 4.1.3 Molecular mechanisms of indirubins ...................................................................................... 77 4.1.4 Indirubin-3-monoxime as an anti-cancer agent ...................................................................... 79 4.1.5 Additional applications of I3M ............................................................................................... 79 4.1.6 Rationale ................................................................................................................................. 80

4.2 Materials and Methods ................................................................................................................... 81 4.2.1 Preparation of drugs and reagents ........................................................................................... 81 4.2.2 Zebrafish strains and husbandry ............................................................................................. 81 4.2.3 Cell lines ................................................................................................................................. 81 4.2.4 Zebrafish developmental angiogenesis assay ......................................................................... 81 4.2.4 In vivo quantification of EC number ...................................................................................... 81 4.2.5 Fin regeneration assay ............................................................................................................. 83 4.2.6 Embryonic tumour xenograft model ....................................................................................... 83

4.3 Results .............................................................................................................................................. 86 4.3.1 I3M decreases ISV number in developing zebrafish embryos ............................................... 86 4.3.2 I3M inhibits endothelial cells migration in vivo ..................................................................... 87 4.3.3 I3M inhibits larval and adult fin regeneration ........................................................................ 88 4.3.4 Development of zebrafish tumour angiogenesis model and preliminary test of I3M ............. 89 4.3.5 DAPT partially rescues the I3M chemical genetic phenotype ................................................ 93

4.4 Discussion ......................................................................................................................................... 95 4.4.1 I3M demonstrates specific inhibition of developmental angiogenesis ................................... 95 4.4.2 I3M inhibits fin regeneration .................................................................................................. 96 4.4.3 I3M efficacy in tumour models ............................................................................................... 97 4.4.4 I3M inhibits zebrafish ISV development partially through the notch pathway ...................... 97 4.4.5 Conclusions and future directions ......................................................................................... 100

Appendix .............................................................................................................................................. 102

CHAPTER 5: THE ROLE OF DHM IN ANGIOGENESIS ................................................ 105 5.1 Introduction ................................................................................................................................... 105

5.1.1 Rotenoids .............................................................................................................................. 105 5.1.2 The anti-cancer activities of rotenoids .................................................................................. 106 5.1.3 Dihydromunduletone ............................................................................................................ 108

5.2 Materials and Methods ................................................................................................................. 109 5.2.1 Preparation of drugs and reagents ......................................................................................... 109 5.2.2 Zebrafish strains and husbandry ........................................................................................... 109 5.2.3 Zebrafish developmental angiogenesis assay ....................................................................... 109 5.2.4 In vivo quantification of EC number .................................................................................... 109 5.2.5 Fin regeneration assay ........................................................................................................... 109

5.3 Results ............................................................................................................................................ 110 5.3.1 DHM decreases ISV number in developing zebrafish embryos ........................................... 110

Page 6: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

vi

5.3.2 DHM alters the number of endothelial cells in ISVs ............................................................ 111 5.3.3 DHM inhibits adult fin regeneration ..................................................................................... 112

5.4 Discussion ....................................................................................................................................... 114 5.4.1 DHM inhibits developmental angiogenesis .......................................................................... 114 5.4.2 DHM inhibits adult fin regeneration ..................................................................................... 114 5.4.3 DHM toxicity ........................................................................................................................ 114 5.4.4 DHM future studies ............................................................................................................... 115 5.4.5 Conclusions and future directions ......................................................................................... 116

5.5 Appendix ........................................................................................................................................ 117

CHAPTER 6 – GENERAL DISCUSSION AND FUTURE DIRECTIONS ....................... 118

CHAPTER 7 – GENERAL CONCLUSIONS ........................................................................ 122

Page 7: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

vii

LIST OF ABBREVIATIONS

4HPR 4-hydroxyphenyl retinamide (or fenretinide)

ADME absorption, distribution, metabolism and excretion

ALS amyotrophic lateral sclerosis

AMD age-related macular degeneration

Ang-1 angiopoietin-2

Ang-2 angiopoietin-2

ANOVA analysis of variance

ATP adenosine triphosphate

bFGF basic fibroblast growth factor

BSA bovine serum albumin

CAD coronary artery disease

CAM chorioallantoic membrane

CDKs cyclin-dependent kinases

cDNA complementary DNA

CF cystic fibrosis

CLSM confocal laser scanning microscopy

CNV copy number variation

COPD chronic obstructive pulmonary diseases

CRABP cellular retinoic acid binding proteins

DA dorsal aorta

DAPT γ-secretase (notch) inhibitor

DHM dihydromunduletone

DLAV dorsal longitudinal anastomatic vessel

Page 8: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

viii

Dll4 delta-like ligand 4 (notch ligand)

DMEM Dulbecco’s modified eagle medium

DMSO dimethyl sulfoxide

DNA deoxyribonucleic acid

Dpf days post fertilization

Dpa days post amputation

Dpi days post injection

DsRed red fluorescence protein

DU145 prostate cancer cell line

ECs endothelial cells

ECM extracellular matrix

EDTA ethylene diamine tetraacetic acid

EGF epidermal growth factor

eNOS endothelial nitric oxide synthase

FBS fetal bovine serum

FDA food and drug administration (American)

FGF fibroblast growth factor

FGF2 fibroblast growth factor 2

FGFR1a fibroblast growth factor receptor 1 alpha

G-CSF granulocyte colony-stimulating growth factor

GFP green fluorescence protein

GSK-3 glycogen synthase kinase-3

HIF-1 hypoxia inducible factor 1

HM hybridization mix

Hpf hours post fertilization (age of zebrafish)

Page 9: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

ix

HSCs hematopoietic stem cells

Hsp90 heat shock protein 90

HTS high-throughput system

HUVECs human umbilical vein endothelial cells

I3M indirubin-3-monoxime

IC50 half maximal inhibitory concentration

IGF-1 insulin-like growth factor-1

IHC immunohistochemistry

IL-6 interleukin-6

IL-8 interleukin-8

ISH in situ hybridization

ISVs intersegmental vessels

iTRAQ isobaric tags for relative adn absolute quantitation

LB luria broth

LOPAC library of pharmacologically active compounds

mCherry red fluorescence protein

MDA435 breast cancer cell line

MMPs matrix metalloproteinases

MO morpholino

MRI magnetic resonance imaging

MTT methylthiazol tetrazolium

MyHC myosin heavy chain

NBT-BCIP nitro blue tetrazolium with 5-bromo-4-chloro-3-indoylphosphate

nEGFP nuclear enhanced green fluorescence protein

NF-H2O nuclease-free water

Page 10: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

x

NF-κB nuclear factor kappa B

NICD notch intracellular domain

NO nitric oxide

PAD peripheral artery disease

PBS phosphate buffered saline

PBT phosphate buffer saline with Triton-X

PC-3 prostate cancer cell line

PCR polymerase chain reaction

PCV posterior cardinal vein

PDGF platelet-derived growth factor

PFA paraformaldehyde

PI propidium iodide

PlGF placental growth factor

PPC-1 prostate cancer cell line

PTK787 vatalanib (VEGFR2 inhibitor)

PTU phenylthiourea

qRT-PCR real-time polymerase chain reaction

RA retinoic acid

RAR retinoic acid receptor

RALDH retinaldehyde dehydrogenases

RXR retinoic acid X receptors

RBP retinol binding protein

RNA ribonucleic acid

ROS reactive oxygen species

RPMI Roswell Park Memorial Institute (type of cell media)

Page 11: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

xi

RTKIs receptor tyrosine kinase inhibitors

SCORE specimen in a corrected optical rotational enclosure (imaging)

SILAC stable isotope labeling by amino acids in cell culture

SIVs subintestinal vessels

SMCs smooth muscle cells

SNP single nucleotide polymorphism

SU5416 semaxanib (VEGR2 inhibitor)

SU5402 FGF inhibitor

TALEN transcription activator-like effector nuclease

TCM traditional Chinese medicine

TGFα transforming growth factor alpha

TGF-β transforming growth factor beta

TILLING targeting induced local lesions in genomes

TIMP-3 tissue inhibitor of metalloproteinase

TSP-1 thrombospondin-1

UTR untranslated region

VEGF vascular endothelial growth factor

VEGFR2 vascular endothelial growth factor receptor 2 (or Flk1)

ZFIN zebrafish model organism database

ZIRC zebrafish international resource center

Page 12: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

xii

LIST OF FIGURES

Figure 1.1.1 Cells and factors in physiological angiogenesis..........................................4

Figure 1.1.3 The angiogenic balance in pathological angiogenesis.................................6

Figure 1.3.2 Fluorescent transgenic zebrafish lines and key vessels.............................18

Figure 3.2.3 Typical protocol for zebrafish developmental angiogenesis assay............47

Figure 3.2.5 The procedure for a fin regeneration assay................................................49

Figure 3.3.1 Effect of fenretinide on ISV development in developing zebrafish..........57

Figure 3.3.2 Average cell number in ISVs of fenretinide-treated zebrafish..................58

Figure 3.3.3.1 Effect of fenretinide on larval zebrafish fin regeneration..........................59

Figure 3.3.3.2 Effect of fenretinide on adult zebrafish fin regeneration...........................60

Figure 3.3.4 Labeling of somite and vessels in 4HPR and SU5416-treated embryos.. 62

Figure 3.3.5 Preliminary ISH of RARg labeling ...........................................................63

Figure 4.1.2 The structures of indirubin and its derivatives...........................................77

Figure 4.2.4 Images generated from MATLAB cell counting program........................83

Figure 4.2.6 Workflow of a zebrafish embryonic tumour xenograft assay....................85

Figure 4.3.1 Effect of I3M on ISV development in developing zebrafish.....................86

Figure 4.3.2 Average cell number in ISVs and DA of I3M-treated zebrafish...............87

Figure 4.3.3.1 Effect of I3M on larval zebrafish fin regeneration....................................88

Figure 4.3.3.2 Effect of I3M on adult zebrafish fin regeneration.....................................89

Figure 4.3.4 Zebrafish embryonic tumour xenograft model..........................................92

Figure 4.3.5 Effect of DAPT and I3M on cell numbers in DLAVs.............................. 94

Figure 5.1.3 Chemical structure of dihydromunduletone and related rotenoids..........105

Figure 5.3.1 Effect of DHM on ISV development in developing zebrafish................111

Figure 5.3.2 Average cell number in ISVs and DA of DHM-treated zebrafish...........112

Figure 5.3.3 Effect of DHM on adult zebrafish fin regeneration.................................113

Page 13: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

xiii

LIST OF APPENDICES

Appendix 3.4.2.1 MTT assay of HUVEC cells treated with fenretinide................................73

Appendix 3.4.2.2 Fenretinide decreases zebrafish fin regeneration in adults........................73

Appendix 3.4.3.1 Albino 2.5 dpf zebrafish treated with 4HPR and 0.5% DMSO.................74

Appendix 4.3.4.1 Xenograft of MDA435 breast cancer cells..............................................102

Appendix 4.3.4.2 Xenograft of PPC-2 prostate cancer cells................................................103

Appendix 4.3.4.3 Xenograft of DU145 prostate cancer cells...............................................104

Appendix 4.4.1 Bright field image of I3M-treated zebrafish embryos.............................104

Appendix 5.4.3 Bright field images of control and DHM-treated zebrafish embryos......117

Page 14: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

1

CHAPTER 1 – GENERAL INTRODUCTION

1.1 Angiogenesis: physiology and pathology

Angiogenesis, the formation of new blood vessels from existing ones, is a physiological

process that occurs during embryonic development. Soon after vasculogenesis establishes a

primary circulatory system formed from differentiating cells of mesodermal origin, angiogenesis

commences to support the metabolic demands of the rapidly growing embryo. Angiogenesis is

mostly quiescent throughout adulthood, though it plays a significant role in female reproduction

and tissue repair. Pathological angiogenesis underlies a variety of conditions, such as obesity,

asthma, diabetes, endometriosis and macular degeneration [1]. Angiogenesis is also a hallmark

of cancer and metastasis – tumour growth cannot be supported beyond a few millimetres if the

tumour cannot establish a link to a blood supply [2] . Considering that over 500 million people

could benefit from pro- and anti-angiogenic therapy [3], a great deal of research has been

undertaken in hopes of developing therapeutic strategies to target this process.

1.1.1 Mechanisms of angiogenesis

Studies of aberrant angiogenesis in these disorders have led to an understanding of its

mechanisms, which facilitate the development of disease treatment methods. Early during

embryonic development, the primary vascular plexus forms from the differentiation of precursor

angioblast cells in the process of vasculogenesis [4]. Secondary vessels arise from these

primitive vessels via angiogenesis; sprouting, branching and recombining in a variety of ways to

form an intricate vascular network. During development, angiogenesis is stimulated to meet the

increasing metabolic demands of growing tissues. Endothelial cells (ECs) from an existing vessel

will destabilize, proliferate and migrate to form a sprout. The cells in the stalk of the sprout

elongate and join lumens to form a tube while a basement membrane reforms. Smooth muscle

cells (SMCs) or pericytes are recruited to encircle nascent ECs. Genetic studies performed in

Page 15: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

2

vitro and in vivo in mice (Mus musculus) and zebrafish (Danio rerio) have elucidated the major

players involved in the sequence of events that occur during angiogenesis [5].

The process of angiogenesis is tightly regulated by a balance between stimulatory and

inhibitory factors in the cell environment (Fig.1.1.1). Angiogenesis is activated by a shift in the

concentration of growth factors that favour the activity of angiogenesis promoters. Vascular

endothelial growth factor (VEGF) is the predominant stimulator of angiogenesis and VEGF

signalling has been identified as a rate-limiting component of the angiogenic in states of health

and disease [6].

Angiogenesis commences with the destabilization of ECs and their subsequent invasion

into the stroma of surrounding tissues. Proteolytic enzymes plasmin and matrix

metalloproteinases (MMPs) are secreted to degrade the extracellular matrix (ECM) of ECs [7].

The vessel is further destabilized by the loosening of cell-cell contacts through VEGF-mediated

dissociation of VE-cadherin and angiopoietin-2 (Ang2)-mediated detachment of SMCs and

pericytes [7]. Vascular permeability is potentiated with VEGF activation of endothelial nitric

oxide synthase (eNOS) leading to vasodilation and EC migration [7]. Plasma proteins (such as

fibrin) extravasate to form a temporary matrix for migrating ECs [7]. Integrins found on the

provisional ECM scaffold mediate the adhesion of ECs to the ECM while EC proliferation is

stimulated by growth factors such as VEGF [8], fibroblast growth factor (FGF), platelet derived

growth factor (PDGF), transforming growth factor alpha (TGFα), epidermal growth factor (EGF)

and granulocyte colony-stimulating growth factor (G-CSF) [7].

Interactions between the VEGF and notch pathways are responsible for the coordination

of vessel sprouting which involves two types of endothelial cells; tip cells and stalk cells [9]. Tip

cells express the notch ligand Dll4 and VEGF receptor 2 (VEGFR2) while another notch ligand,

Page 16: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

3

Jagged1, is expressed in the proliferating stalk cells that form the body of the elongating sprout.

Tip cells guide the migration of the developing sprout along a gradient of soluble and matrix-

bound VEGF isoforms produced by stromal cells [10]. The reproducible vascular pattern

established during development is also driven by guidance signals that attract and repel

migrating ECs to their destination: UNC-5, plexin, slit, ephrin, neuropilin, netrin, and

semaphorin [11]. In neuronal development, Eph receptors inhibit axonal growth [12]. In vascular

development, repulsive ephrin signalling demarcates artery-vein boundaries and navigates tip

cells through somite boundaries in the trunk [11]. Neuronal and vascular cell migration share

many guidance signalling molecules, which is valid considering the similarities in the circuitry of

the two systems [11].

A lumen forms from the fusion of intracellular vacuoles and extracellular channels to

produce a patent vessel that permits blood flow [13]. Stabilization of the growing vessel occurs

through TGF-β and VE-cadherin activity, which inhibits EC proliferation [14]. TGF-β also

stimulates the ECM scaffold during tube formation and VE-cadherin strengthens the junctions

between cells to maintain a stable layer of ECs that make up the vessel wall [15]. Lastly, PDGF

and Ang-1 promote the recruitment and proliferation of pericytes and SMCs respectively, which

actively maintain the structure, survival and perfusion of the mature vessel [6].

Page 17: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

4

Figure.1.1.1 A) Cells and factors involved in the process of physiological angiogenesis [7]. B) Cells and factors involved in stabilization of the nascent vessel. Reprinted by permission from Macmillan Publishers Ltd: [Nature Medicine], [6], copyright (2003).

1.1.2 Physiological angiogenesis

Blood vessel development is a major component of embryogenesis but also has critical

roles in wound healing and female reproduction.

Wound healing is dependent on neovascularisation, as vessels are required to sustain the

metabolic demands of regenerating tissue. Wound healing occurs in four stages: coagulation,

Page 18: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

5

inflammation, proliferation and remodeling [16]. Angiogenesis is initiated immediately

following tissue damage that is augmented by inflammation and suppressed once the nascent

vasculature is stabilized. Basic fibroblast growth factor (bFGF), an angiogenesis stimulator

normally sequestered inside a cell, spills out into the ECM when a cell has been damaged [17].

Platelets and thrombin, early responders to blood clotting, promote the release and expression of

angiogenic growth factors and their receptors: VEGF, PDGF, bFGF, TGF and Ang-1 [16].

Phagocytes arrive at the wound site as part of the inflammatory response, and release more

angiogenic growth factors and cytokines such as interleukin-8 (IL-8) and tumour necrosis factor

alpha (TNF-α) to activate angiogenesis [17]. Angiogenesis is prolonged by the hypoxic state of

injured tissue, leading to the production of VEGF and NO to induce EC proliferation and

enhance local blood flow respectively [18]. Once inflammation has been resolved and oxygen

levels are normal, the levels of pro-angiogenic factors decrease and angiogenesis inhibitors are

secreted [17].

Physiological angiogenesis routinely takes place in the endometrium of the uterus during

menstruation and pregnancy. The endometrium synthesizes and secretes VEGF in a spatially and

temporally regulated manner [19]. FGF, EGF, placental growth factor (PlGF) and angiopoietins

also contribute, though VEGF is considered to be the main mediator of endometrial angiogenesis

[20]. Angiogenesis is carefully modulated during the phases of endometrial growth and

regression through changes in the levels of growth factors (e.g VEGF, PlGF, etc) and hormones

(progesterone, estrogen and human chorionic gonadotropin) [19]. An imbalance of these

components can lead to abnormal bleeding, miscarriage, defective placentation and increased

risk of numerous pregnancy disorders [19, 21].

Page 19: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

6

The complexity of physiological angiogenesis is evident from the spatial and temporal

coordination of growth factors, proteins and cell types in embryogenesis, tissue repair and

reproduction. Normal angiogenesis is tightly regulated so that it only proceeds to the extent that a

tissue requires. In fact, endothelial cells are one of the longest-lived cells in the body and EC

turnover is normally on the scale of years [22]. Angiogenesis pathologies arise from the

deregulation of elements involved in the physiological angiogenesis.

1.1.3 Pathologies with insufficient angiogenesis

Abnormal angiogenesis is a signature feature in a plethora of health conditions and can be

categorized into two classes; diseases characterized by an insufficiency or excess of angiogenesis

(Fig.1.1.3).

Figure 1.1.3 The angiogenic balance in physiological angiogenesis. Pathologies characterized by excessive and insufficient angiogenesis correspond to a change in this relative amounts of angiogenic regulators [6, 23].

Page 20: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

7

Angiogenesis can be insufficient in two ways: not enough vessel growth or abnormal

vessel deterioration. Ischemic diseases such as stroke, peripheral artery and coronary artery

diseases (PAD, CAD) are characterized by the former. Ischemia occurs in tissues that receive

inadequate blood flow, most commonly by means of physical injury or a narrowing of blood

vessels due to atherosclerosis [24]. HIF-1 expression in hypoxic tissues activates VEGF

expression, initiating the angiogenic cascade. However, the HIF-VEGF response is reduced due

to a deficiency in VEGF expression and sensitivity that is likely attributed to age [25], genetics

[26] and co-morbidity with diabetes [27].

Similarly, chronic ulcers often develop from conditions such as diabetes, varicose veins,

atherosclerosis and pressure ulcers [24]. As a result, the pathophysiology of conditions with

vascular insufficiency appears to be multifactorial and complex. For instance, wound healing

angiogenesis is disrupted in foot ulcers because of a variety of physiological states associated

with diabetes: leukocyte abnormality, bacterial infection leading to a prolonged inflammatory

response, arterial insufficiency and microangiopathy caused by increased glucose uptake in ECs

[28].

The impairment of angiogenic processes contributes to the pathophysiology of a variety

of diseases. The recession of bone formation observed in osteoporosis may develop, in part, due

to an age-dependent decline in VEGF [29]. Glomerular damage in the kidney is associated with

decreased VEGF and increased expression of the angiogenic inhibitor, glycoprotein

thrombospondin-1 (TSP-1) [30]. Endothelial dysfunction in the kidney and placenta is thought to

mediate the pathophysiology of hypertension in pre-eclampsia [31]. EC apoptosis caused by

cytokines and oxidative stress contributes to the loss of alveolar cells in chronic obstructive

pulmonary diseases (COPD) like emphysema [32]. VEGF is significantly lower in patients with

Page 21: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

8

amyotrophic lateral sclerosis (ALS) [33]. Hypoxic neural tissue produces insufficient quantities

of VEGF, leading to neuron degradation [33]. Microvascular degeneration of ECs in the brain

also occurs in Alzheimer’s disease. Accumulations of amyloid-β diminishes nitric oxide levels

and EC toxicity develops into vascular abnormalities that cause neurodegeneration [34].

Therapeutic management of these disorders may be possible through the development of

interventions that induce angiogenesis, which are discussed in section 1.2. Although a lot of

progress has been made, there remains a need for the effective treatment of millions of people

affected with insufficient angiogenesis worldwide.

1.1.4 Pathologies with excessive angiogenesis

Pathological angiogenesis occurs with the loss of regulation in angiogenic homeostasis.

In disorders featuring excessive angiogenesis, the balance is shifted towards a surplus of

angiogenic activators accompanied sometimes by the suppression or reduction of angiogenic

inhibitors.

Angiogenesis is sometimes activated in tissues that do not require it. The growth of new

blood vessels in the eye occurs in a variety of ocular diseases that result in severe vision loss

[35]. Neovascular retinal vessels displace normal retinal tissue and cause leakages in the blood-

retinal barrier, which has analogous functions to the blood-brain-barrier [35]. The resultant

ischemia and inflammation leads to considerable tissue damage [35]. VEGF, PlGF, PDGF, Ang-

2, NO and IL-6 have all been implicated in the pathogenesis of neovascularisation in the retina

and choroid of the eye [36-38]. Ocular neovascularisation can also occur from the loss of

angiogenic inhibition. For instance, mutations in a tissue inhibitor of metalloproteinase (TIMP-3)

is reported in patients with choroidal neovascularisation [39].

Page 22: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

9

Other disorders in this category feature abnormalities in the progression of angiogenesis.

Teleangiectasia, a condition characterized by severe bleeding in multiple organs, is caused by

genetic mutations in receptors of TGF-β. Decreased TFG-β signalling allows for the continuous

activation of angiogenic factors that result in the development of fragile blood vessels [40].

Studies are also underway to explore the possibility of using anti-angiogenic therapy to impede

the progression of obesity and endometriosis [41, 42].

The most prominent disease in this category is cancer, one of the leading causes of death

worldwide. Consequently, tumour angiogenesis has been extensively studied to characterize its

mechanisms and to develop targeted and more effective methods of therapy. Although cancer

comes in many forms, angiogenesis is a process to which all cancer progression is dependent.

Two distinguishing features of cancer, uncontrollable cell growth and metastasis, cannot be

sustained in the absence of neovascularisation [2].

Tumour cells induce vessel growth in several ways. Oxygen and nutrient deprivation

result in the release of factors involved in angiogenic signalling; VEGF, PDGF, FGF, Ang, and

SD1α [43]. Oncogenes such as src [44] and Kras [45] upregulate growth factors like VEGF and

HIF-1. Oncogenes also enhance the production of cytokines and proteolytic enzymes [46]. Cell

irregularity and necrosis within a developing tumour is detected as a perceived wound, and an

inflammatory response is mounted. Immune cells are recruited to the site and secrete cytokines

that further amplify the angiogenic signal [43]. Endogenous inhibitors of angiogenesis, such as

TSP-1, are also suppressed in human cancers. In a mouse overexpression model, TSP-1

suppresses tumourigenesis [47] while a null model demonstrates increased vessel density and

tumour cell proliferation [48]. This correlates to the expression levels of TSP-1 in melanoma

and breast and lung carcinomas, which are inversely proportional to malignancy [49].

Page 23: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

10

The vessels derived from excessive, tumour-induced angiogenesis are immature and

poorly constructed. The characteristic morphology of tumour vasculature is leaky and

convoluted, producing inconsistent blood flow and oxygen delivery [50]. The irregularities in

perfusion produce pockets of hypoxic tissue, which results in more angiogenic signalling. The

constant presence of angiogenesis-stimulating factors in the tumour environment not only

enhances vessel growth but also prevents its stabilization. Persistently high levels of VEGF

disrupts the development of a stable vascular barrier by producing fenestrations in ECs,

loosening cell-cell adhesion, reducing pericyte coverage and activating proteases that degrade the

basement membrane [50].

1.2 Models of angiogenesis

The investigation of the physiological and pathological processes of angiogenesis and the

identification of its modulators could not have been accomplished without the development of in

vitro and in vivo bioassays.

1.2.1 In vitro assays of angiogenesis

In vitro studies are versatile and allow for the isolation and manipulation of specific

processes and molecular events. The actions of a newly identified growth factor or anti-

angiogenic agent is routinely tested in standard assays of proliferation, migration and tube

formation in cultures of human umbilical vein endothelial cells (HUVECs), though other cell

types (e.g. fibroblasts, smooth muscle cells) can also be used.

Colourimetric MTT assays are commonly employed to evaluate relative EC growth in

microwell culture plates. The tetrazolium compound MTT is reduced in metabolically active

cells to an insoluble purple dye that is measured with a spectrophotometer [51]. The readout of

Page 24: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

11

an MTT assay allows you to quickly infer dose-dependent effects of an agent on EC viability.

Alternative assays such as Ki-67 antibody labelling, 3H-thymidine or 5-bromo-2’-deoxyuridine

(BrdU) incorporation provide more accurate measurements of cell proliferation by quantifying

DNA content in cells.

Cell migration is often studied in scratch assays and Boyden chambers. Scratch assays are

simple, inexpensive assays that evaluate the migratory response stimulated by denuding a region

on a monolayer of ECs. Migration is measured by monitoring the rate at which the region is

repopulated [52]. Comparatively, Boyden chamber assays require less time and allow

investigators to clearly distinguish between cell proliferation and migration [53]. ECs are loaded

in a chamber and migrate across a porous membrane towards chemoattractants in the adjacent

compartment.

Tube formation can be modelled in vitro by culturing ECs on 2 or 3-dimensional matrices

comprised of fibrin, collagen or Matrigel, a gelatinous protein mixture secreted from mouse

sarcoma cells. The presence of the matrix scaffold, growth factors, and (sometimes) stromal cells

stimulate ECs to attach, migrate and differentiate into a capillary-like network of tubules [54].

1.2.2 Ex vivo assays of angiogenesis

The aortic ring assay is an organ-culture system widely used in angiogenesis research. An

explant cross-section of the aorta is imbedded in an extracellular substrate containing fibrin or

Matrigel. Vessels proliferate from the ring as ECs and stromal cells in the aorta respond to

dissection-induced injury and factors administered in the culture [55].

1.2.3 In vivo assays of angiogenesis

In vivo assays are used to study angiogenesis in a manner that incorporates the complex

regulatory environment of an organism. In the corneal micropocket assay, a polymer pellet

Page 25: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

12

containing the bioactive agent of interest is implanted into the cornea of rodents [56]. Corneas

are transparent and normally avascular. The development of new vessels can be easily observed

and attributed to the agent that diffuses from the pellet. The chick embryo chorioallantoic

membrane (CAM) assay works in a similar manner. Pro- or anti-angiogenic agents are applied

to the CAM and vascularisation is either stimulated or suppressed. Modifications of the CAM

assay have embedded the test agent into nylon mesh to differentiate between developmental and

agent-induced angiogenesis (which grow vertically through the mesh) [57].

Matrigel plug assays incorporate elements of micropocket and CAM assays. A

preparation of Matrigel containing the test agent is injected subcutaneously and solidifies into an

avascular plug. Matrigel itself induces vessel growth into the plug (as Matrigel contains some

growth factors), so the ability of the test agent to induce or inhibit angiogenesis must be

relatively assessed. A major drawback to this technique is caused by variation in the three-

dimensional structure of the Matrigel plug, though a Plexiglas-nylon chamber has been

developed to improve the reproducibility of the assay [58].

In vivo tumour angiogenesis is mainly studied in animal transplant models. Human or

murine tumour cell lines are grafted into immune-suppressed rodents, usually mice or rats

(Rattus norvegicus). The host animal’s entire angiogenic machinery (i.e. extracellular matrix,

growth factors, stromal cells, immune and inflammatory components) is available to interact

with the tumour. Parameters such as microvascular density and tumour size are used to measure

the effect of the test agent on tumour-induced angiogenesis. Xenograft models can incorporate

transgenic animals or cell lines to study specific processes. For example, luminescent OKD48

transgenic mice can be employed to assess the oxidative stress response in vivo [59]. Anti-

angiogenic drugs are routinely assessed preclinically in xenograft models. However, the efficacy

Page 26: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

13

of xenograft models is challenged by “species differences and inherent circumstances” that are

responsible in part, for the difficulties in translating preclinical benefits to patients [55].

Organism-scale angiogenesis can also be studied in zebrafish embryos and tadpoles

(Xenopus laevis), though the former is predominantly used. Test agents can be added directly

into the water or injected into the yolk sac in the zebrafish developmental angiogenesis assay.

The optical transparency of the embryos facilitates visual analysis of the progression of

angiogenesis and potential toxicities. Transgenic zebrafish with fluorescently-labelled ECs are

used in high-throughput screening for angiogenic modulators [60]. Models of tumour and retinal

angiogenesis have also been developed in zebrafish (see section 1.3) [61, 62]. Additionally, a

variety of transgenic or gene knockout strains and technologies such as morpholino gene

knockdown are available for mechanism studies.

Mouse and other mammalian animal models are useful in validating compounds that

modulate angiogenesis. However, because of their size and relatively small number of offspring,

mammals are unfavourable to use in large-scale screens. Zebrafish has recently emerged as an

advanced in vivo model organism for high throughput screens (HTS) for angiogenesis drug

discovery.

1.3 Zebrafish angiogenesis and cancer models in drug discovery1

1.3.1 Zebrafish as an advanced model for chemical genetic screens

The conventional process of drug development is time-consuming and expensive. The

low success rate of traditional cell-based screens demonstrates a need for whole-organism

screening strategies. Zebrafish combine the biological complexity of a whole animal model with

1 Portions of this section have been published in a review that is currently in press. Tat, J., Liu, M., Wen, XY. Zebrafish cancer and metastasis models for in vivo drug discovery. Drug Discovery Today Technologies. 2012. Figures and excerpts reprinted with permission.

Page 27: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

14

the versatility and throughput of in vitro models. Zebrafish demonstrate many conserved

vertebrate characteristics, sharing genes and physiology with mammals despite the 400 million

years that set us apart in evolution [63]. In fact, many genes and regulatory processes involved in

angiogenesis appear to be conserved in zebrafish and mammals [64]. Yet, zebrafish retain many

invertebrate features that facilitate experimental study: small size, fecundity (high number of

offspring), oviparity (eggs hatch outside of the mother), rapid development (major organs form

within three days of hatching) and relatively low maintenance costs. Hundreds of optically

transparent embryos can be generated per mating pair and development occurs externally so that

embryos can be easily (noninvasively) observed and manipulated (using techniques such as

microinjection, cell transplantation). Small molecules can be added directly into the fish water of

multiwell plates and absorbed via diffusion, while proteins can be injected directly into the yolk

sac. The quantity of the tested drug, and technical expertise required for its evaluation are much

less than that of an equivalent study performed in mice [65]. Chemical libraries can be applied to

a large number of embryos to systematically screen for a phenotype of interest. Employing a

phenotype-based approach permits the identification of new drug candidates for diseases in

which therapeutic targets are not known.

Zebrafish are highly accessible to genetic manipulation, and a collection of genetic tools

has been developed as a consequence. Transgenesis has been particularly useful in generating

reporter lines and disease models that are used in screening. Transgenic gene overexpression,

TALEN gene knockout and morpholino knockdown technologies permits in vivo evaluation of

gene functions in specific physiological processes, which are helpful in characterizing the

mechanisms of action and target of a compound [66].

Page 28: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

15

The zebrafish genome has almost been completely sequenced by the Sanger Center and

has been extensively annotated by the Trans-National Institutes of Health Zebrafish Genome

Initiative. There is high genetic variability, in the form of copy number variants (CNVs) and

single nucleotide polymorphisms (SNPs) in zebrafish [67]. This may be advantageous for drug

screening, as genetic factors are known to contribute to phenotypic differences. In a behavioural

zebrafish screen, the basal locomotor activity of embryos and their locomotor response to alcohol

were significantly different depending on the genetic background (strain) of the fish [68].

Conventional preclinical animal models are often inbred and may not be able to predict drug

responses that occur in a more genetically heterogeneous human population [69]. Zebrafish can

be advantageously employed in screening, since they are observed to be four times more

genetically diverse than mice [67]. However, the genetic variation in zebrafish actually exceeds

the percentage observed in humans as well [67]. While genetic diversity may be desirable in

regards to screening, it has the potential to contribute to phenotypic differences in screens

employing disease models [67]. Fortunately, highly homogeneous zebrafish (such as the inbred

India strain) [70] and genetically uniform zebrafish lines [71] are available for these applications.

Thousands of mutant lines, generated through genome-wide mutagenesis such as

chemical, targeting induced local lesions in genomes (TILLING) [72], and insertional gene

trapping, have been catalogued and are readily accessible on online databases such as Sanger,

ZFIN, ZIRC and zfishbook.org. Zebrafish gene expression can also be analyzed using tools such

as real-time polymerase chain reaction (qRT-PCR) [73] and in situ hybridization [74]. RNA

extraction, followed by analysis with DNA microarray is available for gene expression profiling

[75].

Page 29: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

16

Additionally, assessments of drug pharmacology can be determined at an earlier stage of

drug development. Within a whole-organism system, the effects of a drug and its metabolites can

be simultaneously evaluated. Assays of teratogenicity, cardiotoxicity and neuro-sensory organ

toxicity have been also developed in zebrafish and have demonstrated good predictive ability of

compound toxicology in humans [76]. While zebrafish pharmacokinetics for absorption,

distribution, metabolism and excretion (ADME) are not well characterized, efforts are currently

underway to assess the similarities and differences in drug metabolism and bioactivation between

animal models [77]. Preliminary findings have demonstrated shared gene expression and liver

enzyme activity between human and zebrafish in response to a panel of drug compounds,

suggesting conservation in drug pharmacodynamics [78, 79].

In addition to its utility in screening, zebrafish chemical genetics can also help analyze

the target and mechanism of action of a test compound through chemical rescue experiments,

knockdowns and phenotype comparisons.

Zebrafish are uniquely qualified for use in large-scale screening and the availability of

numerous genetic tools facilitates the detailed study of candidate drug effects in vivo, prior to

preclinical testing in mammalian models. The zebrafish chemical genetic screen is a time and

cost-effective method for direct in vivo drug discovery and serves as an advanced system in drug

development.

1.3.2 Drug Mechanisms and drug target deconvolution in zebrafish

Computational and experimental methods can be employed to identify the mechanisms of

action of a compound. The computational approach uses similarities between phenotypes or

chemical structures of compounds [80]. In a proof of concept study, Chan et al. studied the

signalling pathways disrupted by VEGFR2 inhibitor vatalanib (PTK787) by performing rescue

Page 30: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

17

experiments with AKT/PKB and eNOS mRNA injections [81]. Experimental comparisons of the

phenotypes of an unknown drug with compounds of specific, known targets (like PTK787) allow

investigators to discern the implicated pathways of drug action. Subsequent rescue experiments

of specific pathway members, alongside in vitro assays, can be performed in zebrafish to identify

the effected pathways. Genistein, a phytoestrogen isolated from soybeans, potently inhibits

subintetinal vessel (SIV) formation in a manner that phenocopies VEGFR2 inhibitors PTK787

and semaxanib (SU5416) [82]. This led the authors to state that genistein was likely to be

exerting its inhibitory angiogenic effects through VEGF signalling [82].

Other target deconvolution techniques include affinity-based approaches, which use

mammalian or zebrafish protein lysates to evaluate potential binding partners of a small-

molecule [83]. PPA was identified as a stimulator of pigmentation from a zebrafish embryo

screen of a triazine small-molecule library [84]. Affinity chromatography was able to identify its

target as the mitochondrial F1F0-ATP synthase [84]. Proteomic technologies such as mass

spectrometric-based methods SILAC (stable isotope labelling) [85] and iTRAQ (tags of equal

mass) [86] are capable of simultaneously identifying all the binding partners of a compound,

though additional experimental verification of the candidate targets is required.

1.3.3 Zebrafish angiogenesis: process and models

Zebrafish angiogenesis occurs shortly after vasculogenesis, in a manner that is conserved

in most vertebrates. Intersegmental vessels (ISVs) sprout dorsally from the dorsal aorta (DA) at

the boundary of each somite and join to form the dorsal longitudinal anastomatic vessel (DLAV)

above the neural tube along the trunk of the embryo [64]. The temporal and spatial expression of

zebrafish homologs for essential molecules involved in the process of angiogenesis (VEGF,

angiopoietins, ephrins) is comparable to their mammalian counterparts [87-89].

Page 31: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

18

Figure 1.3.2. Three fluorescent transgenic zebrafish lines with key vessels labelled. [A] A 2.5 dpf Tg(flk1:EGFP) embryo with green fluorescence expressed in the cytoplasm of ECs. [B] A 2.5 dpf Tg(fli1:nEGFP) embryo expressing green fluorescence in the nuclei of ECs. [C] A 6 dpf Tg(GATA1:dsRed) embryo with red fluorescence in erythrocytes.

The generation of transgenic zebrafish lines with fluorescent vasculature has enabled

real-time study of angiogenesis within an intact organism throughout its vascular development

(Fig. 1.3.2). Enhanced green fluorescent protein (EGFP) is expressed under the control of

promoters that are specifically expressed in ECs (e.g. Fli, Flk, mTie2) enabling visualization of

the entire developing vasculature under fluorescent microscopy [90-92]. The number of ECs in

specific vessels can be determined by employing the Tg(Fli:nEGFP)y7 line, which has nuclear-

specific expression of EGFP [93]. The quantification of EC number allows investigators to

resolve the specific process of angiogenesis being affected by a drug. The functionality of the

blood vessels can also be evaluated in real-time using a transgenic line with fluorescence

expressed under the GATA1 promoter, which is expressed in circulating erythrocytes.

Alternatively, blood flow can be assessed using microangiography, with the injection of

fluorescent labeling agents such as quantum dots [94]. The effects of a drug on vessel patterning

Page 32: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

19

and blood flow can be tested in isolation, or together, in the double transgenic line

Tg(Fli:EGFP;GATA1-dsRED) [95]. The zebrafish developmental angiogenesis assay is simpler,

faster and easier to quantify than CAM, corneal micropocket or other in vivo angiogenesis

assays. Consequently, these embryos have been used in chemical screens for compounds

modulating angiogenesis [96].

In addition to optical transparency and accessibility, zebrafish have an advantage over

small rodent models, in that the heart rate of zebrafish embryos is closer to humans than the high

heart rate of mice [97]. Zebrafish embryos also have the remarkable capacity to survive for

several days in the absence of heart function or circulation [97]. These features have bolstered

the vigorous and detailed study of zebrafish cardiovascular development. For example, confocal

time-lapse microscopy of double transgenic zebrafish embryos permitted in vivo monitoring of

ECs during stages of angiogenesis in a study conducted by Jakobsson et al. The investigators

were able to observe ECs dynamically competing for the role of a tip cell during angiogenic

sprouting, in a process that is governed by VEGF-notch signalling [98]. Zebrafish research has

made substantial contributions to cellular, molecular and genetic studies of vascular development

in vertebrates, which has been reviewed elsewhere [97].

States of pathological angiogenesis can also be studied in zebrafish. Zebrafish can be

raised in fish water that is perfused with nitrogen gas, creating systemic hypoxia in tissues [99].

This has led to the development of an embryonic metastasis model [100] and an adult

retinopathy model [62]. Myocardial infarction (MI) can also be studied in adult zebrafish. MI

can be induced by cryoinjury and subsequent cardiac regeneration can be analyzed with

histology, immunostaining and in situ hybridization (ISH) [101]

Page 33: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

20

Angiogenesis can also be studied during zebrafish fin regeneration. Zebrafish have much

greater regenerative capacity than humans, but the underlying mechanisms (e.g.

functional role of Pu.1 gene) of tissue regeneration appear to be conserved [102]. In this

assay, half of the caudal fin is amputated and allowed to grow back. Test agents are

applied to the fish water during the allotted time for regeneration (3 days). The

percentage of fin regeneration in adults is an indirect measure of physiological

angiogenesis, as only 1 mm of tissue can be regenerated with the inhibition of

angiogenesis [103]. Chemical screening for fin regeneration can be performed in larval

[102] and adult zebrafish [104]. This assay and the developmental angiogenesis assay are

highly flexible, as there are a variety of options to select for the phenotypic readout.

Transgenic lines or immunostaining can be used to label specific cell types and

morpholinos or chemicals can be applied to block the function of specific genes and

pathways. The major disadvantages to fin regeneration models is that they are not very

amenable to large-scale screening and must be validated with additional studies, as

angiogenesis cannot be differentiated from inhibition of other parts of the regenerative

process.

1.3.4 Zebrafish cancer models

Zebrafish has also emerged as a promising experimental system for modelling human

cancers, through genetic manipulation or cell transplantation (refer to Liu and Leach 2011 for a

thorough review on zebrafish models of cancer [105]).

Transgenic and mutant zebrafish lines have been developed to mimic many key features

of tumourigenesis. A Gal4-UAS system produces HRAS oncogene overexpression in

melanocytes and faithfully reproduces phenotypes of human melanoma in zebrafish larvae [106].

Page 34: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

21

Nguyen et al. have developed a system to induce and track the growth of fluorescent-labelled,

RAS-overexpressing tumours in the livers of larval zebrafish [107]. Mutant lines are also

available to model cancers with loss-of-function mutations. TP53, is a tumour suppressor gene

that is frequently mutated in the majority of human cancers [108]. A zebrafish line containing a

mutation in tp53 exhibits abnormal apoptosis and cell-cycle phenotypes and develops neural

sheath tumours at 8-9 months of age [109]. Conducting compound screens on mutant lines can

lead to the identification of therapeutic agents that can restore gene function and prevent

subsequent tumourigenesis [107].

Cell transplantation models have been used to study tumour angiogenesis and metastasis

in zebrafish. An embryonic xenograft model has been developed to investigate tumour

angiogenesis in vivo. The injection of cancer cells into the yolk sac of 2-day-old embryos

stimulates vessel growth from the subintestinal vessels (SIVs). This process is monitored by

fluorescence microscopy, since vessels and tumour cells are fluorescently labelled. Tumour

angiogenesis can be abrogated by the application of chemical inhibitors [61].

Injected cancer cells can be genetically modified to overexpress FGF2 or TGF-β to

stimulate angiogenic and metastatic behaviour [110, 111]. Metastasis and cell invasion can also

be studied by injecting cancer cells overexpressing the pro-metastatic gene twist directly into the

circulation of embryos [112]. The embryo strain and its environmental conditions can also be

altered. Vasculature-labelled transgenic embryos are commonly used in these studies, and they

can also be treated with morpholinos to inhibit specific pathways. Nicoli et al. treated transgenic

embryos with a morpholino targeting VE-cadherin and observed a decrease in tumour vascularity

[110]. Hypoxia-induced tumour activity can also be modelled by incubating xenografted

embryos in normoxic or hypoxic water. Under hypoxic conditions, there is increased

Page 35: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

22

neovascularisation, and tumour cells disseminate and invade neighbouring tissues [113]. The

hypoxic system models the early stages of metastasis and it would be valuable to study and

develop therapies for this particular process.

The strength and weakness of embryo xenograft experiments is its duration of study,

which is limited to one week post fertilization. Embryos younger than a week old tolerate

xenograft transplants due to the immaturity of the immune system [114]. However, adult

zebrafish permit long-term evaluation of tumour development. Immunocompromised zebrafish

lines are not available at present, though immunosuppression is possible through sub-lethal doses

of radiation [115].

The prevalent use of zebrafish embryos in research faces some criticism. Embryo models

may depict processes that are specific to developmental biology, which may not adequately

translate to adult physiology. Juvenile and adult fish are not routinely used for practical reasons:

tissue opacity, longer experimental timeline, and the maturity of their nervous and immune

system hinder experimental study. A number of techniques can be employed to overcome these

issues, to improve the use of older zebrafish in cardiovascular research. Pigment formation can

be pharmacologically inhibited with propylthiouracil, though transparency can only be

maintained for several weeks [99]. Additionally, a doubly mutant zebrafish line (casper) has

been created to prolong tissue transparency into adulthood [116]. In zebrafish tumour xenograft

models, this has allowed for the in vivo assessment of tumour grafts for up to 5 weeks post-

transplant [116].

Zebrafish offer a diversity of options for modeling cancers. Different stages of cancer

progression can be recapitulated in transgenic and xenograft zebrafish. Genetically engineered

Page 36: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

23

zebrafish also depict disease- and pathway-specific models of cancer. Zebrafish have great

potential in contributing to the advancement of cancer research and therapeutics.

1.3.5 Technological advances in zebrafish drug discovery

Zebrafish embryonic screens can greatly streamline the process of drug discovery by

automating the laborious phases of drug screening; sorting, sample processing, drug dosing,

image acquisition, analysis and interpretation. The COPAS XL (Union Biometrica) is a large

particle flow cytometer that is capable of sorting embryos and hatchlings based on optical light

and fluorescence (up to 3 fluorophores at once) and dispensing them into multiwell plates [117].

This device can significantly accelerate embryo selection and sorting for drug treatment by

identifying embryos positive for fluorescence. Automating slow and tedious tasks like drug

dosing and microinjection can further increase throughput. Liquid handling robots such as

Sciclone G3 (Caliper Life Sciences) can dispense reagents and drug compounds in series or in

parallel into multiwell plates [118]. With automated batch microinjection, up to 15 embryos can

be injected per minute with genetic material such as MO [119]. Although automated

microinjection is not substantially faster than manual microinjection, it reduces the variability

between injections and errors due to operator fatigue [119].

The transparency and small size of zebrafish embryos allows ease of light-based imaging.

Stereo- and confocal microscopies have been widely used in zebrafish studies. Confocal

microscopy is capable of high resolution cellular imaging with well-developed software analysis

tools, but its application is limited by tissue penetration and the transparency and size of the

zebrafish being analyzed [120]. Most zebrafish screens are performed in multiwell plates, and

phenotypic analysis within the plates is possible using Confocal Laser Scanning Microscopy

(CLSM), such as the ImageXpress Ultra (Molecular Devices), with point-by-point image

Page 37: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

24

acquisition allowing for three-dimensional reconstructions. MicroMRI has been applied to detect

and characterize melanomas in adult zebrafish [121], and microscopic ultrasound has been

utilized to assess liver tumours in adult zebrafish in response to chemotherapeutic treatment

[122].

A number of methods have been devised to expedite image processing outside of

multiwell plates to overcome the problems caused by the large working distance and random

movement and orientation of zebrafish [117]. Capillaries [123], agarose-coated plates [124],

round-bottom plates (Corning COSTAR) and rectangular microplates with prisms (Physical

Sciences Inc) are several strategies that have been conceived to manage embryo orientation for

quick image capture. High-throughput histology is possible for larval and adult zebrafish using

several methods to accelerate sample handling: agarose arrays, automated tissue processors,

rotary microtomes and automated slide stainers [125]. An automated imaging system developed

by Gehrig et al. combines embryo recognition software with a high content microscope such that

fluorescent gene expression patterns in up to 2,000 embryos may be acquired within 4 hours

[124]. Analysis programs have been built to handle the quantity of data produced by rapid image

acquisition technologies while also reducing the burden of visual scoring and eliminating

observation bias [126]. Image analysis software such as Cognitive Network Technology

(Definiens) and MetaMorph software application modules (Molecular Devices) can be custom-

designed to detect and quantify specific structures such as intersegmental vessel number in

Tg(fli1:EGFP) zebrafish [126]. The development of these technologies has greatly increased the

efficiency of screening. However, in most zebrafish screenings reported thus far, automation is

not continuous and embryos must be manually manipulated at several steps (semi-automation).

Although zebrafish are an emerging model organism in biomedical research, it is a

Page 38: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

25

powerful system for in vivo drug discovery that is being employed in many disciplines. The

increasing popularity of zebrafish has led to the development of numerous technologies to

improve handling, imaging and processing. Techniques for molecular, histological, behavioural

and genetic analyses are improving, and as a result the body of literature continues to grow. The

utility and versatility of zebrafish genetic tools and disease models will allow researchers to

continue to make waves in biomedical research and accelerate the process of drug development.

1.4 Angiogenesis-targeted therapies

1.4.1 Pro-angiogenic therapies

Traditionally, the clinical interventions for disorders with impaired circulation have

largely been surgical in nature. Clinical treatment of ischemic diseases involve lifestyle changes,

management of co-morbidities and macrovascular interventions such as surgical

revascularization, angioplasty and amputation [127]. Chronic wounds are managed similarly,

with the removal of necrotic tissue, surgical bypass, subcutaneous angioplasty and amputation

[16]. Microvascular therapies have been recently developed to directly promote angiogenesis in

these tissues.

Therapeutic angiogenesis induces the growth of blood vessels, to restore blood perfusion

and enhance tissue repair in disorders that feature a deficiency in angiogenesis [128].

Therapeutic induction of angiogenesis is achieved through the application of growth factors and

pharmacological agents. Cell-based therapies [129, 130], tissue engineered products [131] and

mechanochemical technologies (such as negative pressure [132], low-frequency ultrasound [133]

and hyperbaric oxygen systems [134]) are alternative strategies to stimulate angiogenesis.

However, I will focus on the development of bioactive agents.

Page 39: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

26

Several of these therapies are based on the growth factors FGF, VEGF and PDGF, and

are administered locally as protein-laden formulations [135-137] or incorporated into vectors for

gene therapy delivery [138-140]. Recombinant PDGF (Becaplermin gel) is the only pro-

angiogenic agent approved for clinical use [136]. It has significantly improved the healing rates

of chronic diabetic ulcers [136] and has been successfully used off-label for great variety of

wounds [24]. Angiogenesis stimulation has also been achieved with VEGF and FGF gene

therapy in ischemic heart disease [141]. While gene therapy offers longer term treatment of

ischemia, it has not demonstrated the level of efficacy in clinical trials necessary for approved

usage [142].

Pharmacological agents in the form of peptides [143] and small molecules [144] are also

in development as stimulators of angiogenesis. These agents act as antagonists or agonists,

activating or inhibiting components of the angiogenic cascade to promote angiogenesis. One

method used by the peptide PR39 and synthetic compound TM6008 activates the HIF pathway

through inhibition of HIF degradation, to induce angiogenesis through hypoxic signalling [145].

Applications of therapeutic angiogenesis have mostly been studied in chronic wounds

and ischemic diseases in the heart and limbs. The efficacy of these agents is currently being

evaluated for relevance in other conditions with insufficient or faulty angiogenesis such as

infertility [19], neurodegeneration [146] and sepsis [147]. Substantial work has also been

conducted to stimulate neovascularisation in tissue engineering [148].

1.4.2 Anti-angiogenic therapy

Traditionally, general strategies have been employed to destroy defective tissues in

diseases with excessive angiogenesis. Routine cancer treatment involves chemotherapy, radiation

or surgical excision of a cancerous mass. Photodynamic therapy (PDT) uses a similar but more

Page 40: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

27

refined approach to treat ocular tumours and neovascularisation. Photosensitized agents are

systemically injected and sequester in abnormal ocular vessels until a laser activates them,

resulting in local damage that seals off the vessel. [149]. The side effects and shortcomings of

these strategies have produced a great need for treatment modalities that are more specific, safe

and effective. In the past few decades, a multitude of agents have been developed to specifically

target the molecular mechanisms of the aberrant angiogenesis that characterizes these conditions

[150]. Anti-angiogenesis strategies are employed to inhibit or normalize abnormal angiogenesis.

Inhibitory agents are available in the following varieties: antibodies, growth factors, peptides and

small molecule compounds. The efficacy of these therapies has primarily been studied in ocular

and tumour angiogenesis.

VEGF has been well characterized as a critical component of the angiogenesis cascade in

normal and pathological tissues. Consequently, many anti-angiogenic strategies have been

developed to target VEGF signalling. The first anti-angiogenic agent to be approved for clinical

usage, bevacizumab, is a monoclonal antibody that targets and neutralizes VEGF protein.

Bevacizumab demonstrated improved patient survival in a number of cancers when used in

conjunction with chemotherapy [151]. A high affinity fusion protein for VEGF, aflibercept

(VEGF-Trap), is currently in Phase III clinical trials for colorectal cancer, retinal vein occlusions

and diabetic macular edema [152]. The aptamer (synthetic oligonucleotide), pegaptanib was the

first anti-VEGF agent to be approved for clinical use in age-related macular degeneration (AMD)

[153]. The long-term application of both protein and small-molecule VEGF-based therapies

revealed that positive responses were transient and tumours would develop resistance and

continue to grow vessels even with the loss of VEGF signalling [151].

Page 41: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

28

While these agents target a single growth factor and its downstream effectors, second-

generation agents, are capable of targeting multiple angiogenic signalling pathways at once.

Some investigators have referred to this strategy as “magic shrapnel” in reference to the “magic

bullet” of single-target therapies [154]. RTKIs are a class of small molecule compounds that are

able to block signalling via receptor tyrosine kinases. RTKIs target receptors of VEGF, FGF,

PDGF and some oncogenes. Sunitinib is a RTKI that blocks VEGFR2, PDGFR-α and β, raf

kinase, FLT2 and c-Kit [155]. Another RTKI, sorafenib blocks VEGFR2, PDGF-β, FLT3 and c-

Kit [156]. Sunitinib and sorafenib have both been approved for clinical use for first-line

monotherapy of kidney and liver cancer [155-157]. However, their potency and systemic

administration (as oral agents) can cause serious toxicities related to the disruption vascular

homeostasis. A fraction of patients treated with bevacizumab, sunitinib or sorafenib have

reported higher incidences of bleeding, thrombotic events, hypertension, edema and delayed

post-operative wound healing [158].

Broad-spectrum angiogenesis agents, like angiostatin and endostatin, are endogenous

protein fragments produced by proteolytic processing [159]. Primary tumours are able to keep

distal metastases dormant by blocking angiogenesis and tumour growth through the action of

endogenous circulating inhibitors [160, 161]. Angiostatin and endostatin potently suppresses

angiogenesis and tumour growth in mice without detectable toxicity [162, 163]. Endostar,

recombinant human endostatin, is currently approved in China for the first-line treatment of lung,

gastric and colorectal cancer [164]. Although the mechanism of action is not known, endostatin

prevents ECs from responding to angiogenic signals by downregulating VEGF and FGF

signalling, EGFR, MMPs, c-myc, HIF-1α and simultaneously upregulating anti-angiogenic

factors such as TSP-1 and maspin [165, 166]. The range of this promising class of inhibitors has

Page 42: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

29

allowed it to succeed where other anti-angiogenic therapies have failed. Animals treated with

endostatin do not appear to develop drug-induced resistance [167] and Endostar treatment

demonstrates the potential to reverse multi-drug resistance in a pilot study in patients with

colorectal and gastric cancer [164]. However, there are practical limitations to protein-based

therapy. Angiostatin and endostain are physically difficult to produce in active form, require

high-dose local administration and have short half-lives [168].

Anti-angiogenic agents have also been developed to block specific steps in the

angiogenesis process. Vitaxin is an antibody directed against αvβ3 integrin, which is involved in

cell adhesion and exclusively found in activated ECs [169]. Phase I clinical trials in patients with

advanced cancer determined that while Vitaxin was unable to cause tumours to recede, it did

delay tumour progression [170]. Broad and selective MMP inhibitors are also available to

impede proteolysis in the process of EC migration and tumour cell invasion and metastasis.

Clinical trials show increased survival in long-term use of marimastat [171], a broad spectrum

MMP inhibitor, though treatment benefits were offset by musculoskeletal toxicities [172].

1.4.3 Limitations and future directions in angiogenesis-targeting therapies

Presently, there are eight anti-angiogenic drugs approved for cancer therapy [150], three

approved for the treatment of AMD [173], and one pro-angiogenic drug approved for diabetic

ulcers [136]. Several dozen angiogenesis-targeting agents are making their way through the drug

development pipeline. Targeting angiogenesis is a rational approach to treating cancer, ocular

and ischemic diseases. However, a substantial body of clinical research have accumulated

evidence that the potent benefits observed in preclinical models do not translate to same level of

efficacy in the clinic. Not all cancer patients respond to anti-angiogenic therapy and resistance

quickly develops in those who do [174]. The effectiveness of bevacizumab has been modest; it

Page 43: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

30

prolongs life on the order of months and that is only when it is used in combination with

chemotherapy [175]. Pro-angiogenic therapy has not fared any better, with no clinical benefit

observed in double-blind randomized placebo-controlled trials in ischemic cardiovascular

disease [176]. Assuming the validity of the premise, there are a number of issues that are

contributing to the overall lack of efficacy of these agents: associated side effects, drug

resistance, and the timing and specificity of the drug.

One of the major concerns that must be considered for the next generation of

angiogenesis-modulating therapies is drug resistance. So far, investigators have reasoned, “all

that needs to be done is initiate [or limit] the process of vascular growth and nature will take its

course. Thus far nature has perversely refused to cooperate [177].” It has become clear that a

systems biology approach must be considered. The short-lived efficacies of VEGF inhibitors

suggest that resistance stems from the ability of tumours to use alternative pathways of

angiogenesis such as the FGF, PDGF, PIGF, notch, TIE2/angiopoietin, and integrin pathways

[178, 179]. Breast cancer, and the majority of human cancers, has demonstrated the upregulation

of 5-6 proangiogenic proteins [180].

One method to overcome angiogenic redundancy is to employ broad-spectrum

angiogenesis inhibitors like angiostatin or endostatin. Another way to overpower built-in

compensatory mechanisms is to combine the use of multiple inhibitors to block many pathways

at once or in sequence [178]. Combinatorial strategies should be more effective in countering the

interacting effects of a numerous cell types and signalling pathways. Preclinical studies have

recently demonstrated that combination therapy may be more effective than monotherapy.

Combined delivery of VEGF and Ang-1 was able to increase the capillary density of a hindlimb

ischemia model greater than either agent alone [181]. Co-administration of systemic (angiostatin)

Page 44: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

31

and local (bevacizumab) anti-angiogenic inhibitors has demonstrated additive effects in animal

models, strongly inhibiting angiogenesis while reducing bevacizumab-induced tumour invasion

[182]. Another potential benefit to employing combination therapy is the ability to reduce the

dosage of agents, which are known to produce side effects. Therefore, identification of novel

compounds targeting various pathways of angiogenesis is very important in both pro- and anti-

angiogenesis therapies.

Some pharmacologically active compounds could produce effects comparable to

combinatorial therapy. Established drugs such as celecoxib (Celebrex) [183] and rosiglitazone

[184] are able to upregulate endogenous anti-angiogenesis proteins (such as endostatin and TSP-

1 respectively) and are currently undergoing clinical trials for the treatment of cancer [185, 186].

Finding new functions for old drugs expedites the process of drug development since their

pharmacology and toxicology profiles are already known. The next generation of angiogenesis

inhibitors and stimulators could come from the identification of new small molecules that are

able to exert similar effects.

The efficacy of a drug may depend on the time at which it is administered. MMP and

VEGF inhibitors and endostatin are most successful at treating early-stage cancers due to their

roles in switching off tumour angiogenesis in the initial stages of tumourigenesis [22, 171, 187].

Comparably, an inhibitor of EC growth and SMC migration TNP470 is more suited for the

treatment of late-stage tumours [188]. Multi-target inhibitors are necessary to override the

numerous pathways activated by aggressive, end-stage tumours. Our knowledge of the

physiology of tumour development and angiogenesis should be considered when selecting and

applying appropriate anti-angiogenic agents for therapy.

Page 45: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

32

It is also important to conduct more detailed studies of pathological angiogenesis. The

identification of therapeutic targets will enable the development of angiogenesis therapies that

target specific components of the problematic physiology. The stage, treatment history and

genotype of a tumour may eschew molecular resistance to certain treatments [189]. Ideally, one

would employ an inhibitor specific to the angiogenic profile of a patient’s tumour for optimized,

personalized treatment, though the lack of biomarkers and patient screening limits this strategy at

present [179]. The discovery of novel angiogenesis modulators and thorough investigation of

their molecular mechanisms of action will greatly contribute to efforts in improving treatment

outcomes and future applications in general and personalized medicine.

As research in angiogenesis therapies is still in its infancy, it is imperative that we

continue to find ways to improve its study and clinical application. The zebrafish model system

presents an opportunity to enhance drug discovery in the field of angiogenesis. Judah Folkman,

the researcher credited as the father of angiogenesis therapy, once asserted that the outcomes of

angiogenesis therapy would one day reclassify cancer as a “chronic manageable disease” [190].

Therapeutic modulation of angiogenesis could lead to momentous changes in the management of

an expansive list of diseases that significantly affect human health worldwide.

Page 46: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

33

CHAPTER 2 – RATIONALE AND HYPOTHESES

2.1 Rationale

2.1.1 The need for new drugs targeting angiogenesis

Pathological angiogenesis underlies a variety of conditions, such as obesity, asthma,

diabetes, endometriosis, macular degeneration and cancer. The critical role of aberrant

angiogenesis in these disorders has led to a detailed investigation of its mechanisms to facilitate

the development of treatments that stimulate or abrogate it.

Angiogenesis is tightly regulated by a balance of pro- and anti-angiogenic factors in the

cell environment. Many therapeutic strategies have been devised to rectify the imbalance of

angiogenic factors present in disease conditions. VEGF is a well characterized growth factor

responsible for switching on the angiogenesis cascade and many angiogenesis-targeting therapies

have been developed to target VEGF signalling. However, the long-term application of both

protein and small-molecule based VEGF-targeting therapies has demonstrated short-lived

efficacies due to the development of drug resistance caused by multiple angiogenesis pathways.

Angiogenesis therapeutics must accommodate the physiology of angiogenic redundancy.

Stimulating or inhibiting multiple pathways, through the use of broad-spectrum agents or the

application of multiple single-target agents in combination, may overcome drug resistance.

Therefore, there is an urgent need for the identification of novel compounds targeting different

components and processes of angiogenesis.

The conventional process of drug development is time consuming and has a low rate of

success. In the initial phase of classic in vitro-based drug discovery, high-throughput screening

of compound libraries is performed in biochemical targets or cell-based assays in microtiter plate

formats that range from the standard 96- to 3456-well plates [191]. Several thousand of

Page 47: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

34

compounds can be screened within a week at a typical pharmaceutical company such as Pfizer

[191]. Yet, many drug candidates do not enter clinical trials due to poor translation of in vitro

findings into animal models. To address this particular bottleneck, HTS methods were developed

to evaluate the effect of screened compounds on ADME targets such as microsomal P450

enzymes, protein binding, serum stability and cytotoxicity [191], to predict adverse drug

reactions. Despite the advances in drug profiling and the availability of sophisticated resources in

pharmaceutical technology, the number of FDA-approved drugs has declined in the past two

decades [192]. Scannell et al documented a reduction in the total number of new drugs approved

between 1996 and 2010 and diminishing cost return in respect to the billions of dollars expended

in research and development [192].

2.1.2 Zebrafish as an advanced model system for drug discovery

Screening candidate compounds in a whole-organism allows for the evaluation of drug

specificity and toxicity at an earlier stage of drug development. Zebrafish have numerous

qualities that are well suited to drug screening: accessibility for manipulation and observation,

high fecundity, small size, rapid development, and a physiology and pharmacology that is

analogous to mammals. The optical transparency of zebrafish embryos, coupled with the

availability of transgenic zebrafish lines with fluorescent labelling of specific tissues or organs,

facilitates real-time visualization of drug-induced, cardiovascular phenotypes in a high-

throughput format.

Zebrafish screens are able to identify new hits from libraries of small compounds that

have already been heavily screened with mammalian cell-based assays. For example, Murphey et

al demonstrated the sensitivity of zebrafish screening by identifying 14 small molecule cell cycle

inhibitors not previously identified from in vitro mitotic screens of a 16320-compound library

Page 48: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

35

[193]. Cell-based approaches are limited in their capacity to recapitulate the complexities of the

targeted physiological process and cannot model the metabolism of a drug in the way a whole-

organism model system does. Furthermore, drug metabolites may contribute to the observed

efficacy of a drug in vivo.

Zebrafish screening is able to quickly bring compounds into clinical trials. Zon and

colleagues identified prostaglandin signalling (PGE2) as a regulator of hematopoietic stem cell

(HSC) homeostasis through a small molecule zebrafish embryo screen [194]. Further analysis in

embryo knockdown and rescue experiments and subsequent studies in adult zebrafish and murine

in vitro and in vivo models supported PGE2 as an essential and druggable target for regulating

HSC number [194]. A bioactive PGE2 derivative has been developed and is currently being

evaluated as a therapeutic patients receiving cord blood transplants [194]. It only took two years

for the Zon lab to move the PGE2 compound to a (human) Phase I clinical trial since the first

article was published in 2007 [194].

The zebrafish model possesses specific advantages over existing cell-based and

mammalian models in respect to its use in vascular drug development. As a phenotype-based

screening model, it allows for the identification of compounds without prior knowledge of the

therapeutic target. Although VEGF has been implicated as a critical component of the angiogenic

cascade, it has become evident from existing VEGF-targeted therapies that it alone is not an ideal

therapeutic target. A phenotype-based approach is a practical method for discovering potent

modifiers of angiogenesis and may help identify unknown therapeutic targets for angiogenesis

pathologies.

The molecular and genetic processes of developmental angiogenesis are well understood

in zebrafish. Many of the mechanism studies routinely performed in vitro can be accomplished

Page 49: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

36

within the context of a whole organism, though validation is often necessary in mammalian

models. The physiology of angiogenesis is conserved in vertebrates and a diverse array of

genetic tools is available for target validation and pharmacological studies within the zebrafish

model.

2.1.3 Our pilot zebrafish angiogenesis screens and my personal research interest

The advent of chemical genetic screens using zebrafish has enabled an efficient and

physiologically relevant method of identifying novel therapeutic compounds [195]. In addition,

new applications for established drugs may also be discovered using this screening platform. Our

lab has previously performed chemical genetic screens of two compound libraries using

endothelial-cell labelled Tg(Flk1:EGFP) zebrafish embryos.

The Spectrum Collection contains 2000 compounds, including existing drugs (60%),

compounds derived from natural plant products (25%) and other bioactive compounds (15%).

The screen of the Spectrum Collection produced seven hits that were classified into three groups:

rotenoids (isorotenone, dihydromunduletone; a class of natural pesticides), statins (simvastatin,

mevastatin, lovastatin, rosuvastatin; drugs used in the management of high cholesterol), and the

carcinogen aristolochic acid [96]. Rosuvastatin was selected for detailed cellular and animal

studies because of the potency of its effects in the zebrafish assay, its status as a clinically

established drug, and its controversial role in angiogenesis. My predecessor Chunyang Wang

evaluated the anti-angiogenic and anti-tumour effects of rosuvastatin and his findings were

published in European Urology in 2010 [96].

The LOPAC library contains 1280 bioactive compounds and our screen identified three

anti-angiogenic (SP600123, indirubin-3-monoxime, and mevastatin) and three pro-angiogenic

(retinoic acid, 4-hydroxyphenyl retinamide, and arotinoid acid) compounds (unpublished).

Page 50: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

37

I have selected three compounds for validation and mechanism studies to evaluate their

potential as drugs for therapies targeting angiogenesis. The compound 4-hydroxyphenyl

retinamide (4HPR) was selected due to its incongruous ability to promote angiogenesis in

zebrafish when it is currently in clinical trials for use as an anti-cancer agent with reported anti-

angiogenic activities. Indirubin-3-monoxime (I3M) was chosen for study because it was the most

potent and well tolerated of the anti-angiogenic lead compounds. My last compound of study,

dihydromunduletone (DHM) was picked for its complete novelty as a therapeutic compound.

The unique qualities of the zebrafish model provide us with an advanced and cost-

effective method of screening and dissecting the molecular mechanisms of compounds identified

from high-throughput screening for therapeutic applications in angiogenesis pathologies.

Page 51: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

38

2.2 Objective and General Hypothesis

The zebrafish angiogenesis screen has the capacity to efficiently identify new compounds

that can modulate angiogenesis. My studies will validate the efficacy of three small molecule

compounds selected from our pilot zebrafish angiogenesis screens. I will also utilize the

versatility of the zebrafish model to investigate the molecular mechanisms of drug action of these

compounds in vivo.

I hypothesize that the whole-organism model system of my zebrafish studies provides

useful information for drug testing across different developmental stages. When used in

conjunction with transgenic techniques and other interventions, this system provides potential

mechanisms of the therapeutic functions as well as the side effects of potential drugs that may

affect angiogenesis process in vivo.

The overall objective of this study is to use zebrafish as an in vivo model to expedite the

development of therapeutic compounds that will effectively treat angiogenesis-dependent

pathologies.

Page 52: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

39

2.3 Specific Hypotheses

Since I chose to study three compounds and each of them may have different mechanisms

in modulating angiogenesis activities, I have formed individual hypotheses for each compound.

Hypothesis 1 – 4HPR

4-hydroxyphenyl retinamide (4HPR; fenretinide) was identified in our developmental

angiogenesis screen as a stimulator of angiogenesis. I hypothesize that 4HPR achieves this effect

by stimulating somitogenesis and that angiogenesis is secondary to somitogenesis.

Hypothesis 2 – I3M

Indirubin-3-monoxime (I3M) is a potent inhibitor of angiogenesis and has great potential

for development as an anti-cancer agent. I hypothesize that it affects the process of EC migration

in vivo by acting on the notch-VEGF pathway.

Hypothesis 3 - DHM

Dihydromunduletone (DHM) is a novel compound that is identified from our screen as an

inhibitor of angiogenesis. I hypothesize that it suppresses angiogenesis by modulating EC

function in vivo.

Page 53: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

40

CHAPTER 3: ROLES OF 4-HYDROXYPHENYL RETINAMIDE IN ANGIOGENESIS AND ITS POTENTIAL CELLULAR AND MOLECULAR

MECHANISMS

3.1 Introduction

3.1.1 Retinoids: Physiology and pharmaceutical development

Vitamin A (retinol) is an essential nutrient involved in physiological development and

homeostasis. The World Health Organization has documented weight loss, and eye and immune

disorders in impoverished countries as a consequence of insufficient Vitamin A intake [196].

Vitamin A is a small lipophilic molecule found naturally in foods such as carrots,

potatoes, poultry and fish. It is absorbed in the small intestine where it binds to retinol binding

protein (RBP) and enters the circulation. Retinol-RBP is transported into the liver for storage,

and delivered to tissues where it is metabolized into its functional forms: retinaldehyde and

retinoic acid (RA) [197]. Retinaldehyde is a chromatophore with critical roles in low-light and

colour vision and RA regulates a number of developmental and homeostatic processes [198].

Unlike most signalling molecules, RA is transported into the nucleus by cellular retinoic acid

binding proteins (CRABP) and interacts directly with nuclear retinoic acid receptors (RARs) and

retinoic acid X receptors (RXRs) to regulate gene transcription in various tissues [198].

Retinol is provided from the mother during mammalian development. It crosses the

placenta and is metabolized to retinaldehyde in the embryo [199]. In zebrafish, maternally

derived retinaldehyde is stored in the yolk [200], though both mammals and zebrafish rely on

retinaldehyde dehydrogenases (RALDH) to oxidize retinaldehyde into its active form. RA exerts

its actions in a paracrine manner during embryogenesis, repressing the action of growth factor

signalling pathways (such as FGF and Wnt) in a spatiotemporal manner [201]. RA signalling

Page 54: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

41

also governs many developmental processes: neurogenesis, cardiogenesis, body axis extension,

developmental patterning and organogenesis [202].

RA signalling has important functions in maintaining normal function in organs such as

the heart, lungs and skin [203]. The role of RA in the differentiation and growth of epithelial

tissues was documented as early as 1925, when metaplasias were observed in animals deficient

in RA. These morphological changes, induced by RA-deficiency or chemical carcinogenesis,

could be offset with dietary RA treatment [204] [205]. The link between RA and cancer has

additionally been validated in an epidemiological study. Individuals reporting less Vitamin A

intake in their diet have a higher risk of developing cancer [206]. In the past few decades, RA

has been evaluated as a therapeutic agent for the treatment and prevention of cancer.

RA’s poor bioavailability and tendency to render chemotherapy-resistance to tumours has

limited its application as an anti-cancer agent [207]. RA is also associated with a number of

toxicities, which is to be expected when we consider the essential functions of RA in physiology

and development. These side effects include anemia, weight loss, birth defects and bone fractures

[208].

Nonetheless, a closely related derivative of RA is commonly used as a topical agent for

dermatologic disorders such as acne, under the generic name isotretinoin [209]. It is also

uniquely effective as a primary treatment for acute premyelocytic leukemia, a fatal blood

disorder caused by the inability of hematopoietic stem cells (HSCs) to mature. Oral

administration of isotretinoin is able to induce terminal differentiation of HSCs, resulting in 90-

95% complete remission rate [210]. This is a fantastic example demonstrating the potential of

molecular-targeted therapies to not only treat a disease, but also cure it.

Page 55: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

42

Retinoids are a group of natural and synthetic analogs of Vitamin A. Natural analogs of

Vitamin A, such as retinoic acid (RA) are produced from metabolic processes in the body.

Synthetic retinoids are created by modifying functional groups on RA. Over 10 000 retinoids

have been identified for a variety of biomedical applications [211]. Synthetic retinoids have

greater potency and more favourable pharmacokinetic and toxicity profiles than their parent

compounds [204]. My first compound of study, 4-hydroxyphenyl retinamide (4HPR or

fenretinide) is a derivative of RA that was synthesized in the late seventies by the pharmaceutical

company Johnson and Johnson [212]. It is currently being investigated for applications in the

management of cancer.

3.1.2 Fenretinide as an anti-cancer agent Fenretinide is a cytotoxic agent, arresting proliferation and inducing cell death in many

cancer cell lines: breast, ovarian, cervical, prostate, lung, renal, bladder, glioma, head and neck,

lymphoma, neuroblastoma and Ewing’s sarcoma [213].

The proposed mechanisms for fenretinide’s apoptotic effects can be divided into

receptor-mediated and receptor-independent mechanisms. RAR expression is increased in

fenretinide-induced cell differentiation and apoptosis in some cell types [214]. Curiously,

fenretinide has been shown to bind poorly to RARs [215]. It is possible that fenretinide is

increasing RA release from intracellular stores or is converted to a metabolite that may activate

RARs [216]. The apoptotic effect of 4HPR appears to be distinct from classical retinoids. It is

mainly driven by receptor-independent mechanisms, which involve the increase of signalling

molecules: ceramides, caspases, NO and reactive oxygen species (ROS) [217] [218].

Fenretinide-mediated release of ROS promotes apoptosis through the activation of JNK, p38,

ERK and PKC [219]. In addition, fenretinide acts in synergy with chemotherapeutic agents to

Page 56: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

43

induce apoptosis in vitro [220]. Fenretinide shows the ability to complement the action of

chemotherapy agents by activating apoptosis independently of the p53 pathway, which is

commonly mutated in most cancers [221].

Another mechanism by which fenretinide is able to reduce tumour growth is through the

inhibition of angiogenesis. Studies have demonstrated that fenretinide is able to reduce in vivo

neovascularisation in CAM and Matrigel plug assays [222, 223]. Fenretinide disrupts the

function of ECs in a number of ways that affect the progression of angiogenesis: downregulation

of components of the VEGF and FGF pathways (proliferation), upregulation of TGF-β (to induce

apoptosis), decreased MMP activity (migration), and repression of differentiation (required in

tube formation) [223, 224].

The investigation of fenretinide’s mechanism to reduce tumour growth is an ongoing but

necessary process in determining the optimal applications of the drug.

The anti-proliferative and anti-angiogenic effects of fenretinide contributes to the

reduction of tumour growth observed in animal models of colon, prostate, bladder, pancreas,

liver and skin cancer [225]. However, the route of administration affects the absorption and

distribution of the drug, as mice with ovarian carcinomas survived much longer when fenretinide

was administered intraperitoneally rather than orally [226]. Additionally, not all cancers are

responsive to treatment. Fenretinide treatment did not inhibit total tumour formation in a

chemoprevention model of lung cancer [227]. Fenretinide’s potential to cause abnormal

embryonic development has been assessed in rats and rabbits (Oryctolagus cuniculus) during the

early stages of the drug’s development, where it was classified as a weak teratogen [228].

Page 57: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

44

The results of preclinical studies demonstrated that fenretinide was an effective candidate

for chemoprevention and was less toxic than alternative treatments. These factors encouraged the

evaluation of fenretinide in clinical trials. A 15-year clinical study has demonstrated the

sustained ability of fenretinide to significantly reduce the incidence of second breast cancer

[229]. The use of fenretnide for treatment and chemoprevention has also been tested in other

cancers; brain cancer [230], kidney cancer [231], prostate cancer [232] and bladder cancer [233].

While the drug is generally well tolerated, there does not appear to be any beneficial effect in the

chemoprevention of most cancers. The chemoprotective effect of fenretinide observed with

breast cancer is dependent on age and menopausal status, suggesting that fenretinide acts in

concert with hormones such as IGF-1 [234]. To make use of this window of opportunity,

fenretinide is presently undergoing clinical evaluation as prophylactic therapy for young women

with high-risk for breast cancer [235]. At present, there are over 25 clinical trials in progress to

evaluate the efficacy of fenretinide in chemoprevention and intervention of a variety of cancers

[224]. Long term evaluations of fenretinide in the clinic have established it to be relatively safe.

Eye conditions (diminished dark adaptation), dermatologic disorders, and gastrointestinal

symptoms have been reported as adverse events associated with 4HPR [236].

3.1.3 Additional applications of fenretinide The application of fenretinide is currently being explored in the treatment of a variety of

conditions. Fenretinide has recently been proposed for cystic fibrosis (CF) treatment. Fenretinide

administration was shown to normalize ceramide deficiencies that lead to lipid imbalance and

osteoporosis in a mouse model of CF [237]. Fenretinide is also being considered for the

treatment of diabetes and dyslipidemia. Intraperitoneal fenretinide administration reduced insulin

resistance and decreased plasmid lipid levels in genetically obese mice fed with a high-fat diet

Page 58: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

45

[238]. The outcome of a recent Phase II clinical trial has established the potential use of

fenretinide in the treatment of late stage age-related macular degeneration. The accumulation of

retinol in the eye causes toxic effects that can lead to blindness. Fenretinide competes with

retinol to bind with RBP thereby reducing retinol’s mode of transport to the eye. Results of a

clinical trial demonstrated that oral fenretinide treatment reduced lesion growth and decreased

incidences of choroidal neovascularization in patients in two years [239].

3.1.4 Rationale New applications are constantly being explored to take advantage of the fact that

fenretinide has proven itself as a safe and effective therapeutic agent after decades of

pharmaceutical research. Fenretinide was identified in our pilot zebrafish screen as a compound

promoting angiogenesis, which contradicts published data supporting the anti-angiogenic

properties of fenretinide as an anti-cancer agent. The inhibitory effects of fenretinide on

angiogenesis have been studied in vitro and in vivo; in CAM and Matrigel plug assays. I intend

to examine the pro-angiogenic effects of fenretinide and explore its potential mechanism of

action within the zebrafish model system. I hope to either reconcile these contradictory findings

or provide evidence for a potentially novel application of fenretinide as a promoter of

angiogenesis.

Page 59: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

46

3.2 Materials and Methods

3.2.1 Preparation of drugs and reagents

Fenretinide (4HPR) and semaxanib (SU5416) were purchased from Tocris Bioscience

(Ellisville, MI, USA). Phenylthiourea (PTU) was purchased from Sigma-Aldrich (Oakville, ON,

Canada). Drugs were dissolved in 100% DMSO (Sigma-Aldrich, Oakville, Canada) and serially

diluted to a 10x stock solution stored at -20°C. Embryos and larvae were treated with 4HPR at

final concentrations of 1, 2, 4, 8, and 16 µM and adults were treated with 5 and 10 µM. Embryos

were not exposed to a concentration of DMSO that exceeded 1%. Embryo medium, containing

Millipore water and a mixture of salts, was prepared at half the strength of the protocol described

by Nusslein-Volhard and Dahm [240]. The anesthetic was prepared by mixing eugenol in

embryo water, containing 1% ethanol to a stock concentration of 10 parts per million (ppm).

2.5% methyl cellulose used in SCORE imaging was prepared by combining methyl cellulose and

clove oil in heated nanopure water, mixed until dissolution and left to stand in ice for several

hours to remove bubbles that formed.

3.2.2 Zebrafish strains and husbandry

The following strains were employed for experimental study: wild-type striped and

spotted adult zebrafish (mixed genetic background) and transgenic lines: Tg(flk1:EGFP),

Tg(GATA1:dsRED) both provided by Dr. Ian Scott (University of Toronto), Tg(fli1:nEGFP)y7

provided by Dr. Sarah Childs (University of Calgary), and Tg(fli1:EGFP;casper) provided by

Christian Lawrence (Dr. Leonard Zon, Harvard University) and a mutant albino line produced

through selective breeding in the Wen lab. Fish were maintained at 28°C in a circulating

aquaculture system and fed twice daily at our facilities in the Li Ka Shing Knowledge Institute at

Page 60: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

47

St. Michael’s Hospital. Embryos were obtained using standard mating conditions and incubated

in embryo medium.

3.2.3 Zebrafish developmental angiogenesis assay

Figure 3.2.3 Typical protocol for a zebrafish developmental angiogenesis assay. Eggs produced by natural breeding were collected and dechorionated 10 hpf. The eggs were arrayed into 96-well plate and drugs were added directly into the fish water at 12 hpf. After 2 days of incubation, fish were imaged, and ISV number was used to evaluate the drug’s effect on angiogenesis.

The transgenic zebrafish lines Tg(flk1:EGFP) and Tg(fli1:EGFP;casper) were utilized in

this assay of developmental angiogenesis. Increasing concentrations of the selected compound

were used to evaluate dose-dependent effects on ISV development. Both lines express enhanced

green fluorescence protein (EGFP) in the cytoplasm of endothelial cells (ECs), allowing for

visualization of the entire vasculature under fluorescence microscopy. Following natural

breeding, the chorions of healthy embryos were manually removed (dechorionated) using

Page 61: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

48

Almedic #7 forceps at the bud stage (10 hpf) and 4-5 embryos were dispensed into a single well

of a 96-well plate, with each well containing 90 µL of 0.5x E2 embryo medium. 10 µL of a 10x

stock solution of fenretinide was added into embryo medium at the 6-somite stage (12 hpf), prior

to the commencement of angiogenesis. Control embryos were treated with 0.5% DMSO. After

48 hours of incubation at 28°C, the embryos were anesthetized with 100 ppm eugenol, mounted

on a 1% agarose-coated, 100 mm tissue culture plate and imaged with a Leica fluorescence

stereomicroscope at 32-40X magnification. Samples were analyzed with the following

specifications: only fully formed intersegmental vessels (ISVs) with attachment to the dorsal

longitudinal anastomotic vessel (DLAV) were counted and embryos without a heart beat were

excluded from analysis. Data was compiled in Microsoft excel and statistically analyzed using

ANOVA and a two-tailed student t-test. Error bars denote standard error (SE).

3.2.4 In vivo quantification of EC number Tg(fli1:nEGFP)y7

expresses EGFP in the nucleus of ECs, permitting the quantification of

EC number in specific vascular structures like the DA, DLAV, and ISV. Embryos were treated

as indicated in the angiogenesis screen. 0.22 mM of propylthiourea (PTU) was added into media

at 12 hpf to inhibit pigment formation to improve resolution for cell counting. Images of

embryos were acquired at a magnification of 40X after 2 days of incubation, using a Leica

fluorescent stereomicroscope and prepared using agarose or Specimen in a Corrected Optical

Rotational Enclosure (SCORE) mounting techniques. Embryos were immersed in a solution of

2.5% methylcellulose and 100 ppm eugenol, drawn into a capillary, and rotated in an optimal

orientation. For the manual counting of cell number in the ISV and DA, nuclei were counted and

averaged across 10 segments of each embryo, then expressed as an average of all samples of

each condition. Manual cell counting in the DLAV determined a total number of DLAV cells in

Page 62: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

49

each fish, which was averaged for each condition. The data was compiled in Microsoft Excel and

statistically analyzed using ANOVA and a two-tailed student t-test. Error bars denote SE.

3.2.5 Fin regeneration assay

Figure 3.2.5. The procedure of a fin regeneration assay. Larval and young adult zebrafish were anesthetized and half of the caudal fin was transected with a blade. Fish were incubated in cups or wells with drugs dissolved in the fish water for 3 days. Images before and after amputation, and 3 days post amputation were compared to evaluate the amount of fin regenerated. The notochord is used as a reference point for larval fish. Newly regenerated tail fins are not pigmented in adult zebrafish. [241, 242].

Larval model: I collected the eggs produced by natural breeding of Tg(flk1:EGFP)

vascular-labelled zebrafish. I anaesthetized embryos aged 2 dpf with 100 ppm of eugenol and

transected the tail primordium, just posterior to the end of the notochord with a 26-gauge needle

Page 63: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

50

tip. Each embryo was dispensed into a single well of a 96-well plate and a quantity of drug was

added into the well water. After 3 days of incubation, the larvae were imaged and the amount of

fin regenerated was measured using ImageJ (National Institutes of Health, Bethesda, Maryland)

with the notochord as a reference point. Data was compiled and statistically analyzed in

Microsoft Excel using ANOVA and a two-tailed student t-test. Error bars denote SE.

Adult model: Wild-type young adult zebrafish (3 months old) were anaesthetized with

10 ppm of eugenol and amputated midway through the caudal fin with a clean razor blade. Fish

were imaged at 12-15X magnification before and after amputation using a stereomicroscope.

After 15 minutes of recovery, the fish were distributed into containers each with 2-3 fish and 100

mL solution of water and drug/DMSO. The fish were incubated at 28°C, fed once a day, and

imaged three days post-amputation (3 dpa). Original, amputated, and regenerated fin lengths

were measured using ImageJ and analyzed using Microsoft Excel. The compiled and averaged

data were statistically analyzed using ANOVA and a two-tailed student t-test. Error bars denote

SE.

3.2.6 Whole-mount immunohistochemistry (IHC)

Tg(flk1:EGFP) embryos were treated with 4 µM 4HPR, 7.5 µM SU5416 (alone and

together), and 0.5% DMSO (vehicle) as indicated in the angiogenesis screen. 0.22 mM of PTU

was added to the well solution to inhibit pigmentation. 2.5 dpf embryos were fixed overnight in

4% paraformaldehyde (PFA) and processed using the IHC protocol provided by Dr. Jason Fish.

Embryos were washed 2-3 times (5 minutes each) with PBSTw (1x PBS containing 0.1%

Tween-20) to remove PFA. For permeabilization, the embryos were washed 3 times (10 minutes

each) in PBST solution, 1x PBS containing 1% Triton-X. An additional 20 minutes of washing

(10 minutes each) was performed with PBT solution, which contains 1xPBS, 1% Triton-X and

Page 64: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

51

2% bovine serum albumin (BSA). The embryos were left at room temperature in blocking

solution (containing 1x PBS, 1% Triton-X, 2% BSA, 10% goat or sheep serum, and 1% DMSO)

for 3-4 hours.

The mouse F59 (Developmental Studies Hybridoma Bank, Iowa City, IO, USA) antibody

recognizes slow myosin heavy chain (MyHC) in somites and the rabbit α-GFP (Molecular

Probes, Invitrogen, Burlington, ON, Canada) antibody recognizes the GFP contained in ECs of

transgenic embryos. These primary antibodies were added (at 1:5 and 1:400 respectively) and

left overnight at 4°C. On the second day, the embryos were washed extensively with PBT for at

least 2 hours (10-15 minutes each wash). The secondary antibodies, anti-mouse Alexa-Fluor 647

and anti-rabbit Alexa-Fluor 488 (both raised in goat; Invitrogen, Burlington, Canada) were used

to label F59 in red and α-GFP in green. A 1:200 solution of secondary antibodies were added

into blocking solution and left to incubate overnight at 4°C.

On the third day, embryos were transferred to a new tube and washed extensively in PBT

at least 10 times, 10 minutes each wash. Embryos were cleared in a glycerol solution and

mounted on glass plates with a thin layer of 1% agarose to restrict motion during image capture.

At least 5 embryos were treated in each condition. Representative images of fully intact embryos

were acquired using a Leica confocal microscope and stacked to generate a 3-dimensional image

of the tail at 25X magnification.

3.2.7 Whole-mount RNA in situ hybridization (ISH) Primer preparation

RARg-a: A pCR4-TOPO plasmid containing RARg was provided by Ellie Melancon

(Postlethwait lab, University of Oregon). The plasmid contained a T3 RNA polymerase promoter

Page 65: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

52

fused to a RARg gene fragment and was sequenced to confirm the identity of the RARg-a gene.

The plasmid was dissolved into nuclease-free water (NF-H2O) and OneShot TOP-10 competent

E-coli cells were transformed for the purification of more plasmid DNA. 5 µL of plasmid was

added to competent cells, gently mixed and incubated on ice for 30 minutes. The cells were heat-

shocked in a 42°C waterbath and placed in ice. 250 µL of pre-warmed LB medium was added

and incubated in for an hour at 225 revolutions per minute (rpm) in a 37°C rotary shaker-

incubator. 50 µL of the transformation mix was spread onto LB /ampicillin agar plates and

incubated overnight at 37°C. Three colonies were selected and cultured in LB/amp media

overnight in a 37°C 225 rpm rotary shaker-incubator. Plasmid DNA was isolated from

transformants using a minpreparation kit (Qiagen). 20 µL (6 µg) of purified DNA was digested

with 5 µL of the restriction enzyme NotI for 2 hours at 37°C. The digested product was purified

using a PCR purification kit (Qiagen). In vitro transcription using T3 polymerase and a DIG

RNA labelling kit (MEGAscript, Roche) was employed under the stringent RNAse-free

conditions of a Captair rolling fume hood. The labelling mixture combined 2 µL of each

nucleotide (ATP, GTP, CTP, DIG-UTP), 2 µL of 10x reaction buffer, 1 µL of SuperR (RNAse

inhibitor), 2 µL of T3 RNA polymerase and NF-H2O up to a total volume of 20 µL. The mixture

was incubated for 2 hours at 37°C. The RNA was precipitated with 30 µL of LiCl precipitation

solution and chilled for 45 minutes at -80°C. The sample was centrifuged for 15 minutes at 4°C

to pellet the RNA, washed with 70% ethanol and re-centrifuged. The pellet was then resuspended

in 30 µL of NF-H2O, aliquoted and stored at -20°C in either NF-H2O or hybridization mix until

usage. Nanodrop and 1% agarose gel electrophoresis of the product confirms RNA probes 500bp

in length and of good quality.

Page 66: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

53

FGFR1a: PCR primers were designed using the electronic Primer3 program to flank the

5’ and 3’ untranslated regions (UTRs) of the FGFR1a and BLASTed for specificity. DNA

fragments were PCR amplified using Taq polymerase and a cDNA template generated from 7

day old embryo mRNA by reverse transcriptase (obtained from Dingyan Wang of the Gramolini

lab, University of Toronto). The solution was run in a thermocycler (Eppendorf Mastercycler

gradient) under the following conditions: 93°C for 3 minutes, *93°C for 30 seconds, 58°C for 45

seconds, 70°C for 1 minute,* with the asterisked settings repeating 40x, then 70°C for 10

minutes and 4°C for short term storage. Products were purified using a PCR purification kit

(Qiagen) and ligated into a TOPO 2.1 vector containing a T7 RNA polymerase promoter. 1 µL of

the PCR purified product was added to 1 µL of TOPO vector in a total volume of 7 µL NF-H2O

at room temperature for 15 minutes. Plasmids were cloned by transforming OneShot TOP-10

competent E-coli cells using a modified heat shock protocol. 1 µL of ligation mix was added to

thawed competent cells and incubated on ice for 15 minutes. The mixture was heat-shocked at

42°C for 45 seconds and iced for 2 minutes. 250 µL of pre-warmed LB media was added and

cells were incubated for 1 hour in a 37°C, 225 rpm rotary shaker-incubator. 150 µL of the

transformation mix was spread onto LB/ampicillin plates coated with 40 µL of 2% X-gal and

incubated overnight at 37°C. 3-4 isolated white colonies were selected to be cultured in

LB/ampicillin media overnight in a 37°C, 225 rpm rotary shaker-incubator. Vector DNA was

isolated from transformants using a minpreparation kit (Qiagen). A sample of the DNA was

digested with restriction enzyme EcoRI to confirm ligation of the insert by 1% agarose gel

electrophoresis. The remainder of the vector was linearized using restriction enzyme EcoRV.

RNA probes were generated by in vitro transcription using T7 polymerase and a DIG RNA

Page 67: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

54

labelling kit (MEGAscript, Roche) under the stringent RNAse-free conditions of a Captair

rolling fume hood.

Embryo preparation

Embryos of the albino strain were treated with 4 µM 4HPR, 4 µM retinoic acid (RA) or

0.5% DMSO at 90% epiboly (9 hpf) and incubated overnight at 28°C. 24 hpf embryos were

fixed in 4% paraformaldehyde (PFA) overnight at 4°C, washed with PBS and dehydrated in

methanol for at least 24 hours at -20°C.

In situ hybridization

RNA in situ hybridization was performed as described by Thisse et al. (2010) with some

deviations. Dehydrated embryos stored at -20°C were rehydrated with successive dilutions of

methanol in 1x PBS, then washed 4 times, 5 minutes per wash, in PBT. Embryos were carefully

dechorionated using Almedic #7 forceps after rehydration to decrease tissue damage. Embryos

were permeabilized by digestion with 10 µg/mL Proteinase K for 10 minutes at room

temperature. This is followed with a 20 minute 4% PFA-PBS wash to stop the digestion. The

embryos are washed with PBT 4 times, 5 minutes per wash to remove PFA and transferred to

new 1.5 mL eppendorf tubes. Embryos are prehybridized in 300 µL of Hybridization Mix (HM)

containing tRNA and heparin for 3 hours in a 70°C hybridization chamber. HM is removed and

replaced with 200 µL of HM containing 1 ng of the DIG-labelled RNA probe, then left overnight

in the 70°C hybridization chamber. On day 2, HM (that does not contain tRNA and heparin) is

successively diluted to a solution of 2x SSC with a series of 10 minute washes at 70°C: 75%,

50%, 25% HM and 100% 2x SSC. Two 0.2x SSC washes, 30 minutes each, were performed at

70°C. 0.05x SSC was progressively replaced with PBT with a series of 10 minute washes at

Page 68: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

55

room temperature: 75%, 50%, 25% of 0.05x SSC and 100% PBT. Embryos were incubated in

preincubation buffer (containing 2mg/mL BSA and 2% sheep serum) for 3 hours at room

temperature under agitation. The solution was replaced with 300 µL preincubation buffer

containing a 1:5000 dilution of anti-DIG antibody, and incubated overnight at 4°C under

agitation. On day 3, embryos were transferred to new 1.5 mL tubes and additional PBT washes

were performed at room temperature, 10-15 minutes each wash, at least 20 times. Embryos were

left in PBT to wash at 4°C overnight to decrease background staining caused by residual anti-

DIG antibodies. On day 4, the alkaline tris buffer and NBT-BCIP staining solutions were freshly

prepared without MgCl2 to prevent salt formation and the staining solution was kept in the dark.

Embryos were washed 3 times; 5 minutes each wash, in alkaline tris buffer at room temperature

with agitation. After adding staining solution, the embryos are periodically monitored, with

limited exposure to light, until the appropriate amount of staining is present. The embryos were

rinsed in stop solution (containing 5 mM EDTA) and cleared in glycerol for another 30 minutes.

A coverslip bridge platform was constructed to facilitate imaging by rolling the “sandwich-ed”

embryos into a planar orientation. Embryos were mounted and images were captured the same

day using z-stack compilation on a Leica stereomicroscope.

Page 69: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

56

3.3 Results 3.3.1 Fenretinide increases ISV number in developing zebrafish embryos

In our pilot angiogenesis screen, fenretinide was identified to promote the development

of additional intersegmental vessels (ISVs) in developing zebrafish embryos. To validate this

result and establish a dose-response curve, I administered a range of 4HPR concentrations to

developing zebrafish embryos. Increasing concentrations of 4HPR in transgenic vasculature-

labelled embryos Tg(Flk1:EGFP) resulted in an overall increase in the number of ISVs of each

fish (Fig. 3.3.1). Control embryos treated with vehicle (0.5% DMSO) develop an average of 27.5

± 0.1 ISVs. Embryos treated with 1 µM 4HPR develop 30.2 ± 0.2 ISVs and 4 µM 4HPR develop

32.8 ± 0.3 ISVs (p<0.01). The additional ISVs arise by the formation of extra ISVs in the tail

region, an area absent of ISVs in control embryos.

Page 70: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

57

Figure 3.3.1 [A] Dose-dependent effect of 4HPR on intersegmental vessel (ISV) development in Tg(flk1:EGFP) zebrafish embryos (treated with PTU) after 48h of drug treatment. 4HPR increases the amount of ISV that develop in 2.5 dpf zebrafish embryos. Fluorescent images are shown of embryos treated with [B] a control of 0.5% DMSO (27.5 ± 0.1 ISVs), and [C] 4 µM 4HPR (32.6 ± 0.4 ISVs). 4HPR-treated embryos characteristically form ISVs that extend into the tail tip. *p < 0.05. ** p < 0.01. N=11-14. Error bars denote SE.

3.3.2 Fenretinide alters the number of endothelial cells in blood vessels

Tg(Fli1:nEGFP)y7 embryos were employed to observe changes in the number of

endothelial cells in specific vascular structures in response to 4HPR treatment (Fig.3.3.2).

Tg(Fli1:nEGFP)y7 is a transgenic zebrafish line with nuclear-localized GFP expression [93].

Embryos were treated with PTU to inhibit pigment formation to facilitate cell counting. The

average cell number decreases in each ISV from 7.5 ± 0.2 cells in control embryos to 3.8 ± 0.4

cells in embryos treated with 4 µM 4HPR. The average cell number in each segment of the DA

Page 71: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

58

increases though it is difficult to quantify, as the increase in cell number causes cells to overlap

to the extent that manual and digital cell counting is not possible. There is also a diffusion of the

nuclear EGFP signal along the trunk of 4HPR-treated embryos, which may indicate cellular

apoptosis.

Figure 3.3.2. The average cell number in the [A] intersegmental vessel (ISV) in Tg(fli1:nEGFP) zebrafish embryos after 48h of IO treatment. Fluorescent images of zebrafish embryos at 2.5 dpf show embryos treated with [B] a control of 0.5% DMSO and [C] 4 µM 4HPR. Treatment with 4HPR decreases cell number in the ISV and increases cell number in the dorsal aorta. *p < 0.05. ** p < 0.01. N=6-14. Error bars denote SE.

3.3.3 Fenretinide inhibits fin regeneration Fin regeneration assays were performed in adult zebrafish to assess the effect of 4HPR on

adult angiogenesis. The process of adult tissue regeneration is dependent on angiogenesis and

cell proliferation. Larval fin regeneration is an avascular process, so the assay was performed in

larval and adult zebrafish to differentiate between drug effects on angiogenesis and cell

proliferation. 4HPR dose-dependently inhibits fin regeneration in larval (Fig. 3.3.3.1) and adult

Page 72: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

59

zebrafish (Fig.3.3.3.2) as measured 3 days post amputation (dpa). Larval fish were treated with a

greater range of 4HPR as they can tolerate higher concentrations. At 16 µM, larval fish were able

to regenerate an average of 2 µm relative to the 86 µm regenerated by control-treated fish

(p<0.01). Adult fish treated with 0.5% DMSO regenerate 13.8 ± 0.6 % of their original fin length

3 days post amputation. 5 µM of 4HPR reduces the amount of fin regenerated to 9.8 ± 0.6 %

(p<0.01) and 10 µM decreases regeneration to 8.1 ± 0.7% of the original fin length (p<0.01).

Figure 3.3.3.1 [A] Effect of 4HPR on larval zebrafish fin regeneration. Fins were transected at the end of the notochord and fish were incubated in drug for 3 days. Bright field images of caudal fins 3 days post amputation reveal that fish treated with [B] 0.5% DMSO regenerate an average of 86 ± 6 µm of fin bud while fish treated with [C] 16 µM 4HPR regenerate 2 ± 2 µm of the original fin length. 4HPR dose-dependently inhibits fin regeneration. *p < 0.05. ** p < 0.01. N=6-9. Error bars denote SE.

Page 73: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

60

Figure 3.3.3.2 [A] Effect of 4HPR on adult zebrafish fin regeneration. Fins are transected at the site of the red arrow and fish are incubated in drug for 3 days. Bright field images of caudal fins 3 days post amputation reveal that fish treated with [B] 0.5% DMSO regenerate an average of 13.8 ± 0.6% of the original fin length while fish treated with [C] 10 µM 4HPR regenerate 8.1 ± 0.7% of the original fin length. 4HPR dose-dependently inhibits fin regeneration. *p < 0.05. ** p < 0.01. N=9. Error bars denote SE.

Page 74: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

61

3.3.4 Fenretinide affects somitogenesis during development

The presence of abnormal somites in the trunk and tail were observed in bright field

images of fenretinide-treated fish (appendix 3.4.3.1). Immunohistochemistry (IHC) labeling of

blood vessels (anti-GFP antibodies) and somites (F59, labeling myosin filaments) allows us to

clearly visualize the changes in these structures to distinguish the specificity of fenretinide’s

effects on somitogenesis or angiogenesis. Control embryos have a consistent phenotype:

chevron-shaped somite bundling and a tail tip region that contains a forked somite structure that

is absent of blood vessels (Fig. 3.3.4A). IHC confirmed the presence of somites and vessels in

the tail tip in embryos treated with 4HPR (Fig 3.3.4B). The drug phenotype characteristically

includes a tail curvature phenotype visible in bright field, with a distinct somite bundling pattern

to the tail tip observed with IHC labeling (Fig 3.3.4A and 3.3.4D). Embryos treated with 7.5 µM

of the VEGFR inhibitor, SU5416, develop fewer vessels but have a forked somite pattern in the

tail that is similar to control embryos (Fig. 3.3.4C). The combined treatment of 4HPR and

SU5416 is tested to evaluate the effects of 4HPR while angiogenesis is being suppressed with

inhibition of VEGF signaling. The presence of the somite pattern in the tail tip is apparent with

the combined dosing of these agents (Fig. 3.3.4D). The ability of 4HPR to extend the somite

boundaries into the tail tip persists in a reproducible manner (100% incidence observed).

Page 75: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

62

Figure 3.3.4. Somite and vessel IHC labeling of 2.5 dpf Tg(flk1:EGFP) zebrafish embryos treated with [A] 0.5% DMSO, [B] 4 µM 4HPR, [C] 7.5 µM SU5416 and [D] co-treated with 10 µM SU5416 and 4 µM 4HPR. Images are confocal stacks that show fluorescent immunodetection of slow MyHC (F59, somite-specific marker) in red and GFP (endothelial cells) in green. 4HPR appears to affect somite patterning in the tail tip region independently of angiogenic vessel development. Representative images are shown. N=4-5.

3.3.5 Fenretinide increases RAR expression and may decrease FGFR expression

The curved tail phenotype of 4HPR is similar to that of its parent compound, RA, which

exerts its actions on somitogenesis through the RA receptor (RAR). I have performed a RNA in

situ hybridization experiment with a labeled RNA probe of RAR to examine the changes in RAR

expression in response to 4HPR treatment, compared to RA and vehicle treatments. My

preliminary experiment showed an increase in RAR in fenretinide- and RA-treated fish as

Page 76: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

63

compared to the control (Fig.3.3.5). The intensity of RAR signal is greater in fenretinide-treated

fish than RA-treated fish, though the concentrations of 4HPR and RA are not equivalent in this

preliminary experiment.

Figure 3.3.5. Preliminary ISH of retinoid acid receptor G (RARg) performed in 24 hpf albino embryos. [A] RARg staining of 24 hpf embryo published in literature [243]. I performed an ISH for RARg in [B] embryos treated with 0.5% DMSO, [C] 4 µM 4HPR and [D] 0.5 µM RA.

Page 77: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

64

3.4 Discussion

3.4.1 Fenretinide promotes developmental angiogenesis but inhibits EC migration

Fenretinide has been studied as an anti-cancer drug with documented anti-proliferative

and anti-angiogenic properties. Thus, it was surprising to discover in the initial screen of the

LOPAC compound library that it did not act as an angiogenic inhibitor but as a promoter. In this

study, we confirmed that fenretinide dose-dependently increased the number of ISVs that

developed in zebrafish embryos (Fig.3.3.1). More specifically, it stimulated the growth of ISVs

into the tail tip, an area that is normally absent of ISVs in control fish.

Nascent ISVs are generated from the proliferation and migration of ECs in the DA. To

evaluate the changes in the number of ECs in certain compartments, I performed an angiogenesis

assay with Tg(Fli1:nEGFP)y7 embryos, which have EGFP expression localized to the nucleus of

each EC. The quantification of cell number in the DA was difficult, as the high density of ECs

made it almost impossible to accurately count by manual or digital means. Nonetheless, the

relative increase in fluorescent signals in the DA and decrease in the ISVs (Fig 3.3.2) suggested

that fenretinide was inhibiting the process of cell migration in vivo. Qualitatively, I was also able

to observe EC toxicity in some embryos treated with 4 µM of 4HPR. The expression of EGFP in

the nucleus of 4HPR-treated embryos (Fig.3.3.2C) was much more diffuse than the distinct

nuclear-localized signal of controls (Fig.3.3.2B), suggesting 4HPR-induction of EC death. These

results correlate with in vitro experiments that demonstrate fenretinide’s ability to reduce cell

proliferation [244] and migration [223] in HUVECs.

Page 78: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

65

3.4.2 Fenretinide inhibits the process of fin regeneration

To observe the effects of fenretinide on tissue regeneration, I performed a fin

regeneration assay in larval and adult zebrafish. I noted a significant decrease in the amount of

fin regenerated in both assays (Figs.3.3.3.1 and 3.3.3.2). In the larval assay, I was able evaluate a

greater number of fish and wider range of concentrations. I also observed more variability, which

can be attributed to the high regenerative capacity of larval fish.

Although the process of wound healing depends on the initiation of physiological

angiogenesis, it also depends on the proliferation and differentiation of other regenerating tissues

(such as epithelial cells). Normally, it is difficult to distinguish on the basis of a tissue

regeneration assay if a drug is achieving this effect through the inhibition of angiogenesis, cell

proliferation, cell differentiation or all of the above. However, the larval assay specifically

evaluates avascular tissue regeneration, since regeneration of the larval fin bud does not involve

blood vessel growth [104]. Fenretinide is able to inhibit proliferative processes involved in

avascular larval fin regeneration and angiogenesis-dependent adult fin regeneration. The most

probable mechanism utilized by fenretinide is the inhibition of FGF signalling, which has been

documented in vitro [224]. The FGF signalling pathway induces proliferation of many cell types;

stimulating the proliferation of ECs as well as mesenchymal cells during wound healing [245].

A MTT assay was performed in HUVEC cells by my predecessor, who noted a biphasic

response in cell viability with increasing concentrations of fenretinide (appendix 3.4.2.1). I

expanded the range of my assay to determine if it was possible for fenretinide to promote fin

regeneration at low concentrations (as low as 0.1 µM; appendix 3.4.2.2). Beneath a concentration

of 2.5 µM, there was no distinguishable difference between fenretinide treatment and the control.

Page 79: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

66

I repeated the fin regeneration experiment with transgenic vascular-labelled fish

Tg(Flk1:EGFP) to try to visualize the growth of new blood vessels. I was unable to observe any

fluorescence in the tail in either the control or the treated fish due to the limitations of the

microscope and the opacity of the tissue. Using transparent adult fish Tg(fli1:EGFP;casper) in

this assay may ameliorate these problems. Extending the endpoint to 7 dpa may also improve the

assay, since adult fish require 7 days to completely grow back their tail fins [104]. Nonetheless,

the fin regeneration and EC migration results are in line with literature reports of the anti-

angiogenic and anti-proliferative actions of fenretinide [223]. These findings dismiss the use of

fenretinide in stimulating tissue regeneration but demonstrate that fenretinide’s ability to

promote of angiogenesis is limited to a particular developmental window.

3.4.3 Fenretinide promotes angiogenesis through somitogenesis

The chevron-like arrangement of somites in the trunk of developing zebrafish is a distinct

and invariable pattern. Control fish can be easily distinguished from fenretinide-treated fish due

to the unusual curved-tail phenotype of the latter, even without the aid of a microscope (appendix

3.4.3.1). Under high magnification (bright field microscopy), the presence of small chevrons can

be seen in the tail bud of fenretinide-treated embryos (appendix 3.4.3.2). An

immunohistochemistry (IHC) stain was performed on embryos to compare the arrangements of

somites (Fig.3.3.4). The segmental bundling of fibers coincides with the presence of blood

vessels in control embryos (Fig.3.3.4A). The avascular region of the tail bud is normally

unsegmented and muscle fibers are split into two branches. Fenretinide-treated embryos have

somite segments and blood vessels extending to the tail tip (Fig.3.3.4B and D). Furthermore, this

aberrant pattern prevails even with the inhibition of blood vessel growth, via the application of

Page 80: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

67

VEGF inhibitor SU5416 (Fig.3.3C and D). These results suggest that fenretinide’s promotion of

angiogenesis in developing embryos is actually secondary to somitogenesis.

3.4.4 Fenretinide’s effect on somitogenesis

If fenretinide is able to stimulate somitogenesis in the tail bud, the most plausible

mechanism is through RA signaling, which coordinates embryo patterning during development.

Developmental patterning is conserved in vertebrates and has been studied in detail. In

zebrafish, somites form in pairs from 10-24 hpf, with about 30 somite pairs forming in total

[246]. RA signaling stimulates differentiation and FGF signaling promotes the proliferation of

stem cells in the anterior and posterior regions of the zebrafish trunk respectively [247]. There is

mutual, lateral inhibition between these two pathways. FGF8 represses RA synthesis by

downregulating the expression of the raldh2 enzyme and upregulating cyp26-mediated RA

degradation [248]. Likewise, RA has been shown to attenuate the expression levels of FGF8

[249]. The sequential formation of somite pairs is coordinated by timed fluctuations in the

gradients of RA and FGF.

Disturbing the dimensions of RA and FGF signaling can easily alter the segmentation

program. Increasing FGF or decreasing RA leads to the “caudalization” of the embro.

Implantation of FGF-soaked beads [250] and RALDH2 knockdown [251] leads to the production

of smaller somites and the enlargement of the undifferentiated presomitic mesoderm. In

comparison, decreasing FGF and increasing RA lead to “anteriorization” of the embryo. The

inhibition of FGF signaling shifts the boundary of anterior signaling lower, resulting in the

development of large somites [252]. Embryos treated with exogenous RA during somitogenesis

Page 81: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

68

end up with severely truncated bodies, with the complete loss of the posterior features beyond

the yolk sac [253].

The formation of extra somites I observed in fenretinide-treated embryos is compatible

with the “anteriorizing” effect of excess RA. It is likely that fenrentinide is acting as an

exogenous source of RA to promote somitogenesis in the tail. By extending somite development

into the tail tip, fenretinide is also causing vessels to develop along the extended somites

boundaries. If fenretinide is assuming the role of RA, I should be able to detect the changes in

the expression levels of receptors, RA-metabolizing enzymes and target genes.

I intended to assess the localization and intensity of RAR and FGFR expression in

fenretinide-treated embryos with RNA in situ hybridization (ISH). Normally, FGF signaling in

the posterior trunk excludes RA activity [248]. ISH labeling of FGFR should depict a decrease in

the tail in response to fenretinide treatment [254]. The corresponding RAR expression, which is

absent in control embryos [243], should increase in the tail of treated embryos. In my

preliminary ISH for RAR, I observed an increase in RAR in fenretinide- and RA-treated fish

relative to the control (Fig.3.3.5). The RAR expression is most intense in fenretinide-treated fish,

which may be attributed to the higher concentration, or the potency of the drug. This experiment

is to be repeated with equal concentrations of RA and fenretinide. Additionally, a glycerol-based

mounting method will be employed to improve the resolution and transparency of the ISH.

Fenretinide has reportedly hydrolyzed to RA in vivo [255]. If fenretinide (an amide) is

hydrolyzed to RA then we should be able observe an increase in RALDH expression in treated

embryos. The change in RALDH expression can be compared in response to systemic drug

treatment or localized implants of fenretinide- and RA-soaked beads. If RALDH expression does

Page 82: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

69

not change, then it is possible that fenretinide is exerting its effects through other metabolites

such as N-(4-Methoxyphenyl)retinamide (4MPR) or 4-oxo-4HPR and their associated enzymes

[256]. Alternatively, I could also investigate the RA-degrading enzyme cyp26, which is one of

the gene targets of RAR activation [198]. If fenretinide is able to bind to RAR, a relative increase

in cyp26 should be observed in treated embryos.

The expression levels of cyclic genes and FGF target genes after fenretinide treatment can

also be examined to see whether they will be changed in a manner comparable to exogenous RA

treatment. Excess RA in the tail (knockdown cyp26a or exogenous application of RA)

contributes to asymmetric expression of all cyclic genes such as her1 and her7 [257, 258].

Her13.2 has been shown to be regulated by FGF signalling [258]. Using ISH we expect to see

reduced or absent expression of her13.2 in the tailbud of fish treated with 4HPR and RA

compared to vehicle treatment. Likewise, we expect to see a reduction in the expression of FGF

target genes (e.g. Hox genes c6-10) that are involved in caudalization signalling of body

extension during embryogenesis [247].

3.4.5 The teratology of retinoids and fenretinide

Levels of RA are maintained within a specific range during normal embryogenesis and

the wrong concentration at the wrong period in development can produce congenital

malformations in vertebrates [259]. Rats, whose mothers are Vitamin A-deficient, are born with

malformations in the eye, urogential tract, diaphragm, heart and lung [260]. Excess vitamin A

during gestation can cause resorption of the embryo, and craniofacial and ocular defects [261,

262]. These effects are likely mediated by RAR activation, since RAR was found to be induced

following RA treatment in a mouse developmental model [262]. There is a 40% risk for

spontaneous abortion and 26% risk for major malformations (of craniofacial, cardiac, thymic and

Page 83: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

70

neural structures) reported in cases of embryonic exposure to exogenous RA during early

pregnancy in humans [263, 264].

During the preclinical phase of its development, the teratogenicity of fenretinide was

tested in rats and rabbits. Fenretinide was characterized as a weak teratogen, with low incidences

of craniofacial malformations such as hydrocephalus, micropthalmia and misshapen skull bones

[228]. However, pharmacokinetic studies reveal that fenretinide did not reach significant levels

in fetuses until 10 days of dosing, on gestational day 15 [228]. Considering that somite formation

and many other RA-dependent processes commence at gestational day 9-10 in rats, different

findings may be observed if the dosing regimen commenced at an earlier stage of development

[265]. In a clinical setting, 200 mg of 4HPR is administered orally per day [234]. A series of

maternal oral intake dosages of fenretinide should be studied to determine if RA-like teratogenic

effects would develop in humans.

A zebrafish teratogenicity assay has recently been developed by a group of investigators

affiliated with the pharmaceutical company Bristol-Myers Squibb. Guidelines for numerical

morphological scoring have been established, enabling the quantification of the teratogenicity of

a compound [266]. The authors of the study have tested a number of chemicals in this assay and

were able to identify 87% of the teratogens correctly [267]. RA was one of the compounds to be

scored and identified as a teratogen. The protocol for embryo treatment is virtually the same as

the developmental angiogenesis assay, although wild-type embryos are employed and embryos

are assessed later in development, at 5 dpf. This assay is a simple and convenient method of

assessing the teratogenicity of fenretinide. Using the bright-field images I have obtained from the

angiogenesis assays, I can already identify abnormalities in a number of the structures and organ

systems (e.g. eyes, skull, notochord, somites, and tail) in 2.5 dpf fenretinide-treated embryos

Page 84: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

71

(appendix 3.4.3.2). It would be interesting to investigate the teratogen score of fenretinide

relative to RA and other retinoids.

Isotretinoin (a close relative of RA) is regarded as the biggest teratogenic threat since

thalidomide [268]. When you are dealing with teratogenicity, it is not the same as other

potentially adverse side effects. “There is no safe minimal dose” and health practitioners exercise

caution in their use of retinoids as a result [208]. By extension, all systemically administered

retinoids are considered to be teratogenic, especially if exposure occurs during the first trimester

[208]. Retinoid treatment presents a high risk for spontaneous abortion and the development of

retinoid embryopathy, a term used to describe the collection of craniofacial, cardiovascular,

thymic, nervous and limb anomalies that develop in children exposed to retinoids in utero [269].

Consequently, the clinical use of oral retinoids has been restricted against women with positive

pregnancy tests. In clinical trials for fenretinide, pregnancy is listed among the exclusion criteria

[231]. In addition, female patients of child-bearing age are advised to use contraceptives during

the course of RA treatment and programs are being implemented to raise awareness and increase

compliance [270]. In America, a FDA-mandated program (iPledge) has been employed to

monitor and prevent pregnancy in patients being treated with isotretinoin [270].

3.4.6 Conclusions and future directions

In a zebrafish angiogenesis chemical genetic screen, we identified fenretinide as a pro-

angiogenic agent. Through compound validation and drug mechanism studies, we characterized

that the “pro-angiogenic effect” of fenretinide is secondary to its effect in promoting

somitogenesis, and fenretinide is a potential teratogen

Page 85: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

72

So far, fenretinide’s clinical application has involved a patient population where

pregnancy is not a common occurrence (i.e. cancer patients). As its applications extend to

chemoprevention and the treatment of other diseases (e.g. diabetes, cystic fibrosis), the detailed

characterization of fenretinide’s teratogenic potential becomes a priority. If fenretinide

demonstrates the capacity to act as an exogenous source of RA during critical periods of

development and shows teratogenicity in an animal model of development, then it should be

regarded as a dangerous teratogen as RA. My studies in zebrafish models strongly support that

fenretinide is a teratogen and detailed studies in other mammalian model systems are needed to

validate this finding.

Page 86: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

73

3.5 Appendix

Appendix 3.4.2.1. MTT assay performed on HUVEC cells treated with fenretinide (Hit #4). HUVEC cells were seeded in 96-well plates and viable cells were measured in triplicate. A biphasic growth response was observed, with increased cell viability at low concentration and decreased viability at high concentration of fenretinide. These assays were performed by Chunyang Wang (unpublished).

Appendix 3.4.2.2. Fenretinide decreases adult zebrafish fin regeneration. At lower concentrations of fenretinide, the amount of fin regeneration is similar to the control. Contrary to the MTT results, a low concentration of fenretinide is unable to improve cell survival in vivo. N=2-6

**  

**  

0  

5  

10  

15  

20  

0   0.1   0.25   0.5   1   2.5   5   10  

%  Fin  Regen

erated

 

4HPR  Concentra9on  (μM)  

Page 87: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

74

Appendix 3.4.3.1. [A] Bright field image of a 2.5 dpf albino zebrafish embryo treated with 0.5% DMSO, under high magnification (40x). [B] Bright field image of 2.5 dpf albino zebrafish embryo treated with 4 µM 4HPR under high magnification (40x). Note the curvature of the tail, smaller eyes and skull, and abnormalities in the somite and notochord of the tail.

Page 88: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

75

CHAPTER 4: ANTI-ANGIOGENIC ACTIVITY AND POTENTIAL MOLECULAR MECHANISMS OF INDIRUBIN-3-MONOXIME

4.1 Introduction Historically, natural products have been a rich source of therapeutic compounds [271]:

73% of cancer therapeutics approved to date are derived from natural sources [272]. In the

1970’s and 1980’s this trend surged as pharmaceutical companies modified many naturally-

derived compounds for a variety of therapeutic applications [273]. Two of the compounds I am

studying can trace their origin to this era.

4.1.1 Traditional Chinese medicine and the discovery of indirubin

Traditional Chinese medicine (TCM) has been practiced for millennia and is still used

today. Many of the herbal remedies used in TCM have been investigated within the paradigm of

Western medical research. The active components of medicinal plants have proven to be an

excellent resource for the development of new drugs.

Dang Gui Long Hui Wan is a traditional Chinese medical preparation employed in the

treatment of myelocytic leukemia. The powder derived from indigo naturalis was identified as

the active constituent out of the eleven herbal ingredients contained within this prescription,

[274]. I. naturalis has hemostatic, anti-pyretic, anti-inflammatory, sedative, anti-bacterial and

anti-viral properties [275]. The anti-leukemic property of the powder was attributed to indirubin;

a red indole (aromatic heterocyclic compound) found in minute quantities (0.11%) in the leaves

of indigo-producing plants such as indigofera tinctoria [276].

Page 89: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

76

4.1.2 Applications and properties of indirubin Indirubin was able to reduce the tumour burden of mice xenografted with Lewis lung

carcinoma and Walker carcinoma with minor toxicity [277]. Another preclinical study in dogs

assessed the relative safety of indirubin using 25 times the therapeutic dosage for half a year.

Although diarrhea and liver damage were noted side effects, there was no apparent

cardiovascular or renal toxicity [278]. In clinical trials, indirubin was able to produce beneficial

effects in patients with chronic myelocytic leukemia. Complete remission was observed in 26%

of patients and partial remission in 33% [279]. Indirubin was well tolerated with minor

gastrointestinal effects such as diarrhea, nausea, and vomiting [279].

Derivatives of indirubin have been synthesized to retain its useful properties while

improving its limitations (i.e. poor solubility and absorption and mild gastrointestinal toxicities

[280]. Among these analogues is my second compound of study, indirubin-3-monoxime (I3M;

Fig. 4.1.2).

Page 90: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

77

Figure 4.1.2 The discovery of indirubin and chemical structures indirubin and its derivative, I3M. Indirubin was isolated from Qing Dai, a medicinal powder contained within Dang Gui Long Hui Wan, a Traditional Chinese Medicinal preparation used in the treatment of myelocytic leukemia. Indirubin undergoes imine formation to produce I3M.

4.1.3 Molecular mechanisms of indirubins The anti-leukemic properties of indirubin have largely been attributed to two mechanisms

of action: cell cycle disruption and induction of cell death.

Indirubin demonstrated early in its development, the ability to block cell proliferation by

inhibiting DNA synthesis of leukemia cells in vitro [281] and in vivo [282]. Indirubin and I3M

selectively inhibit the phosphorylation of cyclin-dependent kinases (CDKs) [276]. The activity of

CDKs is essential to the regulation of the cell cycle; progression through all the phases of the cell

cycle is coordinated through the activation and inhibition of CDKs [283]. The advancement of a

normal cell cycle is regulated at three checkpoints to evaluate the integrity of DNA and the

Page 91: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

78

preparedness of the cell for imminent cell division [275]. Many cells that lack control

mechanisms for these checkpoints end up with deregulated expression of CDKs, deviant cell

cycle progression and uncontrollable cell proliferation that is characteristic of cancer [275]. The

ability of indirubins to inhibit CDKs allows them to target a specific feature that enables cancer

growth. Although the mechanisms are not well understood, indirubin and its derivatives have

demonstrated the ability to induce cell cycle arrest in a variety of cancer cell lines [284, 285].

Several studies have also demonstrated I3M’s ability to induce apoptosis. Tumour cells

treated with I3M experienced cell death, evident from the change in cell morphology (membrane

blebbing, chromatin condensation and DNA fragmentation), caspase activation, PARP cleavage,

and cytochrome c release [286]. I3M inhibition of the NF-κB pathway appears to sensitize cells

to apoptosis in response to TNF-activation or the application of chemotherapeutic agents such as

taxol [287].

I3M exerts effects on several other processes that are involved in tumourigenesis. It

inhibits c-Src kinase, which is involved in the STAT signalling that mediates cell proliferation,

tumourigenesis and metastasis [288]. Another intracellular target of I3M is the tyrosine kinase,

FGFR1. I3M-induced inhibition of FGF signalling reduces cell proliferation of a leukemia cell

line just as potently as FGFR1 inhibitor SU5402 [289]. Both indirubin and I3M have

demonstrated the ability to inhibit angiogenesis in vitro and in vivo [290, 291]. I3M also inhibits

glycogen synthase kinase-3 (GSK-3), a kinase involved in the regulation of apoptosis, cell

growth, differentiation, and inflammation [292, 293]. I3M’s negative regulation of cell

proliferation, inflammation and angiogenesis translates to its efficacy in vivo, as all three

processes are overactive during tumourigenesis.

Page 92: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

79

4.1.4 Indirubin-3-monoxime as an anti-cancer agent The 3’-monoxime modification of indirubin enhances its solubility, kinase inhibition and

cell permeability [276, 294]. Consequently, I3M is more potent and more soluble than indirubin

[284]. I3M inhibits the proliferation of many types of cancer cells with minimal toxicity to

normal cells [295]. I3M is a strong and selective inhibitor of CDKs and GSK-3, in addition to

many other intracellular kinases [296]. It has the ability to inhibit neovascularisation in the aortic

ring and Matrigel plug assay through the inhibition of VEGFR activity, though the mechanism is

currently unknown [291]. Furthermore, oral I3M treatment has demonstrated the capacity to

reduce tumour growth in a mouse model of lung cancer [297]. Short-term animal studies of I3M

have not observed any toxic side effects [298]. Collectively, literature regarding I3M indicates

that it is able to selectively target multiple pathological processes in cancer and is relatively safe

to use. Thus, it is a promising anti-cancer agent.

4.1.5 Additional applications of I3M Although I have indicated that I3M promotes the induction of apoptosis, it is also able to

prevent apoptosis through the inhibition of cellular kinases. CDK activation is implicated in the

induction of neuron death in Alzheimer’s disease. I3M, a selective CDK inhibitor, demonstrates

neuroprotective effects by preventing tau phosphorylation in vitro, through inhibition of

amyloid-β induced apoptosis of neuroblastoma cells [299]. These effects translate in vivo, as

I3M treatment was shown to reduce memory impairment and neuropathology in a transgenic

mouse model of Alzheimer’s disease [300]. I3M can also downregulate apoptosis through the

inhibition of glycogen synthase kinase-3 (GSK-3), to exert protective effects in renal and hepatic

injury [301]. The inhibition of c-Jun NH2-terminal protein kinase (JNK) is another means of

preventing apoptosis, as demonstrated in cerebellar granule neurons [302].

Page 93: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

80

Indirubin and I3M have also demonstrated immunomodulatory activities. Indirubins

decrease production of the chemokine RANTES, implicated in inflammatory cell recruitment

during the pathogenesis of influenza [303]. Indirubin treatment suppresses the production of a

number of factors involved in the inflammatory response such as interferon γ, IL-6, and NF-κB

[287, 304, 305]. Similarly, I3M acts on NFκB and JNK to lower levels of NO, prostaglandin E2,

IL-1β and IL-6 in cells with lipopolysaccharide-induced inflammation [306]. These findings

support the potential application of I3M as a therapeutic agent for various inflammatory diseases.

Many of I3M’s kinase targets are implicated in the pathogenesis of a diversity of

diseases. Among the most recently proposed applications of I3M are its uses in treating type 2

diabetes and mood disorders [292]. Additionally, a brominated derivative of I3M has shown the

capacity to maintain pluripotency in embryonic stem cells through Wnt activation [307].

Indirubins can be used to prolong self-renewal in stem cell cultures, thereby overcoming a major

limitation in stem cell-related biomedical research and regenerative medicine [308].

4.1.6 Rationale I3M was identified in our initial zebrafish screen as a negative modulator of

angiogenesis. A literature review of indirubin activity has indicated that I3M has demonstrated

anti-angiogenic properties in HUVEC and Matrigel plug assays. However, the mechanism by

which I3M inhibits angiogenesis has not yet been identified. In my studies, I will evaluate the

anti-angiogenic properties and investigate the mechanisms of its actions in zebrafish models of

angiogenesis. These studies will help demonstrate I3M’s value as an anti-angiogenic agent in

vivo for future applications in the treatment of diseases characterized by excessive angiogenesis.

Page 94: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

81

4.2 Materials and Methods

4.2.1 Preparation of drugs and reagents

Indirubin-3-monoxime (I3M) and γ-secretase inhibitor DAPT were purchased from

Tocris Bioscience (Ellisville, MI, USA). Refer to text and section 3.2.1 for source and

preparation of other drugs and reagents respectively. Embryos and larvae were treated with 2, 4,

8, and 16 µM of I3M and/or 100 µM of DAPT and adults were treated with 5 and 10 µM of I3M.

4.2.2 Zebrafish strains and husbandry Refer to 3.2.1

4.2.3 Cell lines

Several human cancer cell lines were used in the zebrafish xenograft assay. Breast cancer

MDA435 DsRed-expressing cells that express normal and excess amounts of the gene RhoC

were provided by Dr. Konstantin Stoletov (University of San Diego). Several prostate cancer cell

lines (PPC-1, PC3 and DU145) were modified to express mCherry by James Wang (Dr. Jeff

Medin lab, University of Toronto). These cells were cultured in DMEM or RPMI media

containing 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (Multicell, Wisent Inc)

and passaged biweekly.

4.2.4 Zebrafish developmental angiogenesis assay Refer to 3.2.3

4.2.4 In vivo quantification of EC number The transgenic zebrafish line, Tg(fli1:nEGFP)y7

expresses EGFP in the nucleus of ECs,

permitting the quantification of EC number in specific vascular structures like the DA, DLAV,

Page 95: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

82

and ISV. Embryos were treated with I3M and DAPT (alone and in combination) as described in

section 3.2.4 with the following modifications; cell counting of “good” fluorescent images (non-

blurry, with low background fluorescence) were performed with digital assistance and lower

quality images were analyzed by manual counting.

Cell number in the ISV and DA were manually counted and averaged across 10 segments

of each embryo, and then expressed as an average of all samples treated with each drug

concentration. Manual cell counting in the DLAV determined a total number of DLAV cells in

each fish, which was averaged for each condition.

A custom algorithm was developed to analyze and batch process the number of nuclei in

each vessel type within the Matlab software (MatLab V.7.6, The MathWorks, Natick, MA,

USA). The Matlab program analyzed each image under two specifications recognizing high or

low thresholds for fluorescent intensity. The average of the conservative and non-conservative

outputs produced cell counts that were within 10 nuclei of the manual count. The final cell

numbers were manually corrected to include missed nuclei (e.g. overlapping nuclei) by visually

assessing each Matlab output. The data was compiled in Microsoft Excel and statistically

analyzed using ANOVA and a two-tailed student t-test. Error bars denote SE.

Page 96: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

83

Figure 4.2.4. Sample images generated from MATLAB’s semi-automated cell counting algorithm, which was able to quantify the number of cells (overlay in blue) in the ISV, DA and DLAV.

4.2.5 Fin regeneration assay Refer to 3.2.5

4.2.6 Embryonic tumour xenograft model

Newly fertilized eggs were collected following natural breeding of Tg(Flk1:EGFP) or

Tg(Fli1:nEGFP)y7 fish. At 12 hpf, propylthiouracil (PTU) was added to inhibit the development

of embryonic pigmentation. Tg(fli1:EGFP;casper) transparent embryos were also used. At 2-3

dpf, embryos were immobilized in embryo medium containing 100ppm eugenol. In preparation

for microinjection, human cancer cells were washed with D-PBS and treated with non-enzymatic

cell-dissociation solution (Sigma-Aldrich, Oakville, ON, Canada) to remove adherent cells from

the surface of the tissue culture flask. Cell concentration was calculated by staining cells with

Page 97: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

84

0.4% trypan blue (Medstore, Toronto, Canada) and counted with the aid of a hematocytometer

(VWR Scientific, 15170-208) and a hand-held tally counter. The cell suspension was iced or

maintained at room temperature until microinjection.

Embryos were microinjected in the perivitelline space (yolk sac) using a Femtojet

microinjector (Eppendorf) and permitted to recover in embryo medium for 15-30 minutes.

Control embryos were injected with vehicle, containing cell dissociation solution. Healthy

embryos were injected with a moderate amount of cancer cells (between 10-100 cells) were

selected, anesthetized, mounted onto a layer of agarose, imaged and dispensed into 96-well

plates. Each well housed one embryo to prevent contamination in the event of embryo mortality.

I3M and DMSO were added into the embryo water and incubated at 28 or 32°C. Two days post-

injection, the embryos were imaged with a fluorescence stereomicroscope using the SCORE

mounting method. SIVs, endothelial and cancer cell numbers were compared to assess tumour

angiogenesis and metastasis.

Page 98: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

85

Figure 4.2.6 Workflow of a zebrafish embryonic tumour xenograft assay. Newly fertilized eggs are collected and dosed with PTU 12 hpf to block pigment formation. At 2-3 dpf, embryos are microinjected with cancer cells and dispensed into wells of a 96-well plate. Drugs are added directly into the fish water. The embryos are imaged with fluorescence 2 days post injection (dpi). SIV morphology was analyzed to evaluate the drug effect on tumour angiogenesis. Figure reprinted with permission from Elsevier Limited (review paper in press).

Page 99: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

86

4.3 Results

4.3.1 I3M decreases ISV number in developing zebrafish embryos

I3M was the most potent inhibitory compound identified in the pilot screen of the

LOPAC library. I administered increasing concentrations of I3M in the zebrafish angiogenesis

assay. I3M dose-dependently inhibits vessel development in Tg(Flk1:EGFP) zebrafish embryos

resulting in an decrease in the number of ISVs (Fig. 4.3.1). Control embryos (0.5% DMSO)

develop an average of 27.5 ± 0.1 ISVs. Embryos treated with 4 µM I3M develop 16.3 ± 1.2

ISVs, treatment with 8 µM results in 3.3 ± 1.1 ISVs (p<0.01). With 16 µM I3M treatment, vessel

development is almost completely inhibited, with 0.7 ± 0.4 ISVs developing (p<0.01).

Figure 4.3.1. [A] Dose-dependent effect of I3M on ISV development in Tg(flk1:EGFP) zebrafish embryos after 48h of treatment. I3M decreases ISV development in 2.5 dpf zebrafish embryos. Fluorescent images are shown of embryos treated with [B] a control of 0.5% DMSO (27.5 ± 0.1 ISVs), and [C] 4 µM I3M (16 ± 1.2 ISVs).*p < 0.05. ** p < 0.01. N=9-13. Error bars denote SE.

Page 100: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

87

4.3.2 I3M inhibits endothelial cells migration in vivo

Tg(Fli1:nEGFP)y7 embryos (nuclear-localized GFP expression in ECs) were treated with

I3M to evaluate changes in the number of endothelial cells in each vessel (Fig. 4.3.2). The

average cell number dose-dependently decreases in the ISVs from 7.4 ± 0.2 in controls to 3.6 ±

0.5 in 8 µM and 1.8 ± 0.3 in 16 µM of IO (p<0.01). Average cell number increases in the

segments of DA from 8.2 ± 0.2 in controls to 10.6 ± 0.4 in 8 µM and 12.8 ±0.6 in 16 µM of I3M

(p<0.01). It is interesting to note that the sum of DA and ISV ECs are similar between control

and various concentrations of I3M-treated embryos (Fig. 4.3.2 A&B), suggesting that I3M

affects endothelial cell migration from DA to ISV.

Figure 4.3.2. The average cell number in the [A] intersegmental vessel (ISV) and [B] dorsal aorta (DA) in Tg(fli1:nEGFP) zebrafish embryos after 48h of I3M treatment. Fluorescent images of zebrafish embryos at 2.5 dpf show embryos treated with [C] 0.5% DMSO (control) and [D] 16 µM I3M. Treatment with I3M decreases ISV cell number and increases DA cell number. *p < 0.05 **p< 0.01. N=9-14. Error bars denote SE.

Page 101: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

88

4.3.3 I3M inhibits larval and adult fin regeneration

A fin regeneration assay was performed in adult zebrafish to examine the anti-angiogenic

effect of I3M in an adult organism. Because tissue generation is dependent on other processes

(proliferation, migration, differentiation) as well as angiogenesis, a larval (avascular) model was

also employed. I3M dose-dependently inhibits fin regeneration in larvae (Fig. 4.3.3.1) and in

adults (Fig.4.3.3.2). 16 µM of I3M reduced the length of fin regenerated in the control, by more

than half (50 and 86 µm respectively; p<0.01). In the adult model, 5 µM reduces fin regeneration

to 7.5 ± 0.6% of the original fin length, and 10 µM decreases regeneration to 3.1± 0.4 %

compared to a control regeneration of 13.8 ± 0.6% (p<0.01).

Figure 4.3.3.1 [A] Effect of I3M on larval zebrafish fin regeneration. Fins are transected at the end of the notochord and fish are incubated in drug for 3 days. Bright field images of caudal fins 3 days post amputation reveal that fish treated with [B] 0.5% DMSO regenerate an average of 86 ± 6 µm of fin bud while fish treated with [C] 16 µM I3M regenerate 50 ± 10 µm of the original fin length. *p < 0.05. ** p < 0.01. N=9-12. Error bars denote SE.

Page 102: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

89

Figure 4.3.3.2 [A] Effect of I3M on adult zebrafish fin regeneration. Fins are transected at the site of the red arrow and fish are incubated in drug for 3 days. Bright field images of caudal fins 3 days post amputation reveal that fish treated with [B] a control of 0.5% DMSO regenerate an average of 13.8 ± 0.5% of their original fin length and fish treated with [C] 10 µM I3M regenerate an average of 3.1± 0.4%. IO dose-dependently inhibits fin regeneration. *p < 0.05. ** p < 0.01. N=9-10. Error bars denote SE.

4.3.4 Development of zebrafish tumour angiogenesis model and preliminary test of I3M

Tumour development is dependent on angiogenesis and consequently, many cancer cells

have the property to induce angiogenesis. Previously our lab used the mouse xenograft cancer

model to study cancer angiogenesis but this assay is time-consuming (2-3 months for each

experiment) and expensive (as it requires large quantities of chemical compounds). Therefore, I

intended to establish a zebrafish xenograft cancer angiogenesis model and test the drug efficacy

against cancer angiogenesis in 96-well plate format. This will facilitate the compound validation

process of drug development in our lab.

Page 103: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

90

I conducted transplantation experiments, grafting fluorescent cancer cells into embryo

recipients possessing fluorescent vasculature to observe the effect of a drug on cancer cell-

induced angiogenesis. The assay was time-consuming and technically demanding. Only 10-40

embryos could be injected and imaged per experiment. Embryo mortality was extremely high as

there were numerous technical challenges and variables to manipulate in this model.

Injected cells had the tendency to aggregate in the injection needle. I was able to solve

this problem by substituting a PBS injection solution with a non-enzymatic cell-dissociation

solution. The strain and age of zebrafish receiving the graft was another variable to consider. Of

the three strains used, Tg(fli1:EGFP;casper) was most convenient as PTU did not need to be

administered to inhibit pigment formation. However, Tg(Fli1:nEGFP)y7 was most effective as

nuclear fluorescent signals produced distinct visualization of the SIVs (which were intensely

fluorescent and more difficult to visualize in casper and flk embryos). The ideal age for injection

was 3 dpf, when SIVs have already formed and neovascular SIV growth is reported to occur

directionally towards the injection site. This was not observed experimentally. Instead, embryos

were injected at 2 dpf amidst SIV development. Embryos were observed 2-3 days post injection;

though at least half the injected embryos died 24 hours after injection. Comparatively, only about

a quarter of embryos injected with vehicle would die. Embryo mortality could be attributed to

three causes; injury during injection, infection of the injection site and inability to accommodate

the cancer cells injected. Embryos can reportedly tolerate injections of up to 200 cells for 3 days

of observation. The optimal cell number was 50-100 cells, such that there was an adequate

number to induce angiogenesis but not so much as to cause embryo mortality. The temperature

of incubation was also a variable factor; zebrafish are normally maintained at 28°C and cells at

36°C. I compromised between the two and incubated injected embryos at 33°C. Usually injected

Page 104: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

91

cells remain at the site of injection, though approximately 1% of embryos would show cancer

cells disseminated throughout the circulatory system.

Several cancer cell lines were evaluated to find a suitable cell type for xenografting.

MDA435 expressed red fluorescence poorly, was not particularly angiogenic and caused

significant embryo mortality (very few untreated embryos survived with MDA435; appendix

4.3.4.1). PPC-1 cells proved to be too malignant as well. Even though very small quantities of

PPC-1 were injected, the majority of embryos did not survive past 1.5 dpi (appendix 4.3.4.2).

PC3 cells were cultured but not used since cell growth was slow and media was contaminated

with particulate matter secreted by the cells. DU145 cells demonstrated the best qualities of the

cell lines used: strong mCherry expression, moderate proliferation and lower embryo mortality

(appendix 4.3.4.3).

Page 105: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

92

Figure 4.3.4.Zebrafish embryonic tumour xenograft model. [A] SCORE fluorescence imaging of Flk1:EGFP 5 day old zebrafish (vessels in green), 2 days after injection with mcherry labelled human prostate cancer cells (DU145; red). [B] Control injection with vehicle (cell dissociation solution) and treated with 0.5% DMSO. [C] Xenograft embryo treated with 0.5% DMSO. [D] Xenograft embryo treated with 8 µM IO. Note the inhibition of vessel growth from the subintestinal vessels (arrow). N=1 due to high embryo mortality.

I was able to observe an inhibitory effect of I3M treatment in a number of injected fish

(Fig. 4.3.4). However, statistical analysis was not an option considering the extremely small

sample size. After months of experimentation and over a thousand embryo injections, I stopped

trying to develop this assay. There were too many challenges to the xenograft; problems in

maintaining consistency in the injection, large variability in angiogenic response (which was

confounded by infection-induced angiogenesis) and high mortality of embryos (ranging from 50-

95%).

Page 106: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

93

4.3.5 DAPT partially rescues the I3M chemical genetic phenotype

Since I3M was shown to affect EC migration in our zebrafish cell counting assay, I

investigated a candidate pathway involved in angiogenic sprouting to elucidate I3M’s

mechanism of action. The interaction between the notch and VEGF pathways coordinates the

process of vessel sprouting during angiogenesis [98]. Notch activation has been shown to block

sprouting of ISVs in zebrafish [309] to produce a similar phenotype to a VEGF morphant [310].

The notch-activated embryos are also characterized by immature cytoplasmic extensions along

the dorsal aorta, which suggests that some ECs are able to sprout from the DA but not complete

their migration. This is reminiscent of I3M’s chemical genetic phenotype. The notch inhibitor

DAPT was co-administered with I3M in zebrafish embryos to determine if I3M is exerting its

effects through the activation of the notch pathway.

Nuclear fluorescent Tg(fli1:nEGFP)y7 zebrafish were used to quantify total endothelial

cell number in of 2.5dpf embryos after drug treatment (Fig. 4.3.5). Control embryos treated with

0.5% DMSO have an average total of 105 ± 2.7 cells in the DLAV, while 100 µM DAPT

treatment produces an average total of 100.4 ± 7.8 (p>0.05). Embryos treated with 4 µM and 8

µM I3M have an average total of 30.5 ± 4.4 and 22.2 ± 4.1 cells in the DLAV. Combined dosing

of I3M with 100 µM of the notch inhibitor DAPT significantly increased the average total

DLAV cell number to 44.3 ± 3.2 and 33.5 ± 3.4 at 4 µM and 8 µM of I3M respectively (p<0.05).

Embryos treated with I3M alone, DAPT alone and co-treated with I3M and DAPT did not have

statistically different cell numbers in the DA and ISV from those of the controls (p>0.05).

Page 107: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

94

Figure 4.3.5 Effect of notch inhibitor DAPT on cell numbers in the ISV and [A] DLAV in I3M-treated Tg(fli1:nEGFP) y7 zebrafish embryos after 48h of treatment. Fluorescent images of zebrafish embryos are shown at 2.5 dpf treated with [B] 0.5% DMSO, [C] 100 µM DAPT, [D] 4 µM I3M and [E] 4 µM I3M +100 µM DAPT. The treatment of DAPT increases the average cell number in the DLAV of I3M-treated embryos. *p < 0.05. ** p< 0.01. N=10-25. Error bars denote SE.

Page 108: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

95

4.4 Discussion

4.4.1 I3M demonstrates specific inhibition of developmental angiogenesis I3M was identified from our initial screen of the LOPAC library as an angiogenesis

inhibitor. I observed a potent, dose-dependent inhibition of angiogenesis in my assay of zebrafish

developmental angiogenesis (Fig.4.3.1). The blood vessels that make up the primary circulatory

loop, such as the DA and PCV, remained intact and functional with I3M treatment. This was

apparent even at the highest dose of I3M (16 µM), when very few angiogenesis-derived vessels

were observed. Thus, I3M may selectively affect the process of angiogenesis rather than

vasculogenesis, and the viability of ECs does not appear to be affected.

This assay was repeated using Tg(Fli1:nEGFP)y7 embryos to discern changes in EC

number within specific vessels. I observed a dose-dependent decrease in the number of ECs in

the ISV and a corresponding increase in DA cells in response to I3M treatment (Fig.3.3.2). The

increase in DA cells indicates that ECs proliferate but do not migrate from their original location.

I3M may be inhibiting developmental angiogenesis in zebrafish by acting on mechanisms that

prevent cell migration. I3M’s ability to inhibit this particular stage of angiogenesis has also been

observed in vitro. In an endothelial cell scratch assay, I3M treatment dose-dependently hindered

the migration of HUVECs [291].

During the course of these studies, I was also able to coarsely evaluate the safety of I3M.

I did not observe the diffusion of nuclear-GFP expression in Tg(Fli1:nEGFP)y7 embryos

(Fig.4.3.2D). Thus, I3M may not be inhibiting angiogenesis through generalized induction of cell

death. I also did not observe any obvious morphological malformations relative to control

embryos during the first 3 days of embryonic development. There was no curved body shape,

Page 109: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

96

enlarged yolk sac, no changes in skull shape or delayed pigmentation in I3M-treated embryos at

2.5 dpf (appendix 4.4.1). The teratogenicity of I3M can be more accurately evaluated with a

morphological scoring assay [266]. The embryo size decreases at high concentrations of I3M,

possibly due to a delay in development (toxicity). However, at its effective range of dosages, the

development of I3M-treated embryos is relatively normal, its effects on angiogenesis

notwithstanding. Additional assays for organ-specific toxicology can also be used to assess I3M

effects the function of the heart, liver, kidney, central nervous system, muscles and

gastrointestinal tract [76].

4.4.2 I3M inhibits fin regeneration Reportedly, adult zebrafish can only regenerate up to 1 mm of fin tissue when

angiogenesis is inhibited [104]. The results of my adult fin regeneration assay are indicative of

I3M’s ability to halt regenerative angiogenesis (Fig.4.3.3.2). However, as I noted in section

3.4.2, adult fin regeneration involves several other processes. I3M’s inhibition of fin regeneration

can be attributed to more than just the inhibition of physiological angiogenesis. The results of the

larval assay demonstrate that I3M is also able to affect avascular processes of regeneration, such

as cell proliferation, differentiation and migration (Fig. 4.3.3.1). It has been reported in literature

that I3M is able to directly target FGFR tyrosine kinase to inhibit cell proliferation as potently as

the FGFR1 inhibitor SU5402 [289]. The blockade of FGF, a prominent signalling pathway

involved in the proliferation of many tissue types, is the most likely mechanism of action for

I3M’s suppression of tissue regeneration. FGF is also a potent angiogenic factor that stimulates

angiogenesis.

Page 110: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

97

4.4.3 I3M efficacy in tumour models Despite the problems I encountered in setting up my xenograft assay, I was able to

observe that I3M treatment blocked SIV growths in embryos injected with DU145 (Fig. 4.3.3).

However, these findings must be interpreted with caution considering the numerous limitations

of my assay. The sample size was very small, due to substantial mortality of injected fish,

variability in the site of injection and presence of infection (which would stimulate angiogenesis

even in fish injected with injection solution). In addition, the timing of the assay (injection at 2

dpf) made it difficult to differentiate tumour-induced angiogenesis from developmental

angiogenesis. While it would be ideal to inject cells after SIV formation is complete (at 3dpf), I

was unable to observe tumour-induced angiogenesis from mature SIVs. Perhaps this would be

possible if I employed a cancer cell line that has been genetically modified to secrete

angiogenesis factors such as FGF [110].

I3M has demonstrated the ability to selectively inhibit the growth of cancer cells in vitro,

in a variety of cell lines [295]. I3M’s in vivo efficacy has also been evaluated in one study of a

[B(α)P]-induced (tobacco carcinogen) lung cancer model in mice. Oral I3M treatment dose-

dependently reduced the number of pulmonary tumours and alveolar damage by inducing

apoptosis in cancer cells [297]. While these results are encouraging, I3M needs to be tested in

more animal models of cancer to verify if I3M’s wide spread in vitro efficacy translates to anti-

tumour effects in vivo.

4.4.4 I3M inhibits zebrafish ISV development partially through the notch pathway The combined administration of I3M with the notch inhibitor DAPT increases the

number of ECs in the DLAV, partially restoring the vascular network of developing zebrafish

Page 111: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

98

embryos (Fig. 4.3.5). When notch ligands (e.g. Dll4, Jagged1) bind to a transmembrane notch

receptor, it undergoes a conformational change from a series of enzymatic cleavages. The

enzyme γ-secretase releases the intracellular domain of the notch receptor (NICD), which

translocates to the nucleus to activate the transcription of target genes. DAPT inhibits the notch

pathway in an early part of the signalling cascade by inhibiting γ-secretase activity. DAPT

partially rescues the I3M phenotype, which suggests that I3M may be achieving its effects

through the activation of the notch pathway. This can be verified by investigating relative

changes in the expression levels of notch ligands, receptors or target genes.

However, it is also possible that notch activation is indirectly stimulated by I3M. Notch

plays such an important role in the developmental process, as indicated by the role of notch

genes in somitogenesis, that a stimulator of notch would be expected to produce more deleterious

effects in embryos. This was observed in a transgenic zebrafish line that utilized a heat-shock

triggered Gal4-UAS system to overactivate the notch pathway. Heat-shock was applied to

stimulate the overexpression of NICD. Although vessel sprouting was inhibited, it was

accompanied by numerous morphological defects [309, 311]. In my angiogenesis assay, I did not

observe any obvious defects in I3M treated embryos (appendix 4.4.1). Curiously, other

investigators have reported inhibitory effects of I3M on notch signalling. I3M has been shown to

suppress the activation of the notch1 receptor, through GSK-3β [312]. This is not supported by

my in vivo results as the vascular phenotype of mutant zebrafish with deficient notch signalling

has increased vessel sprouting (with multiple filopodia resembling tree branches) [313].

I3M has been well characterized as a kinase inhibitor and there are numerous kinases

involved in angiogenic cascade. It would be most reasonable to speculate that I3M’s anti-

angiogenic effects occur through kinase inhibition. I3M acts on a variety of kinases because it is

Page 112: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

99

able to interact with the ATP-binding site of a kinase. Co-crystallization experiments have

revealed that I3M forms 3 hydrogen bonds with glutamate and leucine residues of CDK2’s ATP-

site [276]. The complementary shape and H-bonding allows I3M to fit into the ATP-binding

pocket more snugly than indirubin or ATP [276]. Similarly, co-crystal structures of I3M and

GSK-3β indicate that I3M also tightly fits into the ATP-site [314]. I3M forms 4 H-bonds with

aspartate, valine and glutamine residues and orients its apolar ring into the apolar residues found

within the hinge region of GSK-3’s binding pocket [314].

It is reasonable to speculate that I3M may also competitively inhibit ATP binding at the

catalytic sites of other kinases. I3M is able to inhibit the phosphorylation of FGFR1 in fibroblast

cells under FGF-1 stimulation, which subsequently blocked the phosphorylation of several

downstream effectors (e.g. ERK1, Src, STAT3) [289].

A recently published paper identified another tyrosine kinase receptor involved in

angiogenic signalling as a target of I3M. The phosphorylation of VEGFR2 was reported to be

dose-dependently reduced by I3M treatment in a kinase assay performed in HUVEC cells [291].

Furthermore, Kim et al (2011) were able to show that I3M inhibition of angiogenesis requires

VEGFR2-mediated signalling. I3M was unable to inhibit EC migration and tube formation in

HUVECs treated with VEGFR2-targeted siRNA [291]. The requirement of VEGFR2 in this

study strongly supports that I3M’s anti-angiogenic effects are attributed to the direct inhibition of

VEGFR2 activity.

A kinase assay can be performed with varying concentrations of I3M and ATP to verify

the kinetic mechanism of competitive inhibition. In addition, the rescue strategy Chan et al used

to study the VEGF inhibitor PTK787 can also be effectively employed by performing AKT/PKB

Page 113: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

100

or eNOS mRNA injections in I3M-treated embryos [81]. If the anti-angiogenic effects of I3M

diminish with the restoration of VEGFR2 downstream signaling in zebrafish, we can further

validate its role as a VEGFR2 inhibitor in vivo. The co-crystallization of I3M with VEGFR2 is

needed to ascertain if the structural-activity relationship of I3M’s inhibition occurs through the

ATP-site, as it is with CDK2 and GSK-3β. It is probable that the potency of I3M’s anti-

angiogenic effects is due to its ability to inhibit multiple kinase signalling pathways at once.

I believe that the findings of the DAPT rescue experiment can be re-evaluated in

consideration of I3M’s function as a competitive inhibitor of VEGFR2. There is considerable

crosstalk between the VEGF and notch pathways in the process of vessel sprouting during

angiogenesis. Notch activation has been shown to limit vessel branching through negative

regulation of VEGF [315]. This is achieved through a downstream target of notch signalling,

transcription factor HESR-1, which represses the transcription of VEGFR2 [316]. DAPT

treatment increases the expression of VEGFR2 by inhibiting notch signalling [98].

Consequently, in my experiment, DAPT may be able to overcome I3M’s competitive inhibition

and restore VEGF-mediated angiogenesis by increasing the amount of substrate (VEGFR2)

available. In essence, it is a systems biology illustration of enzyme-inhibitor kinetics. Within this

proposed framework, it would be plausible to observe full restoration of angiogenesis by

adjusting the relative quantities of DAPT and I3M (increasing and decreasing respectively).

4.4.5 Conclusions and future directions

At present, I3M is at an early point in drug development, with the majority of it studies

conducted in vitro. The next step is to test its efficacy in vivo, in animal models. Although rodent

studies remain the gold standard for preclinical testing, zebrafish are uniquely capable of

expediting drug development by permitting in vivo mechanism studies.

Page 114: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

101

The results of my studies in zebrafish have supported the efficacy of I3M as a therapeutic

agent for the treatment of cancer and angiogenesis disorders. I3M inhibition of cell proliferation

and angiogenesis allows it to target numerous aspects of tumour pathophysiology, rendering it

more resistant to drug resistance. I3M’s ability to inhibit angiogenesis occurs through

competitive inhibition of the kinase VEGFR2. Co-crystallization experiments will be able to

confirm the precise interaction of I3M with VEGFR2.

The abundance of VEGF-based angiogenesis agents and their lack of clinical efficacy

limit the utility of I3M in angiogenic therapy. However, I3M’s capacity to inhibit several

aberrant cell processes as a broad kinase inhibitor can be powerfully applied in anti-cancer

therapy. Thus far, I3M has demonstrated a protective effect in the progression of lung cancer in a

mouse model. Its utility in other cancers will need to be further characterized. The safety status

of I3M in mammalian models will also need to be confirmed in the future, though I have

observed that I3M is well tolerated in adult and larval zebrafish.

The culmination of I3M’s studies thus far reveals that it shows great potential as

therapeutic agent for applications in the treatment of cancer, angiogenesis disorders, and

neurodegenerative and immune diseases. The development of I3M and its potential successes are

additionally significant because it reflects positively on the combined efforts of traditional

medicine and modern biomedical science.

Page 115: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

102

Appendix

Appendix 4.3.4.1. [A] Bright field image and [B] fluorescent image of human breast cancer cells MDA435, expressing DsRed and the metastatic gene RhoC. Note the levels of DsRed expression. [C] Fluorescent image of 5 dpf Tg(fli1:nEGFP) y7 embryos. [D] Fluorescent image of 5 dpf Tg(fli1:nEGFP) y7 embryo treated with 16 µM of I3M. Most of the fish injected with MDA435 did not survive. MDA435 cells would proliferate/migrate, bursting out of the yolk sac and causing embryo death.

Page 116: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

103

Appendix 4.3.4.2. [A] Bright field image and [B] fluorescent image of human prostate cancer cells PPC-1, expressing mcherry. [C] Fluorescent image of 5 dpf Tg(fli1:nEGFP) y7 embryos mounted in a SCORE imaging apparatus to allow rotation of the embryo [D] Fluorescent image of 5 dpf Tg(fli1:nEGFP) y7 embryo injected with PBS. [E] Fluorescent image of 5 dpf Tg(fli1:nEGFP) y7 embryo injected with PPC-1. Most of the fish injected with PPC-1 did not survive.

Page 117: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

104

Appendix 4.3.4.3. [A] Bright field image and [B] fluorescent image of human prostate cancer cells DU145, strongly expressing mcherry. Fluorescent image of 5 dpf Tg(fli1:nEGFP) y7 embryo injected with DU145, treated with [C] 0.5% DMSO and [D] 8 µM of I3M

Appendix 4.4.1. Bright field image of 2.5 dpf Tg(fli1:nEGFP)y7 embryo treated with 16 µM of I3M, at 40x magnification.

Page 118: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

105

CHAPTER 5: THE ROLE OF DHM IN ANGIOGENESIS

5.1 Introduction

5.1.1 Rotenoids Rotenoids are a group of natural and synthetic heterocyclic compounds with insecticide

properties. The core structure of a rotenoid is comprised of a linked ring system

(tetrahydrochromeno[3,4b] chromene rings) [317] (Fig. 5.1.3). Natural rotenoids have been

isolated from the seeds, fruits, leaves and stems of leguminous plants native to the southern

regions of Asia, America and Africa [318]. These plants have been historically utilized by

indigenous people for applications in traditional medicine and as pesticides [319].

Figure 5.1.3. Chemical structure of dihydromunduletone and related rotenoids.

Page 119: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

106

Rotenoids have been employed as household and industrial organic pesticides. Reports of

its bioactivity in mammals have stimulated the scientific community to investigate the potential

environmental dangers posed by rotenoids, as well as its potential use in drug discovery. The

World Health Organization has classified a common rotenoid, rotenone, as a moderately

hazardous chemical and many countries have placed restrictions on its use to curtail the

possibility of environmental damage [318]. Yet, rotenoids present a great opportunity for drug

discovery. Many extracted and modified rotenoids have been evaluated for applications in anti-

bacterial, anti-malarial, anti-fungal and anti-cancer therapies [317].

5.1.2 The anti-cancer activities of rotenoids Mundulone is a compound extracted from the plant Mundulea chaperlieri that is native to

Madagascar [320]. Mundulone was able to exert moderate cytotoxic effects (IC50 = 13 µg/ml) in

an ovarian cancer cell line [320], though further characterization of its anti-cancer activities have

not been evaluated.

Rotenone has been applied traditionally, environmentally and industrially as a natural

piscicide, a fish-killing substance. It is a lipophilic compound that is easily absorbed into the gills

of fish and enters the bloodstream to quickly produce toxic effects. Rotenone toxicity is

attributed to its ability to interfere with the transfer of electrons in the mitochondrial membrane

during cellular respiration [321]. Although its use is prohibited in several countries, its

environmental hazards are minor because it is light-sensitive and the mode of chemical exposure

(skin, gastrointestinal tract) in mammals curbs its toxicity [318]. Interestingly, rotenone has been

successfully applied in biomedical research to model Parkinson’s disease (PD). Rotenone is

injected into rats, crosses the blood-brain-barrier and damages dopaminergic neurons in a

manner that reproduces the symptoms and pathogenesis of PD [322]. While rotenone does not

Page 120: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

107

show carcinogenic or seriously toxic effects in mammals, it still poses a health risk. A recent

epidemiological study has demonstrated an association between rotenone use and the incidence

of Parkinson’s disease [323].

Deguelin is another naturally occurring rotenoid that is commercially used as an

insecticide. Actually, deguelin and rotenone can both be found in Indigofera tinctoria, which if

you recall, also contains small quantities of indirubin [324]. This plant appears to be a bountiful

source of anti-cancer chemicals since deguelin, like indirubin, exhibits anti-cancer properties.

Deguelin is able to limit the growth of breast, skin and colon cancer cells through apoptotic and

anti-proliferative effects [325, 326].

Deguelin was found to induce apoptosis in colon and gastric cancer cells by inhibiting

AKT and NF-κB signalling [327]. Deguelin’s reported inhibition of angiogenesis has also been

tested in vitro and in vivo. Deguelin is able to reduce angiogenesis in a scratch assay, tube

formation assay, a CAM assay [328]. Its anti-angiogenic effects may be attributed to the

inhibition of AKT and downregulation of VEGF and HIF-1α [329]. Deguelin also inhibits heat-

shock 90 (Hsp90), by directly binding to its ATP-site [330]. Hsp90 is overexpressed in many

cancer cells, so by targeting it, deguelin is able to downregulate Hsp-driven tumour progression,

angiogenesis and drug-induced resistance [331].

A reduction in tumour progression has also been observed in rodent models of cancer

(xenograft and carcinogen treatment) after the short term application of deguelin [330].

Deguelin’s activities appear to be selective, as pre-malignant and malignant cells respond more

sensitively than normal cells to its effects [329]. Additionally, no toxic effects have been

observed at therapeutic doses in animal models [330]. Unfortunately, long term or high doses of

Page 121: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

108

deguelin have produced neurotoxic effects resembling the PD-like symptoms produced by

rotenone [332]. Biomedical research continues to develop new rotenoid derivatives to search for

the balance between the efficacy and potential toxicity of deguelin and rotenone [333].

5.1.3 Dihydromunduletone My last compound of study, dihydromunduletone, is a rotenoid derivative identified from

the Spectrum Collection library as an inhibitor of angiogenesis in zebrafish [96].

DHM is a novel, semi-synthetic compound that has not been previously studied as an

anti-angiogenic or anti-cancer agent. In fact, a literature search on DHM only produces two hits.

Wetli et al identified DHM as a weak inhibitor of iron uptake in a cell-based screen and did not

characterize it any further [334]. The second paper, published from our lab, identified DHM as

an inhibitor of angiogenesis [96].

Although next to nothing is known about DHM, the documented activities of the

aforementioned, structurally related rotenoids could provide insights into DHM’s potential

biological activities (Fig 5.1.3).

I would like to evaluate DHM’s anti-angiogenic properties in order to assess its potential

uses in the treatment of cancer and other diseases characterized by excessive angiogenesis.

Page 122: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

109

5.2 Materials and Methods

5.2.1 Preparation of drugs and reagents DHM was purchased from MicroSource Discovery Systems Incorporated (Gaylordsville, CT,

USA). Embryos and larvae were treated with DHM at the final concentrations of 2, 4 and 6 µM,

and adults were treated with 0.5, 0.75 and 1 µM.

5.2.2 Zebrafish strains and husbandry Refer to 3.2.1

5.2.3 Zebrafish developmental angiogenesis assay Refer to 3.2.3

5.2.4 In vivo quantification of EC number Refer to 3.2.4

5.2.5 Fin regeneration assay Refer to 3.2.5

Page 123: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

110

5.3 Results

5.3.1 DHM decreases ISV number in developing zebrafish embryos

DHM reduced the number of ISVs that developed in zebrafish embryos in the pilot

screen so I administered a range of concentrations to study the dose responsiveness of the drug.

DHM dose-dependently inhibits ISV development in transgenic Tg(Flk1:EGFP) zebrafish

embryos resulting in an decrease in the number of mature ISVs (Fig. 5.3.1). Control embryos

treated with vehicle (0.5% DMSO) develop an average of 27.5 ± 0.1 ISVs. Embryos treated with

2 µM DHM develop 25.7.1 ± 0.7 ISVs (p<0.05), 4 µM develop 18.1 ± 2.1 ISVs (p<0.05), and

treatment with 6 µM results in 13.6 ± 2.6 ISVs (p<0.01). Survival of embryos treated with 2 µM

DHM was similar to controls, whereas treatment with 4 µM and 6 µM DHM resulted in the

death of 33% and 67% of treated embryos respectively.

Page 124: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

111

Figure 5.3.1. [A] Dose-dependent effect of DHM on ISV development in Tg(flk1:EGFP) zebrafish embryos after 48h of treatment. Fluorescent images are shown of 2.5 dpf zebrafish embryos treated with [B] a control of 0.5% DMSO (27.5 ± 0.1 ISVs), [C] 4 µM DHM (18.1 ± 2.1 ISVs). *p < 0.05. ** p< 0.01. N=6-19. Error bars denote SE.

5.3.2 DHM alters the number of endothelial cells in ISVs

Tg(Fli1:nEGFP) zebrafish embryos were treated with DHM and manually counted to

detect any changes in the number of endothelial cells in each vessel. The average cell number

dose-dependently decreases in the ISVs (Fig.5.3.2). Embryos treated with 2 µM of DHM have an

average of 5.4 ± 0.4 cells per ISV and 4 µM have 4.0 ± 0.4 compared to the control average of

7.5 ± 0.2 cells per ISV (p<0.01). Average cell number increases in the caudal segments of DA.

Cells were digitally counted using Matlab software with manual corrections. Embryos treated

with 4 µM of DHM have an average of 11.8 ± 0.3 compared to the control average of 8.2 ± 0.2

Page 125: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

112

cells per DA (p<0.05).

Figure 5.3.2. The average cell number in the [A] intersegmental vessel (ISV) and [B] dorsal aorta (DA) in Tg(fli1:nEGFP) zebrafish embryos after 48h of DHM treatment. Average cell number decreases in the ISV and increases in DA segments. Control embryos treated with 0.5% DMSO develop an average of 7.5 ± 0.2 cells per ISV and of 8.2 ± 0.2 cells per DA. Embryos treated with 4 µM DHM have an average of 4.0 ± 0.4 cells per ISV and 11.8 ± 0.3 cells per DA. *p < 0.05. ** p < 0.01. N=7-14. Error bars denote SE.

5.3.3 DHM inhibits adult fin regeneration A fin regeneration assay was performed in adult zebrafish to examine the anti-angiogenic

effect of DHM in an adult organism. For my other two compounds, I also assessed larval fin

regeneration to distinguish between angiogenesis and other tissue regenerative processes (such as

proliferation). I was not able to complete larval fin regeneration experiments for DHM due to

high embryo mortality. DHM dose-dependently inhibited adult fin regeneration (Fig.5.3.3). After

Page 126: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

113

3 days of drug treatment, 0.5 µM reduces fin regeneration to 9.8 ± 0.6% of the original fin

length, 0.75 µM decreases regeneration to 8.6 ± 0.7 % and 1 µM decreases regeneration to 5.9 ±

0.8 % compared to a control regeneration of 13.8 ± 0.5% (p<0.01). Survival of fish treated with

less than 1 µM DHM was similar to controls, whereas treatment with 1 µM DHM resulted in the

death of 5 fish.

Figure 5.3.3. [A] Effect of DHM on adult zebrafish fin regeneration. Fins are transected at the site of the red arrow and fish are incubated in drug for 3 days. Bright field images of caudal fins 3 days post amputation reveal that fish treated with [B] a control of 0.5% DMSO regenerate an average of 13.8 ± 0.5% of their original fin length and fish treated with [C] 10 µM 4HPR regenerate 8.1 ±0.7%. DHM dose-dependently inhibits fin regeneration. *p < 0.05 ** p < 0.01. N=9-10. Error bars denote SE.

Page 127: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

114

5.4 Discussion

5.4.1 DHM inhibits developmental angiogenesis DHM moderately inhibits angiogenesis, reducing the number of ISVs that develop in

zebrafish embryos (Fig 5.3.1). Vessels derived through vasculogenesis are intact, so DHM’s

action may be specific to the process of angiogenesis.

DHM treatment in Tg(Fli1:nEGFP)y7embryos demonstrated a decrease in the number of

ECs in the ISV and increase in the DA (Fig. 5.3.2). DHM may be inhibiting the process of EC

migration during developmental angiogenesis. The EGFP expression of control embryos is

localized to the nucleus, producing a distinct fluorescent signal (Fig.5.3.2c). I observed a diffuse

fluorescent signal in a proportion of embryos treated with 4 µM of DHM (Fig.5.3.2d), indicating

that DHM treatment has the potential to produce EC apoptosis at a higher concentration.

5.4.2 DHM inhibits adult fin regeneration DHM is able to inhibit fin regeneration in adult zebrafish as measured at 3 dpa

(Fig.3.4.2). This may be achieved through the inhibition of regenerative angiogenesis, or other

processes involved in fin regeneration, such as mesenchymal cell proliferation. On the basis of

these findings alone, it is difficult to delineate if a particular process or cell type is being affected

by DHM.

5.4.3 DHM toxicity

I observed a substantial amount of drug toxicity during the course of my experiments

with DHM in zebrafish. Although embryos can be treated with 4-6 µM of DHM, adults appear to

be more sensitive to the toxic effects of DHM. Very low concentrations of DHM were tolerated

Page 128: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

115

in adult zebrafish, and half of the fish would perish before the end of the assay. About half of

DHM-treated embryos exhibited features of toxicity and delayed development; less

pigmentation, change in body shape and enlarged yolk sacs (appendix 5.4.3). The therapeutic

window of this drug is very small in both embryos and adults, and it is accompanied by toxic and

possibly teratogenic effects.

The observed fatalities in DHM-treated fish could be associated with rotenoid fish-

specific toxicity. Compounds that are structurally related to DHM (rotenone, deguelin) have been

applied as piscicides due to the ease of chemical absorption - fish uptake rotenoids dissolved in

water more readily than mammals which absorb rotenoids through the skin and gastrointestinal

tract [318]. Rotenone inhibition of mitochondrial complex I reduces ATP and enhances ROS

production in fibrosarcoma cells [335][335]. If DHM is acting through the same mechanism, it

should produce similar changes in ATP and ROS levels in vitro.

5.4.4 DHM future studies

To assess its suitability for therapeutic use, I would first determine the specificity of

DHM’s toxicity, pertaining to its specificity of action in fish and particular cell types.

Mundulone, DHM’s most closely related compound, has demonstrated cytotoxic effects in

cancer cells, although its limited study does not distinguish between general and selective

toxicity [320]. To evaluate DHM’s anti-cancer, anti-angiogenic and cytotoxic properties, I would

propose a comparative cell survivability assay (e.g. MTT) in ECs, cancer cells and non-ECs (e.g.

epithelial cells or fibroblasts). If the in vitro studies determine DHM’s activity to be specific to

angiogenesis or cancer cells, it is necessary to validate these findings in a whole-organism

model. I would suggest a mammalian model, as previous studies performed with rotenone have

reported minimal toxicity in non-fish models, due to absorption and rapid metabolism [336].

Page 129: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

116

Several assays can be performed to validate the effect of DHM on angiogenesis. An in

vitro characterization of DHM using MTT, Ki-67 labelling or thymidine incorporation assays

can test EC viability and proliferation. PI staining and flow cytometry can analyze cell cycle

progression, and TUNEL staining can evaluate apoptosis in ECs. Cell viability or proliferation

can also be assessed in vivo in zebrafish using methods such as acridine orange staining TUNEL,

Ki-67 labeling, or a transgenic reporter line for apoptosis [337]. Additionally, a scratch assay and

tube formation assay can be performed in HUVECs to evaluate DHM effects on in vitro

angiogenesis. The next step involves the in vivo characterization of DHM’s anti-angiogenic

capacity, in a rodent Matrigel plug assay or corneal micropocket assay.

A commercially available PCR microarray specific to angiogenesis (containing 113

angiogenesis related genes) could help identify particular pathways affected by DHM (SA

Biosciences). I propose to treat HUVECs with DHM and vehicle and analyze the differential

gene expression to identify candidate pathways using whole genome microarray. Proteins in

affected pathways can then be examined as potential drug targets using target deconvolution

strategies such as affinity binding or mass spectrometry-based proteomic techniques.

Foreknowledge of DHM’s related compound, deguelin, can lead to more focused experiments

such as western blot analyses of the phosphorylation states of AKT, MAPK, NF-κB, VEGF and

Hsp90 [327, 329, 330].

5.4.5 Conclusions and future directions

DHM has demonstrated potential as an inhibitor of angiogenesis in my experiments with

zebrafish. A proper evaluation of its toxicity and specificity of action is warranted before more

detailed mechanism studies are conducted. To be considered as a potential therapeutic agent,

DHM must be well tolerated and selective in its effects. Chemical modification of the compound

Page 130: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

117

may be necessary to increase DHM’s anti-angiogenic efficacy while decreasing its general

toxicity.

5.5 Appendix

Appendix 5.4.3. Bright field images of 2.5 dpf Tg(Fli1:nEGFP)y7 treated with [A] 0.5% DMSO and [B] 4 µM DHM with 25x magnification. Note the morphological differences in the pigmentation, yolk sac size and body shape.

Page 131: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

118

CHAPTER 6 – GENERAL DISCUSSION AND FUTURE DIRECTIONS

Numerous drugs in use today were discovered through happenstance. Serendipity is well

documented in the history of medicine [338], though happy accidents are not a reliable means of

drug discovery. As the scientific community improved their understanding of the mechanisms of

disease, target-based drug discovery became the prevailing albeit ineffective strategy for the past

few decades [339]. This has been evident in the field of angiogenesis as VEGF monotherapies

(strategies directed only at VEGF signalling) have produced disappointing outcomes in the clinic

[340]. Phenotype-based approaches are able to identify new chemical candidates without prior

knowledge of a known target. Traditional cell-based phenotype assays can efficiently screen

large numbers of chemicals. However, a very limited number of drugs can translate their efficacy

from in vitro to in vivo systems. Performing screens in animal models allow for a more

comprehensive assessment of drug efficacy on the systems biology of a phenotype or disease.

The success rates of in vivo screens are higher than cell-based screens, which are unable to

simulate the complex physiology of a disease [341].

The advantage to employing zebrafish rather than mice in whole-organism chemical

screening lies in its amenability for high-throughput analysis. A large number of samples can be

evaluated at once and the phenotypic readouts are more simple and accessible. Compare the

angiogenesis assays of mice and zebrafish; Matrigel plugs and corneal micropocket assays

compared to the developmental angiogenesis assay. The former assays have more extensive

sample preparations (as they are invasive techniques), and require larger quantities of drug and

maintenance costs. ISV development in zebrafish is easy to quantify and visualize, with the

availability of transgenic and transparent lines, and hundreds of embryos can be handled together

in one assay.

Page 132: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

119

I encountered a few limitations to zebrafish screening in the course of my studies.

Incorporating a whole-organism into a chemical screen allowed for the combined assessment of

drug efficacy and toxicity. However, sometimes these phenotypes can overlap, making it

difficult to distinguish between good and bad drug candidates. Furthermore, the developmental

stage of the zebrafish may also contribute to the discovery of undesirable hits. Compounds like

fenretinide can modulate angiogenesis through the disruption of developmental processes. Other

compounds may impede angiogenesis by inducing nonspecific toxicity. DHM may be achieving

its effects this way or through species-specific rotenoid toxicity. Neither of these effects are

desired qualities of therapeutic compounds.

A thorough characterization of embryo phenotypes can help improve the identification of

hits that are poor candidates for angiogenesis therapy. Bristol-Myers Squibb has developed a

zebrafish teratogenicity assay that uses morphological scoring to quantify the teratogenic

potential of a compound. These studies have revealed a good correlation between zebrafish and

human teratogenicity [267]. A post-hoc evaluation of fenretinide-treated embryos reveals several

morphological features indicative of teratogenicity (eyes, skull, notochord, somites, and tail).

Zebrafish have also demonstrated the capacity to predict organ-specific toxicities of

environmental and pharmacological agents [76]. Large-scale genetic screens have employed a

phenoclustering strategy to computationally search for patterns between phenotypes of known

and unknown chemicals in invertebrates such as C. elegans [342]. Methods of automating

phenotype recognition have been developed to assess zebrafish angiogenesis [126] and toxicity

[343] more efficiently. Phenotype annotation will aid the characterization of drug activities and

help predict toxicities as well as mechanisms of action.

Page 133: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

120

My studies have demonstrated that zebrafish phenotypic screens can accelerate the

process of drug discovery by identifying in vivo efficacy and toxicity earlier in a drug’s

developmental timeline. The appropriate identification of toxic and teratogenic phenotypes will

be essential to distinguish the promising hits (such as I3M) from the less desirable ones (4HPR

and DHM). Although additional validation is required in cell-based assays and mouse models,

my studies support the relevance of the model in the development of human angiogenesis

therapeutics.

Chemical and genetic screens performed in zebrafish have demonstrated strong

correlations to the physiology observed in mice and men [76]. Zebrafish absorption, distribution,

metabolism and excretion (ADME) have been representative of higher vertebrates but systemic

characterization of ADME is necessary to improve the comparative analysis of drug

pharmacology. New technologies are continually being developed to improve the speed and

precision of zebrafish handling, imaging and processing.

Fully automated HTS platforms represent the pinnacle of efficiency for drug screening.

Our lab at St. Michael’s Hospital has recently acquired a fully automated zebrafish screening

facility that is capable of performing high-throughput screens for angiogenesis modulating drugs.

The system is able to automate embryo sorting and dispensing, drug dosing, imaging and image

analysis. Based on the estimated hit rate of the pilot screens, we hope to identify over 100

angiogenesis-modulating compounds in the next few years.

The strategies I have employed in my investigation of 4HPR, I3M and DHM will

hopefully serve as a guide for more detailed characterizations of new compounds identified in

future angiogenesis screens. The characteristics of a drug’s activity on a particular physiological

Page 134: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

121

process (such as wound healing or somitogenesis) or molecular pathway can also be probed in

zebrafish assays and methods of phenotype analysis (phenoclustering or morphological scoring).

The zebrafish model is a robust tool for drug discovery and its utility is continually being

improved and validated against other animal models. Zebrafish are expected to contribute

extensively to preclinical drug development, complementing data found in other model systems.

The power of the zebrafish model will allow biomedical science to optimize and “systematize the

serendipity of drug discovery” [344].

Page 135: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

122

CHAPTER 7 – GENERAL CONCLUSIONS

The traditional process of drug discovery has a very low success rate. In oncology, only

5% of preclinical anti-cancer agents are approved for clinical usage [345]. Drug attrition is

mainly caused by the lack of efficacy and safety of drug candidates [346]. Studying the effects of

a compound within the physiological context of a whole-organism allows for the characterization

of a drug’s efficacy and toxicity earlier in the drug development pipeline [347]. Small molecules

isolated from natural sources, and their derivatives, are a promising class of compounds for drug

discovery. These compounds are more likely to possess bioactivities that can be exploited for

therapeutic applications [273]. I have used zebrafish to characterize three angiogenesis-

modulating compounds identified through a small molecule screen of two compound libraries.

Although fenretinide was identified in our screen as a promoter of angiogenesis, I was

able to determine that it achieved this effect by interfering with somitogenesis in zebrafish.

Fenretinide is currently being evaluated in clinical trials for prophylactic and therapeutic

applications in a variety of health conditions. If these findings translate to mammalian models,

then patients undergoing fenretinide treatments should be strongly advised about the risk of

developmental defects in fetuses.

I3M was identified in our screen as a potent inhibitor of angiogenesis. My studies in

zebrafish angiogenesis and fin regeneration have characterized it as an agent that produces anti-

angiogenic effects. I have identified notch as a potential pathway involved in I3M’s anti-

angiogenic chemical genetic phenotype. Based on I3M’s kinase activity in the literature, it is also

likely that the anti-angiogenic activity and DAPT rescue I observed were produced through

modulation of the VEGF pathway, as the literature has documented cross-talk between these two

Page 136: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

123

pathways. Although additional studies are required to validate this theory, my studies in

zebrafish have significantly contributed to the small body of research of I3M’s in vivo effects.

My last compound of study, DHM, was found to inhibit developmental angiogenesis and

fin regeneration in zebrafish. Due to time constraints, I did not have the opportunity to explore its

mechanisms or specificity of its actions. However, by studying DHM’s effects in a whole

organism, I have been able to identify anti-angiogenic and anti-proliferative activity as well as

indications of cellular, tissue and general toxicities. If DHM is confirmed as a generic cytotoxic

agent, then my zebrafish studies have assisted and streamlined the identification of an

incompetent drug early in the drug development pipeline.

Comprehensively, my studies have demonstrated the utility of zebrafish as a convenient

and versatile tool for advanced drug discovery. With 4HPR, I was able to learn new qualities

about a drug in clinical development. I was also able to contribute to the validation of a proposed

mechanism of action for the promising anti-cancer agent, I3M. Lastly, with DHM, I was able to

discover a novel use for a novel compound while simultaneously identifying its potential

toxicity. I hope that I have demonstrated in the course of my studies, that zebrafish are uniquely

suited to provide an efficient and effective systems-biology approach to improving the outcomes

of angiogenesis drug discovery.

Page 137: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

124

REFERENCES

1. Bushue, N. and Y.J. Wan, Retinoid pathway and cancer therapeutics. Adv Drug Deliv Rev, 2010. 62(13): p. 1285-98.

2. Verheul, H.M., E.E. Voest, and R.O. Schlingemann, Are tumors angiogenesis-dependent? . Journal of National Cancer Institute, 2004. 202: p. 13.

3. (USA), N.C.I., Cancer Bullentin. 2006.

4. Eichmann, A., Pardanaud, L., Li, Y., and Moyon, D, Vasculogenesis and the search for the hemangioblast. Journal of Hematotherapy and Stem Cell Research 2002. 11: p. 207-214.

5. Carmeliet, P., Angiogenesis in life, disease and medicine. Nature, 2005. 438(6): p. 936.

6. Carmeliet, P., Angiogenesis in health and disease. Nature Medicine, 2003. 9(6): p. 653-660.

7. Clapp, C., et al., Peptide hormone regulation of angiogenesis. Physiol Rev, 2009. 89(4): p. 1177-215.

8. Ferrera, N., Molecular and biological properties of vascular endothelial growth factor. Journal of Molecular Medicine, 1999. 77: p. 527-543.

9. Phng, L.K. and H. Gerhardt, Angiogenesis: a team effort coordinated by notch. Developmental Cell, 2009. 16: p. 196-208.

10. Gerhardt, H., et al., VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia. J Cell Biol, 2003. 161(6): p. 1163-77.

11. Carmeliet, P. and M. Tessier-Lavigne, Common mechanisms of nerve and blood vessel wiring. Nature, 2005. 436: p. 193-200.

12. Pasquale, E.B., Eph receptor signalling casta a wide net on cell behaviour. Nature Reviews Molecular Cell Biology, 2005. 6: p. 462-475.

13. Egginton, S. and M. Gerritsen, Lumen formation: in vivo versus in vitro observations. Microcirculation, 2003. 10(1): p. 45-61.

14. Papetti, M. and I.M. Herman, Mechanisms of normal and tumor-derived angiogenesis. Am J Physiol Cell Physiol, 2002. 282(5): p. C947-70.

Page 138: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

125

15. Lampugnani, M.G., Resnati, M., Raiteri, M., Pigott, R., Pisacane, A|., Houen, G., Ruco, L.P., Dejana, E. , A novel endothelial-specific membrane protein is a marker of cell-cell contacts. Journal of Cell Biology, 1992. 118.

16. Bauer, S.M., R.J. Bauer, and O.C. Velazquez, Angiogenesis, vasculogenesis, and induction of healing in chronic wounds. Vasc Endovascular Surg, 2005. 39(4): p. 293-306.

17. Barrientos, S., et al., Growth factors and cytokines in wound healing. Wound Repair Regen, 2008. 16(5): p. 585-601.

18. Bao, P., et al., The role of vascular endothelial growth factor in wound healing. J Surg Res, 2009. 153(2): p. 347-58.

19. Nardo, L.G., Vascular endothelial growth factor expression in the endometrium during the menstrual cycle, implantation window and early pregnancy. Current Opinion in Obstetrics and Gynecology, 2005. 17(4): p. 419-423.

20. Demir, R., A. Yaba, and B. Huppertz, Vasculogenesis and angiogenesis in the endometrium during menstrual cycle and implantation. Acta Histochem, 2010. 112(3): p. 203-14.

21. Ferrera, N., Chen, H., Davis-Smyth, T., Gerber, H.P., Nguyen, T.N., Peers, D., Chisolm, D., Hillan, K.J., Schwall, R.H., Vascular endothelial growth factor is essential for corpus luteum angiogenesis. Nature Medicine, 1998. 4(3): p. 336-40.

22. Bergers, G. and L.E. Benjamin, Tumorigenesis and the angiogenic switch. Nat Rev Cancer, 2003. 3(6): p. 401-10.

23. Wicki, A., Christofori, G., ed. The angiogenic switch in tumorigenesis. Tumor angiogenesis: basic mechanisms and cancer therapy, ed. D. Marme, Fusenig, N. 2008, Springer: Hiedelberg, Germany. 67-88.

24. Li, W.W., Li, V.W, Therapeutic angiogenesis: using growth factors to restore circulation in damaged tissues. Supplement to Contemporary Surgery, 2003: p. 19-32.

25. Rivard, A., et al., Age-dependent defect in vascular endothelial growth factor expression is associated with reduced hypoxia-inducible factor 1 activity. J Biol Chem, 2000. 275(38): p. 29643-7.

26. Schultz, A., et al., Interindividual heterogeneity in the hypoxic regulation of VEGF: significance for the development of the coronary artery collateral circulation. Circulation, 1999. 100(5): p. 547-52.

Page 139: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

126

27. Chou, E., et al., Decreased cardiac expression of vascular endothelial growth factor and its receptors in insulin-resistant and diabetic States: a possible explanation for impaired collateral formation in cardiac tissue. Circulation, 2002. 105(3): p. 373-9.

28. Mulder, G.D., Diabetic foot ulcers: old problems--new technologies. Nephrol Dial Transplant, 2001. 16(4): p. 695-8.

29. Martinez, P., et al., Age-related changes in parathyroid hormone-related protein and vascular endothelial growth factor in human osteoblastic cells. Osteoporos Int, 2002. 13(11): p. 874-81.

30. Kang, D.H., et al., Impaired angiogenesis in the aging kidney: vascular endothelial growth factor and thrombospondin-1 in renal disease. Am J Kidney Dis, 2001. 37(3): p. 601-11.

31. Lamarca, B., Endothelial dysfunction. An important mediator in the pathophysiology of hypertension during pre-eclampsia. Minerva Ginecol, 2012. 64(4): p. 309-20.

32. Taraseviciene-Stewart, L. and N.F. Voelkel, Molecular pathogenesis of emphysema. J Clin Invest, 2008. 118(2): p. 394-402.

33. Rodrigues, M.C., et al., Neurovascular aspects of amyotrophic lateral sclerosis. Int Rev Neurobiol, 2012. 102: p. 91-106.

34. de la Torre, J.C., Vascular basis of Alzheimer's pathogenesis. Ann N Y Acad Sci, 2002. 977: p. 196-215.

35. Das, A. and P.G. McGuire, Retinal and choroidal angiogenesis: pathophysiology and strategies for inhibition. Prog Retin Eye Res, 2003. 22(6): p. 721-48.

36. Praidou, A., et al., Vitreous and serum levels of vascular endothelial growth factor and platelet-derived growth factor and their correlation in patients with non-proliferative diabetic retinopathy and clinically significant macula oedema. Acta Ophthalmol, 2011. 89(3): p. 248-54.

37. Ando, A., et al., Nitric oxide is proangiogenic in the retina and choroid. J Cell Physiol, 2002. 191(1): p. 116-24.

38. Hackett, S.F., et al., Angiopoietin 2 expression in the retina: upregulation during physiologic and pathologic neovascularization. J Cell Physiol, 2000. 184(3): p. 275-84.

39. Fariss, R.N., et al., Tissue inhibitor of metalloproteinases-3 is a component of Bruch's membrane of the eye. Am J Pathol, 1997. 150(1): p. 323-8.

40. Cirulli, A., et al., Vascular endothelial growth factor serum levels are elevated in patients with hereditary hemorrhagic telangiectasia. Acta Haematol, 2003. 110(1): p. 29-32.

Page 140: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

127

41. Seeley, R.J., Treating obesity like a tumor. Cell Metab, 2012. 15(1): p. 1-2.

42. Becker, C.M. and R.J. D'Amato, Angiogenesis and antiangiogenic therapy in endometriosis. Microvasc Res, 2007. 74(2-3): p. 121-30.

43. Grivennikov, S.I., F.R. Greten, and M. Karin, Immunity, inflammation, and cancer. Cell, 2010. 140(6): p. 883-99.

44. Jiang, B.H., et al., V-SRC induces expression of hypoxia-inducible factor 1 (HIF-1) and transcription of genes encoding vascular endothelial growth factor and enolase 1: involvement of HIF-1 in tumor progression. Cancer Res, 1997. 57(23): p. 5328-35.

45. Okada, F., et al., Impact of oncogenes in tumor angiogenesis: mutant K-ras up-regulation of vascular endothelial growth factor/vascular permeability factor is necessary, but not sufficient for tumorigenicity of human colorectal carcinoma cells. Proc Natl Acad Sci U S A, 1998. 95(7): p. 3609-14.

46. Arbiser, J.L., et al., Oncogenic H-ras stimulates tumor angiogenesis by two distinct pathways. Proc Natl Acad Sci U S A, 1997. 94(3): p. 861-6.

47. Streit, M., et al., Overexpression of thrombospondin-1 decreases angiogenesis and inhibits the growth of human cutaneous squamous cell carcinomas. Am J Pathol, 1999. 155(2): p. 441-52.

48. Lawler, J., et al., Thrombospondin-1 gene expression affects survival and tumor spectrum of p53-deficient mice. Am J Pathol, 2001. 159(5): p. 1949-56.

49. Zabrenetzky, V., et al., Expression of the extracellular matrix molecule thrombospondin inversely correlates with malignant progression in melanoma, lung and breast carcinoma cell lines. Int J Cancer, 1994. 59(2): p. 191-5.

50. Weis, S.M. and D.A. Cheresh, Pathophysiological consequences of VEGF-induced vascular permeability. Nature, 2005. 437(7058): p. 497-504.

51. Morgan, D.M.L., ed. Tetrazolium (MTT) assay for celllular viability and activity. Methods in Molecular Biology: Polyamine Protocols, ed. D.M.L. Morgan. 1998, Humana Press Inc: Totowa, New Jersey, USA. 179-184.

52. Liang, C.C., A.Y. Park, and J.L. Guan, In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc, 2007. 2(2): p. 329-33.

53. Chen, H., ed. Boyden chamber assay. Methods in Molecular Biology: Cell Migration, ed. J. Guan. 2005, Humana Press, Inc.: Totowa, New Jersey, USA. 15-22.

Page 141: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

128

54. Donovan, D., et al., Comparison of three in vitro human 'angiogenesis' assays with capillaries formed in vivo. Angiogenesis, 2001. 4(2): p. 113-21.

55. Norrby, K., Drug testing with angiogenesis models. Expert Opinion on Drug Discovery, 2008. 3(7): p. 533-549.

56. Rogers, M.S., A.E. Birsner, and R.J. D'Amato, The mouse cornea micropocket angiogenesis assay. Nat Protoc, 2007. 2(10): p. 2545-50.

57. Nguyen, M., Y. Shing, and J. Folkman, Quantitation of angiogenesis and antiangiogenesis in the chick embryo chorioallantoic membrane. Microvasc Res, 1994. 47(1): p. 31-40.

58. Kragh, M., et al., In vivo chamber angiogenesis assay: an optimized Matrigel plug assay for fast assessment of anti-angiogenic activity. Int J Oncol, 2003. 22(2): p. 305-11.

59. Oikawa, D., et al., A transgenic mouse model for monitoring oxidative stress. Sci Rep, 2012. 2: p. 229.

60. Peal, D.S., R.T. Peterson, and D. Milan, Small molecule screening in zebrafish. J Cardiovasc Transl Res, 2010. 3(5): p. 454-60.

61. Nicoli, S. and M. Presta, The zebrafish/tumor xenograft angiogenesis assay. Nat Protoc, 2007. 2(11): p. 2918-23.

62. Cao, Z., et al., Hypoxia-induced retinopathy model in adult zebrafish. Nat Protoc, 2010. 5(12): p. 1903-10.

63. Serbedzija, G.N., E. Flynn, and C.E. Willett, Zebrafish angiogenesis: a new model for drug screening. Angiogenesis, 1999. 3(4): p. 353-9.

64. Vogel, A.M. and B.M. Weinstein, Studying vascular development in the zebrafish. Trends Cardiovasc Med, 2000. 10(8): p. 352-60.

65. Staton, C.A., M.W. Reed, and N.J. Brown, A critical analysis of current in vitro and in vivo angiogenesis assays. Int J Exp Pathol, 2009. 90(3): p. 195-221.

66. Howe, D.G., et al., ZFIN, the Zebrafish Model Organism Database: increased support for mutants and transgenics. Nucleic Acids Res, 2013. 41(D1): p. D854-60.

67. Brown, K.H., et al., Extensive genetic diversity and substructuring among zebrafish strains revealed through copy number variant analysis. Proc Natl Acad Sci U S A, 2012. 109(2): p. 529-34.

68. Lockwood, B., et al., Acute effects of alcohol on larval zebrafish: a genetic system for large-scale screening. Pharmacol Biochem Behav, 2004. 77(3): p. 647-54.

Page 142: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

129

69. Liao, G., et al., A genomic "roadmap" to "better" drugs. Drug Metab Rev, 2008. 40(2): p. 225-39.

70. Shinya, M. and N. Sakai, Generation of highly homogeneous strains of zebrafish through full sib-pair mating. G3 (Bethesda), 2011. 1(5): p. 377-86.

71. Mizgirev, I. and S. Revskoy, Generation of clonal zebrafish lines and transplantable hepatic tumors. Nat Protoc, 2010. 5(3): p. 383-94.

72. Wienholds, E., et al., Target-selected inactivation of the zebrafish rag1 gene. Science, 2002. 297(5578): p. 99-102.

73. Lan, C.C., et al., Quantitative real-time RT-PCR (qRT-PCR) of zebrafish transcripts: optimization of RNA extraction, quality control considerations, and data analysis. Cold Spring Harb Protoc, 2009. 2009(10): p. pdb prot5314.

74. Jing, L., et al., In situ hybridization assay-based small molecule screening in zebrafish. Curr Protoc Chem Biol, 2012. 4(2).

75. Ton, C., et al., Construction of a zebrafish cDNA microarray: gene expression profiling of the zebrafish during development. Biochem Biophys Res Commun, 2002. 296(5): p. 1134-42.

76. Peterson, R.T. and C.A. Macrae, Systematic approaches to toxicology in the zebrafish. Annu Rev Pharmacol Toxicol, 2012. 52: p. 433-53.

77. Sukardi, H., et al., Zebrafish for drug toxicity screening: bridging the in vitro cell-based models and in vivo mammalian models. Expert Opin Drug Metab Toxicol, 2011. 7(5): p. 579-89.

78. Jones, H.S., et al., Oxidative and conjugative xenobiotic metabolism in zebrafish larvae in vivo. Zebrafish, 2010. 7(1): p. 23-30.

79. Kurogi, K., et al., Sulfation of Drug Compounds by the Zebrafish Cytosolic Sulfotransferases (SULTs). Drug Metab Lett, 2010. 4(2): p. 62-68.

80. Keiser, M.J., et al., Relating protein pharmacology by ligand chemistry. Nat Biotechnol, 2007. 25(2): p. 197-206.

81. Chan, J., et al., Dissection of angiogenic signaling in zebrafish using a chemical genetic approach. Cancer Cell, 2002. 1(3): p. 257-67.

82. Bakkiyanathan, A., Joseph, A.M., Tharani, L., Malathi, R., Genistein, the phytoestrogen induces heart-and-soul (has) phenotypes in zebrafish embryo. Journal of Developmental Biology and Tissue Engineering, 2010. 2(2): p. 18-22.

Page 143: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

130

83. Khersonsky, S.M., et al., Facilitated forward chemical genetics using a tagged triazine library and zebrafish embryo screening. J Am Chem Soc, 2003. 125(39): p. 11804-5.

84. Jung, D.W., et al., Identification of the F1F0 mitochondrial ATPase as a target for modulating skin pigmentation by screening a tagged triazine library in zebrafish. Mol Biosyst, 2005. 1(1): p. 85-92.

85. Ong, S.E., et al., Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol Cell Proteomics, 2002. 1(5): p. 376-86.

86. Ross, P.L., et al., Multiplexed protein quantitation in Saccharomyces cerevisiae using amine-reactive isobaric tagging reagents. Mol Cell Proteomics, 2004. 3(12): p. 1154-69.

87. Durbin, L., et al., Eph signaling is required for segmentation and differentiation of the somites. Genes Dev, 1998. 12(19): p. 3096-109.

88. Liang, D., et al., Cloning and characterization of vascular endothelial growth factor (VEGF) from zebrafish, Danio rerio. Biochim Biophys Acta, 1998. 1397(1): p. 14-20.

89. Lyons, M.S., et al., Isolation of the zebrafish homologues for the tie-1 and tie-2 endothelium-specific receptor tyrosine kinases. Dev Dyn, 1998. 212(1): p. 133-40.

90. Lawson, N.D. and B.M. Weinstein, Arteries and veins: making a difference with zebrafish. Nat Rev Genet, 2002. 3(9): p. 674-82.

91. Motoike, T., et al., Universal GFP reporter for the study of vascular development. Genesis, 2000. 28(2): p. 75-81.

92. Cross, L.M., et al., Rapid analysis of angiogenesis drugs in a live fluorescent zebrafish assay. Arterioscler Thromb Vasc Biol, 2003. 23(5): p. 911-2.

93. Roman, B.L., et al., Disruption of acvrl1 increases endothelial cell number in zebrafish cranial vessels. Development, 2002. 129(12): p. 3009-19.

94. Schmitt, C.E., M.B. Holland, and S.W. Jin, Visualizing vascular networks in zebrafish: an introduction to microangiography. Methods Mol Biol, 2012. 843: p. 59-67.

95. Gray, C., et al., Ischemia is not required for arteriogenesis in zebrafish embryos. Arterioscler Thromb Vasc Biol, 2007. 27(10): p. 2135-41.

96. Wang, C., et al., Rosuvastatin, identified from a zebrafish chemical genetic screen for antiangiogenic compounds, suppresses the growth of prostate cancer. Eur Urol, 2010. 58(3): p. 418-26.

Page 144: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

131

97. Chan, J. and J.D. Mably, Dissection of cardiovascular development and disease pathways in zebrafish. Prog Mol Biol Transl Sci, 2011. 100: p. 111-53.

98. Jakobsson, L., et al., Endothelial cells dynamically compete for the tip cell position during angiogenic sprouting. Nat Cell Biol, 2010. 12(10): p. 943-53.

99. Jensen, L.D., et al., Zebrafish models to study hypoxia-induced pathological angiogenesis in malignant and nonmalignant diseases. Birth Defects Res C Embryo Today, 2011. 93(2): p. 182-93.

100. Rouhi, P., et al., Hypoxia-induced metastasis model in embryonic zebrafish. Nat Protoc, 2010. 5(12): p. 1911-8.

101. Chablais, F. and A. Jazwinska, Induction of myocardial infarction in adult zebrafish using cryoinjury. J Vis Exp, 2012(62).

102. Mathew, L.K., et al., Unraveling tissue regeneration pathways using chemical genetics. J Biol Chem, 2007. 282(48): p. 35202-10.

103. Wood, J.M., et al., PTK787/ZK 222584, a novel and potent inhibitor of vascular endothelial growth factor receptor tyrosine kinases, impairs vascular endothelial growth factor-induced responses and tumor growth after oral administration. Cancer Res, 2000. 60(8): p. 2178-89.

104. Bayliss, P.E., et al., Chemical modulation of receptor signaling inhibits regenerative angiogenesis in adult zebrafish. Nat Chem Biol, 2006. 2(5): p. 265-73.

105. Liu, S. and S.D. Leach, Zebrafish models for cancer. Annu Rev Pathol, 2011. 6: p. 71-93.

106. Santoriello, C., et al., Kita driven expression of oncogenic HRAS leads to early onset and highly penetrant melanoma in zebrafish. PLoS One, 2010. 5(12): p. e15170.

107. Nguyen, A.T., et al., An inducible kras(V12) transgenic zebrafish model for liver tumorigenesis and chemical drug screening. Dis Model Mech, 2012. 5(1): p. 63-72.

108. Soussi, T. and C. Beroud, Significance of TP53 mutations in human cancer: a critical analysis of mutations at CpG dinucleotides. Hum Mutat, 2003. 21(3): p. 192-200.

109. Berghmans, S., et al., tp53 mutant zebrafish develop malignant peripheral nerve sheath tumors. Proc Natl Acad Sci U S A, 2005. 102(2): p. 407-12.

110. Nicoli, S., et al., Mammalian tumor xenografts induce neovascularization in zebrafish embryos. Cancer Res, 2007. 67(7): p. 2927-31.

111. Marques, I.J., et al., Metastatic behaviour of primary human tumours in a zebrafish xenotransplantation model. BMC Cancer, 2009. 9: p. 128.

Page 145: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

132

112. Stoletov, K., et al., Visualizing extravasation dynamics of metastatic tumor cells. J Cell Sci, 2010. 123(Pt 13): p. 2332-41.

113. Lee, S.L., et al., Hypoxia-induced pathological angiogenesis mediates tumor cell dissemination, invasion, and metastasis in a zebrafish tumor model. Proc Natl Acad Sci U S A, 2009. 106(46): p. 19485-90.

114. Stoletov, K. and R. Klemke, Catch of the day: zebrafish as a human cancer model. Oncogene, 2008. 27(33): p. 4509-20.

115. Smith, A.C., et al., High-throughput cell transplantation establishes that tumor-initiating cells are abundant in zebrafish T-cell acute lymphoblastic leukemia. Blood, 2010. 115(16): p. 3296-303.

116. White, R.M., et al., Transparent adult zebrafish as a tool for in vivo transplantation analysis. Cell Stem Cell, 2008. 2(2): p. 183-9.

117. Yanik, M.F., C.B. Rohde, and C. Pardo-Martin, Technologies for micromanipulating, imaging, and phenotyping small invertebrates and vertebrates. Annu Rev Biomed Eng, 2011. 13: p. 185-217.

118. Zhang, X., Lu, Z., Gelinas, D., Ciruna, B., Sun, Y., Batch transfer of zebrafish embryos into multiwell plates. Automation Science and Engineering, 2011. 8(3): p. 625-631.

119. Wang, W., et al., A fully automated robotic system for microinjection of zebrafish embryos. PLoS One, 2007. 2(9): p. e862.

120. Cheng, K.C., et al., Whole-animal imaging, gene function, and the Zebrafish Phenome Project. Curr Opin Genet Dev, 2011. 21(5): p. 620-9.

121. Kabli, S., et al., In vivo magnetic resonance imaging to detect malignant melanoma in adult zebrafish. Zebrafish, 2010. 7(2): p. 143-8.

122. Goessling, W., T.E. North, and L.I. Zon, Ultrasound biomicroscopy permits in vivo characterization of zebrafish liver tumors. Nat Methods, 2007. 4(7): p. 551-3.

123. Petzold, A.M., et al., SCORE imaging: specimen in a corrected optical rotational enclosure. Zebrafish, 2010. 7(2): p. 149-54.

124. Gehrig, J., et al., Automated high-throughput mapping of promoter-enhancer interactions in zebrafish embryos. Nat Methods, 2009. 6(12): p. 911-6.

125. Sabaliauskas, N.A., et al., High-throughput zebrafish histology. Methods, 2006. 39(3): p. 246-54.

Page 146: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

133

126. Vogt, A., et al., Automated image-based phenotypic analysis in zebrafish embryos. Dev Dyn, 2009. 238(3): p. 656-63.

127. Abdulhannan, P., D.A. Russell, and S. Homer-Vanniasinkam, Peripheral arterial disease: a literature review. Br Med Bull, 2012.

128. Li, W.W., Tsakayannis, D., Li, V.W., ed. Lessons to be learned from clinical trials of angiogenesis modulators in ischemic diseases. Angiogenesis in health and disease, ed. G. Rubanyi. 1999, Marcel Dekker: New York. 519-536.

129. Brenes, R.A., et al., Cell-based interventions for therapeutic angiogenesis: review of potential cell sources. Vascular, 2012.

130. Silvestre, J.S., Pro-angiogenic cell-based therapy for the treatment of ischemic cardiovascular diseases. Thromb Res, 2012. 130 Suppl 1: p. S90-4.

131. Papanas, N., et al., Advances in the topical treatment of diabetic foot ulcers. Curr Diabetes Rev, 2012. 8(3): p. 209-18.

132. Desai, K.K., et al., Negative pressure wound therapy: an algorithm. Clin Plast Surg, 2012. 39(3): p. 311-24.

133. Voigt, J., et al., Low-frequency ultrasound (20-40 kHz) as an adjunctive therapy for chronic wound healing: a systematic review of the literature and meta-analysis of eight randomized controlled trials. Int J Low Extrem Wounds, 2011. 10(4): p. 190-9.

134. Wang, C., et al., Hyperbaric oxygen for treating wounds: a systematic review of the literature. Arch Surg, 2003. 138(3): p. 272-9; discussion 280.

135. Simons, M., et al., Pharmacological treatment of coronary artery disease with recombinant fibroblast growth factor-2: double-blind, randomized, controlled clinical trial. Circulation, 2002. 105(7): p. 788-93.

136. Papanas, N. and E. Maltezos, Becaplermin gel in the treatment of diabetic neuropathic foot ulcers. Clin Interv Aging, 2008. 3(2): p. 233-40.

137. Henry, T.D., et al., The VIVA trial: Vascular endothelial growth factor in Ischemia for Vascular Angiogenesis. Circulation, 2003. 107(10): p. 1359-65.

138. Hedman, M., et al., Safety and feasibility of catheter-based local intracoronary vascular endothelial growth factor gene transfer in the prevention of postangioplasty and in-stent restenosis and in the treatment of chronic myocardial ischemia: phase II results of the Kuopio Angiogenesis Trial (KAT). Circulation, 2003. 107(21): p. 2677-83.

Page 147: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

134

139. Mulder, G., et al., Treatment of nonhealing diabetic foot ulcers with a platelet-derived growth factor gene-activated matrix (GAM501): results of a phase 1/2 trial. Wound Repair Regen, 2009. 17(6): p. 772-9.

140. Grines, C.L., et al., A randomized, double-blind, placebo-controlled trial of Ad5FGF-4 gene therapy and its effect on myocardial perfusion in patients with stable angina. J Am Coll Cardiol, 2003. 42(8): p. 1339-47.

141. Kastrup, J., Gene therapy and angiogenesis in patients with coronary arter disease. Expert Review of Cardiovascular Therapy, 2010. 8: p. 1143.

142. Lavu, M., S. Gundewar, and D.J. Lefer, Gene therapy for ischemic heart disease. J Mol Cell Cardiol, 2011. 50(5): p. 742-50.

143. D'Andrea, L.D., et al., Peptide-based molecules in angiogenesis. Chem Biol Drug Des, 2006. 67(2): p. 115-26.

144. Wieghaus, K.A., et al., Small molecule inducers of angiogenesis for tissue engineering. Tissue Eng, 2006. 12(7): p. 1903-13.

145. Li, J., et al., PR39, a peptide regulator of angiogenesis. Nat Med, 2000. 6(1): p. 49-55.

146. Correia, S.C. and P.I. Moreira, Hypoxia-inducible factor 1: a new hope to counteract neurodegeneration? J Neurochem, 2010. 112(1): p. 1-12.

147. Goldenberg, N.M., et al., Broken barriers: a new take on sepsis pathogenesis. Sci Transl Med, 2011. 3(88): p. 88ps25.

148. Sefcik, L.S., C.E. Petrie Aronin, and E.A. Botchwey, Engineering vascularized tissues using natural and synthetic small molecules. Organogenesis, 2008. 4(4): p. 215-27.

149. Rivellese, M.J. and C.R. Baumal, Photodynamic therapy of eye diseases. J Ophthalmic Nurs Technol, 2000. 19(3): p. 134-41.

150. Folkman, J., Angiogenesis: an organizing principle for drug discovery? Nat Rev Drug Discov, 2007. 6(4): p. 273-86.

151. Rogosin, S. and A.B. Sandler, Beyond bevacizumab: antiangiogenesis agents. Clinical Lung Cancer, 2012.

152. Stewart, M.W., Aflibercept (VEGF-TRAP): the next anti-VEGF drug. Inflamm Allergy Drug Targets, 2011. 10(6): p. 497-508.

153. Gragoudas, E.S., et al., Pegaptanib for neovascular age-related macular degeneration. N Engl J Med, 2004. 351(27): p. 2805-16.

Page 148: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

135

154. Chen, X., et al., Database of traditional Chinese medicine and its application to studies of mechanism and to prescription validation. Br J Pharmacol, 2006. 149(8): p. 1092-103.

155. Motzer, R.J., et al., Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. . The New England Journal of Medicine, 2007. 356: p. 124.

156. Llovet, J.M., Ricci, S., Mazzaferro, V., Hilgard, P., Gane, E., Blank, J., de Oliviera, A., Santoro, A., Raoul, J., Forner, A., Schward, M., Porta, C., Zeuzem, S., Bolondi, L., Greten, T.F., Galle, P.R., Seitz, J., Borbath, I., Haussinger, D., Giannaris, T., Shan, M., Moscovici, M., Voliotis, D., Bruix, J., Sorafenib in advanced hepatocellular carcinoma. . The New England Journal of Medicine, 2008. 359: p. 390.

157. Jain, R.K., et al., Lessons from phase III clinical trials on anti-VEGF therapy for cancer. Nat Clin Pract Oncol, 2006. 3(1): p. 24-40.

158. Verheul, H.M. and H.M. Pinedo, Possible molecular mechanisms involved in the toxicity of angiogenesis inhibition. Nat Rev Cancer, 2007. 7(6): p. 475-85.

159. Ferreras, M., et al., Generation and degradation of human endostatin proteins by various proteinases. FEBS Lett, 2000. 486(3): p. 247-51.

160. O'Reilly, M.S., Angiostatin: an endogenous inhibitor of angiogenesis and of tumor growth. EXS, 1997. 79: p. 273-94.

161. O'Reilly, M.S., et al., Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell, 1997. 88(2): p. 277-85.

162. Goodwin, J.M., et al., Angiogenesis inhibition using an oncolytic herpes simplex virus expressing endostatin in a murine lung cancer model. Cancer Invest, 2012. 30(3): p. 243-50.

163. O'Reilly, M.S., et al., Angiostatin induces and sustains dormancy of human primary tumors in mice. Nat Med, 1996. 2(6): p. 689-92.

164. Zhou, J.F., et al., Endostar combined with chemotherapy for treatment of metastatic colorectal and gastric cancer: a pilot study. Chin Med J (Engl), 2011. 124(24): p. 4299-303.

165. Abdollahi, A., et al., Endostatin's antiangiogenic signaling network. Mol Cell, 2004. 13(5): p. 649-63.

166. Folkman, J., Antiangiogenesis in cancer therapy - endostatin and its mechanisms of action. Exp Cell Res, 2006 312: p. 594-607.

167. Boehm, T., et al., Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance. Nature, 1997. 390(6658): p. 404-7.

Page 149: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

136

168. Kim, K.S., et al., Inhibition of angiogenesis and tumor progression by hydrodynamic cotransfection of angiostatin K1-3, endostatin, and saxatilin genes. Cancer Gene Ther, 2006. 13(6): p. 563-71.

169. Eliceiri, B.P. and D.A. Cheresh, The role of alphav integrins during angiogenesis: insights into potential mechanisms of action and clinical development. J Clin Invest, 1999. 103(9): p. 1227-30.

170. Patel, S.R., et al., Pilot study of vitaxin--an angiogenesis inhibitor-in patients with advanced leiomyosarcomas. Cancer, 2001. 92(5): p. 1347-8.

171. Coussens, L.M., B. Fingleton, and L.M. Matrisian, Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science, 2002. 295(5564): p. 2387-92.

172. Rudek, M.A., J. Venitz, and W.D. Figg, Matrix metalloproteinase inhibitors: do they have a place in anticancer therapy? Pharmacotherapy, 2002. 22(6): p. 705-20.

173. Park, Y.G., et al., New Approach of Anti-VEGF Agents for Age-Related Macular Degeneration. J Ophthalmol, 2012. 2012: p. 637316.

174. Loges, S., T. Schmidt, and P. Carmeliet, Mechanisms of resistance to anti-angiogenic therapy and development of third-generation anti-angiogenic drug candidates. Genes Cancer, 2010. 1(1): p. 12-25.

175. Hurwitz, H., et al., Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med, 2004. 350(23): p. 2335-42.

176. Simons, M. and J.A. Ware, Therapeutic angiogenesis in cardiovascular disease. Nat Rev Drug Discov, 2003. 2(11): p. 863-71.

177. Simons, M., Angiogenesis: where do we stand now? Circulation, 2005. 111(12): p. 1556-66.

178. Grepin, R., Pages, Gilles. , Molecular Mechanisms of Resistance to Tumour Anti-Angiogenic Strategies. Journal of Oncology, 2010. 2010: p. 8.

179. Teicher, B.A., Antiangiogenic agents and targets: a perspective. Biochemical Pharmacology, 2011. 81: p. 12.

180. Relf, M., et al., Expression of the angiogenic factors vascular endothelial cell growth factor, acidic and basic fibroblast growth factor, tumor growth factor beta-1, platelet-derived endothelial cell growth factor, placenta growth factor, and pleiotrophin in human primary breast cancer and its relation to angiogenesis. Cancer Res, 1997. 57(5): p. 963-9.

Page 150: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

137

181. Shyu, K.G., H. Chang, and J.M. Isner, Synergistic effect of angiopoietin-1 and vascular endothelial growth factor on neoangiogenesis in hypercholesterolemic rabbit model with acute hindlimb ischemia. Life Sci, 2003. 73(5): p. 563-79.

182. Zhang, W., et al., Bevacizumab with angiostatin-armed oHSV increases antiangiogenesis and decreases bevacizumab-induced invasion in U87 glioma. Mol Ther, 2012. 20(1): p. 37-45.

183. Ma, L., P. del Soldato, and J.L. Wallace, Divergent effects of new cyclooxygenase inhibitors on gastric ulcer healing: Shifting the angiogenic balance. Proc Natl Acad Sci U S A, 2002. 99(20): p. 13243-7.

184. Panigrahy, D., et al., PPARgamma ligands inhibit primary tumor growth and metastasis by inhibiting angiogenesis. J Clin Invest, 2002. 110(7): p. 923-32.

185. Pruthi, R.S., et al., Phase II trial of celecoxib in prostate-specific antigen recurrent prostate cancer after definitive radiation therapy or radical prostatectomy. Clin Cancer Res, 2006. 12(7 Pt 1): p. 2172-7.

186. Smith, M.R., et al., Rosiglitazone versus placebo for men with prostate carcinoma and a rising serum prostate-specific antigen level after radical prostatectomy and/or radiation therapy. Cancer, 2004. 101(7): p. 1569-74.

187. Kindler, H.L., et al., Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: phase III trial of the Cancer and Leukemia Group B (CALGB 80303). J Clin Oncol, 2010. 28(22): p. 3617-22.

188. Yeh, J.R., R. Mohan, and C.M. Crews, The antiangiogenic agent TNP-470 requires p53 and p21CIP/WAF for endothelial cell growth arrest. Proc Natl Acad Sci U S A, 2000. 97(23): p. 12782-7.

189. Bergers, G. and D. Hanahan, Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer, 2008. 8(8): p. 592-603.

190. Ezzell, C., Starving tumors of their lifeblood. Sci Am, 1998. 279(4): p. 33-4.

191. Pereira, D.A. and J.A. Williams, Origin and evolution of high throughput screening. Br J Pharmacol, 2007. 152(1): p. 53-61.

192. Scannell, J.W., et al., Diagnosing the decline in pharmaceutical R&D efficiency. Nat Rev Drug Discov, 2012. 11(3): p. 191-200.

193. Murphey, R.D., et al., A chemical genetic screen for cell cycle inhibitors in zebrafish embryos. Chem Biol Drug Des, 2006. 68(4): p. 213-9.

Page 151: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

138

194. Lord, A.M., T.E. North, and L.I. Zon, Prostaglandin E2: making more of your marrow. Cell Cycle, 2007. 6(24): p. 3054-7.

195. Crawford, A.D., C.V. Esguerra, and P.A. de Witte, Fishing for drugs from nature: zebrafish as a technology platform for natural product discovery. Planta Med, 2008. 74(6): p. 624-32.

196. WHO, Global prevalence of vitamin A defiiciency in populations at risk 1995-2005., in WHO Global Database on Vitamin A Deficiency. 2009, World Health Organization: Geneva.

197. D'Ambrosio, D.N., R.D. Clugston, and W.S. Blaner, Vitamin A metabolism: an update. Nutrients, 2011. 3(1): p. 63-103.

198. Kam, R.K., et al., Retinoic acid synthesis and functions in early embryonic development. Cell Biosci, 2012. 2(1): p. 11.

199. Rhinn, M. and P. Dolle, Retinoic acid signalling during development. Development, 2012. 139(5): p. 843-58.

200. Levi, L., et al., Insight into molecular pathways of retinal metabolism, associated with vitellogenesis in zebrafish. Am J Physiol Endocrinol Metab, 2012. 302(6): p. E626-44.

201. Kudoh, T., S.W. Wilson, and I.B. Dawid, Distinct roles for Fgf, Wnt and retinoic acid in posteriorizing the neural ectoderm. Development, 2002. 129(18): p. 4335-46.

202. Duester, G., Retinoic acid synthesis and signaling during early organogenesis. Cell, 2008. 134(6): p. 921-31.

203. Ross, C.A., ed. Vitamin A. 2nd ed. Encyclopedia of Dietary Supplements, ed. P.M. Coats, Betz, J.M., Blackman, M.R. 2010, Informa Healthcare: London/New York. 778-791.

204. Sporn, M.B., et al., Prevention of chemical carcinogenesis by vitamin A and its synthetic analogs (retinoids). Fed Proc, 1976. 35(6): p. 1332-8.

205. De Luca, L.M., D. Roop, and F.L. Huang, Vitamin A: a key nutrient for the maintenance of epithelial differentiation. Acta Vitaminol Enzymol, 1985. 7 Suppl: p. 13-20.

206. Sun, S.Y. and R. Lotan, Retinoids and their receptors in cancer development and chemoprevention. Crit Rev Oncol Hematol, 2002. 41(1): p. 41-55.

207. Lasorella, A., A. Iavarone, and M.A. Israel, Differentiation of neuroblastoma enhances Bcl-2 expression and induces alterations of apoptosis and drug resistance. Cancer Res, 1995. 55(20): p. 4711-6.

Page 152: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

139

208. Chapman, M.S., Vitamin a: history, current uses, and controversies. Semin Cutan Med Surg, 2012. 31(1): p. 11-6.

209. Thielitz, A., et al., Topical retinoids in acne--an evidence-based overview. J Dtsch Dermatol Ges, 2008. 6(12): p. 1023-31.

210. Zhou, G.B., et al., Treatment of acute promyelocytic leukaemia with all-trans retinoic acid and arsenic trioxide: a paradigm of synergistic molecular targeting therapy. Philos Trans R Soc Lond B Biol Sci, 2007. 362(1482): p. 959-71.

211. Collins, M.D. and G.E. Mao, Teratology of retinoids. Annu Rev Pharmacol Toxicol, 1999. 39: p. 399-430.

212. Gander, R.J., Gurney, J.A, All-trans retinoic acid esters and amides, in Chemical Abstracts, U.P. Office, Editor. 1978, Johnson & Johnson: USA.

213. Cheung, E., et al., Oral Fenretinide in Biochemically Recurrent Prostate Cancer: A California Cancer Consortium Phase II Trial. Clinical Genitourinary Cancer, 2009. 7(1): p. 43-50.

214. Kazmi, S.M., et al., Comparison of N-(4-hydroxyphenyl)retinamide and all-trans-retinoic acid in the regulation of retinoid receptor-mediated gene expression in human breast cancer cell lines. Cancer Res, 1996. 56(5): p. 1056-62.

215. Sheikh, M.S., et al., N-(4-hydroxyphenyl)retinamide (4-HPR)-mediated biological actions involve retinoid receptor-independent pathways in human breast carcinoma. Carcinogenesis, 1995. 16(10): p. 2477-86.

216. Clifford, J.L., et al., Retinoid receptor-dependent and -independent effects of N-(4-hydroxyphenyl)retinamide in F9 embryonal carcinoma cells. Cancer Res, 1999. 59(1): p. 14-8.

217. Ruvolo, V.R., et al., Role for PKC Delta in Fenretinide-Mediated Apoptosis in Lymphoid Leukemia Cells. Journal of Signal Transduction, 2010. 2010.

218. Simeone, A.M. and A.M. Tari, How retinoids regulate breast cancer cell proliferation and apoptosis. Cell Mol Life Sci, 2004. 61(12): p. 1475-84.

219. Kim, H.J., et al., N-(4-hydroxyphenyl)retinamide-induced apoptosis triggered by reactive oxygen species is mediated by activation of MAPKs in head and neck squamous carcinoma cells. Oncogene, 2006. 25(19): p. 2785-94.

220. Lovat, P.E., et al., Distinct properties of fenretinide and CD437 lead to synergistic responses with chemotherapeutic reagents. Med Pediatr Oncol, 2000. 35(6): p. 663-8.

Page 153: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

140

221. Corazzari, M., et al., Fenretinide: a p53-independent way to kill cancer cells. Biochem Biophys Res Commun, 2005. 331(3): p. 810-5.

222. Ribatti, D., et al., Fenretinide as an anti-angiogenic agent in neuroblastoma. Cancer Lett, 2003. 197(1-2): p. 181-4.

223. Ferrari, N., et al., The transforming growth factor-beta family members bone morphogenetic protein-2 and macrophage inhibitory cytokine-1 as mediators of the antiangiogenic activity of N-(4-hydroxyphenyl)retinamide. Clin Cancer Res, 2005. 11(12): p. 4610-9.

224. Sogno, I., et al., Angioprevention with fenretinide: targeting angiogenesis in prevention and therapeutic strategies. Crit Rev Oncol Hematol, 2010. 75(1): p. 2-14.

225. Ulukaya, E. and E.J. Wood, Fenretinide and its relation to cancer. Cancer Treat Rev, 1999. 25(4): p. 229-35.

226. Formelli, F. and L. Cleris, Synthetic retinoid fenretinide is effective against a human ovarian carcinoma xenograft and potentiates cisplatin activity. Cancer Res, 1993. 53(22): p. 5374-6.

227. Conaway, C.C., et al., Chemopreventive potential of fumaric acid, N-acetylcysteine, N-(4-hydroxyphenyl) retinamide and beta-carotene for tobacco-nitrosamine-induced lung tumors in A/J mice. Cancer Lett, 1998. 124(1): p. 85-93.

228. Kenel, M.F., et al., Teratogenicity of N-(4-hydroxyphenyl)-all-trans-retinamide in rats and rabbits. Teratog Carcinog Mutagen, 1988. 8(1): p. 1-11.

229. Veronesi, U., et al., Randomized trial of fenretinide to prevent second breast malignancy in women with early breast cancer. J Natl Cancer Inst, 1999. 91(21): p. 1847-56.

230. Puduvalli, V.K., et al., Phase II study of fenretinide (NSC 374551) in adults with recurrent malignant gliomas: A North American Brain Tumor Consortium study. J Clin Oncol, 2004. 22(21): p. 4282-9.

231. Vaishampayan, U., et al., Phase II trial of fenretinide in advanced renal carcinoma. Invest New Drugs, 2005. 23(2): p. 179-85.

232. Moore, M.M., et al., A phase II study of fenretinide in patients with hormone refractory prostate cancer: a trial of the Cancer Therapeutics Research Group. Cancer Chemother Pharmacol, 2010. 66(5): p. 845-50.

233. Sabichi, A.L., et al., Phase III prevention trial of fenretinide in patients with resected non-muscle-invasive bladder cancer. Clin Cancer Res, 2008. 14(1): p. 224-9.

Page 154: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

141

234. Decensi, A., et al., Fenretinide and risk reduction of second breast cancer. Nat Clin Pract Oncol, 2007. 4(2): p. 64-5.

235. Cazzaniga, M., et al., Fenretinide (4-HPR): A Preventive Chance for Women at Genetic and Familial Risk? J Biomed Biotechnol, 2012. 2012: p. 172897.

236. Camerini, T., et al., Safety of the synthetic retinoid fenretinide: long-term results from a controlled clinical trial for the prevention of contralateral breast cancer. J Clin Oncol, 2001. 19(6): p. 1664-70.

237. Guilbault, C., et al., Cystic Fibrosis Fatty Acid Imbalance Is Linked to Ceramide Deficiency and Corrected by Fenretinide. American Journal of Respiratory Cell and Molecular Biology, 2009. 41(1): p. 100-106.

238. Koh, I.U., et al., Fenretinide ameliorates insulin resistance and fatty liver in obese mice. Biol Pharm Bull, 2012. 35(3): p. 369-75.

239. Mata, N.L., et al., Investigation of Oral Fenretinide for Treatment of Geographic Atrophy in Age-Related Macular Degeneration. Retina, 2012.

240. Nusslein-Volhard, C., Dahm, R., ed. Zebrafish: a practical approach. 2002, Oxford University Press. 322.

241. Zon, L.I., Intrinsic and extrinsic control of haematopoietic stem-cell self-renewal. Nature, 2008. 453(7193): p. 306-13.

242. Yoshinari, N. and A. Kawakami, Mature and juvenile tissue models of regeneration in small fish species. Biol Bull, 2011. 221(1): p. 62-78.

243. Waxman, J.S. and D. Yelon, Comparison of the expression patterns of newly identified zebrafish retinoic acid and retinoid X receptors. Dev Dyn, 2007. 236(2): p. 587-95.

244. Ferrari, N., et al., Inhibition of Kaposi's sarcoma in vivo by fenretinide. Clin Cancer Res, 2003. 9(16 Pt 1): p. 6020-9.

245. Poss, K.D., M.T. Keating, and A. Nechiporuk, Tales of regeneration in zebrafish. Dev Dyn, 2003. 226(2): p. 202-10.

246. Kimmel, C.B., et al., Stages of embryonic development of the zebrafish. Dev Dyn, 1995. 203(3): p. 253-310.

247. Diez del Corral, R. and K.G. Storey, Opposing FGF and retinoid pathways: a signalling switch that controls differentiation and patterning onset in the extending vertebrate body axis. Bioessays, 2004. 26(8): p. 857-69.

Page 155: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

142

248. Abu-Abed, S., et al., The retinoic acid-metabolizing enzyme, CYP26A1, is essential for normal hindbrain patterning, vertebral identity, and development of posterior structures. Genes Dev, 2001. 15(2): p. 226-40.

249. Diez del Corral, R., et al., Opposing FGF and retinoid pathways control ventral neural pattern, neuronal differentiation, and segmentation during body axis extension. Neuron, 2003. 40(1): p. 65-79.

250. Dubrulle, J., M.J. McGrew, and O. Pourquie, FGF signaling controls somite boundary position and regulates segmentation clock control of spatiotemporal Hox gene activation. Cell, 2001. 106(2): p. 219-32.

251. Vermot, J. and O. Pourquie, Retinoic acid coordinates somitogenesis and left-right patterning in vertebrate embryos. Nature, 2005. 435(7039): p. 215-20.

252. Sawada, A., et al., Fgf/MAPK signalling is a crucial positional cue in somite boundary formation. Development, 2001. 128(23): p. 4873-80.

253. Martin, B.L. and D. Kimelman, Brachyury establishes the embryonic mesodermal progenitor niche. Genes Dev, 2010. 24(24): p. 2778-83.

254. Rohner, N., et al., Duplication of fgfr1 permits Fgf signaling to serve as a target for selection during domestication. Curr Biol, 2009. 19(19): p. 1642-7.

255. Anding, A.L., et al., 4-Hydroxybenzyl modification of the highly teratogenic retinoid, 4-[(1E)-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-2-naphthalenyl)-1-propen-1-yl]b enzoic acid (TTNPB), yields a compound that induces apoptosis in breast cancer cells and shows reduced teratogenicity. Chem Res Toxicol, 2011. 24(11): p. 1853-61.

256. Villani, M.G., et al., Identification of the fenretinide metabolite 4-oxo-fenretinide present in human plasma and formed in human ovarian carcinoma cells through induction of cytochrome P450 26A1. Clin Cancer Res, 2004. 10(18 Pt 1): p. 6265-75.

257. Echeverri, K. and A.C. Oates, Coordination of symmetric cyclic gene expression during somitogenesis by Suppressor of Hairless involves regulation of retinoic acid catabolism. Dev Biol, 2007. 301(2): p. 388-403.

258. Kawamura, A., et al., Zebrafish hairy/enhancer of split protein links FGF signaling to cyclic gene expression in the periodic segmentation of somites. Genes Dev, 2005. 19(10): p. 1156-61.

259. Sporn, M.B. and A.B. Roberts, Interactions of retinoids and transforming growth factor-beta in regulation of cell differentiation and proliferation. Mol Endocrinol, 1991. 5(1): p. 3-7.

Page 156: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

143

260. Wilson, J.G. and J. Warkany, Cardiac and aortic arch anomalies in the offspring of vitamin A deficient rats correlated with similar human anomalies. Pediatrics, 1950. 5(4): p. 708-25.

261. Cohlan, S.Q., Congenital anomalies in the rat produced by excessive intake of vitamin A during pregnancy. Pediatrics, 1954. 13(6): p. 556-67.

262. Huang, F.J., et al., Adverse effects of retinoic acid on embryo development and the selective expression of retinoic acid receptors in mouse blastocysts. Hum Reprod, 2006. 21(1): p. 202-9.

263. Lammer, E.J., et al., Retinoic acid embryopathy. N Engl J Med, 1985. 313(14): p. 837-41.

264. Dai, W.S., J.M. LaBraico, and R.S. Stern, Epidemiology of isotretinoin exposure during pregnancy. J Am Acad Dermatol, 1992. 26(4): p. 599-606.

265. Suckow, M.A., Weisbroth, S.H., Franklin, C.L, ed. Embryology and teratology. The laboratory rat, ed. M.A. Suckow, Weisbroth, S.H., Franklin, C.L. 2006, Elselvier academic press: New York city.

266. Panzica-Kelly, J.M., et al., Morphological score assignment guidelines for the dechorionated zebrafish teratogenicity assay. Birth Defects Res B Dev Reprod Toxicol, 2010. 89(5): p. 382-95.

267. Brannen, K.C., et al., Development of a zebrafish embryo teratogenicity assay and quantitative prediction model. Birth Defects Res B Dev Reprod Toxicol, 2010. 89(1): p. 66-77.

268. De Santis, M., et al., The need for restricted prescription of retinoic acid derivative isotretinoin to prevent retinoid teratogenicity. Prev Med, 2007. 45(2-3): p. 243-4.

269. Lammer, E.J., et al., Isotretinoin dose and teratogenicity. Lancet, 1988. 2(8609): p. 503-4.

270. Maloney, M.E. and S.P. Stone, Isotretinoin and iPledge: a view of results. J Am Acad Dermatol, 2011. 65(2): p. 418-9.

271. Mishra, B.B. and V.K. Tiwari, Natural products: an evolving role in future drug discovery. Eur J Med Chem, 2011. 46(10): p. 4769-807.

272. Newman, D.J. and G.M. Cragg, Natural products as sources of new drugs over the last 25 years. J Nat Prod, 2007. 70(3): p. 461-77.

273. Koehn, F.E. and G.T. Carter, The evolving role of natural products in drug discovery. Nat Rev Drug Discov, 2005. 4(3): p. 206-20.

Page 157: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

144

274. Han, J., Traditional Chinese medicine and the search for new antineoplastic drugs. J Ethnopharmacol, 1988. 24(1): p. 1-17.

275. Eisenbrand, G., et al., Molecular mechanisms of indirubin and its derivatives: novel anticancer molecules with their origin in traditional Chinese phytomedicine. J Cancer Res Clin Oncol, 2004. 130(11): p. 627-35.

276. Hoessel, R., et al., Indirubin, the active constituent of a Chinese antileukaemia medicine, inhibits cyclin-dependent kinases. Nat Cell Biol, 1999. 1(1): p. 60-7.

277. Ji, X.J., et al., [Studies on the antineoplastic action and toxicity of synthetic indirubin (author's transl)]. Yao Xue Xue Bao, 1981. 16(2): p. 146-8.

278. Medicine, S.I.o.T.C., Subacute toxicity of indirubin in dogs, in Chinese Traditional and Herbal drugs. 1981. p. 27-29.

279. Gan, W.J., Yang, T., Wen, S., Liu, Y., Tan, Z., Deng, C., Wu, J., Liu, M., Studies on the mechanism of indirubin action in the treatment of chronic myelocytic leukemia. Chinese Academy of Medical Sciences Beijing, 1985. 6: p. 611-613.

280. Ji, X.J., Zhang, F.R., Antineoplastic effect of indirubin derivatives and their structure-activity relationship. Acta Pharmacological Sinica, 1985. 20: p. 137-139.

281. Wu, G.Y. and F.D. Fang, [Studies on the mechanism of indirubin action in the treatment of chronic granulocytic leukemia. II. Effects of indirubin on nucleic acid and protein synthesis in animal transplantable tumor cells and normal proliferating cells in vitro (author's transl)]. Zhongguo Yi Xue Ke Xue Yuan Xue Bao, 1980. 2(2): p. 83-7.

282. Du, D.J., Ceng, Q.T, Effect of indirubin on the incorporation of isotope labelled precursors into nucleic acid and protein of tumor tissues. Chinese Traditional and Herbal drugs, 1981. 12: p. 406-409.

283. Morgan, D.O., Cyclin-dependent kinases: engines, clocks, and microprocessors. Annu Rev Cell Dev Biol, 1997. 13: p. 261-91.

284. Marko, D., et al., Inhibition of cyclin-dependent kinase 1 (CDK1) by indirubin derivatives in human tumour cells. Br J Cancer, 2001. 84(2): p. 283-289.

285. Damiens, E., et al., Anti-mitotic properties of indirubin-3'-monoxime, a CDK/GSK-3 inhibitor: induction of endoreplication following prophase arrest. Oncogene, 2001. 20(29): p. 3786-97.

286. Shi, J. and H.M. Shen, Critical role of Bid and Bax in indirubin-3'-monoxime-induced apoptosis in human cancer cells. Biochem Pharmacol, 2008. 75(9): p. 1729-42.

Page 158: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

145

287. Sethi, G., et al., Indirubin enhances tumor necrosis factor-induced apoptosis through modulation of nuclear factor-kappa B signaling pathway. J Biol Chem, 2006. 281(33): p. 23425-35.

288. Nam, S., et al., Indirubin derivatives inhibit Stat3 signaling and induce apoptosis in human cancer cells. Proc Natl Acad Sci U S A, 2005. 102(17): p. 5998-6003.

289. Zhen, Y., et al., Indirubin-3'-monoxime inhibits autophosphorylation of FGFR1 and stimulates ERK1/2 activity via p38 MAPK. Oncogene, 2007. 26(44): p. 6372-85.

290. Alex, D., et al., Indirubin shows anti-angiogenic activity in an in vivo zebrafish model and an in vitro HUVEC model. J Ethnopharmacol, 2010. 131(2): p. 242-7.

291. Kim, J.-K., et al., Indirubin-3'-monoxime, a derivative of a chinese antileukemia medicine, inhibits angiogenesis. Journal of Cellular Biochemistry, 2011. 112(5): p. 1384-1391.

292. Jope, R.S., C.J. Yuskaitis, and E. Beurel, Glycogen synthase kinase-3 (GSK3): inflammation, diseases, and therapeutics. Neurochem Res, 2007. 32(4-5): p. 577-95.

293. Huang, W.C., et al., Glycogen synthase kinase-3 negatively regulates anti-inflammatory interleukin-10 for lipopolysaccharide-induced iNOS/NO biosynthesis and RANTES production in microglial cells. Immunology, 2009. 128(1 Suppl): p. e275-86.

294. Leclerc, S., et al., Indirubins inhibit glycogen synthase kinase-3 beta and CDK5/p25, two protein kinases involved in abnormal tau phosphorylation in Alzheimer's disease. A property common to most cyclin-dependent kinase inhibitors? J Biol Chem, 2001. 276(1): p. 251-60.

295. Perabo, F.G.E., et al., Antiproliferative and apoptosis inducing effects of indirubin-3′-monoxime in renal cell cancer cells. Urologic Oncology: Seminars and Original Investigations, 2009. 29(6): p. 815-820.

296. Polychronopoulos, P., et al., Structural basis for the synthesis of indirubins as potent and selective inhibitors of glycogen synthase kinase-3 and cyclin-dependent kinases. J Med Chem, 2004. 47(4): p. 935-46.

297. Ravichandran, K., A. Pal, and R. Ravichandran, Effect of indirubin-3-monoxime against lung cancer as evaluated by histological and transmission electron microscopic studies. Microscopy Research and Technique, 2010. 73(11): p. 1053-1058.

298. Sugihara, K., et al., Comparative study of aryl hydrocarbon receptor ligand activities of six chemicals in vitro and in vivo. Arch Toxicol, 2008. 82(1): p. 5-11.

299. Zhang, S., et al., Indirubin-3'-monoxime inhibits beta-amyloid-induced neurotoxicity in neuroblastoma SH-SY5Y cells. Neurosci Lett, 2009. 450(2): p. 142-6.

Page 159: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

146

300. Ding, Y., A. Qiao, and G.H. Fan, Indirubin-3'-monoxime rescues spatial memory deficits and attenuates beta-amyloid-associated neuropathology in a mouse model of Alzheimer's disease. Neurobiol Dis, 2010. 39(2): p. 156-68.

301. Dugo, L., et al., GSK-3beta inhibitors attenuate the organ injury/dysfunction caused by endotoxemia in the rat. Crit Care Med, 2005. 33(9): p. 1903-12.

302. Xie, Y., et al., Indirubin-3'-oxime inhibits c-Jun NH2-terminal kinase: anti-apoptotic effect in cerebellar granule neurons. Neurosci Lett, 2004. 367(3): p. 355-9.

303. Mak, N.K., et al., Inhibition of RANTES expression by indirubin in influenza virus-infected human bronchial epithelial cells. Biochem Pharmacol, 2004. 67(1): p. 167-74.

304. Kunikata, T., et al., Indirubin inhibits inflammatory reactions in delayed-type hypersensitivity. Eur J Pharmacol, 2000. 410(1): p. 93-100.

305. Suzuki, K., et al., Indirubin, a Chinese anti-leukaemia drug, promotes neutrophilic differentiation of human myelocytic leukaemia HL-60 cells. Br J Haematol, 2005. 130(5): p. 681-90.

306. Kim, J.K. and G.M. Park, Indirubin-3-monoxime exhibits anti-inflammatory properties by down-regulating NF-kappaB and JNK signaling pathways in lipopolysaccharide-treated RAW264.7 cells. Inflamm Res, 2012. 61(4): p. 319-25.

307. Sato, N., et al., Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling by a pharmacological GSK-3-specific inhibitor. Nat Med, 2004. 10(1): p. 55-63.

308. Wen, J., et al., Effects of 6-bromoindirubin-3'-oxime on the maintenance of pluripotency of porcine embryonic germ cells in combination with stem cell factor, leukemia inhibitory factor and fibroblast growth factor. Reproduction, 2010. 139(6): p. 1039-46.

309. Siekmann, A.F. and N.D. Lawson, Notch signalling limits angiogenic cell behaviour in developing zebrafish arteries. Nature, 2007. 445(7129): p. 781-4.

310. Nasevicius, A., J. Larson, and S.C. Ekker, Distinct requirements for zebrafish angiogenesis revealed by a VEGF-A morphant. Yeast, 2000. 17(4): p. 294-301.

311. Ozbudak, E.M. and J. Lewis, Notch signalling synchronizes the zebrafish segmentation clock but is not needed to create somite boundaries. PLoS Genet, 2008. 4(2): p. e15.

312. Lee, M.J., et al., Indirubin-3'-monoxime, a derivative of a Chinese anti-leukemia medicine, inhibits Notch1 signaling. Cancer Lett, 2008. 265(2): p. 215-25.

Page 160: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

147

313. Therapontos, C. and N. Vargesson, Zebrafish notch signalling pathway mutants exhibit trunk vessel patterning anomalies that are secondary to somite misregulation. Dev Dyn, 2010. 239(10): p. 2761-8.

314. Bertrand, J.A., et al., Structural characterization of the GSK-3beta active site using selective and non-selective ATP-mimetic inhibitors. J Mol Biol, 2003. 333(2): p. 393-407.

315. Sainson, R.C., et al., Cell-autonomous notch signaling regulates endothelial cell branching and proliferation during vascular tubulogenesis. FASEB J, 2005. 19(8): p. 1027-9.

316. Holderfield, M.T., et al., HESR1/CHF2 suppresses VEGFR2 transcription independent of binding to E-boxes. Biochem Biophys Res Commun, 2006. 346(3): p. 637-48.

317. Sangthong, S., et al., Synthesis of rotenoid derivatives with cytotoxic and topoisomerase II inhibitory activities. Bioorg Med Chem Lett, 2011. 21(16): p. 4813-8.

318. Robertson, D.R., Smith-Vaniz, W.F, Rotenone: An essential but demonized tool for assessing marine fish diversity. BioScience, 2008. 58(2): p. 165-170.

319. Fukami, H., Nakajima, M, ed. Naturally occurring insecticides. ed. M. Jacobson, Crosby, D.G. 1971, Dekker: New York. 71-97.

320. Cao, S., et al., Cytotoxic compounds from Mundulea chapelieri from the Madagascar Rainforest. J Nat Prod, 2004. 67(3): p. 454-6.

321. Singer, T.P. and R.R. Ramsay, The reaction sites of rotenone and ubiquinone with mitochondrial NADH dehydrogenase. Biochim Biophys Acta, 1994. 1187(2): p. 198-202.

322. Xiong, N., et al., Mitochondrial complex I inhibitor rotenone-induced toxicity and its potential mechanisms in Parkinson's disease models. Crit Rev Toxicol, 2012. 42(7): p. 613-32.

323. Tanner, C.M., et al., Rotenone, paraquat, and Parkinson's disease. Environ Health Perspect, 2012. 119(6): p. 866-72.

324. Kamal, R., Mangia, M., In vitro and in vivo investigations on rotenoids from indigofera tinctoria and their bioefficacy against the larvae of Anopheles stephensi and adults of Calmlosobruchus chinesis. Journal of Biosciences, 1993. 18(1): p. 93-101.

325. Chun, K.H., et al., Effects of deguelin on the phosphatidylinositol 3-kinase/Akt pathway and apoptosis in premalignant human bronchial epithelial cells. J Natl Cancer Inst, 2003. 95(4): p. 291-302.

326. Murillo, G., et al., Deguelin inhibits the growth of colon cancer cells through the induction of apoptosis and cell cycle arrest. Eur J Cancer, 2002. 38(18): p. 2446-54.

Page 161: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

148

327. Kang, H.W., et al., Deguelin, an Akt Inhibitor, Down-Regulates NF-kappaB Signaling and Induces Apoptosis in Colon Cancer Cells and Inhibits Tumor Growth in Mice. Dig Dis Sci, 2012. 57(11): p. 2873-82.

328. Lee, J.H., et al., Deguelin inhibits human hepatocellular carcinoma by antiangiogenesis and apoptosis. Oncol Rep, 2008. 20(1): p. 129-34.

329. Lee, H., et al., Deguelin promotes apoptosis and inhibits angiogenesis of gastric cancer. Oncol Rep, 2010. 24(4): p. 957-63.

330. Oh, S.H., et al., Structural basis for depletion of heat shock protein 90 client proteins by deguelin. J Natl Cancer Inst, 2007. 99(12): p. 949-61.

331. Isaacs, J.S., W. Xu, and L. Neckers, Heat shock protein 90 as a molecular target for cancer therapeutics. Cancer Cell, 2003. 3(3): p. 213-7.

332. Caboni, P., et al., Rotenone, deguelin, their metabolites, and the rat model of Parkinson's disease. Chem Res Toxicol, 2004. 17(11): p. 1540-8.

333. Kim, W.Y., et al., A novel derivative of the natural agent deguelin for cancer chemoprevention and therapy. Cancer Prev Res (Phila), 2008. 1(7): p. 577-87.

334. Wetli, H.A., P.D. Buckett, and M. Wessling-Resnick, Small-molecule screening identifies the selanazal drug ebselen as a potent inhibitor of DMT1-mediated iron uptake. Chem Biol, 2006. 13(9): p. 965-72.

335. Li, N., et al., Mitochondrial complex I inhibitor rotenone induces apoptosis through enhancing mitochondrial reactive oxygen species production. J Biol Chem, 2003. 278(10): p. 8516-25.

336. Ling, N., Rotenone - a review of its toxicity and use for fisheries management, in Science for Conservation. 2003 Department of Conservation: Wellington, New Zealand.

337. van Ham, T.J., et al., Live imaging of apoptotic cells in zebrafish. FASEB J, 2010. 24(11): p. 4336-42.

338. Ban, T.A., The role of serendipity in drug discovery. Dialogues Clin Neurosci, 2006. 8(3): p. 335-44.

339. Brown, D. and G. Superti-Furga, Rediscovering the sweet spot in drug discovery. Drug Discov Today, 2003. 8(23): p. 1067-77.

340. Hoff, P.M. and K.K. Machado, Role of angiogenesis in the pathogenesis of cancer. Cancer Treat Rev, 2012. 38(7): p. 825-33.

Page 162: VALIDATION AND MECHANISM STUDIES OF NOVEL THERAPEUTIC COMPOUNDS MODULATING ANGIOGENESIS · 2013-07-18 · Discovering novel compounds that stimulate or abrogate angiogenesis can lead

149

341. Zon, L.I., Peterson, Randall. T, In vivo drug discovery in the zebrafish. . Nature Reviews Drug Discovery, 2005. 4: p. 44.

342. Gunsalus, K.C., et al., Predictive models of molecular machines involved in Caenorhabditis elegans early embryogenesis. Nature, 2005. 436(7052): p. 861-5.

343. Liu, R., et al., Automated phenotype recognition for zebrafish embryo based in vivo high throughput toxicity screening of engineered nano-materials. PLoS One, 2012. 7(4): p. e35014.

344. Schlueter, P.J. and R.T. Peterson, Systematizing serendipity for cardiovascular drug discovery. Circulation, 2009. 120(3): p. 255-63.

345. Hutchinson, L. and R. Kirk, High drug attrition rates--where are we going wrong? Nat Rev Clin Oncol, 2011. 8(4): p. 189-90.

346. Kola, I. and J. Landis, Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug Discov, 2004. 3(8): p. 711-5.

347. Baker, M., Screening: the age of fishes. Nat Methods, 2011. 8(1): p. 47-51.