the use of endogenous and synthetic cannabinoids in prostate cancer therapy · 2018-11-15 ·...

147
The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy by Domenica Roberto A thesis submitted in conformity with the requirements for the degree of Master of Science Institute of Medical Science University of Toronto © Copyright by Domenica Roberto, 2018

Upload: others

Post on 03-Aug-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy

by

Domenica Roberto

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

Institute of Medical Science University of Toronto

© Copyright by Domenica Roberto, 2018

Page 2: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

ii

The Use of Endogenous and Synthetic Cannabinoids in Prostate

Cancer Therapy

Domenica Roberto

Master of Science

Institute of Medical Science

University of Toronto

2018

Abstract

Cannabinoids are mainly used as an antiemetic and for cancer-related pain, however, studies

have implicated an anti-proliferative role in various cancer models. This thesis examines the

therapeutic potential of cannabinoids anandamide and WIN55,212-2 in prostate cancer.

In vitro studies on prostate cancer cells showed that the cannabinoids significantly reduce

proliferation, migration, invasion, and induce apoptosis in a dose-dependent manner. Inhibition

of cannabinoid receptor 2 resulted in a reversal of the anti-proliferative effects. Cell cycle

analysis revealed that WIN55,212-2 caused arrest in G1/2 phases, and mechanistic studies

demonstrated that these effects were mediated through alterations in key cell cycle regulators.

Based on these studies, the effect of WIN55,212-2 was assessed using a xenograft model,

resulting in a reduction in tumor volume compared to control.

Evidence from this thesis provides a framework for future studies and provides a more in-depth

understanding of the potential benefit of cannabinoid use in prostate cancer.

Page 3: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

iii

Acknowledgments

First and foremost, I would like to thank my primary supervisor Dr. Vasundara Venkateswaran

and my co-supervisor Dr. Laurence Klotz. They accepted me into the lab with very little

experience and helped me to identify my passion for research. They have consistently mentored

and supported me throughout my degree, provided me with numerous opportunities to grow as a

scientist, and pushed me to reach my potential.

My sincere thanks must also go to my fellow lab members both past and present. Michelle

Mayer who introduced me to the lab, Dr. Natalie Venier for all her advice and detailed protocol

notes, Dr. Azik Hoffman who greatly contributed to my understanding of urology and provided

guidance throughout my degree, and Dr. Roman Bass for his positive influence towards the end

of my degree.

I would like to acknowledge the amazing people I have met at Sunnybrook for inspiring me and

making my experience as a graduate student a positive one. These individuals offered their time

and expertise so generously and have become great friends that I am truly lucky to have met.

I am grateful to my Program Advisory Committee Members Dr. Urban Emmenegger and Dr.

David Ma for lending me their expertise and intuition, instrumental for the completion of my

degree. I would like to recognize my examiners Dr. Stanley Liu, Dr. Marianne Koritzinsky, and

Dr. Sanjay Gupta who took the time to review and edit my thesis.

I would like to acknowledge the IMS department for awarding me the Entrance Award and the

Open Fellowship Award which has provided me with financial support throughout my degree.

I deeply thank and dedicate this thesis to my wonderful parents for their unconditional love and

endless patience. I have learned so much from them and I appreciate all of the sacrifices that they

have made for me. My brother, Adriano who has encouraged me to pursue my passion and who

has always supported me when times were challenging. My Nonna, for the knowledge and

values she has instilled in me; I know that she would be proud of the person I have become. I

cannot thank them enough for everything they have done for me and would not be where I am

today without them.

Page 4: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

iv

At last, I would like to express my deepest gratitude to my best friend and soul mate, Kogulan

who has provided me continuous support, love and encouragement not only throughout my

degree, but throughout every aspect of my life. His positive outlook on life has given me the

strength to accomplish anything. I am blessed to have someone so kind, loving, and selfless in

my life; he has taught me so much about myself and continuously inspires me to become a better

person.

Page 5: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

v

Table of Contents

Table of Contents

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

Table of Contents .............................................................................................................................v

List of Abbreviations ..................................................................................................................... ix

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

List of Figures ............................................................................................................................... xii

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

Introduction .................................................................................................................................1

1.1 Prostate Anatomy .................................................................................................................1

1.2 Prostate Cancer Epidemiology .............................................................................................2

1.3 Prostate Cancer Pathophysiology ........................................................................................2

1.4 Prostate Cancer Detection and Diagnosis ............................................................................4

1.5 Treatment Strategies ............................................................................................................6

1.5.1 Active Surveillance ..................................................................................................6

1.5.2 Surgery .....................................................................................................................6

1.5.3 Androgen Deprivation Therapy ...............................................................................7

1.5.4 Radiotherapy ............................................................................................................7

1.5.5 Adjuvant and Neo-Adjuvant Therapies ...................................................................8

1.5.6 Chemotherapy ..........................................................................................................8

1.5.7 Chemopreventive and Novel Agents .......................................................................9

1.6 Cannabinoids......................................................................................................................10

1.6.1 Phytocannabinoids .................................................................................................11

1.6.2 Endocannabinoids ..................................................................................................12

1.6.3 Synthetic Cannabinoids .........................................................................................17

Page 6: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

vi

1.7 Molecular Targets of Cannabinoids ...................................................................................19

1.7.1 Endocannabinoid System .......................................................................................19

1.7.2 Cannabinoids and Non-Cannabinoid Receptors ....................................................21

1.7.3 Endoplasmic Reticulum Stress Response ..............................................................25

1.7.4 Oxidative Stress .....................................................................................................28

1.7.5 Rho GTPase Signalling ..........................................................................................32

1.7.6 Apoptosis ...............................................................................................................35

1.7.7 Cell- Cycle Regulation ...........................................................................................37

1.8 Preclinical Models of Prostate Cancer ...............................................................................46

1.8.1 In Vitro Models ......................................................................................................46

1.8.2 In Vivo Models .......................................................................................................48

1.8.2.1 Xenograft Mouse Models ........................................................................48

1.8.2.2 TRAMP Mouse Model ............................................................................49

1.8.2.3 Lady Transgenic Model ...........................................................................50

1.8.2.4 The Phosphatase and tensin homolog deleted on chromosome ten

(PTEN) Model .........................................................................................50

1.8.2.5 c-MYC Model .........................................................................................51

1.8.2.6 NK3 Homeobox (1NKX3.1) Model ........................................................51

Chapter 2 Rationale, Hypothesis, and Aims .................................................................................52

Rationale, Hypothesis, and Aims ..............................................................................................52

2.1 Rationale ............................................................................................................................52

2.2 Hypothesis..........................................................................................................................52

2.3 Aims ...................................................................................................................................53

Chapter 3 Materials and Methods .................................................................................................54

Materials and Methods ..............................................................................................................54

3.1 Cell Culture ........................................................................................................................54

Page 7: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

vii

3.2 Chemicals ...........................................................................................................................54

3.3 MTS Cell Proliferation Assay ............................................................................................54

3.4 Wound Healing (Scratch) Assay ........................................................................................55

3.5 Matrigel Invasion Assay ....................................................................................................56

3.6 Flow Cytometry .................................................................................................................57

3.6.1 Cell Cycle Distribution ..........................................................................................57

3.6.2 Apoptosis ...............................................................................................................58

3.7 Western Blot Analysis .......................................................................................................58

3.8 Xenograft Studies...............................................................................................................59

3.8.1 Animals and Housing .............................................................................................59

3.8.2 Establishment of Xenografts ..................................................................................59

3.8.3 Administration of WIN 55,212-2 ...........................................................................60

3.9 In Vitro Mitogenicity Assay...............................................................................................62

3.10 Statistical Analysis .............................................................................................................62

Chapter 4 Results ...........................................................................................................................63

Results .......................................................................................................................................63

4.1 Differential growth inhibitory effect of anandamide on prostate cancer cell lines ............63

4.2 Treatment with WIN-55,212-2 reduces prostate cancer cell proliferation ........................67

4.3 Anandamide and WIN 55,212-2 treatment reduces the migration and invasion

capacity of prostate cancer cells ........................................................................................71

4.4 Anandamide treatment does not significantly alter the cell cycle distribution in

DU145 and LNCaP cells ....................................................................................................75

4.5 WIN 55,212-2 treatment causes cell cycle arrest in DU145 and PC3 cells .......................78

4.6 Anandamide reduces proliferation and induces apoptosis in LNCaP cells but not in

DU145 cells .......................................................................................................................81

4.7 WIN 55,212-2 significantly induces apoptosis in PC3 and DU145 cells but not in

LNCaP cells .......................................................................................................................84

4.8 Cannabinoid receptor 2 antagonist AM630 does not alter prostate cancer cell growth ....88

Page 8: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

viii

4.9 Treatment with CB2 antagonist AM630 abrogates the anti-proliferative effects of

anandamide in DU145 and LNCaP cell lines ....................................................................90

4.10 Treatment with CB2 antagonist AM630 abrogates the anti-proliferative effects of

WIN 55,212-2 in prostate cancer cell lines ........................................................................92

4.11 WIN 55,212-2 treatment alters expression of pRb, Cdk4, and p27 in PC3 cells ...............94

4.12 WIN 55,212-2 treatment reduces tumor growth in a mouse xenograft model ..................96

4.13 Serum containing WIN 55,212-2 reduces PC3 cell proliferation ......................................99

Chapter 5 Discussion ...................................................................................................................103

Discussion ...............................................................................................................................103

Chapter 6 Future Directions and Overall Conclusion ..................................................................108

Future Directions and Overall Conclusion ..............................................................................108

6.1 Potential In Vivo Studies ..................................................................................................108

6.2 Current and Potential Clinical Trials ...............................................................................109

6.3 Overall Conclusion ..........................................................................................................110

References ....................................................................................................................................112

Appendix ......................................................................................................................................133

Page 9: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

ix

List of Abbreviations

ACPA: Arachidonoyl cyclopropamide

ADT: Androgen Deprivation Therapy

AEA: Anandamide

2AG: 2-arachidonoyl glycerol

ANOVA: One-Way Analysis of Variance

AOM: Azoxymethane

AR: Androgen Receptor

BCL-2: B-cell lymphoma 2 (oncogene)

BPH: Benign prostatic hyperplasia

BrdU: BromodeoxyUridine

CB: Cannabinoid Receptor

CBC: Cannabichromene

CBD: Cannabidiol

CBDA: Cannabidiolic acid

CBG: Cannabigerol

CBL: Cannabicyclol

CBN: Cannabinol

COX: Cyclooxygenase

CRC: Colorectal cancer

CRPC: Castration-Resistant Prostate Cancer

DAG: Diacylglycerol

DAGL: Diacylglycerol lipase

DMSO: Dimethyl Sulfoxide

DR5: Death Receptor 5

DRE: Digital Rectal Examination

DSS: Dextran sulfate sodium

DU145: Human Prostate Cancer Cell Line

EAU: European Association of Urology

EDTA: Ethylenediaminetetraacetic Acid

ER: Endoplasmic Reticulum

FAAH: Fatty Acid Amide Hydrolase

FITC: Fluorescein Isothiocyanate

FBS: Fetal Bovine Serum

GGG: Gleason Grade Group

GPCR: G-Protein Coupled Receptor

GW: GW405833

HCl: Hydrochloric Acid

HGPIN: High-Grade Prostatic Intraepithelial Neoplasia

5-HT: 5-hydroxytryptamine

LHRH: Luteinizing Hormone Releasing Hormone

LNCaP: Lymph Node Carcinoma of the Prostate

MAGL: Monoacylglycerol lipase

MAPK: Mitogen-activated Protein Kinase

MET: Methanandamide

Met-F-AEA: 2-methyl-2’-F- anandamide

Page 10: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

x

MMP: Metalloproteinase

mTOR: Mammalian Target of Rapamycin

NAPE: N-arachidonoyl-phosphatidylethanolamine

NAPE-PLD: NAPE-specific phospholipase D

NAT: N-acyl transferase

NCCN: National Comprehensive Cancer Network

NPC: Nutrition Prevention of Cancer Trial

OEA: N-oleoylethanolamide

p53: Tumor Suppressor Protein 53

PARP: Poly (ADP-ribose) polymerase

PBS: Phosphate Buffered Saline

PC3: Human Prostate Cancer Cell Line

PCa: Prostate cancer

pCB: Phytocannabinoid

PDX: Patient-derived

PEA: N-palmitoylethanolamide

PI: Propidium Iodide

PIN: Prostatic Intraepithelial Neoplasia

PIP2: Phosphatidylinositol-4,5-bisphosphate

PKA: Protein kinase A

PLC: Phospholipase C

PPAR: Nuclear Peroxisome Proliferator-Activated Receptor

PSA: Prostate Specific Antigen

PTEN: Phosphatase and tensin homolog deleted on chromosome ten

QOL: Quality of life

Rb: Retinoblastoma (tumor suppressor gene)

ROS: Reactive Oxygen Species

SC: Synthetic Cannabinoid

SD: Standard deviation

SDS: Sodium Dodecyl Sulfate

SELECT: Selenium and Vitamin E Cancer Prevention Trial

SRI: Sunnybrook Research Institute

∆8-THC: delta-8-tetracannabinol

THC: delta-9-tetrahydrocannabinol

TNF: Tumor necrosis factor

TRAMP: Transgenic Adenocarcinoma Mouse Prostate

TRB3: Tribbles Homolog 3

TRP: Transient Receptor Potential

TRPV1: Transient Receptor Potential cation channel Vanilloid Type 1

TRUS: Transrectal Ultrasound

UPR: Unfolded Protein Response

WIN: WIN 55,212-2

Page 11: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

xi

List of Tables

Table 1: Effects of cannabinoids on cell viability, migration, and invasion in prostate and various

cancers.

Table 2: General characteristics of common immortalized human prostate cancer cell lines.

Table 3: Summary of in vitro results.

Table 4: Summary of in vivo results.

Page 12: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

xii

List of Figures

Figure 1: The prostate anatomy.

Figure 2: The process of carcinogenesis.

Figure 3: The biosynthesis of anandamide.

Figure 4: The biosynthesis of 2-arachidonoylglycerol.

Figure 5: The chemical structure of anandamide.

Figure 6: The chemical structure of WIN 55,212-2.

Figure 7: Cannabinoid receptor localization.

Figure 8: Proposed endoplasmic reticulum stress signalling pathway.

Figure 9: Proposed oxidative stress signalling pathway.

Figure 10: Proposed RhoA GTPase signalling pathway.

Figure 11: Proposed cell cycle regulation pathway.

Figure 12: MTS cell proliferation assay.

Figure 13: Wound healing (scratch) assay.

Figure 14: Matrigel invasion assay.

Figure 15: Xenograft establishment and group assignment.

Figure 16: Xenograft experiment timeline.

Figure 17: Effect of anandamide treatment on proliferation of PC3 cells.

Figure 18: Effect of anandamide treatment on proliferation of LNCaP cells.

Figure 19: Effect of anandamide treatment on proliferation of DU145 cells.

Page 13: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

xiii

Figure 20: Effect of WIN 55,212-2 treatment on proliferation of PC3 cells.

Figure 21: Effect of WIN 55,212-2 treatment on proliferation of LNCaP cells.

Figure 22: Effect of WIN 55,212-2 treatment on proliferation of DU145 cells.

Figure 23: Effect of 24hr treatment of anandamide on DU145 cell migration and invasion.

Figure 24: Effect of 24hr treatment of WIN 55,212-2 on DU145 cell migration and invasion.

Figure 25: Effect of 24hr treatment of WIN 55,212-2 on PC3 cell migration and invasion.

Figure 26: Effect of anandamide treatment on cell cycle distribution in DU145 cells.

Figure 27: Effect of anandamide treatment on cell cycle distribution in LNCaP cells.

Figure 28: Effect of WIN 55,212-2 treatment on cell cycle distribution in DU145 cells.

Figure 29: Effect of WIN 55,212-2 treatment on cell cycle distribution in PC3 cells.

Figure 30: Effect of anandamide on proportion of live versus apoptotic DU145 cells using

Annexin V flow cytometry.

Figure 31: Effect of anandamide on proportion of live versus apoptotic LNCaP cells using

Annexin V flow cytometry.

Figure 32: Effect of WIN 55,212-2 on proportion of live versus apoptotic PC3 cells using

Annexin V flow cytometry.

Figure 33: Effect of WIN 55,212-2 on proportion of live versus apoptotic DU145 cells using

Annexin V flow cytometry.

Figure 34: Effect of WIN 55,212-2 on proportion of live versus apoptotic LNCaP cells using

Annexin V flow cytometry.

Figure 35: Effect of Cannabinoid receptor 2 antagonist AM630 on viability of prostate cancer

cell lines.

Page 14: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

xiv

Figure 36: Effect of anandamide after treatment with cannabinoid receptor 2 antagonist AM630

on proliferation of DU145 and LNCaP cell lines.

Figure 37: Effect of WIN 55,212-2 after treatment with cannabinoid receptor 2 antagonist

AM630 on cell proliferation.

Figure 38: Effect of WIN 55,212-2 on expression of cell cycle regulator proteins.

Figure 39: WIN 55,212-2 significantly reduces tumor growth rate.

Figure 40: Lack of effect of WIN 55,212-2 treatment on animal weight.

Figure 41: Representative images of mice tumors before and after excision.

Figure 42: Effect of WIN 55,212-2 containing serum on proliferation of PC3 cells.

Page 15: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

1

Chapter 1 Introduction

Introduction

1.1 Prostate Anatomy

The prostate is a gland of the male reproductive system (Figure 1), located anterior to the rectum

and inferior to the bladder. It is the size and shape of a walnut but increases in size when men

reach their late forties and early fifties. It surrounds the urethra, the tube that carries urine and

semen through the penis.

The main functions of the prostate are to produce prostatic fluid for semen and to contribute to

urinary continence. Prostatic fluid is rich in enzymes, proteins and minerals that help protect and

nourish sperm. Hormones, including testosterone and those made by the adrenal and pituitary

glands, help control the function of the prostate gland.

Figure 1: The Prostate Anatomy. The location of the prostate gland below the bladder and

surrounding the urethra.

Page 16: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

2

1.2 Prostate Cancer Epidemiology

Prostate cancer (PCa) is the second most commonly diagnosed cancer, with 1.1 million new

cases reported globally (Zhou et al., 2016). It is one of the leading causes of morbidity and

mortality worldwide and thus considered as an important public health issue. Risk factors for

prostate cancer include age, family history, race, diet, and certain genetic polymorphisms,

amongst many others (Fradet et al 2009). The incidence rates of PCa vary by more than 50-fold

worldwide, with highest rates observed in Australia, North America, and Western Europe, and

lowest in South Central Asia and China (Wong et al., 2016). The practice of prostate specific

antigen (PSA) screening and subsequent biopsy in developed countries has led to a rise in PCa

incidence rates. However, earlier diagnosis comes at the expense of potentially treating a

significant proportion of men who are at little risk of symptom development or associated

disease complications during their lifetime (Fradet, Klotz, Trachtenberg, & Zlotta, 2009).

In comparison to incidence and detection, the trends in mortality are less clear. With over

300,000 deaths worldwide, PCa is the fifth leading cause of death from cancer in men (Wong et

al 2016). PSA testing substantially increases prostate cancer incidence rates. However, the effect

of this detection tool on mortality reduction is less clear. There is a ten-fold variation in mortality

rates worldwide (Ferlay et al., 2013). The lowest mortality rates reported are in Asia and North

Africa, and the highest death rates are seen in the Caribbean (Wong et al 2016). The variation in

PCa mortality rates can be attributed to various factors. It is accounted for by differences in

genetic predisposition, as well as variations in accuracy of recording mortality

causes and differences in treatment strategies (Wong et al 2016).

1.3 Prostate Cancer Pathophysiology

As is the case with other cancers, PCa development depends on cardinal characteristics such as

sustained proliferative signalling, evasion of growth suppressors, avoiding immune destruction,

resistance to cell death, replicative immortality, promotion of inflammation, angiogenesis,

invasion and migration, genome instability and deregulation of cellular energetics (D Hanahan &

Weinberg, 2000)(,Douglas Hanahan & Weinberg, 2011).

Prostate cancer development occurs in three stages (Figure 2). In the initiation stage, normal cells

acquire irreversible mutations. Overtime, these mutations accumulate, further deregulating cell

Page 17: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

3

growth and leading to a tumor large enough to become detectable. Eventually, cells enter the

promotion phase, where they can undergo biological effects without metabolic activation. In the

final stage of progression, tumors acquire a malignant phenotype. These may remain localized to

the prostate, or evolve the ability to secrete proteases and other mediators of invasion that allow

infiltration and metastasis (Abel & DiGiovanni, 2011),(Pitot, 1993).

Figure 2: The Process of Carcinogenesis. Prostate cancer develops in three stages: initiation,

promotion and progression. Figure was adapted from (Siddiqui, Sanna, Ahmad, Sechi, &

Mukhtar, 2015).

Greater than 95% of prostate cancers are classified as adenocarcinoma. Within this classification,

there is profound molecular and phenotypic heterogeneity. This heterogeneity underlies the

distinction between latent and clinical disease, as well as the correlation between PCa

progression and aging. Prostate cancer develops commonly with age. It is ubiquitous in aging

men, and common in autopsies on young men dying of other unknown causes. This supports the

view that prostatic carcinogenesis is initiated early in life (Yatani, Kusano, Shiraishi, Hayashi, &

Stemmermann, 1989). High-grade prostatic intraepithelial neoplasia (HGPIN), defined as a

neoplastic growth of epithelial cells within pre-existing prostatic acini or ducts, is considered to

be a precursor for PCa development, with more than 40% of HGPIN patients clinically

diagnosed with PCa within 3 years of HGPIN diagnosis (S. H. Lee et al., 2016). Prostatic

intraepithelial neoplasia (PIN) spreads through the prostatic ducts causing cell proliferation and

cytologic changes similar to those of cancer. PIN is associated with progressive phenotypic and

genotypic abnormalities that are an intermediate between normal prostate epithelium and

prostate cancer, with more than 36 genetic and molecular alterations reported (Klink,

Miocinovic, Galluzzi, & Klein, 2012). A current area of uncertainty is whether high grade,

aggressive cancer develops in most cases from low grade cancer, or has a separate phylogeny

derived directly from PIN.

Page 18: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

4

1.4 Prostate Cancer Detection and Diagnosis

Throughout the developed world there are efforts to improve the outcome of prostate cancer

through early detection. The concept is to detect the cancer at a point where it is more amenable

to cure, resulting in improved survival and quality of life (QOL). Currently, the main screening

measures that exist to help detect PCa at an earlier stage include serum concentration of prostate-

specific antigen, digital rectal examination (DRE), and the transrectal ultrasound (TRUS)-guided

biopsy (Tenke, Horti, Balint, & Kovacs, 2007).

PSA is a protein produced by the cells of the prostate and is secreted into seminal fluid in high

concentrations. Trace amounts of PSA can be detected in circulating blood of healthy men,

which allows PSA to be measured using a blood test. Although the adoption of PSA screening

since the early 1990s has dramatically increased the detection of PCa, this has also contributed to

presumptive overdiagnosis and overtreatment, resulting in unfavourable effects on patient’s QOL

(E. H. Kim & Andriole, 2015).

DRE is a procedure in which a healthcare professional inserts a gloved finger into the rectum to

check for abnormalities in the size and shape of the prostate. DRE has low sensitivity when used

alone. However, when used together with PSA testing, the detection rate can be improved. Of the

three, TRUS guided biopsy is the most invasive and is associated with higher costs. The DRE

aids not only in diagnosing PCa but also in determining its clinical stage.

TRUS-guided biopsy has become the standard way to obtain material for histopathologic

examination. Web based nomograms allow for the integration of multiple risk factors such as

PSA level, age, family history, positive DRE, among others, to provide a risk prediction of the

likelihood cancer considered to be of significant nature. This level of risk drives the decision to

perform a biopsy. In this procedure, a biopsy needle is inserted into the prostate gland and small

samples of tissue are removed for pathological examination. In general, for a prostate gland

volume of 30-40ml, 10-12 cores should be sampled (Heidenreich et al., 2011). The cores are then

examined to evaluate microscopic features of potential cancer cells and the Gleason grade(s) of

the tumor are reported, as well as extent of cancer, presence of lymphovascular or perineural

invasion, if any.

The Gleason system remains one of the most powerful prognostic predictors in prostate cancer,

Page 19: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

5

and accurate grading is crucial for predicting a patient’s prognosis and treatment options (Epstein

et al., 2016). Depending on morphological PCa growth features, a score between 1-5 is applied,

with higher numbers indicating worse prognosis. In the current application, Gleason patterns of 1

and 2 are no longer assigned on needle core biopsy, however, consist of single closely-packed

glands with well-defined edges (Gleason 1), and simple, round, loosely-packed glands (Gleason

2). Gleason pattern 3 consists of well-formed, individual glands of varying sizes. Gleason

pattern 4 includes poorly-formed glands, fused into chords or chains. Gleason pattern 5 consists

of sheets of tumor, individual cells, and cords of cells with no glandular differentiation

(Gordetsky & Epstein, 2016).

The Gleason score is a combination of two tissue grades; the primary grade, and the secondary

grade. The primary grade is assigned to the dominant pattern of the tumor, whereas the

secondary grade is assigned to the next-most frequent pattern. The sum of the primary and

secondary grades results in the final Gleason score, which ranges from 6-10. Based on these

grading scores, tumors can be categorized into groups that show similar biologic behavior.

Scores of 6 (3+3) are now referred to as Gleason grade group (GGG) 1, Gleason scores 3+4 are

GGG II, Gleason score 4+3 is GGG III, Gleason score 4+4 is GGG IV, and Gleason scores of 9-

10 are GGG V. This new classification makes it easier for counselling patients on disease status

in comparison to the Gleason risk stratification groups (≤6, 7, 8–10) (Pierorazio, Walsh, Partin,

& Epstein, 2013).

Aside from Gleason score, tumor staging is another useful technique to predict the pathological

stage of prostate cancer. Currently, the TNM staging is the most widely used system for prostate

cancer staging. It aims to determine the extent of the primary tumor (T), the absence or presence

of regional lymph node (N) involvement, and the presence or absence of distant metastases (M).

Once the T, N, and M are determined, a stage of I, II, III, or IV can be assigned, with stage I

being early disease and stage IV being advanced disease (Cosma et al., 2016).

After diagnosis with prostate cancer, the National Comprehensive Cancer Network (NCCN),

recommends further classification of the cancer into one of four risk categories- very low risk,

low risk, intermediate risk, or high risk. As with staging and grading, several factors are taken

into consideration when determining risk category, including PSA level, size of the prostate,

biopsy results, and stage of disease. Very low risk prostate cancers include stage T1c, PSA less

Page 20: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

6

than 10 ng/mL, and Gleason score 6 or less. Low risk cancers include stage T1c or T2a, PSA less

than 10 ng/mL, and Gleason score 6 or less. Patients classified as intermediate risk include stage

T2b-T2c, or PSA 10 to 20 ng/mL, or Gleason score 7. Lastly, prostate cancers classified as high

risk include stage T3a, PSA 20 ng/mL or higher, and Gleason score 8 or higher (Clinical,

Clinical, Guidelines, & Guidelines, 2009).

1.5 Treatment Strategies

Treatment choices for patients with prostate cancer depend on a combination of patient and

tumor characteristics. In the following section, various management and treatment modalities

will be discussed.

1.5.1 Active Surveillance

Many patients with prostate cancer are estimated to have a prolonged natural history of disease

which poses little threat during their lifetime. In such cases, patients are placed on active

surveillance in an effort to limit unnecessary treatments (Dall’Era et al., 2012). Under active

surveillance, patients with low-risk disease are carefully observed with repeated PSA

assessments, biopsies, and other tests to identify early signs of progression to more advanced

PCa. At the first signs of progression, these patients are placed on an appropriate treatment

regime within a timely window for opportunity in curing the disease (Cooperberg, Carroll, &

Klotz, 2011; Klotz, 2005). Active surveillance for prostate cancer offers an opportunity to delay

active treatment and its associated morbidities until evidence of clinical cancer progression is

discovered, which is never the case in a majority of low risk patients (Klotz et al., 2015).

1.5.2 Surgery

Treating patients with radical prostatectomy, in which the prostate gland and its surrounding

tissue is surgically excised, is a common primary treatment option for patients with intermediate

to high-risk prostate cancer. In a clinical trial comparing radical prostatectomy with observation

in the management of early prostate cancer, results show that radical prostatectomy reduced the

risk of death due to PCa by 50% and the risk of distant metastasis by 37% (Bill-Axelson et al.,

2005, 2011). The primary limitations of surgery include associated risk of incontinence and

erectile dysfunction amongst others, as well as positive surgical margins in patients with locally

advanced disease.

Page 21: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

7

1.5.3 Androgen Deprivation Therapy

Androgens play a key role in prostate cancer development and progression. As such, targeting

the androgen receptor signalling pathway has been considered an effective strategy in PCa

management. Androgen deprivation therapy (ADT) is a treatment strategy wherein the

production or effects of testosterone and other male hormones are blocked. ADT can be achieved

by either surgical removal of both testicles or by medications which inhibit testosterone

production. Surgical castration is rarely used due to its permanent and irreversible nature,

combined with patients’ requests to avoid this procedure. The medications include anti-

androgens, which block ligand binding to the androgen receptor; luteinizing hormone releasing

hormone (LHRH) agonists or antagonists, which reduce luteinizing hormone secretion by the

pituitary, resulting in inhibition of gonadal synthesis of androgens; or new agents such as

Abiraterone, an inhibitor of Cyp 17, a crucial enzyme for androgen production. In addition to the

well-established role of ADT in treating patients with metastatic disease (Fizazi et al., 2012;

Scher et al., 2012), it is also used to treat patients with increasing PSA levels after local

treatment, and acts as an adjuvant therapy for men with localized disease undergoing radiation

therapy (Sharifi, Gulley, & Dahut, 2005).

1.5.4 Radiotherapy

Radiation therapy is a curative and effective therapy for localized prostate cancer. Although

there is a great deal of variability between risk group and treatment centers, a fairly large

percentage of men with localized PCa receive external beam radiation and brachytherapy

(Cooperberg, Broering, & Carroll, 2010). Radiation interacts with DNA in normal and malignant

cells, causing genetic damage and eventual cell death. With the development of highly conformal

techniques to limit the amount of normal tissues exposed to radiation, large cumulative doses can

be delivered in small daily fractions spread out over several weeks of treatment (Martin &

D’Amico, 2014). The conventional regime for prostate radiotherapy includes 70-80 Gy given in

2-2.5 Gy daily fractions. Alternative regimes have been used, including the delivery of fewer,

larger fractions, also known as hypofractionation. In addition to its economic and logistic

advantages, hypofractionation offers great tumor control and reduced normal tissue toxicities

(Stein, Boehmer, & Kuten, 2007). With brachytherapy, multiple radioactive seeds are implanted

into the tumor site under ultrasound guidance. This technique has also yielded favourable results,

Page 22: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

8

with reports of ten-year disease-specific survival rates in favorable patients of greater than 95%

of treated patients (Stock, Cesaretti, & Stone, 2006).

1.5.5 Adjuvant and Neo-Adjuvant Therapies

Adjuvant therapy is often used after primary treatment, such as surgery, in order to reduce the

risk of cancer recurrence. Some of the main modalities used as adjuvant therapies include,

chemotherapy, hormone therapy, and radiation therapy (A V Bono, 2004).

Neo-adjuvant therapy is the administration of therapeutic agents after diagnosis but before

primary treatment with the goal of enhancing the effectiveness of local therapy, down staging

disease and eliminating micrometastasis. Neoadjuvant therapy not only provides the benefit of

assessing the efficacy of a particular treatment in the patient and gauging prognosis, but also

helps to increase the number of patients eligible for local therapy (Kent & Hussain, 2003). There

are several studies that have demonstrated the reduced incidence of positive disease margins with

the use of neo-adjuvant hormone therapy and surgery as treatment for PCa, with no reported

improvement in PSA recurrence rates (bNED survival) (Aldo V. Bono et al., 2001; Soloway et

al., 1995, 2002). Additionally, studies investigating neoadjuvant hormone therapy with

brachytherapy in locally advanced disease have reported improved biochemical outcomes in

high-risk patients (Merrick, Butler, Galbreath, Lief, & Adamovich, 2003).

1.5.6 Chemotherapy

Patients with advanced PCa initially respond to ADT in 90% of cases. However, a significant

proportion of patients develop castrate resistant prostate cancer (CRPC) over the course of time,

leading to the increased use of cytotoxic chemotherapy for management. CRPC is characterized

by a continuous rise in serum PSA levels, or other signs of PCa progression, despite castrate

testosterone levels (Saad & Hotte, 2010). According to the European Association for Urology

(EAU) guidelines on PCa, patients with asymptomatic or mildly asymptomatic metastatic disease

typically should be treated with enzalutamide or abiraterone plus prednisone, whereas patients

with aggressive metastatic CRPC should receive docetaxel in combination with prednisone, at

three week intervals (Heidenreich et al., 2014). Additionally, cabazitaxel is also considered for

its significant increases in overall survival in patients with docetaxel-resistant metastatic CRPC

(Hotte & Saad, 2010). Radium 223 is a bone seeking radioisotope for men with bone metastases

only offering a survival benefit when compared to best supportive care.

Page 23: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

9

1.5.7 Chemopreventive and Novel Agents

Chemoprevention is the use of specific agents to hinder or delay the process of carcinogenesis

(described in prostate cancer pathophysiology), thus preventing the development of advanced

disease (Syed, Khan, Afaq, & Mukhtar, 2007). PCa is considered an ideal disease candidate for

chemoprevention given its extended duration of time for disease inception to clinical diagnosis,

combined with a relatively slow growth rate. Thus, minor delays in the development of PCa may

result in a substantial reduction in the incidence of clinically relevant disease.

Dietary agents, including lycopene, vitamins D, green tea polyphenols, and capsaicin have the

potential to be chemopreventive agents. Several other potentially interesting micronutrients have

failed in prospective phase 3 trials to demonstrate benefit, or have caused harm, despite pre-

clinical evidence supporting their use. These agents interact with a variety of oncogenic

pathways, including cell cycle processes, apoptosis, androgen metabolism, and oxidative stress

(Venkateswaran & Klotz, 2010). The benefits of selenium, for example, were demonstrated in

vitro and in vivo studies, showing activation of apoptosis and inhibition of PCa development (Hu

et al., 2006; S. O. Lee et al., 2006). The Nutrition Prevention of Cancer Trial (NPC) showed a

50% reduction in PCa incidence among men supplemented with selenium. However, the

Selenium and Vitamin E Cancer Prevention Trial (SELECT) demonstrated no benefit from

selenium and found an increase in the rates of diabetes in the selenium arm (Duffield-Lillico et

al., 2003; Lippman et al., 2009). Lycopene has also been shown to reduce the amount of

oxidative DNA damage in cell and animal studies (Matos, Capelozzi, Gomes, Mascio, &

Medeiros, 2001), along with phase II studies demonstrating significant decreases in PSA levels

over one year (Barber et al., 2006). An abundance of evidence has also depicted the role of

vitamin D in the inhibition of proliferation, invasion and PCa metastasis (Kubota et al., 1998;

Donna M Peehl, Krishnan, & Feldman, 2003). This also included epidemiological data

portraying decreases in PCa incidence and PSA levels in patients taking vitamin D

supplementation (Colli & Colli, 2006). Vitamin E is also a potent intracellular antioxidant,

recognized for its ability to inhibit lipid peroxidation and regulate the cell cycle through DNA

synthesis arrest (Israel, Sanders, & Kline, 1995). Vitamin E, which was evaluated in the

SELECT trial, was associated with an increased the risk of PCa development (Lippman et al

2009). In comparison, green tea, with its high polyphenolic content, has been shown to be an

effective chemopreventive agent for a variety of cancers. Studies have shown its potential to

Page 24: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

10

inhibit cell growth, induce apoptosis and inhibit tumor growth, as well as PSA secretion (Vaqar

M Adhami, Ahmad, & Mukhtar, 2003; Vaqar Mustafa Adhami, Siddiqui, Ahmad, Gupta, &

Mukhtar, 2004). However, two small clinical trials which tested green tea in patients with high

grade PIN and advanced prostate cancer reported minimal clinical activity against PCa (Bettuzzi

et al., 2006; Brausi, Rizzi, & Bettuzzi, 2008; Choan et al., 2005). Capsaicin has been shown to

reduce proliferation and induce apoptosis in prostate cells by a mechanism involving reactive

oxygen species (ROS) generation and the dissipation of the mitochondrial membrane potential

(Mori et al., 2006; A. M. Sánchez, Sánchez, Malagarie-Cazenave, Olea, & Díaz-Laviada, 2006),

in addition to its radio-sensitizing properties through the inhibition of NFκB signalling (Venier,

Colquhoun, et al., 2015). Additionally, in vivo studies have demonstrated a significant reduction

in the metastatic burden of mice treated with capsaicin compared to control (Venier, Yamamoto,

et al., 2015). A prospective phase II trial (CAPSAICIN) has been launched to evaluate the

efficacy and safety of capsaicin treatment in men on active surveillance for localized prostate

cancer (ClinicalTrials.gov Identifier: NCT02037464).

1.6 Cannabinoids

Cannabis has been used for medicinal purposes, with its origin dating back more than 5000

years. In 1839, William O’Shaughnessy, a British physician and surgeon discovered the

analgesic, antiemetic, muscle relaxant and anticonvulsant properties of cannabis. These

observations quickly prompted an expanded medical use of cannabis (Ben Amar, 2006). In the

early 1990’s, the cannabinoid receptors and their endogenous ligands were discovered, leading to

a volume of research on the physiology and therapeutic benefit of cannabinoids (Díaz-Laviada,

2011).

Cannabinoids can be classified into three groups based on the source of their production;

phytocannabinoids, endogenous cannabinoids, and synthetic cannabinoids. Cannabinoids exert

their effects by binding to two G-protein coupled receptors: the cannabinoid receptor 1 (CB1),

identified by Devane et al in 1988 (Devane, Dysarz, Johnson, Melvin, & Howlett, 1988), and the

cannabinoid receptor 2 (CB2), discovered by Munro et al in 1993 (Munro, Thomas, & Abu-

Shaar, 1993).

The endogenous cannabinoids, their receptors and the enzymes responsible for their synthesis,

transport and degradation, make up the endocannabinoid system. This system is crucial for

Page 25: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

11

neuromodulation, control of cardiovascular tone, energy metabolism, and immunity, thus making

it a promising target for the management of a variety of diseases (Roberto, Klotz, &

Venkateswaran, 2017).

1.6.1 Phytocannabinoids

Phytocannabinoids (pCBs) are lipid-soluble phytochemicals occurring naturally in the plant,

Cannabis sativa L, and include the main psychoactive constituents, delta-9-tetrahydrocannabinol

(THC) and cannabidiol (CBD). Other cannabinoids present in the plant include delta-8-

tetracannabinol (∆8-THC), cannabinol (CBN), cannabicyclol (CBL), cannabichromene (CBC),

and cannabigerol (CBG). However, these molecules are present in much smaller quantities and

possess no significant psychotropic effects as compared to THC (Ben Amar 2006). THC acts as a

partial agonist at the CB receptors, with most of its psychoactive effects being mediated by

activation of the CB1G-protein coupled receptor. CBD, on the other hand, is not a ligand for the

two CB receptors, but has shown cannabimimetic characteristics attributed to its antioxidant

properties, including inhibiting the degradation of the endogenous cannabinoid anandamide

(ElSohly & Slade, 2005). The therapeutic potential of the rest of the cannabinoids found in C.

Sativa is poorly explored.

The palliative effects of pCBs in the inhibition of nausea and emesis associated with

radiotherapy or chemotherapy, appetite stimulation, mood elation, pain relief, and insomnia in

cancer patients has been recognized for centuries (Pacher et al 2006). Several pCBs have been

reported to bind to and interact with CB receptors at high affinities, appearing as promising

candidates for drug development and cancer therapeutics (Patil, Goyal, Sharma, Patil, & Ojha,

2015). Research has suggested that CBD exerts some of its pharmacological activity through the

inhibition of fatty acid amide hydrolase (FAAH), which subsequently increases the levels of

endogenous cannabinoids. FAAH plays an important role in the endocannabinoid system and the

progression of prostate cancer. Data has shown elevated expression of the FAAH enzyme in

prostate tumor biopsies, corresponding to increasing Gleason grade and poor disease-specific

survival (Endsley et al., 2008).

Sativex, a pharmaceutical product composed of controlled amounts of THC and other plant-

derived cannabinoids, has been approved for its use in controlling nausea in cancer patients

undergoing chemotherapy and is also used as an appetite stimulant and in pain management in

Page 26: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

12

cancer patients (Velasco, Sánchez, & Guzmán, 2012). This drug is currently available as an oral

mucosal spray with a one to one ratio of THC to CBD. Phase I and II clinical trials have recently

been launched for glioblastoma multiforme, whereby Sativex is used in combination with

temozolomide (a standard chemotherapeutic agent in the treatment of brain cancer). In part one

of this study, 6 patients with recurrent glioblastoma multiforme were treated with Sativex adjunct

to dose-intense temozolomide to assess safety of the combination. Part two involved 20 patients

receiving either an individualized dose of Sativex or placebo plus temozolomide. Results of this

two part safety and exploratory study were recently published in the Journal of Clinical

Oncology (Twelves, Short, & Wright Stephen, 2017), and has shown no Grade 3 or 4 toxicities

associated with use of the drug. Phase II results showed a median survival in the placebo group

of 369 days compared to 550 days in the Sativex treatment group, as well as a 1-year survival of

83% in the Sativex group compared to 56% in the placebo group. As the first study to examine

cannabinoids in combination with chemotherapy, this research highlights the lack of potential

side effects or toxicities associated with cannabinoid use, and their interactions with

chemotherapeutic agents, thus paving the way for their use in a variety of cancers.

1.6.2 Endocannabinoids

Endocannabinoids are compounds produced in our body that bind to CB receptors. They act as

neuromodulators, affecting the release of various neurotransmitters in the periphery and play a

vital role in inflammation and fat/energy metabolism (Christie & Vaughan, 2001). Traditional

compounds with cannabinoid activity include the following molecular characteristics; a phenolic

hydroxyl group, a lipophilic side chain and an appropriately oriented carbocyclic ring system

(Razdan, 1986). Several non-classical compounds have been discovered, which contain differing

chemical characteristics, but still bind to the CB receptors. These endocannabinoids are usually a

lipid derived from long chain polyunsaturated fatty acids bound to ethanolamine or glycerol. The

ethanolamine-bound fatty acids include anandamide (AEA), the earliest discovered

endocannabinoid, N-oleoylethanolamide (OEA), and N-palmitoylethanolamide (PEA). While

AEA has a high affinity for the classical cannabinoid receptors, OEA and PEA are structurally

similar, however mainly interact with the transient receptor potential cation channel vanilloid

type 1 (TRPV1) and the nuclear peroxisome proliferator-activated receptor (PPAR) (Alger &

Kim, 2011). These two ethanolamides potentiate the effect of endocannabinoids but bind poorly

to the cannabinoid receptors and are thus referred to as “cannabinoid-like substances”.

Page 27: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

13

Cannabinoids can also bind to glycerol and include 2-arachidonoyl glycerol (2-AG) and 2-

arachidonoyl glycerol ether. These compounds are highly abundant throughout the central

nervous system, the gastrointestinal tract, the spleen, and the pancreas, binding with a high

affinity to the cannabinoid receptors (Hanus et al., 2001).

Several studies have depicted the role of endocannabinoids in cancer through inhibition of cell

proliferation both in vitro and in vivo (Izzo & Camilleri, 2009). In colon cancer, treatment with

endocannabinoids have resulted in the inhibition of colonic inflammation, with this effect

reversed by the deletion of CB receptors (Storr et al., 2008; D. Wang et al., 2008). This

dysregulation in cannabinoid receptors possibly suggests their involvement in the malignant

transformation of the colon. Despite limited clinical use of endocannabinoids due to rapid

metabolism, it remains useful to uncover the dynamics between the endocannabinoid system and

a variety of disease states (Roberto et al 2017).

Two of the best-studied endocannabinoids are AEA and 2-AG. 2-AG is prevalent at relatively

high levels in the nervous system and is stored in intracellular compartments. AEA, on the other

hand, is present at very low levels throughout the body due to high metabolic breakdown rates,

thus is produced on demand rather than stored intracellularly.

Endocannabinoids are typically produced biosynthetically from phospholipids. AEA is produced

through the transfer of arachidonic acid from phosphatidylcholine to the nitrogen atom of

phosphatidylethanolamine by the enzyme N-acyl transferase (NAT), which results in the

formation of N-arachidonoyl-phosphatidylethanolamine (NAPE). NAPE is then converted into

AEA in a one-step hydrolysis reaction (Figure 3). 2-AG is synthesized through the hydrolysis of

phosphatidylinositol-4,5-bisphosphate (PIP2) with arachidonic acid to form diacylglycerol

(DAG). Subsequently, DAG is hydrolyzed to 2-AG by the enzyme diacylglycerol lipase (DAGL)

(Figure 4). The endocannabinoids are metabolized through an enzyme catalyzed hydrolysis

reaction to arachidonic acid, by fatty acid amide hydrolase for AEA, and by monoacylglycerol

lipase (MAGL) for 2-AG.

Endocannabinoids can be enzymatically transformed by other enzymes, such as cyclooxygenase

(COX), lipoxygenase, epoxygenase, or hydroxylases, to generate derivatives including

prostaglandins, prostacyclins, thromboxanes, and leukotriene eicosanoids (Khanapure, Garvey,

Janero, & Letts, 2007). Cytochrome P450 4X1, which is found in the prostate, efficiently

Page 28: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

14

metabolizes anandamide into a single monooxygenated product (Stark, Dostalek, & Guengerich,

2008).

Figure 3: The Biosynthesis of Anandamide. AEA biosynthesis is initiated by the formation of

N-arachidonoyl phosphatidylethanolamine (NAPE), which is formed by the transfer of

arachidonic acid from phosphatidylcholine to phosphatidylethanolamine by N-acyltransferase

(NAT). NAPE is converted to AEA through a hydrolysis reaction catalyzed by NAPE-specific

phospholipase D. Figure was adapted from (Bambang et al., 2010).

Page 29: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

15

Figure 4: The Biosynthesis of 2-Arachidonoylglycerol. 2-AG is produced by the hydrolysis of

phosphatidylinositol 4,5-bisphosphate by phospholipase C (PLC) and subsequent cleavage of the

generated diacylglycerol (DAG) by diacylglycerol lipase. Figure was adapted from (Fonseca,

Costa, Almada, Correia-Da-Silva, & Teixeira, 2013).

Page 30: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

16

Anandamide (cis-5,8,11,14-eicosatetraenoylethanolamide) was the first endogenous ligand of the

CB1 receptor to be discovered and isolated in 1992 (Devane et al., 1992). Its structure is an

ethanolamide with a tetraenic twenty-carbon fatty acid, as depicted in Figure 5. In earlier studies,

it appeared that AEA also exhibited some effects that were not mediated by the cannabinoid

receptors. To date, at least two different G-protein-coupled AEA receptors have been suggested

to exist in the brain and vascular endothelium. However, these receptors have yet to be

characterized (V. Di Marzo, De Petrocellis, Fezza, Ligresti, & Bisogno, 2002). The only

reasonably well characterized, non-cannabinoid site of action for AEA is the transient receptor

potential vanilloid receptor 1, which is a non-selective cation channel gated by capsaicin, protons

and heat.

Figure 5: The Chemical Structure of Anandamide. The structure of anandamide contains an

ethanolamide and a twenty-carbon fatty acid with four double bonds. Structure was obtained

from (Fonseca et al., 2013).

Page 31: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

17

1.6.3 Synthetic Cannabinoids

Synthetic cannabinoids (SC) have been extensively used as research tools to gain insight into the

endogenous cannabinoid system and to assess therapeutic use. In vitro and in vivo studies have

shown that the analgesic, anti-inflammatory and anticancer growth effects of SCs are

approximately two to one hundred times more potent than their phytocannabinoid counterparts

(Castaneto et al., 2014). This is possibly due to their higher binding affinity to the cannabinoid

receptors in comparison to phytocannabinoids. Several studies focus on CP55940, a non-

classical cannabinoid, WIN 55,212-2, an aminoalkylindole, as well as JWH cannabinoids

(synthesized by the John W Huffman research group at Clemson University). This section will

discuss WIN 55,212-2 in more detail below.

CP55940 is an SC that mimics the effects of THC and is currently being used to study the

endocannabinoid system. It was created by Pfizer in 1974 and acts as a full agonist at both the

CB1 and CB2 receptors. Studies have demonstrated the effect of CP55940 on inducing apoptosis

in gastric cancer cells and inducing changes in the cells’ morphology (Ortega et al., 2016).

JWH cannabinoids such as JWH-007, JWH-015, JWH-018, and JWH-030 are from the

naphthoylindole family and act as selective CB receptor agonists (Romero-Sandoval & Eisenach,

2007). Each of these cannabinoids were created to explore the effect of differing binding

affinities for the cannabinoid receptors.

WIN 55,212-2 (WIN) also mimics the effects of THC; however, it has a different chemical

structure and a much higher affinity for the CB2 receptor compared to THC (see Figure 6). This

cannabinoid was synthesized by the Sterling Research Group in New York in the late 1980s

(D’mbra et al., 1992). WIN 55,212-2 is an aminoalkylindole; a member of one of the four

distinct classes of cannabinoid receptor agonists, including fatty acid derivative such as the

endocannabinoids anandamide and 2-AG, the classical cannabinoids such as THC, and the non-

classical cannabinoids such as CP55940. The structure of WIN contains a polar amine, a central

indole ring system, and a lipophilic naphthalene group. Early work with the cannabinoid WIN

55,212-2 showed that this SC produced the typical tetrad effects similar to those elicited by

THC, i.e. hypothermia, hypolocomotion, analgesia, and catalepsy, but does so at lower dosages,

Page 32: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

18

indicating much greater potency (Compton, Gold, Ward, Balster, & Martin, 1992; Fan,

Compton, Ward, Melvin, & Martin, 1994).

To date, there is very little information about the pharmacological properties of synthetic

cannabinoids. Generally, they share similarities in liposolubility, nonpolarity, volatility, and

contain a side chain with a range of four to nine saturated carbons. Unlike phytocannabinoids,

SCs are stable in their active form and can be rapidly absorbed via inhalation.

Figure 6: The Chemical Structure of WIN 55,212-2. The structure of WIN contains a polar

amine, a central indole ring system, and a lipophilic naphthalene group. Structure was obtained

from (Ortega et al., 2015).

Page 33: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

19

1.7 Molecular Targets of Cannabinoids

Many investigators have attempted to elucidate the molecular mechanisms through which

cannabinoids alter tumorigenesis. The following sections will explore proposed mechanisms that

have been brought to light, including those related to endoplasmic reticulum stress, oxidative

stress, Rho GTPase signalling, apoptosis and cell cycle regulation (summarized in Table 1, found

at the end of this section).

1.7.1 Endocannabinoid System

The endocannabinoid system is a biological system composed of cannabinoid receptors, which

are endocannabinoids expressed throughout the central and peripheral nervous systems, and their

endogenous ligands, the endocannabinoids.

The endocannabinoid system is involved in regulating a variety of physiological and cognitive

processes including appetite, energy metabolism, pain and inflammation, mood, memory,

learning, fertility, and in mediating the pharmacological effects of cannabis. The realization of

the complexity of the endocannabinoid system in physiological and pathological conditions has

led to the exploration of its association with conditions such as pain and inflammation,

immunological disorders, neurological diseases, obesity, cardiovascular disorders,

gastrointestinal conditions and cancer (Vincenzo Di Marzo, 2008).

Although the biosynthesis of endocannabinoids by the healthy human prostate tissue has not

been established, several studies have demonstrated the importance of the endocannabinoid

system in prostate function and physiology.

Cannabinoid receptors have been identified in human prostate and in biopsy samples from

patients with benign prostatic hyperplasia (BPH) and prostate cancer. In normal prostate tissue,

the CB1 receptor has been localized in two areas; the parasympathetic afferent nerves and the

acini epithelium. Activation of this receptor results in the inhibition of prostate contraction, as

well as the regulation of prostatic secretory activity (Gratzke et al., 2010; Tokanovic, Malone, &

Ventura, 2007).

The first evidence suggesting a dysregulation of the endocannabinoid system in prostate cancer

was reported in 2005, where it was shown that CB1 expression was higher in cancer-derived cell

Page 34: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

20

lines compared to normal human prostate epithelial cells (Sarfaraz, Afaq, Adhami, & Mukhtar,

2005). Later it was shown that the expression of this receptor was correlated with degree of

malignancy in prostate cancer tissues, with higher expression of CB1 in the most aggressive

samples of PCa as compared to normal prostate tissue (Orellana-Serradell et al., 2015).

A study conducted on a total of 399 human prostate cancer samples demonstrated that the

expression levels of the CB1 receptor were significantly higher in patients with metastases at the

time of diagnosis and those with Gleason scores of 8-10 (Chung et al., 2009). This suggests that

high expression of CB1 in prostate tumor is associated with prostate cancer severity and poor

clinical outcome.

FAAH, the enzyme responsible for the metabolic breakdown of anandamide, is expressed in

normal prostate tissue and is upregulated after puberty (Dhanasekaran et al., 2005). This suggests

that androgens are involved in the regulation of its expression, and that the endocannabinoid

system may be important in the development and growth of the prostate. Additionally, AEA,

PEA, and OEA have all been detected in seminal fluid, where they regulate the fertilizing

potential of human sperm (Schuel et al., 2002).

In the human prostate cancer cell lines PC3, DU145, and LNCaP, endocannabinoids, including

2-AG, as well as several enzymes involved in the synthesis of cannabinoids, including NAPE-

specific phospholipase D (NAPE-PLD) are produced at high concentrations (Endsley et al.,

2007; J. Wang et al., 2008). Additionally, there is also evidence of upregulation of the

cannabinoid receptors CB1 and CB2 (M. G. Sánchez, Ruiz-Llorente, Sánchez, & Díaz-Laviada,

2003). Several receptor agonists, including WIN,55-212-2 and JWH015 have been shown to

inhibit prostate cancer cell growth, induce cell cycle arrest and increase apoptotic rates (Olea-

Herrero, Vara, Malagarie-Cazenave, & Díaz-Laviada, 2009; Sarfaraz et al., 2005). Studies

focusing on the CB2 receptor are of interest, as this receptor limits the psychotropic activity

associated with cannabinoid use and may be more suitable for pharmacological targeting in the

PCa context. The following section will discuss the more commonly known cannabinoid

receptors and briefly describe the non-conventional cannabinoid receptors.

Page 35: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

21

1.7.2 Cannabinoids and Non-Cannabinoid Receptors

The CB1 and CB2 receptors are members of the G protein-coupled receptor (GPCR) family that

were identified almost 30 years ago. These receptors mediate the effects of the primary

psychoactive component of marijuana, THC, as well as the endogenous cannabinoids

anandamide and 2-AG, and numerous synthetic cannabinoids.

The CB1 receptor was initially characterized in rat brains by Devane and colleagues in 1988 and

was later cloned from the human testis. The CB1 receptor is mainly distributed throughout the

central nervous system, particularly the hippocampus, cerebral cortex, cerebellum and basal

ganglia (Galiegue et al., 1995). Expression of this receptor has also been found on peripheral

neurons and in non-neuronal tissues such as the adrenal glands, lungs, testis, ovary, uterus,

prostate and vascular tissues (Liu et al., 2000; Roger G. Pertwee, 1999). The CB1 receptor is

thought to modulate central functions including motor activity, learning and memory,

motivation, emotion, and energy homeostasis (R. G. Pertwee et al., 2010). The CB1 receptor

inhibits adenylyl cyclase and activates mitogen-activated protein kinase (MAPK) by signalling

through Gi/o proteins, although it may also be coupled to Gs proteins or ion channels. This

receptor can also mediate the activation of potassium currents and the inhibition of calcium

currents (Howlett, 2005).

The G-protein coupled cannabinoid receptor CB2 was cloned from rat spleen macrophages. It

was also identified in B cells and natural killer cells, spleen and tonsils, where CB2 modulates

immune cell migration and cytokine release (Galiegue et al 1995). Apart from the immune

system, CB2 receptor is also expressed in the male and female reproductive systems, the

gastrointestinal system, bone, and adipose tissue (Patel, Davison, Pittman, & Sharkey, 2010).

There is also some evidence that the CB2 receptor is expressed on some neurons, both within and

outside of the brain, where it modulates neurotransmitter release (Morgan, Stanford, &

Woodhall, 2009). As with CB1 receptors, CB2 couples to Gi/o proteins to inhibit adenylyl cyclase,

however, does not seem to be coupled to potassium or calcium ion channels. The expression of

the cannabinoid receptors throughout the body can be visualized in Figure 7 below.

Accumulating evidence has demonstrated that the cannabinergic effects of cannabinoids cannot

only be attributed to activation of CB1 and CB2. GPR55 was originally isolated in 1999 as an

Page 36: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

22

orphan GPCR (Sawzdargo et al., 1999), belonging to group of the rhodopsin-like receptors,

and not sharing significant similarities in gene sequence to the CB1 or CB2 receptors, particularly

in the ligand binding site (McPartland, Matias, Di Marzo, & Glass, 2006). Studies have

demonstrated that several cannabinoids, including anandamide, 2-AG, THC, CBD, and the CB1

antagonist AM251, activate GPR55 and cause downstream effects such as ERK1/2

phosphorylation, calcium mobilization, and RhoA activation (Pertwee et al 2010). Additionally,

studies in mice models of colorectal cancer have shown that GPR55 and CB1 play differential

roles in colon carcinogenesis, where GPR55 acts as an oncogene, and CB1 acts as a tumor

suppressor (Hasenoehrl et al., 2017). Due to inconsistency in pharmacological data, the efficacy

of other cannabinoids at this receptor remains unknown. Hence, the classification of GPR55 as a

cannabinoid receptor remains indefinite. Several other orphan receptors including GPR23,

GPR18, GPR120, and GPR84 have also been discovered. However, their role in the

endocannabinoid system remains unknown. Thus, there is a need to test for their responsiveness

on a broad range of cannabinoids in order to help clarify their pharmacological profiles and

physiological roles within the endocannabinoid system.

There is further evidence to demonstrate that CB1 receptor antagonists, rimonabant and

taranabant, including the endocannabinoids AEA and 2-AG, can bind to some types of

adrenergic, dopamine, opioid, adenosine, melatonin, 5-hydroxytryptamine (5-HT), angiotensin,

tachykinin, and prostanoid receptors (Christopoulos & Wilson, 2001; T. M. Fong et al., 2009;

Lane, Beukers, Mulder-Krieger, & IJzerman, 2010) . Their potency at these receptors is

significantly less than the potency at which they bind to the CB1 and CB2 receptors.

Additionally, some of these reactions appear to be allosteric in nature, thus no convincing

evidence can implicate any of these receptors as novel cannabinoid receptors.

The transient receptor potential (TRP) family of cation channels includes six subfamilies:

canonical, vanilloid (TRPV), melastatin (TRPM), polycystin, mucolipin, and ankyrin (TRPA).

Each of these channels contain six-transmembrane domain integral membrane proteins. TRP

channels are involved in the transduction of a range of stimuli, including light, taste, electrical

charge, temperature, mechanical and osmotic stimuli (Venkatachalam & Montell, 2007). Five of

these types of TRP channels have been suggested to interact with cannabinoids: TRPV1,

TRPV2, TRPV4, TRPM8 and TRPA1.

Page 37: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

23

TRPV1 was the first TRP channel to be cloned as a receptor for capsaicin, the component in hot

chili peppers responsible for their pungency (Caterina et al., 1997). This receptor is activated by

stimuli such as temperature, protons, and other natural toxins and colocalizes with the CB1 and

CB2 receptors, suggesting the potential for intracellular crosstalk (Vincenzo Di Marzo &

Cristino, 2008). It has been well established that endocannabinoids, including anandamide, N-

arachidonoyl dopamine, but not 2-AG, act as full agonists to both human and rat TRPV1

channels. Furthermore, phytocannabinoids that do not bind to the cannabinoid receptors and

synthetic CB1 and CB2 ligands, act as full agonists at TRPV1 receptors (Ligresti et al., 2006).

Thus, there is sufficient basis for the classification of TRPV1 as an “ionotropic cannabinoid

receptor”.

Aside from TRPV1, five other TRP channels have been discovered and cloned. TRPV2, -3, and -

4 are all involved in high-temperature sensing and nociception, whereas TRPV5 and -6 are

involved in calcium absorption and reabsorption. There is evidence to show that THC,

cannabinol and CP55940 interact with TRPV2 (Qin et al., 2008), and that anandamide and 2-AG

activate TRPV4 via the formation of cytochrome P450 metabolites of arachidonic acid

(Watanabe et al., 2003). There is insufficient evidence to deduce conclusions about whether or

not TRPV2 and TRPV4 can be considered cannabinoid receptors.

TRPM8 and TRPA1 are both involved in thermosensation, however belong to a different

subfamily than that of the capsaicin (TRPV1) receptor. It has been shown that anandamide, N-

arachidonoyl dopamine, and several nonpsychotropic phytocannabinoids can antagonize the

stimulatory effect of TRPM8 agonists (L. De Petrocellis et al., 2008; Luciano De Petrocellis et

al., 2007). Additionally, TRPA1 has been shown to be activated by phytocannabinoid CB1 and

CB2 agonists, THC, CBN, and the synthetic cannabinoid WIN 55,212-2 (Akopian, Ruparel,

Patwardhan, & Hargreaves, 2008; Jordt et al., 2004). In order to definitively classify TRPA1 and

TRPM8 as cannabinoid receptors, further research is warranted to investigate how these

receptors are able to mediate the pharmacological effects of cannabinoids.

Page 38: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

24

Figure 7: Cannabinoid Receptor Localization. Cannabinoid receptor 1 is primarily located in

the brain, central nervous system, and many other parts of the body. The cannabinoid receptor 2

is found throughout the body on cells associated with the immune system. Figure was adapted

from (Sharma, Murumkar, Kanhed, Giridhar, & Yadav, 2014).

Page 39: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

25

1.7.3 Endoplasmic Reticulum Stress Response

The endoplasmic reticulum (ER) is an organelle responsible for the synthesis, folding

and modification of secreted, membrane-bound and organelle-targeted proteins. In order to

achieve optimum protein folding, several factors are required, including intraluminal calcium

concentrations, ATP availability and an oxidizing environment for disulphide-bond formation. A

range of physiological and pathological conditions such as exposure to anticancer agents,

calcium depletion, and viral infections, may lead to an imbalance between ER protein folding

load and capacity. This causes ER stress, which is an accumulation and aggregation of unfolded

proteins in the ER lumen (Verfaillie, Salazar, Velasco, & Agostinis, 2010). Cells have evolved

strategies to protect against the deleterious effects of ER stress, in which protein translation and

genetic transcription are temporarily halted. This strategy is commonly referred to as the

unfolded protein response (UPR). The UPR is considered a pro-survival response initiated to

reduce the accumulation of unfolded proteins, thereby restoring normal ER functioning

(Schröder & Kaufman, 2005). However, if this transcriptional program fails to re-establish,

persistent ER stress can cause a switch to a pro-apoptotic response.

Recent literature has suggested that cannabinoids exert their anticancer effects through

activation of apoptosis. It is postulated that the production of ceramide may induce ER stress and

initiate apoptosis. The inability to return to ER homeostasis may result in cell death by a

mechanism involving mammalian target of rapamycin (mTOR) pathway inhibition, and

subsequently, autophagy. Studies have demonstrated that activation of the CB2 receptor by the

synthetic cannabinoid JWH-015 induces synthesis of ceramide in PC3 cells, inhibiting the Akt-

mTOR pathway and activating initiation factors involved in autophagy regulation and the ER

stress response (Olea-Herrero et al., 2009). The authors showed that this effect was dependent on

CB2 activation, as combined treatment with CB2 antagonist SR144528 resulted in the prevention

of cell death and a decrease in the synthesis of intracellular ceramide. Additionally, studies have

shown that THC induces ceramide accumulation, activating an ER stress response that promotes

autophagy through tribbles homolog 3 dependent (TRB3-dependent) inhibition of the mTORC1

axis in human and mouse cancer cells (Salazar et al., 2009). An increase in ceramide levels and

ER stress may trigger activation of the caspase cascade leading to apoptosis. The proposed

pathway through which this may occur is depicted below in Figure 8. This idea was

demonstrated in a study where primary cultures of prostate cancer cells treated with the

Page 40: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

26

endocannabinoids 2-AG, AEA and methanandamide (MET) showed increased levels of active

caspase-3, and decreased expression of Bcl-2 and Akt (Orellana-Serradell et al., 2015). This

study suggests that the inhibition in Akt may contribute to the activation of anti-proliferative

pathways, and unlike Olea-Herrero et al., these effects were CB1 dependent, as combination

treatment with the CB1 antagonist SR141716 prevented apoptosis in these cells. Additionally,

studies by Carracedo and colleagues (Carracedo et al., 2006) have shown that THC upregulates

the stress-regulated protein p8, which then mediates its apoptotic effect via the upregulation of

ER stress related genes, including ATF-4, CHOP, and TRB3. This pathway activation is limited

to tumor cells, and does not become activated in nontransformed cells, supporting the notion

previously described by Guzman (Guzman, 2003).

Future studies are warranted to clarify the role of the cannabinoid receptors in the activation of

ER stress related pathways and to elucidate the link between CB receptors and downstream

targets in the ER stress response. Identification of the pathways involved will help to clarify the

molecular events that lead to activation of ER-stress mediated cell death by cannabinoids and

may contribute to the design of novel therapeutic strategies for inhibiting tumor growth and

progression.

Page 41: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

27

Figure 8: Proposed Endoplasmic Reticulum Stress Signalling Pathway. Cannabinoid binding

to cannabinoid receptors results in the accumulation of ceramide and alters the expression of ER

stress related proteins downstream, inducing apoptosis.

Page 42: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

28

1.7.4 Oxidative Stress

The term reactive oxygen species (ROS) is used to describe a number of reactive molecules and

free radicals derived from molecular oxygen. These molecules, produced as byproducts during

the mitochondrial electron transport of aerobic respiration, are generated during every day

metabolic processes in normal cells and play a vital role in cell signalling, including apoptosis,

gene expression, and the activation of cell signalling cascades. They have the potential to cause a

number of deleterious effects. Excessive production of ROS or an inadequate antioxidant defense

system may lead to a phenomenon known as oxidative stress, which has been associated with the

initiation and development of a variety of cancers, including prostate cancer (Khandrika, Kumar,

Koul, Maroni, & Koul, 2009). Oxidative free radicals caused by modulation of androgens,

vitamin D, inflammation, tumor suppressor protein 53 (p53), and antioxidants may initiate

prostate cancer. Specifically, in men with prostate cancer, serum androgens may promote ROS

production and its accumulation in prostate cancer cells. (Minelli, Bellezza, Conte, & Culig,

2009). Supporting evidence has suggested that increasing ROS production in prostate cancer

cells are associated with aggressive phenotype, thus, targeting ROS production might offer a

potential approach in preventing cancer development (Kumar, Koul, Khandrika, Meacham, &

Koul, 2008).

Paradoxically, oxidative stress occurring at the intracellular level can have chemopreventive

effects and thus oxidative stress induction may be used as an anticancer strategy triggering

apoptosis in malignant cells. Various studies have reported that chemopreventive agents work in

some part by generating ROS and disrupting redox homeostasis (Ling, Liebes, Zou, & Perez-

Soler, 2003; Sikka, 2003). Studies have reported that ROS may act as secondary messengers

influencing mitochondrial function, mediating the elevation of intracellular calcium, and thus

activating the caspase cascade. In addition, ROS production may induce pro-apoptotic signals

leading to the release of proteins from the mitochondrial intermembrane space into the cytosol,

thereby promoting apoptosis (Paradies, Petrosillo, Pistolese, & Ruggiero, 2002).

Studies have shown that cannabinoids may induce apoptosis in cancer cells through the

production of ROS (Figure 9). It was reported that intracellular calcium levels were elevated and

ROS production was activated in LNCaP cells after treatment with CBD (De Petrocellis et al

2013). This suggests that both oxidative stress and ER stress are contributing factors in the pro-

Page 43: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

29

apoptotic effect of CBD. These results were also seen in non-AR expressing cells, DU145 and

PC3, indicating that CBD increases oxidative stress and ER stress independent of p53 status or

androgen receptor status. It is speculated that ROS is necessary for the increase in the AMP/ATP

ratio, which mediates the activation of AMPK by cannabinoids, leading to cell death. Studies

have proposed that ROS production by cannabinoids activates a positive feedback loop. Here,

electron transport chain inhibition leads to NADH accumulation and the subsequent inhibition of

oxidative phosphorylation, amplifying the production of ROS (Dando et al., 2013). However, it

is important to note that differing non-THC cannabinoids might produce different effects on the

cell cycle and apoptosis, so an all-encompassing, definitive mechanism may not be possible to

describe.

Cannabinoid-mediated ROS production may also trigger the release of pro-apoptotic proteins

such as cytochrome c, caspase-9, apoptosis inducing factor, and Smac/DIABLO from the inner

mitochondrial membrane space into the cytosol, leading to the activation of apoptosis.

Researchers showed that the endocannabinoid, AEA induced cell death through a pathway

involving mitochondrial uncoupling and cytochrome c release, potentially mediated by oxidative

stress and ROS production through activation of TRP (vanilloid) receptors (Maccarrone,

Lorenzon, Bari, Melino, & Finazzi-Agro, 2000). Similarly, it was reported that CBD exposure to

human glioma cells resulted in an induction of ROS production with a time course preceding

caspase-8 and -9 activations (Massi et al., 2006). This time course suggests that the concomitant

activation of both caspase 8 and 9 is the cause, rather than consequence, of caspase 3 activation,

and that both intrinsic and extrinsic pathways of apoptosis are involved in CBD-related death.

How cannabinoids induce ROS accumulation remains controversial. It has been suggested that

CBD is able to induce apoptosis in a cannabinoid and vanilloid receptor-independent

mechanism, through intercalating into the cell membrane, or by possibly binding to an

unidentified cannabinoid receptor (Roger G. Pertwee, Thomas, Stevenson, Maor, & Mechoulam,

2005). Moreover, due to CBD’s ability to act as a potent modulator of intracellular calcium,

researchers have suggested a role of calcium in driving some aspects of tumor cell death

(Drysdale, Ryan, Pertwee, & Platt, 2006). Another working theory is that CBD signalling may be

mediated by a membrane lipid raft domain, causing apoptosis and triggering a complete caspase

cascade (Sarker & Maruyama, 2003).

Page 44: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

30

Despite increasing evidence associating cannabinoid treatment to increased ROS production and

oxidative stress, the conflicting findings regarding the benefit and/or harm of ROS production

cannot be ignored. Thus, an in-depth analysis of these pathways involving oxidative stress and

ROS production is warranted to develop a deeper understanding of the future use of

cannabinoids as anticancer treatment modalities.

Page 45: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

31

Figure 9: Proposed Oxidative Stress Signalling Pathway. Upon binding to and activating the

cannabinoid receptors, cannabinoids increase ROS production, which induces a decrease in the

mitochondria membrane potential, releasing cytochrome C from the mitochondria into the

cytosol, and results in the activation of the caspase cascade.

Page 46: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

32

1.7.5 Rho GTPase Signalling

Cell migration is an integral process that controls inflammation, wound healing, embryogenesis

and morphogenesis. It is a highly complex process, involving numerous compartments of the

cell, including signalling elements, surface receptors, and the cytoskeleton. Cell migration plays

an essential role in the delivery of protective immune responses to tissues, and aberrant cell

migration is associated with many disease states, including autoimmune syndromes,

developmental defects, chronic inflammation, and cancer invasion and metastasis (KS, 2010;

Wells & Parsons, 2011). Under pathological conditions such as tumor invasion and metastasis,

cells become detached from the primary tumor and enzymatically degrade the extracellular

matrix or basement membrane of tissues to become established in a new location.

A variety of intracellular signalling molecules have been implicated in cell migration and

invasion, including phospholipases, Tyr kinases, lipid kinases, Ser/Thr, and MAPK cascades. Of

these, the protein family most pivotal to the regulation of cell migration and invasion is the Rho

GTPases. The most well studied and highly conserved Rho GTPases include Rho, Rac, and

Cdc42.

Rho family GTPases play key roles in coordinating the cellular responses required for cell

migration. They regulate cell migration through the assembly of actin/myosin filaments, cell

adhesion and spreading, and the establishment of cell polarity (Lambrechts, Van Troys, & Ampe,

2004; Anne J. Ridley, 2015). There exist molecular crosslinks between Rho family proteins and

the actin cytoskeleton, where they act to regulate actin polymerization, depolymerization, and the

activity of actin-associated myosins. Additionally, Rho proteins affect the organization of the

microtubule and intermediate filament networks important for cell migration (A J Ridley, 2001).

Critical downstream components in Rho-GTPase signalling and actin binding proteins have been

linked to metastasis in vivo. In prostate carcinoma cells, activity of RhoA is amplified and

corresponds to an increase in cell migration and invasion. The amplification in RhoA is induced

by the stimulation of multiple G protein coupled receptors for thrombin and thromboxane A2

(Nie et al., 2008; Somlyo et al., 2000).

Page 47: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

33

Stemming from this, other studies have explored the ability of cannabinoid receptor activation to

repress RhoA activity, thereby providing a novel mechanism to diminish migration and invasion

of aggressive prostate carcinoma cells (Figure 10). It was reported that activation of CB1 with

endogenous agonists AEA and 2-AG resulted in the suppression of RhoA activity in PCa cells,

contributing to the suppression of cell migration. This loss of RhoA activity was accompanied by

the loss of actin/myosin microfilaments, reduced cell migration, and decreased cell adhesion

(Nithipatikom et al., 2012). Similarly, studies on highly aggressive breast cancer cells MDA-

MB-231 reported a CB1 mediated inhibition in GTPase activity of RhoA (Laezza, Pisanti,

Malfitano, & Bifulco, 2008; Pillé et al., 2005). These results would suggest that the inhibition of

RhoA by cannabinoids mitigate Rho’s ability to promote invasion by causing a disruption in

RhoA membrane localization, necessary for its interaction with several signalling components.

Several other studies have shown a CB1 dependent inhibition of adenylyl cyclase and protein

kinase A, resulting in a reduction of RhoA activity, and subsequent decreases in prostate and

breast cancer cell invasion (Nithipatikom et al., 2004; Takeda et al., 2012).

A deeper understanding of signalling events that cause CB receptor dependent alterations in Rho

GTPase activity is warranted, despite conclusive evidence regarding CB receptor mediated

reductions in RhoA activity. This knowledge will help to elucidate how RhoA is targeted by

cannabinoid receptor stimulation and whether this pathway is responsible for cannabinoid

induced inhibition of cell migration and invasion.

Page 48: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

34

Figure 10: Proposed RhoA GTPase Signalling Pathway. Cannabinoids binding to the

cannabinoid receptor result in the inhibition of RhoA activity, which causes a loss of actin and

myosin microfilaments, resulting in a reduction in cell migration.

Page 49: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

35

1.7.6 Apoptosis

Apoptosis or programmed cell death is a key regulator of physiological growth and tissue

homeostasis. Most anticancer strategies currently utilized in clinical oncology involve the

activation of apoptosis signal transduction pathways. Thus, understanding the molecular

mechanisms that regulate apoptosis in response to anticancer treatment is crucial to the

development of a more rational approach to drug therapies.

The mechanisms of apoptosis are highly complex and involve an energy-dependent cascade of

molecular events. Research indicates that there are two main apoptotic pathways: the intrinsic

(mitochondrial) pathway, and the extrinsic (death receptor) pathway. Both these pathways

converge onto the same end goal; cleavage of caspase 3, formation of apoptotic bodies, DNA

fragmentation, and lastly, the uptake by phagocytic cells (Elmore, 2007).

The intrinsic mechanism of apoptosis is characterized by a diverse array of non-receptor

mediated stimuli which produce intracellular signals that act directly on targets within the cell.

This mechanism involves mitochondria-initiated events, ROS and excess intracellular calcium.

Stimuli including the absence of certain growth factors or hormones, or the presence of toxins,

free radicals, or viral infections cause changes in the inner mitochondrial membrane, which

results in the loss of the mitochondrial transmembrane potential and the release of cytochrome c

from the intermembrane space into the cytosol (Saelens et al., 2004). This initiates the caspase-

dependent mitochondrial pathway, ultimately leading to apoptosis.

The majority of studies examining the pro-apoptotic effects of cannabinoids on prostate and

other malignant cell lines have reported apoptosis linked to intrinsic mechanisms due to an

increase in ROS. It has been demonstrated that the interaction of cannabinoids with TRPV

receptors causes the activation of overlapping mechanisms, including the mitochondrial

apoptotic pathway, accompanied by an increase in the level of ROS with consequent oxidative

stress. This was shown in a study, where CBD increased the generation of ROS and reduced

mitochondrial membrane potential, released cytochrome c to the cytosol and ultimately led to the

activation of the intrinsic apoptotic pathway in breast cancer cells (Shrivastava, Kuzontkoski,

Groopman, & Prasad, 2011). Similarly, in various experimental tumor models, high ROS levels

induced ER stress (as demonstrated by increases in ER-stress mediators such as p8, CHOP, and

TRIB3), which in turn, led to the activation of mitochondrial intrinsic apoptosis (Malhotra &

Page 50: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

36

Kaufman, 2007). In PC3 prostate cancer cells, the synthetic cannabinoid JWH-015 induced

cytochrome c release into the cytosol, and activated caspase 9, confirming the involvement of

apoptosis, and pointing towards an activation of the intrinsic apoptotic pathway. JWH-015 also

induced an increase in ceramide, leading to ER stress, and activation of autophagy, suggesting

this pathway may also be involved in cannabinoid-induced prostate cell death (Olea-Herrero et

al., 2009).

The extrinsic mechanism of apoptosis is characterized by transmembrane receptor-mediated

interactions. These involve death receptors, which are members of the tumor necrosis factor

(TNF) receptor gene superfamily. To date, the best characterized death receptors include

TNFR1, TRAIL, and DR (Locksley, Killeen, & Lenardo, 2001). These receptors can be activated

upon contact or insult by death signalling molecules, leading to downstream caspase-mediated

apoptosis (Khan, Blanco-Codesido, & Molife, 2014).

In the past decade, only a few studies have reported apoptosis by cannabinoids through extrinsic

mechanisms. In human leukemia cells, THC lead to caspase 8 activation, an event typically

associated with the extrinsic apoptotic pathway (Herrera et al., 2006). In immune cells, it was

demonstrated that treatment with the synthetic cannabinoid JWH-015 resulted in extrinsic

apoptosis activation, as demonstrated by caspase 8 activation (Lombard, Nagarkatti, &

Nagarkatti, 2007). This study suggested a possible cross-talk between the two pathways of

apoptosis, i.e. extrinsic and intrinsic, whereby the caspase 8 inhibitor could almost completely

block JWH-015 induced apoptosis, suggesting that the extrinsic pathway of apoptosis plays a

crucial role in cannabinoid-induced apoptosis. However, the precise death receptor/ligands

involved remains unclear. In hepatocellular carcinoma cells, WIN 55,212-2 treatment sensitized

cells to TRAIL- induced apoptosis, mediated by ER stress proteins p8 and CHOP and Death

Receptor 5 (DR5). The upregulation of TRAIL death receptor DR5 contributed to the

amplification of the caspase cascade, promoting extrinsic apoptosis (Pellerito et al., 2010). To

date, there have been no reports on extrinsic mediated apoptosis in prostate cancer cells.

Page 51: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

37

1.7.7 Cell- Cycle Regulation

Regulation of the cell cycle involves processes that are crucial to the survival of a cell, including

the detection and repair of genetic damage and the prevention of uncontrolled cell division. Loss

of this regulatory cell-cycle control is a hallmark of neoplastic cells.

Common cell cycle abnormalities in PCa involve the RB1 pathway (Figure 11), wherein cyclin

dependent kinase inhibitor, p27 is activated and binds to cyclin D, inhibiting the catalytic activity

of Cdk4. This prevents Cdk4 from adding phosphate residues to the retinoblastoma protein, thus,

preventing pRb from releasing the transcription factor E2F1. In order for progression of a cell

through G1 and S phase, Rb inactivation by phosphorylation is necessary. However, once

phosphorylation of this tumor suppressor gene is inhibited, Rb can directly bind to E2F1 and

actively repress transcription, preventing the progression of cells from G1 into S phase (Harbour

& Dean, 2000).

In addition, several metalloproteinases (MMPs) contain E2F binding sites, thus inhibition of

E2F1 activity may also be linked to the impairment of migration and invasion through the

transcriptional inactivation of MMPs, although this exact mechanism remains unclear and was

not explored in this thesis (Johnson et al., 2012; Z. Wang et al., 2017).

Cannabinoids have been shown to cause alterations in cell cycle distribution or cell cycle arrest

in various cancer cell lines. Anandamide was shown to arrest the proliferation of MDA-MB-231

human breast cancer cells in the S phase of the cell cycle (Laezza, Simona Pisanti, Crescenzi, &

Bifulco, 2006). Additionally, the phytocannabinoid THC inhibited breast cancer cell

proliferation by arresting the progression of cells from the G2 to M phase in a CB2 receptor-

dependent manner (Caffarel, Sarrió, Palacios, Guzmán, & Sánchez, 2006). THC administration

was also shown to elicit G0/G1 cell cycle arrest in glioblastoma cells through suppression of

E2F1 (Galanti et al., 2008). In AGS and MKN-1 human gastric cancer cells, WIN 55,212-2

caused cells to arrest in the G0/G1 phase through the mechanism described in Figure 11 (Park et

al., 2011).

Studies within the field of prostate cancer have demonstrated that WIN 55,212-2 causes a dose-

dependent accumulation of LNCaP human prostate cancer cells in the G0/G1 phase of the cell

cycle through an induction of p27 and a dose-dependent decrease in pRb and E2F1 levels

Page 52: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

38

(Sarfaraz, Afaq, Adhami, Malik, & Mukhtar, 2006). In DU145 cells, treatment with CBD

resulted in the inhibition of G1/S phase transition of the cell cycle, however no changes in G1/S

phase transition were observed in LNCaP cells (Luciano De Petrocellis et al., 2013).

Figure 11: Proposed Cell Cycle Regulation Pathway. Cannabinoids act to inhibit cell cycle

progression in the G0/G1 phase. Upregulation of p27 inhibits Cdk4, thus inhibiting

retinoblastoma phosphorylation and E2F1 activation. Inhibition of E2F1 may also lead to

reductions in migration and invasion through inhibition of MMPs, although the exact mechanism

remains unclear.

Page 53: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

39

Table 1: Effects of cannabinoids on cell viability, migration, and invasion in prostate and

various cancers.

Cell Lines Cannabinoid Anticancer Effect Mechanism of Action References

PC3

prostate

cancer cells

WIN55212-2

Decrease in cell

motility

Activation of CB1 results

in repression of RhoA

activity (suppression of

cell migration)

Nithipaticom

et al 2012

PC3,

DU145,

LNCaP

prostate

cancer cells

JWH-015

MET

Decrease in cell

viability

IN VIVO:

Reduction in tumor

growth

CB2 activation by JWH-

015 inhibits Akt-mTOR

pathway and activates

eIF2α (induction of ER

stress- proapoptotic

effect)

Olea-Herrero

et al 2009

PC3,

DU145

prostate

cancer cells

2AG Inhibition of invasion 2AG activates CB1

receptor, inhibits

adenylyl cyclase and

decreases activity of

PKA (inhibition of

invasion)

Nithipaticom

et al 2004

PC3, and

primary

cultures of

prostate

cancer and

benign

prostatic

AEA, 2-AG,

MET

Decrease in viability

Increase in apoptosis

Activation of CB1

receptor results in

activation of apoptotic

pathway without

modification in cell cycle

or necrosis

Orellana-

Serradell et al

2015

Page 54: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

40

hyperplasia

tissue Endocannabinoids

modulate AKT and ERK

pathways

LNCaP

prostate

cancer cells

WIN, CBD Inhibition of

proliferation

WIN and CBD activate

PARP cleavage and

induce apoptosis

WIN effects are CB

receptor independent and

CBD effects are CB

receptor dependent

Sreevalsan et

al 2011

DU145 and

LNCaP

prostate

cancer cells

CBD Inhibition of viability CBD induces ER stress

and production of ROS

DePetrocellis

et al 2013

LNCaP

prostate

cancer cells

WIN Inhibition of

neuroendocrine

differentiation

Inhibition of PI3K/Akt

pathway results in

activation of mTOR and

inhibition of AMPK

Role of CB2 implicated

but not fully elucidated

Morell et al

2016

LNCaP

prostate

cancer cells

WIN G0/G1 phase cell

cycle arrest

Upregulation of p27,

inactivation of pRb and

E2F1

Safaraz et al

2006

Page 55: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

41

DU145

prostate

cancer cells

CBD G1/S phase cell cycle

arrest

Increased expression of

p27 and p21

DePetrocellis

et al 2013

Primary

cultures of

brain tumor

cells;

cortical

astrocytes

THC Induction of apoptosis

IN VIVO:

Reduction in tumor

growth and increased

apoptosis

Ceramide-dependent

upregulation of stress

protein p8 and several

downstream targets

(ATF-4, CHOP, TRB3)

related with ER stress

proapoptotic pathway

IN VIVO:

Upregulation of p8 in

tumors

Carracedo et

al 2006

Primary

cultures of

brain tumor

cells;

cortical

astrocytes

THC Induction of

autophagy; increased

apoptosis

IN VIVO:

Reduction in tumor

growth by 50%

Upregulation of the p8-

TRB3 pathway leads to

ceramide synthesis and

eIF2 phosphorylation,

promoting autophagy.

Inhibition of the

Akt/mTORC1 axis

IN VIVO:

Increases in TRB3

expression; increases in

LC3 and LC3-II, and

increases in active

caspase 3

Salazar et al

2009

Page 56: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

42

U87 human

glioma

cells; Glial

primary

cultures

CBD Induction of apoptosis Induction of oxidative

stress (through early

production of ROS),

depletion of glutathione,

concomitant activation of

caspase 8 and 9, and the

cleavage of caspase 3

Massi et al

2006

Human

neuroblasto

ma

CHP100

and

lymphoma

U937 cells

AEA Induction of apoptosis Increases in intracellular

calcium, mitochondrial

uncoupling, and

cytochrome c release,

leading to apoptosis

Maccarrone

et al 2000

MDA-MB-

231 human

breast

cancer cells

CBDA, CBD Inhibition of cell

migration

PKA inhibited, which in

turn leads to decreased

levels of phosphorylated

RhoA.

Laezza et al

2005

Human

breast

carcinoma

cell line

MDA-MB-

231

Met-F-AEA Inhibition of cell

migration

Inhibition in the GTPase

activity of RhoA, which

induced a delocalization

of RhoA from cell

membrane to the

cytoplasm, leading to the

disruption of skeleton

actin stress fibers.

RHOA/ROCK signalling

could be involved in the

maintenance of actin

Pille et al

2005

Page 57: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

43

organization and

induction of migration in

the MDA-MB-231 cells.

Human

leukemia

Jurkat cells

THC Induction of apoptosis Mitochondrial intrinsic

pathway activation.

Activation of caspase 8

leading to induction of

apoptosis.

Stimulation of the

extracellular signal-

regulated kinase, c-Jun

N-terminal kinase and

p38 mitogen-activated

protein kinase.

Herrera et al

2006

Human

hepatocellu

lar

carcinoma

cells

HepG2

WIN Induction of apoptosis Early activation of p8

and CHOP cause up-

regulation of TRAIL

receptor DR5, sensitizing

the cells to TRAIL.

Increases in PPAR leads

to down-regulation of

survival factors (pAkt,

Bcl-2, Survivin)

contributing to cell death.

Pellerito et al

2010

C57BL/6

Mice T and

B cells

JWH-015 Induction of apoptosis Activation of both

extrinsic and intrinsic

apoptotic pathways.

Lombard et al

2007

Page 58: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

44

JWH-015 induced a loss

of inner mitochondrial

membrane potential and

lead to activation of

caspase 8, 9, and 3.

MDA-MB-

231 human

breast

cancer cells

CBD Induction of apoptosis

and autophagy

Induction of ER stress,

followed by LC3-II

accumulation.

Generation of ROS.

Inhibition of

AKT/mTOR signalling.

Activation of caspase-8,

the generation and

translocation of t-BID to

the mitochondria, the

release of cytochrome c

and SMAC into the

cytosol, and increased

levels of Fas-L

(mitochondria-mediated

apoptosis).

Shrivastava et

al 2011

MDA-MB-

231 human

breast

cancer cells

AEA Arrest in S phase of

cell cycle

Loss of Cdk2 activity,

upregulation of p21

Laezza et al

2006

Page 59: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

45

Human

breast

cancer cells

THC Blocking progression

in the G2/M phase of

the cell cycle

Downregulation of Cdc2

via CB2 receptor

Caffarel et al

2006

Glioblasto

ma cells

THC G0/G1 cell cycle

arrest

Suppression of E2F1 Galanti et al

2008

AGS and

MKN-1

human

gastric

cancer cells

WIN Cell cycle arrest in

G0/G1 phase

Induction of p27,

decreased Cdk4,

decreased pRb and E2F1

expression

Park et al

2011

Abbreviations: CB: Cannabinoid Receptor; MET: Methanandamide; eIF2: Eukaryotic

Initiation Factor 2 Alpha; mTOR: Mammalian Target of Rapamycin; ER: Endoplasmic

Reticulum; 2AG: 2-arachidonoyl glycerol; PKA: Protein Kinase A; AEA: Anandamide; ERK:

Extracellular Signal-Regulated Kinases; WIN: WIN 55,212-2; CBD: Cannabidiol; PARP: Poly

(ADP-ribose) Polymerase; ROS: Reactive Oxygen Species; AMPK: 5' Adenosine

Monophosphate- Activated Protein Kinase; pRb: phospho- Retinoblastoma Protein; THC:

Tetrahydrocannabinol; ATF-4: Activating Transcription Factor 4; TRB3: Tribbles Homolog 3;

LC3: Light Chain 3; CBDA: Cannabidiolic Acid; Met-F-AEA: 2-Methyl-2'-F-Anandamide;

TRAIL: TNF-Related Apoptosis-Inducing Ligand; DR5: Death Receptor 5; Bcl-2: B Cell

Lymphoma 2; SMAC: Second Mitochondria-Derived Activator of Caspase

Page 60: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

46

1.8 Preclinical Models of Prostate Cancer

Valid experimental models of prostate cancer that accurately reflect disease progression are

crucial in ensuring a proper experimental design and increasing our collective understanding of

the biology of the disease. Recently, more suitable models derived from human specimens have

been developed and are available for use. Although it is unlikely that a single model system

holistically reflects the process of prostate cancer development and progression, the use of these

models permits the study of significant aspects of cancer progression and they are essential tools

for the development of new therapies.

1.8.1 In Vitro Models

The most commonly used in vitro model available in prostate cancer research is cell culture.

Primary and immortalized cell lines are the two main types of cell systems predominately used in

vitro.

Primary cell lines are isolated and cultured from primary tumors and normal prostate tissue and

can be passaged a limited number of times. The use of primary cultures has become more

widespread, as cultures of normal prostate epithelium can be compared to established prostate

cancer cell lines to identify cancer-specific traits (D. M. Peehl, 2005).

Immortalized cell lines, commonly mentioned throughout this thesis, have been continually

passaged over long periods of time and have acquired the ability to proliferate indefinitely. The

first human prostatic tumor epithelial cell lines to become established were the Lymph Node

Carcinoma of the Prostate Cell Line (LNCaP), PC3, and DU145, which remain the most

commonly used PCa cell lines in research (Sampson, Neuwirt, Puhr, Klocker, & Eder, 2013).

While LNCaP cells were derived from a supraclavicular lymph node of a patient whose prostate

cancer was exhibiting androgen independent growth. LNCaP behave like androgen-dependent

cells. PC3 cells were derived from bone metastases of a patient with castration-resistant prostate

cancer, and DU145 cells were derived from a brain metastasis of an untreated prostate cancer.

Out of the three cell lines, LNCaP cells express significant levels of androgen receptor (AR) and

produces PSA, whereas DU145 and PC3 cells are considered androgen receptor negative and do

not produce PSA. For several years, LNCaP cells were the only ones available for the study of

AR signalling in vitro. However, several additional AR positive cell lines have now been

Page 61: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

47

established and are used to investigate the mechanisms underlying castration resistance (Navone

et al., 1997). The table below was adapted from a review by Cunningham and You in 2015 and

provides a summary of general characteristics of common immortalized prostate cancer cell

lines. While these in vitro cell models do not recapitulate all aspects of human prostate cancer,

their use in basic science research increases knowledge about molecular mechanisms underlying

the development and progression of PCa and, with further development, can provide more

biologically relevant platforms for mechanistic studies and drug discovery.

Table 2: General Characteristics of Common Immortalized Human Prostate Cancer Cell

Lines

General Characteristics of Common Immortalized Human Prostate Cancer Cell Lines

LNCaP PC3 DU145

Source Lymph node Vertebral metastasis Brain metastasis

Doubling Time 28-60 hours ~33 hours ~34 hours

PSA Protein Yes No No

AR Status Positive Negative Negative

p53 Wild-type with point

mutation

Null Mutated

Page 62: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

48

1.8.2 In Vivo Models

In order to explore the complex biology of prostate cancer metastasis, pre-clinical in vivo models

are the best available approach, as they can be used to study the biological behavior of tumor

tissue in an environment that cannot be easily mimicked in an in vitro setting. The most widely

used animal models in prostate cancer research include human xenograft mouse models and

transgenic mouse models, each of which will be discussed in the following sections.

1.8.2.1 Xenograft Mouse Models

The human prostate cancer xenograft mouse model functions as an extremely useful alternative

approach for exploring the biology of prostate cancer, including potential interactions between

molecularly and genetically altered tumor cells and their microenvironment (Park et al., 2008;

Park, Kim, McCauley, & Gallick, 2010). Studies have shown that direct comparison of patient

tumor biopsy tissue with xenografts demonstrate a high concordance in gene expression and

similar alterations in the genome when tumors are cultivated in mice (Whiteford et al., 2007).

Additionally, xenograft mouse models are widely used for the investigation of new drugs and

therapeutic strategies. Although the predictive value of xenograft models is rather variable, these

models have been relatively accurate in identifying clinically active agents and effective drug

combinations (Peterson & Houghton, 2004), and have been shown to provide use in predicting

Phase II clinical trial performance of cancer drugs under the right framework (Voskoglou-

Nomikos, Pater, & Seymour, 2003).

Xenograft mouse models are established by inoculating a predetermined number of human tumor

tissues, cell lines or primary cell cultures into immunodeficient mice. Subcutaneous xenograft

mouse models, in which the cells are injected into the flank area, allow tumors to be easily

identified and measured, however lack the ability to metastasize. Orthotopic prostate cancer

xenograft models are developed by injecting human prostate cancer cells directly into the

prostate of the mouse. This model allows for the study of genetic and molecular changes in the

tumor cells and their organ microenvironment, as well as lymph node metastasis (Park et al.,

2008). One drawback of orthotopic models is their failure to lead to spontaneous metastasis to

bone, the most frequent metastatic site in men with prostate cancer. However, orthotopic intra-

tibial implantation of metastatic PCa cells produces homogenous cohorts of tumors in bone, and

allows for the study of these microenvironment interactions (S. J. Kim et al., 2006). In some

Page 63: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

49

situations, tumor xenograft models are established by directly transplanting human tissues into

mice, thus limiting the potential for molecular and epigenetic changes to occur during long

periods of in vitro growth. This approach has shown greater predictability for clinical tumor

responses to various drugs compared to more conventional cell line xenograft models (Garber,

2009; Kerbel, 2003). An emerging model, primarily used to study various aspects of PCa biology

including angiogenesis, the identification of castrate resistant stem-like cells, and the effect of

antiandrogen therapies, is a patient-derived model (PDX) (E. L. S. Fong et al., 2014). PDX

models are established by directly implanting surgically removed tumor tissue from the patient

into an immunocompromised mouse, without in vitro manipulation. However, given the high

cost, rare access to patient tissue, and variable engraftment rates, PDX models are not widely

employed in prostate cancer research, despite their ability to retain more reproducible tumor

features (Siolas & Hannon, 2013; Tentler et al., 2012).

1.8.2.2 TRAMP Mouse Model

The transgenic adenocarcinoma mouse prostate (TRAMP) model was first publicized in 1995 for

its ability to form prostate cancer (Greenberg, 1996) and was later explored for its metastatic

potential (Gingrich et al., 1996). This model was established by transgenic expression of SV40-

Tag early genes, under the prostate-specific rat Probasin promoter. The Probasin promoter

regulates the expression of the both the large and small simian virus (SV40) t antigen, specially

limited to the dorsolateral and ventral lobes of the prostate (Gingrich et al 1996). The large t

antigen inhibits tumour suppressor genes, p53 and Retinoblastoma (Rb), simultaneously, the

small t antigen terminates the function of protein phosphatase 2. Thus, the SV40-Tag oncogene

was selected as a target due to its role in inducing oncogenic progression by binding to and

activating tumor suppressors (Colvin, Weir, Ikin, & Hudson, 2014). TRAMP mice typically

develop epithelial hyperplasia after eight weeks of age, PIN at 18 weeks, and lymph node

metastasis by 28 weeks of age (Rea et al., 2016). The TRAMP model has been used to study the

effect of ADT on the progression of PCa by surgical castration of TRAMP mice, the effects of

several chemopreventive agents (including green tea, retinoic acid, vitamin E, dietary

restriction), and immunotherapy, among others (Zhang, Wang, Zhang, & Lu, 2013). TRAMP

mice have rarely been used to study metastases to distant organs of greater clinical relevance,

including bone, as these types of metastases are quite rate in this model (Hsieh et al., 2007).

Page 64: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

50

However, the TRAMP model provides considerable promise in understanding aspects of PCa

development, and for testing novel therapies.

1.8.2.3 Lady Transgenic Model

The Lady transgenic mouse model was developed in 1998 and, similarly to the TRAMP model,

the Probasin promoter drives SV40 T antigen expression. However, in order to avoid the

problem of variable expression levels of the large and small simian virus (SV40) t antigen in the

TRAMP model, the Lady models target only the large-T antigen (Kasper et al., 1998). In this

model, PIN and high glandular proliferation develop by ten weeks of age, followed by high-

grade epithelial dysplasia and poorly undifferentiated adenocarcinoma by twenty weeks, with

metastasis to the lymph nodes, liver, and lung, reported in several transgenic lines (Masumori et

al., 2001). Although the Lady model is less aggressive than the TRAMP model, they share

several histopathological similarities and have also been used to study progression from initial

androgen-dependent regression to androgen-independent relapse following castration (Kasper et

al., 1998; Klein, 2005). Several studies using the Lady model have explored dietary effects of fat

and antioxidants on PCa progression (Venkateswaran, Fleshner, Sugar, & Klotz, 2004), while

others use this model to assess its interaction with TGF- signalling in promoting metastasis (Tu

et al., 2003). The Lady model more accurately mimics the majority of human PCa due to its slow

growth and mostly epithelial phenotype (Valkenburg & Williams, 2011). This model has helped

to move prostate cancer mouse modeling forward and has allowed researchers to better

understand the progression of the disease.

1.8.2.4 The Phosphatase and tensin homolog deleted on chromosome ten (PTEN) Model

PTEN is frequently lost in a variety of human cancers, including prostate, with deletions

occurring in approximately 23% of HGPIN, 69% of localized PCa, and 86% of metastatic CRPC

(Holcomb et al., 2009; Song, Salmena, & Pandolfi, 2012; Yoshimoto et al., 2006). It is a

significant tumor suppressor demonstrating numerous roles in cell metabolism, polarity, motility,

cancer “stem-ness,” and stromal-epithelial interactions (Cantley & Neel, 1999). Due to these

roles, various transgenic models targeting PTEN have been established. The homozygous

knockout of PTEN is lethal, however, the heterozygous knockout mice slowly develop tumors

and a spectrum of prostate phenotypes (Antonio Di Cristofano, Pesce, Cordon-Cardo, &

Page 65: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

51

Pandolfi, 1998; Podsypanina et al., 1999). Due to the slow progression of PCa in PTEN

knockout models, and the severe health issues they develop, some of these studies combined

PTEN knockout mice with other genes lost in PCa to accelerate disease progression. For

example, p27Kip1 knockout coupled with heterozygous PTEN deletion mice rapidly develop PIN

at 13 weeks and about 25% of mice develop invasive PCa (A. Di Cristofano, De Acetis, Koff,

Cordon-Cardo, & P Pandolfi, 2001).

1.8.2.5 c-MYC Model

c-MYC is a proto-oncogene and its overexpression is correlated with increasing tumor stage in

human prostate cancer samples (Gurel et al., 2008). Thus, targeting Myc over-expression to the

mouse prostate is a valid model of human PCa. The first studies to evaluate Myc overexpression

in the mouse prostate was performed by Ellwood-Yen et al, where both Lo-Myc and Hi-Myc

mice were developed under the control of the PB promoter. Hi-Myc mice progress from PIN at

13 weeks to adenocarcinoma with invasion by 26 weeks, while Lo-Myc mice progress slower

with both PIN and adenocarcinoma developing by about 30 weeks (Ellwood-Yen et al., 2003).

One of the major drawbacks of this model is its inability to progress following castration or its

inability to develop metastasis (Koh et al., 2010). However, due to the early onset of Myc

overexpression during PCa progression, the MYC model is an excellent starting point to assess

additional genetic alterations that drive PCa progression (Gurel et al., 2008).

1.8.2.6 NK3 Homeobox (1NKX3.1) Model

The NKX3.1 transcription factor plays a critical role in urogenital development and function and

is frequently lost in early human PIN and PCa samples (Bhatia-Gaur et al., 1999; Bowen et al.,

2000). Loss of NKX3.1 in mice will cause the development of hyperplasia and some dysplasia,

but its loss alone is not sufficient to induce prostate cancer or HGPIN in mice (Abdulkadir et al.,

2002; Tanaka et al., 2000). Combining NKX3.1 loss with PTEN deletion allows for accelerated

progression to HGPIN and early carcinoma (M. J. Kim et al., 2002).

Page 66: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

Chapter 2 Rationale, Hypothesis, and Aims

Rationale, Hypothesis, and Aims

2.1 Rationale

PCa is the most commonly diagnosed cancer in men and is the second leading cause of cancer-

related death in Canadian men. In 2017, Statistics Canada reported that one in seven men will

develop PCa during their lifetime, and one in twenty-nine will die from the disease. Despite the

wide variety of treatment options available for patients, many of these treatments are associated

with adverse side effects that reduce an individual’s quality of life. Thus, there is a need for

novel treatment options which target cancerous cells as opposed to healthy ones, providing an

advantage over more conventional therapies. Given the anticancer properties that have been

demonstrated for cannabinoids both in vitro and in vivo, it is evident that there is a great need to

demonstrate if cannabinoids have the potential to inhibit varying types of prostate cancer cell

lines and to determine the potential mechanism of action involved. Since cannabinoids may have

psychoactive effects, it is crucial to determine the detailed role of the CB receptors in mediating

anti-cancer versus psychotropic effects. This may allow for approaches that reduce the

psychoactive effects while maintaining anti-cancer benefits. The present research will allow us to

explore the mechanism by which cannabinoids exert their anti-PCa effect and the role of the

cannabinoid receptors in targeting downstream pathways responsible for the inhibition of cell

proliferation, invasion, migration, and induction of apoptosis and cell cycle arrest. A comparison

of the endocannabinoid anandamide to synthetic cannabinoid WIN 55,212-2 will also be

explored, allowing for the discovery of how CB receptor affinity variations lead to alterations in

the anti-cancer properties.

2.2 Hypothesis

It is hypothesized that cannabinoids including Anandamide and WIN 55,212-2 reduce

proliferation, migration and invasion, and induce apoptosis and cell cycle arrest in preclinical

models of prostate cancer through the CB2 receptor mediated pathway.

52

Page 67: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

53

2.3 Aims

The aim of this thesis is to explore the anticancer effect of cannabinoids on different prostate

cancer cell types. To achieve this, the following sub-aims were established.

1. To assess the anticancer properties of AEA and WIN on various prostate cancer cell lines

in vitro.

2. To investigate the mechanism of action of WIN treatment on prostate cancer cells in

vitro.

3. To determine the antitumor effect of WIN in a PC3 xenograft model of prostate cancer.

Page 68: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

Chapter 3 Materials and Methods

Materials and Methods

3.1 Cell Culture

Three human prostate cancer cell lines (PC3, DU145, and LNCaP) were obtained from the

American Type Culture Collection (Rockville, Maryland, USA). DU145 and PC3 cells were

cultured in Dulbecco’s minimal essential medium/F12 (Invitrogen, Burlington, ON, Canada)

with 10% fetal bovine serum (FBS; Gibco, Grand Island, NY, USA), 0.3mg/ml l-glutamine and

100IU/ml penicillin and 100g/ml streptomycin (Invitrogen, Burlington, ON, Canada). LNCaP

cells were cultured in RPMI 1640 medium (Invitrogen, Burlington, ON, Canada) with 10% FBS

supplemented with 0.3mg/ml l-glutamine and 100IU/ml penicillin and 100g/ml streptomycin.

All cells were cultured under sterile conditions at 37C in a 5% CO2 incubator.

3.2 Chemicals

Health Canada approval was obtained for the use of controlled substances for research purposes.

Anandamide was obtained from Bio-Techne (Minneapolis, MN, USA) and was purchased pre-

dissolved in anhydrous ethanol. Stock solutions of 0.1mM were created and stored at -20C.

WIN 55,212-2 and AM630 were obtained from Cayman Chemical (Ann Arbor, MI, USA) and

were dissolved in dimethyl sulfoxide (DMSO; Sigma, USA) to create a stock concentration of

10mM, and stored at -20C. Working solutions of anandamide (5-40M), WIN 55,212-2 (1-

30M), and AM630 (1-10M) were diluted in appropriate medium and prepared fresh daily

prior to treatment. All compounds were prepared and stored with minimal exposure to light to

avoid oxidation. All other chemicals were purchased from Sigma unless otherwise specified.

3.3 MTS Cell Proliferation Assay

Cell proliferation was assessed using the CellTiter 96® AQueous One Solution Cell Proliferation

(MTS) assay (Promega, Madison, WI) as depicted in Figure 12. Cells were plated in 96-well

micro-titre plates at a density of 5x103 (LNCaP) or 4x103 (PC3 and DU145) cells per well and

left to adhere at 37C for 24 hours. After adherence, cells were treated with a range of

54

Page 69: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

55

anandamide concentrations (5, 10, 20, 40M), a range of WIN 55,212-2 concentrations (1, 5, 10,

20, 30M), or a range of AM630 concentrations (1, 5, 10M) for 24, 48, and 72hr time points to

establish dose standardization for each cell line. After evaluating the effect of single agent

treatment on the cell lines, CB2 receptor activation was blocked using CB2 antagonist AM630 in

addition to Anandamide or WIN 55,212-2. The following combinations were selected for

evaluation: 10M WIN 55,212-2 + 5M AM630, 10M WIN 55,212-2 + 10M AM630, 20M

WIN 55,212-2 + 5M AM630, 20M WIN 55,212-2 + 10M AM630, 20M Anandamide +

5M AM630, 20M Anandamide + 10M AM630, 40M Anandamide + 5M AM630, 40M

Anandamide + 10M AM630. Control wells were treated with vehicle alone (DMSO or ethanol

0.01%, respectively). After treatment for 24, 48, and 72hr, 20L of MTS dye was added to each

well and plates were incubated at 37C for 2 hours. The absorbance was recorded at 490nm

using a 96-well plate reader. Each experiment was carried out in triplicate wells and repeated at

least three times.

Figure 12: MTS Cell Proliferation Assay: Cells are plated in a 96-well plate and left to adhere

for 24 hours. Treatment is added into each well and cells are incubated for 24, 48, and 72-hour

time points. At the end of each time point, MTS dye is added to each well and the plate is

incubated for 2 hours at 37°C. Optical density is measured using a 96-well plate reader at an

absorbance of 490nm.

3.4 Wound Healing (Scratch) Assay

Cell migration was assessed in PC3 and DU145 cells using a wound healing assay as depicted in

Figure 13. A total of 5x104 cells were plated per well in a 24-well plate and grown until cells

reached approximately 90% confluence. Mitomycin C was prepared at a concentration of 1mg/L

and added to each well to temporarily inhibit cell proliferation, and cells were incubated for 1hr

at 37C. A vertical scratch was made across each well using a 100l pipette tip, followed by two

Page 70: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

56

washes with phosphate buffered saline (PBS; Invitrogen, Burlington, ON, Canada). Reference

marks for imaging were made on the bottom of each well using a fine-tipped marker.

Anandamide (5, 10, 20, 40M) or WIN 55,212-2 (5, 10, 15, 20M) was then added to each well

and left for 24 hours. Images were captured at 2 different reference points along the wound in

each well at 0hr of treatment to establish a baseline measurement, and at the 24hr time point after

cells were left to migrate. AxioVision SE64 Rel. 4.9.1 software was used to measure the

percentage of wound closure over the 24hr time point.

Figure 13: Wound Healing (Scratch) Assay: Cells are plated in a 24-well plate and left to grow

for approximately 48hrs until 90% confluent. A vertical scratch is made across each well using a

100µL pipette tip. An image is captured of the wound, and cells are treated accordingly. After 24

hours, a second image is captured, and wound closure is quantified.

3.5 Matrigel Invasion Assay

Invasion of PC3 and DU145 cells was assessed using the BD BioCoat™ Matrigel™ Invasion

Chamber 8.0 Micron, obtained from BD Biosciences (Mississauga, ON, Canada). Matrigel was

diluted to a final concentration of 0.125g/L with PBS and added to the upper chamber of each

well, and incubated overnight at 37C. As depicted in Figure 14, 5x104 cells per well were

seeded onto the upper chamber using 6-well plates, and cultured for 24 hours at 37C.

Anandamide (20, 40M) or WIN 55,212-2 (1, 5M) treatment was then added to the bottom

wells, and cells were left to invade for 24hrs at 37C. Following incubation, non-invasive cells

were removed from the upper chamber using a cotton swab. The inserts were fixed in methanol

and stained with a 0.1% crystal violet solution. Cells were counted manually (cells per four

fields) using a microscope, and the number of invading cells were quantified. Each experiment

was carried out in duplicate wells and repeated three times.

Page 71: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

57

Figure 14: Matrigel Invasion Assay: Cells are plated in the upper chamber of a transwell and

treatment is added to the lower chamber. Cells are allowed to invade through Matrigel layer for

24 hours. Non-invasive cells are removed from the upper chamber using a cotton swab, and

invasive cells are fixed in methanol, stained in crystal violet solution, and counted.

3.6 Flow Cytometry

3.6.1 Cell Cycle Distribution

Cell cycle distribution was determined by flow cytometry in LNCaP and DU145 cells treated

with anandamide and in PC3 and DU145 cells treated with WIN 55,212-2. Briefly,

asynchronously growing cells were plated at a density of 1×106 per 10cm petri dish and treated

with anandamide (20, 40M) for 24 hours or WIN 55,212-2 (10, 20M) for 48 hours. Control

plates were treated with vehicle alone (cell culture media or 0.01% ethanol). Cells were pulse

labeled with bromodeoxyuridine (BrdU) for 2 hours prior to harvesting. A no-BrdU control was

included. Following incubation with BrdU, cells were trypsinized, fixed in ice-cold 70% ethanol

and stored at -20C until further analysis. Cells were then washed in PBS buffer with 0.5%

Tween-20 and treated with 2N hydrochloric acid (HCl) for 20 min to denature and expose

labelled DNA. Cells were incubated in the dark on ice for 1 hour with anti-BrdU conjugated

FITC (DAKO, Burlington, ON, Canada). Cells were then washed, centrifuged and resuspended

in 1g/L propidium iodide (PI) and incubated for 30 minutes in the dark on ice. Samples were

filtered through a nylon mesh and cell cycle analysis performed on the FACSCalibur flow

cytometer using Cell Quest Pro software package (Becton-Dickinson, CA, USA). Ten thousand

events were counted for each experiment. Each experiment was carried out in duplicate tubes

and repeated three times.

Page 72: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

58

3.6.2 Apoptosis

The proportion of apoptotic to live cells was assessed using the FITC Annexin V/Dead Cell

Apoptosis Kit (Invitrogen, cat# V13242). PC3, LNCaP and DU145 cells were plated at a density

of 1×106 per 10cm petri dish and treated with anandamide (20, 40M) or WIN 55,212-2 (10,

20M for PC3 and DU145; 20, 30M for LNCaP) for 24 hours. Control plates were treated with

vehicle alone (0.01% ethanol or 0.01% DMSO, respectively). After treatment, cells were

trypsinized, washed with cold PBS and centrifuged at 1200rpm for 5 min. The supernatant was

discarded, and the cells were resuspended in 1ml of cold PBS. The cells were counted with a

Neubauer chamber and diluted in binding buffer to a density of 1x106 cells/ml. Cells were

labelled with 2L fluorescein isothiocyanate (FITC) Annexin V and 1 μL of 100 μg/mL PI and

incubated at room temperature for 15 min. After the incubation period, 400L binding buffer

was added to each cytometry tube, samples were filtered through a nylon mesh and cell analysis

performed with the FACSCalibur flow cytometer using the Cell Quest Pro software package

(Becton Dickinson, San Jose, CA, USA). Each experiment was carried out in duplicate tubes and

repeated three times.

3.7 Western Blot Analysis

Cells were prepared for lysate collection by plating 1x106 cells per 10cm petri dish and allowed

to adhere for 24hrs at 37C. Following incubation, cells were treated with anandamide (20,

40M) and WIN 55,212-2 (10, 20M). Control plates were treated with vehicle alone (0.01%

ethanol or 0.01% DMSO, respectively). After 24hr treatment, cells were lysed using NP-40 lysis

buffer containing protease inhibitors (leupeptin/pepstatin, aprotinin and

phenylmethanesulfonylfluoride), sodium dodecyl sulfate (SDS), deoxycholate and

ethylenediaminetetraacetic acid (EDTA). Protein concentration was quantified using the Pierce

BCA Protein Assay Kit (Thermo Fischer Scientific) prior to loading into 10% SDS gels for

electrophoresis. Antibodies for phospho-retinoblastoma, Cdk4 and p27 were purchased from Cell

Signaling Technology (Beverly, Massachusetts, USA), and antibodies for β-actin were purchased

from Abcam (Cambridge Science Park, UK). ImageJ software (US National Institute of Health,

Bethesda, MA, USA) was used to semi-quantitatively determine protein expression levels,

relative to β-actin. Each experiment was carried out in duplicate wells and repeated three times.

Page 73: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

59

3.8 Xenograft Studies

3.8.1 Animals and Housing

Animal Ethics approval was obtained from the Sunnybrook Research Institute Ethics Board and

all work was conducted in accordance with established institutional guidelines including the Care

and Use of Experimental Animals guidelines of the Canadian Council. Athymic nude (nu/nu) six

to eight-week-old male mice were purchased from Taconic (Charles River Laboratories, USA)

and housed in a laminar airflow cabinet under pathogen-free conditions on a 12-h light/dark

schedule. Mice were provided access to food and water ad libitum. Animals were allowed to

acclimate for at least one week prior to the start of the experiments.

3.8.2 Establishment of Xenografts

PC3 cells were maintained in medium with 10% fetal bovine serum. Using a 7-gauge needle,

1.5 × 106 cells resuspended in 100μL Matrigel solution (BD Biosciences, CA, USA) were

subcutaneously inoculated into the left flank of mice anesthetized with isoflurane (induction with

5% for 5-15sec, maintenance with 1-5%). Mice were monitored daily for tumor growth. When

tumors achieved a volume of 100mm3, mice were randomly assigned to two treatment groups:

control (i.e. DMSO vehicle; n=5), and WIN 55,212-2 (n=5) (Figure 15). Tumor volume was

calculated using the formula: Volume= (Length x Width2) (/6).

Page 74: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

60

Figure 15: Xenograft Establishment and Group Assignment: Mice were subcutaneously

injected with 1.5 million PC3 cells in the flank. Mice were monitored for two weeks, allowing

tumors to reach a volume of 100mm3. Animals were then randomly assigned into two treatment

groups; control (i.e. DMSO vehicle) and 5mg/kg treatment with WIN 55,212-2.

3.8.3 Administration of WIN 55,212-2

WIN 55,212-2 was administered to mice three days per week by intraperitoneal injection at a

final dose of 5mg/kg body weight over the course of three weeks. WIN 55,212-2 in DMSO

(0.01M) was diluted fresh in saline solution prior to administration. Control animals received

only the vehicle. Mice were monitored daily and body weight and tumor measurements were

recorded thrice weekly. Mice with tumors exceeding the maximum permissible diameter of

17mm were euthanized, in accordance with the Canadian Council on Animal Care and Cancer

Endpoint Guidelines. At the experiment termination (21 days), blood was drawn from all mice

by direct cardiac puncture. Serum was separated, aliquoted, and stored at -80C for future

analysis. Tumors were excised, weighed, and processed for histopathologic studies. In addition,

liver samples were obtained and fixed for histological analysis by a pathologist to determine

potential toxicities associated with the study (data not shown). Methods overview is depicted in

Figure 16.

Page 75: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

61

Figure 16: Xenograft Experiment Timeline: Ten athymic nude mice were randomized and left

to acclimatize for one week at the animal facility in Sunnybrook Research Institute (SRI). Mice

were inoculated with PC3 cells and tumors were left to grow for approximately two weeks, until

they reached a volume of 100mm3. Mice were randomized to two groups and treated three times

per week with either WIN 55,212-2 (5mg/kg body weight) or DMSO vehicle control for a period

of 21 days. Tumor volume and body weight was monitored three days per week for the entirety

of the study. At the termination of the experiment, serum and tumors were collected and stored

for further analysis.

Page 76: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

62

3.9 In Vitro Mitogenicity Assay

For measurement of cell growth, the MTS method was employed as previously described. PC3

cells (4x103 cells/well) were plated in 96-well plates. After 24 hours, cells were washed twice

with PBS and treated with serum free media for an additional 24 hours. Following this, serum

free media was removed, and cells were treated for up to 72 hours with 10% animal serum in

DMEM/F12 (100L, filtered through a 0.2m syringe filter) obtained from animals in the

control and treatment groups. Treated cells, containing animal serum, were incubated with the

MTS dye for 2 hours. The ensuing formation of tetrazolium compounds was measured using a

plate reader. All experiments were conducted in duplicate wells and repeated three times per

animal.

3.10 Statistical Analysis

Statistical analysis was completed using Microsoft Excel 2016. All in vitro experiments were

assessed using two-tailed Student’s t-testing. Analysis of the in vivo results were performed

using either Student’s t-testing or repeated measures One-Way Analysis of Variance (ANOVA)

techniques. Statistical analysis was performed using SAS software, version 8 (SAS Institute Inc.,

Cary, NC, USA). Results were considered significant at the 5% level (p<0.05). The data shown

represents the mean standard deviation (SD).

Page 77: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

63

Chapter 4 Results

Results

4.1 Differential growth inhibitory effect of anandamide on prostate cancer cell lines

The MTS cell proliferation assay was used to determine the effect of anandamide treatment on

prostate cancer cell growth. Dose standardization experiments were completed on PC3, DU145,

and LNCaP cells in order to determine optimal doses for further experiments at time points of

24, 48, and 72 hours. Results revealed differences in growth inhibitory effects for each of the cell

lines. In PC3 cells, treatment of anandamide at a concentration of 5-40M did not affect cell

viability during the three time points tested (Figure 17). In the androgen- sensitive cell line,

LNCaP, treatment with 20M and 40M at 24 hours resulted in a 31% and 33% reduction in

proliferation, respectively. At 48 hours, LNCaP cell proliferation was inhibited by 25%, 26%,

and 30% upon treatment with 10M, 20M, and 40M, respectively (Figure 18). Treating

DU145 cells with 40M of anandamide resulted in a significant reduction in proliferation by

34%, 25%, and 16% at 24, 48, and 72 hours, respectively (Figure 19). Subsequent experiments

were conducted using DU145 and LNCaP cells at treatment concentrations of 20-40M

anandamide for 24 hours.

Page 78: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

64

0.00

0.10

0.20

0.30

0.40

0.50

0.60

0.70

0.80

0.90

1.00

Control 5 10 20 40

Op

tica

l Den

sity

(49

0nm

)

Anandamide Treatment Concentrations (µM)

0.00

0.20

0.40

0.60

0.80

1.00

1.20

1.40

1.60

Control 5 10 20 40

Op

tica

l Den

sity

(49

0nm

)

Anandamide Treatment Concentrations (µM)

0.00

0.50

1.00

1.50

2.00

2.50

Control 5 10 20 40

Op

tica

l Den

sity

(49

0nm

)

Anandamide Treatment Concentrations (µM)

A)

B)

C)

Figure 17: Effect of anandamide treatment on proliferation of PC3 cells. PC3 cells were

treated with various concentrations of anandamide for A) 24, B) 48, and C) 72 hours and optical

density at 490nm was recorded.

Page 79: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

65

0.00

0.10

0.20

0.30

0.40

0.50

0.60

0.70

0.80

0.90

1.00

Control 5 10 20 40

Op

tica

l Den

sity

(49

0nm

)

Anandamide Treatment Concentrations (µM)

0.00

0.20

0.40

0.60

0.80

1.00

1.20

1.40

Control 5 10 20 40

Op

tica

l Den

sity

(49

0nm

)

Anandamide Treatment Concentrations (µM)

0.00

0.50

1.00

1.50

2.00

Control 5 10 20 40

Op

tica

l Den

sity

(49

0nm

)

Anandamide Treatment Concentrations (µM)

A)

B)

C)

Figure 18: Effect of anandamide treatment on proliferation of LNCaP cells. LNCaP cells

were treated with various concentrations of anandamide for A) 24, B) 48, and C) 72 hours and

optical density at 490nm was recorded. Differences in optical density relative to control that

reach significance (p<0.05) are denoted with an asterisk (*).

* *

* * *

* *

* * *

Page 80: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

66

0.00

0.50

1.00

1.50

2.00

2.50

Control 5 10 20 40

Op

tica

l Den

sity

(49

0nm

)

Anandamide Treatment Concentrations (µM)

0.00

0.50

1.00

1.50

2.00

2.50

3.00

Control 5 10 20 40

Op

tica

l Den

sity

(49

0nm

)

Anandamide Treatment Concentrations (µM)

0.00

0.50

1.00

1.50

2.00

2.50

3.00

3.50

Control 5 10 20 40

Op

tica

l Den

sity

(49

0nm

)

Anandamide Treatment Concentrations (µM)

A)

B)

C)

Figure 19: Effect of anandamide treatment on proliferation of DU145 cells. DU145 cells

were treated with various concentrations of anandamide for A) 24, B) 48, and C) 72 hours and

optical density at 490nm was recorded. Differences in optical density relative to control that

reach significance (p<0.05) are denoted with an asterisk (*).

*

*

*

*

*

Page 81: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

67

4.2 Treatment with WIN-55,212-2 reduces prostate cancer cell proliferation

Using the MTS cell proliferation assay, the effect of WIN 55,212-2 on prostate cancer growth

was assessed. Results revealed that treatment with WIN 55,212-2 at a concentration of 1-30M

inhibited the growth of PC3, LNCaP, and DU145 cells in a dose-dependent manner. PC3 cells

displayed significant reductions in growth by 50%, 55%, and 64% at 5, 10, and 20M of WIN

55,212-2, respectively, at 24 hours (Figure 20). In DU145 cells, growth was significantly

inhibited by 46%, 51%, and 65% at 5, 10, 20M of WIN 55,212-2, respectively (Figure 22).

LNCaP cells showed a similar trend, however concentrations of 20M and 30M were needed in

order to significantly reduce cell proliferation (Figure 21). Subsequent experiments were

conducted in DU145 and PC3 cells using concentrations of 10-20M WIN 55,212-2, and in

LNCaP cells using concentrations of 20-30M WIN 55,212-2.

Page 82: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

68

0.00

0.20

0.40

0.60

0.80

1.00

1.20

1.40

Control 1 5 10 20

Op

tica

l Den

stiy

(49

0nm

)

WIN 55,212-2 Treatment Concentrations (µM)

0.00

0.20

0.40

0.60

0.80

1.00

1.20

1.40

1.60

1.80

Control 1 5 10 20

Op

tica

l Den

stiy

(49

0nm

)

WIN 55,212-2 Treatment Concentrations (µM)

0.00

0.50

1.00

1.50

2.00

2.50

Control 1 5 10 20

Op

tica

l Den

stiy

(49

0nm

)

WIN 55,212-2 Treatment Concentrations (µM)

A)

B)

C)

Figure 20: Effect of WIN 55,212-2 treatment on proliferation of PC3 cells. PC3 cells were

treated with various concentrations of WIN 55,212-2 for A) 24, B) 48, and C) 72 hours and

optical density at 490nm was recorded. Differences in optical density relative to control that

reach significance (p<0.05) are denoted with an asterisk (*).

* *

*

*

*

*

*

*

*

Page 83: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

69

0.00

0.10

0.20

0.30

0.40

0.50

0.60

0.70

0.80

Control 5 10 20 30

Op

tica

l Den

stiy

(49

0nm

)

WIN 55,212-2 Treatment Concentrations (µM)

0.00

0.20

0.40

0.60

0.80

1.00

1.20

1.40

Control 5 10 20 30

Op

tica

l Den

stiy

(49

0nm

)

WIN 55,212-2 Treatment Concentrations (µM)

0.00

0.20

0.40

0.60

0.80

1.00

1.20

1.40

1.60

1.80

2.00

Control 5 10 20 30

Op

tica

l Den

stiy

(49

0nm

)

WIN 55,212-2 Treatment Concentrations (µM)

A)

B)

C)

Figure 21: Effect of WIN 55,212-2 treatment on proliferation of LNCaP cells. LNCaP cells

were treated with various concentrations of WIN 55,212-2 for A) 24, B) 48, and C) 72 hours and

optical density at 490nm was recorded. The * symbol denotes significance (p<0.05) relative to

control; ** denotes significance (p<0.001) relative to control.

* *

* *

*

*

*

** **

** **

Page 84: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

70

0.00

0.50

1.00

1.50

2.00

2.50

3.00

Control 1 5 10 20

Op

tica

l Den

stiy

(49

0nm

)

WIN 55,212-2 Treatment Concentrations (µM)

0.00

0.50

1.00

1.50

2.00

2.50

3.00

Control 1 5 10 20

Op

tica

l Den

stiy

(49

0nm

)

WIN 55,212-2 Treatment Concentrations (µM)

0.00

0.50

1.00

1.50

2.00

2.50

3.00

3.50

Control 1 5 10 20

Op

tica

l Den

stiy

(49

0nm

)

WIN 55,212-2 Treatment Concentrations (µM)

A)

B)

C)

Figure 22: Effect of WIN 55,212-2 treatment on proliferation of DU145 cells. DU145 cells

were treated with various concentrations of WIN 55,212-2 for A) 24, B) 48, and C) 72 hours and

optical density at 490nm was recorded. The * symbol denotes significance (p<0.05) relative to

control; ** denotes significance (p<0.001) relative to control.

* * **

*

* *

**

*

** **

* * *

*

Page 85: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

71

4.3 Anandamide and WIN 55,212-2 treatment reduces the migration and invasion capacity of prostate cancer cells

The effect of anandamide and/or WIN 55,212-2 treatments on cell migration was assessed using

a wound healing (scratch) assay. After treatment with 20-40M anandamide for 24 hours,

DU145 cells showed a significant decrease in cell migration compared to vehicle control, with

the maximal reduction in migration at 40M (p<0.001) (Figure 23A, B). Treatment with 15M

and 20M WIN 55,212-2 resulted in a significant reduction in migration of PC3 cells by 41%

and 37%, respectively (Figure 25A, B). As for WIN 55,212-2, treatment with 15M and 20M

resulted in a significant reduction in the migration of DU145 cells by 36% and 28%, respectively

(Figure 24A, B).

A Matrigel invasion assay was used to assess the ability of cells to penetrate an extracellular

matrix-like environment. Results from this experiment revealed that anandamide significantly

inhibited the invasion of DU145 cells at 20-40M (p<0.001) (Figure 23C). As for WIN 55,212-2

treatment, as little as 1-5M inhibited PC3 cell invasion by 34% and 39% (Figure 25C), and

DU145 cell invasion by 36% and 41%, respectively (Figure 24C).

Page 86: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

72

0

20

40

60

80

100

120

Control 20 40

% W

ou

nd

Clo

sure

(Rel

ativ

e to

Co

ntr

ol)

Anandamide Treatment Concentrations (µM)

Migration Assay Quantification

0

10

20

30

40

50

60

70

80

90

Control 20 40

% In

vasi

on

Anandamide Treatment Concentrations (µM)

Invasion Assay Quantification

A)

B) C)

Figure 23: Effect of 24hr treatment of anandamide on DU145 cell migration and invasion.

A) Morphological representation of wound healing assay, where DU145 cells were treated with

anandamide (20M and 40M) for 24 hours; B) Quantification of wound healing assay; C)

Quantification of Matrigel invasion assay. The * symbol denotes significance (p<0.05) relative to

control; ** denotes significance (p<0.001) relative to control.

*

** ** **

Page 87: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

73

0

20

40

60

80

100

120

Control 5 10 15 20

% W

ou

nd

Clo

sure

(R

elat

ive

to C

on

tro

l)

WIN 55,212-2 Treatment Concentrations (µM)

Migration Assay Quantification

0

20

40

60

80

100

120

Control 1 5

% In

vasi

on

WIN 55,212-2 Treatment Concentrations (µM)

Invasion Assay Quantification

A)

B) C)

Figure 24: Effect of 24hr treatment of WIN 55,212-2 on DU145 cell migration and invasion.

A) Morphological representation of wound healing assay, where DU145 cells were treated with

WIN 55,212-2 (5-20M) for 24 hours; B) Quantification of wound healing assay; C)

Quantification of Matrigel invasion assay. The * symbol denotes significance (p<0.05) relative to

control; ** denotes significance (p<0.001) relative to control.

* *

* **

Page 88: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

74

0

20

40

60

80

100

120

Control 5 10 15 20

% W

ou

nd

Clo

sure

(R

elat

ive

to C

on

tro

l)

WIN 55,212-2 Treatment Concentrations (µM)

Migration Assay Quantification

0

20

40

60

80

100

120

Control 1 5

% In

vasi

on

WIN 55,212-2 Treatment Concentrations (µM)

Invasion Assay Quantification

A)

B) C)

Figure 25: Effect of 24hr treatment of WIN 55,212-2 on PC3 cell migration and invasion.

A) Morphological representation of wound healing assay, where PC3 cells were treated with

WIN 55,212-2 (5-20M) for 24 hours; B) Quantification of wound healing assay; C)

Quantification of Matrigel invasion assay. The * symbol denotes significance (p<0.05) relative to

control.

* *

* *

Page 89: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

75

4.4 Anandamide treatment does not significantly alter the cell cycle distribution in DU145 and LNCaP cells

In order to determine whether anandamide treatment resulted in alterations in cell cycle

distribution, cells were treated with anandamide (20-40M), collected, fixed, and assessed for

cell cycle alterations at 24 hours. In DU145 and LNCaP cells, treatment with anandamide did not

significantly alter cell cycle distribution. Populations of cells in G1 phase, S phase, and G2/M

phase remained relatively consistent across treatment conditions (Figure 26-27).

Page 90: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

76

0

10

20

30

40

50

60

70

Control 20 40

Pro

po

rtio

n o

f C

ells

(%

)

Anandamide Treatment Concentrations (µM)

G1 S G2

A)

B)

Figure 26: Effect of anandamide treatment on cell cycle distribution in DU145 cells. A) Cell

cycle histogram; B) Quantification. Cells were treated with anandamide, labeled with anti-

bromodeoxyuridine (BrdU) fluorescein isothiocyanate (FITC) and propidium iodide (PI), and

fixed at 24 hours, and then subsequently analyzed by flow cytometry to determine the percentage

of cells in each phase of the cell cycle. Error bars represent the standard deviation (SD). All

experiments were carried out in triplicate.

Page 91: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

77

0

10

20

30

40

50

60

70

Control 20 40

Pro

po

rtio

n o

f C

ells

(%

)

Anandamide Treatment Concentrations (µM)

G1 S G2

A)

B)

Figure 27: Effect of anandamide treatment on cell cycle distribution in LNCaP cells. A)

Cell cycle histogram; B) Quantification. Cells were treated with anandamide, labeled with anti-

bromodeoxyuridine (BrdU) fluorescein isothiocyanate (FITC) and propidium iodide (PI), and

fixed at 24 hours, and then subsequently analyzed by flow cytometry to determine the percentage

of cells in each phase of the cell cycle. Error bars represent the standard deviation (SD). All

experiments were carried out in triplicate.

Page 92: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

78

4.5 WIN 55,212-2 treatment causes cell cycle arrest in DU145 and PC3 cells

In order to determine whether WIN 55,212-2 treatment resulted in alterations in cell cycle

distribution, cells were treated with WIN 55,212-2 (10-20M), collected, fixed, and assessed for

cell cycle alterations at 48 hours. In both DU145 and PC3 cells, treatment with WIN 55,212-2

caused a decrease in the proportion of cells in S phase. In DU145 cells, the percentage of cells in

S phase dose-dependently decreased from approximately 10% in control to 6% and 4% in the

WIN treatment conditions. In addition, the percentage of cells in G1 increased from 46% in

control to 64% and 67% in the 10M and 20M treatment conditions, respectively (Figure 28).

This would indicate cell cycle arrest in G1 phase of the cell cycle. In PC3 cells, the percentage of

cells in S phase dose-dependently decreased from approximately 7% in control to 2% and 0.54%

in the WIN treatment conditions. The percentage of cells in G1 phase increased, and the

percentage of cells in G2 phase dose-dependently increased from 15.2% in control to 17.2% and

22.6% in the 10M and 20M treatment conditions, respectively (Figure 29). This would

indicate an arrest in G2/M phase of the cell cycle.

Page 93: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

79

0

10

20

30

40

50

60

70

80

Control 10µM WIN 20µM WIN

Pro

po

rtio

n o

f C

ells

(%

)

WIN 55,212-2 Treatment Concentrations (µM)

G1 S G2

A)

B)

Figure 28: Effect of WIN 55,212-2 treatment on cell cycle distribution in DU145 cells. A)

Cell cycle histogram; B) Quantification. Cells were treated with WIN, labeled with anti-

bromodeoxyuridine (BrdU) fluorescein isothiocyanate (FITC) and propidium iodide (PI), and

fixed at 24 hours, and then subsequently analyzed by flow cytometry to determine the percentage

of cells in each phase of the cell cycle. Error bars represent the standard deviation (SD). All

experiments were carried out in triplicate. The * symbol represents significance (p<0.05) relative

to control, the ** symbol represents significance (p<0.001) relative to control.

*

**

*

*

*

*

Page 94: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

80

0

10

20

30

40

50

60

70

Control 10 20

Pro

po

rtio

n o

f C

ells

(%

)

WIN 55,212-2 Treatment Concentrations (µM)

G1 S G2

A)

B)

Figure 29: Effect of WIN 55,212-2 treatment on cell cycle distribution in PC3 cells. A) Cell

cycle histogram; B) Quantification. Cells were treated with WIN, labeled with anti-

bromodeoxyuridine (BrdU) fluorescein isothiocyanate (FITC) and propidium iodide (PI), and

fixed at 24 hours, and then subsequently analyzed by flow cytometry to determine the percentage

of cells in each phase of the cell cycle. Error bars represent the standard deviation (SD). All

experiments were carried out in triplicate. The * symbol represents significance (p<0.05) relative

to control.

*

*

*

Page 95: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

81

4.6 Anandamide reduces proliferation and induces apoptosis in LNCaP cells but not in DU145 cells

To analyze the effect of anandamide on prostate cancer cells, and the possibility of a pro-

apoptotic effect, a FITC Annexin V assay was used. As shown in Figure 30, there were no

significant differences in the proportion of apoptotic or live cells in the DU145 cells treated with

anandamide (20-40M) compared to vehicle control. In LNCaP cells, the number of live cells

significantly decreased from 57% to approximately 41% while the number of cells in the

apoptotic state significantly increased from 33% to approximately 49% in the anandamide

treatment conditions compared to untreated control (Figure 31).

Page 96: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

82

0

10

20

30

40

50

60

70

80

Control 20 40

Pro

po

rtio

n o

f C

ells

(%

)

Anandamide Treatment Concentrations (µM)

Live Apoptotic

A)

B)

Figure 30: Effect of anandamide on proportion of live versus apoptotic DU145 cells using

Annexin V flow cytometry. A) Representative flow cytometry images; B) Quantification. Cells

were treated with anandamide for 24 hours, labelled with fluorescein isothiocyanate (FITC) and

propidium iodide (PI), and then subsequently analyzed by flow cytometry to determine the

percentage of live versus apoptotic cells.

Page 97: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

83

0

10

20

30

40

50

60

70

Control 20 40

Pro

po

rtio

n o

f C

ells

(%

)

Anandamide Treatment Concentrations (µM)

Live Apoptotic

A)

B)

Figure 31: Effect of anandamide on proportion of live versus apoptotic LNCaP cells using

Annexin V flow cytometry. A) Representative flow cytometry images; B) Quantification. Cells

were treated with anandamide for 24 hours, labelled with fluorescein isothiocyanate (FITC) and

propidium iodide (PI), and then subsequently analyzed by flow cytometry to determine the

percentage of live versus apoptotic cells. The * symbol represents significance (p<0.05) relative

to control, the ** symbol represents significance (p<0.001) relative to control.

*

*

**

**

Page 98: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

84

4.7 WIN 55,212-2 significantly induces apoptosis in PC3 and DU145 cells but not in LNCaP cells

A FITC Annexin V assay was used to analyze the possible pro-apoptotic effect of WIN 55,212-2

treatment on prostate cancer cells. In DU145 and PC3 cells, the proportion of cells in the

apoptotic state significantly increased in a dose-dependent manner after treatment with 10-20M

WIN 55,212-2, while the number of live cells remained statistically without variation (Figure 32,

33). On the contrary, there were no significant differences in the proportion of live or apoptotic

LNCaP cells upon treatment with 20-30M WIN 55,212-2 compared to vehicle control (Figure

34).

Page 99: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

85

A)

B)

Figure 32: Effect of WIN 55,212-2 on proportion of live versus apoptotic PC3 cells using

Annexin V flow cytometry. A) Representative flow cytometry images; B) Quantification. Cells

were treated with WIN 55,212-2 for 24 hours, labelled with fluorescein isothiocyanate (FITC)

and propidium iodide (PI), and then subsequently analyzed by flow cytometry to determine the

percentage of live versus apoptotic cells. The * symbol represents significance (p<0.05) relative

to control.

0

10

20

30

40

50

60

70

80

90

100

Control 10 20

Pro

po

rtio

n o

f C

ells

(%

)

WIN 55,212-2 Treatment Concentrations (µM)

Live Apoptotic

* *

Page 100: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

86

A)

B)

Figure 33: Effect of WIN 55,212-2 on proportion of live versus apoptotic DU145 cells using

Annexin V flow cytometry. A) Representative flow cytometry images; B) Quantification. Cells

were treated with WIN 55,212-2 for 24 hours, labelled with fluorescein isothiocyanate (FITC)

and propidium iodide (PI), and then subsequently analyzed by flow cytometry to determine the

percentage of live versus apoptotic cells. The * symbol represents significance (p<0.05) relative

to control.

0

10

20

30

40

50

60

70

80

90

Control 10 20

Pro

po

rtio

n o

f C

ells

(%

)

WIN 55,212-2 Treatment Concentrations (µM)

Live Apoptotic

* *

Page 101: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

87

A)

B)

Figure 34: Effect of WIN 55,212-2 on proportion of live versus apoptotic LNCaP cells using

Annexin V flow cytometry. A) Representative flow cytometry images; B) Quantification. Cells

were treated with WIN 55,212-2 for 24 hours, labelled with fluorescein isothiocyanate (FITC)

and propidium iodide (PI), and then subsequently analyzed by flow cytometry to determine the

percentage of live versus apoptotic cells.

0

10

20

30

40

50

60

70

80

Control 20 30

Pro

po

rtio

n o

f C

ells

(%

)

WIN 55,212-2 Treatment Concentrations (µM)

Live Apoptotic

* *

Page 102: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

88

4.8 Cannabinoid receptor 2 antagonist AM630 does not alter prostate cancer cell growth

To determine the role of the cannabinoid 2 receptor in reducing the proliferation of prostate

cancer cells, the cells were pre-treated with AM630, a cannabinoid 2 receptor antagonist, which

acts as an inverse agonist by binding to CB2. The effect of AM630 on cell proliferation was first

assessed using the MTS assay in order to confirm a postulated lack of change in cell viability

upon treatment. As depicted in Figure 35, treatment of AM630 at a concentration of 1-10M had

no effect on the viability of PC3, DU145, and LNCaP cells. This allowed us to move forward

with the use of this compound in combination with anandamide and/or WIN 55,212-2 to

determine their effects after inhibition of CB2.

Page 103: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

89

A)

B)

C)

Figure 35: Effect of Cannabinoid receptor 2 antagonist AM630 on viability of prostate

cancer cell lines. A) PC3 cells; B) DU145 cells; C) LNCaP cells. Cells were treated with various

concentrations of AM630 for 24 hours and optical density at 490nm was recorded. All

experiments were carried out in triplicate.

0.00

0.10

0.20

0.30

0.40

0.50

0.60

0.70

0.80

0.90

Control 1 5 10

Op

tica

l Den

sity

(49

0nm

)

AM630 Treatment Concentrations (µM)

0.00

0.10

0.20

0.30

0.40

0.50

0.60

0.70

Control 1 5 10

Op

tica

l Den

sity

(49

0nm

)

AM630 Treatment Concentrations (µM)

0.00

0.50

1.00

1.50

2.00

2.50

3.00

Control 1 5 10

Op

tica

l Den

sity

(49

0nm

)

AM630 Treatment Concentrations (µM)

Page 104: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

90

4.9 Treatment with CB2 antagonist AM630 abrogates the anti-proliferative effects of anandamide in DU145 and LNCaP cell lines

Using the MTS cell proliferation assay, cell proliferation was assessed for cells treated with

anandamide after the inhibition of the CB2 receptor by AM630. In both DU145 and LNCaP cell

lines, treatment with anandamide and AM630 resulted in an abrogation of the anti-proliferative

effects of anandamide (Figure 36). Treatment with 40M anandamide and 5M AM630 resulted

in a significant increase by more than 200% in the proliferation of DU145 cells compared to

40M anandamide alone. Increasing the concentration of AM630 to 10M resulted in a similar

increase in proliferation by over 200% compared to anandamide alone. In LNCaP cells,

treatment with 20M anandamide and 5M AM630 resulted in a significant increase by

approximately 150% compared to 20M anandamide alone. Similarly, anandamide and 10M

AM630 resulted in an increase by approximately 140% compared to anandamide alone.

Page 105: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

91

A)

B)

Figure 36: Effect of anandamide after treatment with cannabinoid receptor 2 antagonist

AM630 on proliferation of DU145 and LNCaP cell lines. A) DU145 cells; B) LNCaP cells.

Cells were pre-treated with CB2 antagonist AM630 and subsequently treated with anandamide

for 24 hours. Optical density was recorded at 490nm. The symbol * denotes significance

(p<0.05) relative to control, the symbol ** denotes significance (p<0.001) relative to control.

The symbol & denotes significance (p<0.05) relative to anandamide alone, the symbol &&

denotes significance (p<0.001) relative to anandamide alone.

0.00

0.10

0.20

0.30

0.40

0.50

0.60

0.70

0.80

0.90

1.00

Control 20µM AEA 20µM AEA+ 5µM AM630 20µM AEA+ 10µMAM630

Op

tica

l Den

sity

(49

0nm

)

Treatment Concentrations

0.00

0.50

1.00

1.50

2.00

2.50

3.00

3.50

Control 40µM AEA 40µM AEA+ 5µM AM630 40µM AEA+ 10µMAM630

Op

tica

l Den

sity

(49

0nm

)

Treatment Concentrations

*

&

&

*

&& &&

**

&

&

Page 106: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

92

4.10 Treatment with CB2 antagonist AM630 abrogates the anti-proliferative effects of WIN 55,212-2 in prostate cancer cell lines

Cell proliferation was assessed for cells treated with WIN 55,212-2 after inhibition of CB2 using

the MTS cell proliferation assay. In all three cell lines, blockage of the CB2 receptor by CB2

antagonist AM630 resulted in an abrogation of WIN 55,212-2’s anti-proliferative effect.

Treatment with 10M WIN 55-212,2 and 5M AM630 resulted in a significant increase in PC3

cell proliferation by 165% compared to WIN 55,212-2 alone. In DU145 cells, treatment with

10M WIN 55,212-2 and 5M AM630 resulted in an increase in cell proliferation by more than

180%. Following a similar trend, LNCaP cells treated with 30M WIN 55,212-2 and 5M

AM630 resulted in an increase in proliferation by approximately 150% compared to WIN

55,212-2 alone (Figure 37).

Page 107: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

93

A)

B)

C)

Figure 37: Effect of WIN 55,212-2 after treatment with cannabinoid receptor 2 antagonist

AM630 on cell proliferation. A) PC3 cells; B) DU145 cells; C) LNCaP cells. Cells were pre-

treated with CB2 antagonist AM630 and subsequently treated with WIN 55,212-2 for 24 hours.

Optical density was recorded at 490nm. The symbol * denotes significance (p<0.05) relative to

control, the symbol ** denotes significance (p<0.001) relative to control. The symbol & denotes

significance (p<0.05) relative to WIN 55,212-2 alone.

0.00

0.20

0.40

0.60

0.80

1.00

1.20

1.40

Control 10µM WIN 10µM WIN+ 5µMAM630

10µM WIN+10µM AM630

Op

tica

l Den

sity

(49

0nm

)

Treatment Concentrations

0.00

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

Control 30µM WIN 30µM WIN+ 5µMAM630

30µM WIN+ 10µMAM630

Op

tica

l Den

sity

(49

0nm

)

Treatment Concentrations

0.00

0.50

1.00

1.50

2.00

2.50

3.00

Control 10µM WIN 10µM WIN+ 5µMAM630

10µM WIN+ 10µMAM630

Op

tica

l Den

sity

(49

0nm

)

Treatment Concentrations

*

*

*

**

&

&

& &

& &

Page 108: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

94

4.11 WIN 55,212-2 treatment alters expression of pRb, Cdk4, and p27 in PC3 cells

Western blotting was used to investigate changes in the expression of downstream proteins in the

cell cycle pathway, including phosphorylated retinoblastoma protein, Cdk4, and p27 following

treatment with cannabinoid WIN 55,212-2. PC3 cells were treated with 10M and 20M WIN

55,212-2 for a period of 48 hours. Treatment with WIN 55,212-2 resulted in a dose-dependent

decrease in the expression of pRb and Cdk4, and a dose-dependent increase in the expression of

p27 compared to vehicle control (Figure 38). Quantification of the Western blot results are

provided in Figure 38.

Page 109: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

95

0

0.2

0.4

0.6

0.8

1

1.2

Control 10µM WIN 20µM WIN

Rel

ativ

e D

ensi

ty

Densitometric Analysis of pRb

0

0.5

1

1.5

2

2.5

3

3.5

4

Control 10µM WIN 20µM WIN

Rel

ativ

e D

ensi

ty

Densitometric Analysis of p27

0

0.2

0.4

0.6

0.8

1

1.2

Control 10µM WIN 20µM WIN

Rel

ativ

e D

ensi

ty

Densitometric Analysis of Cdk4

A) B)

Figure 38: Effect of WIN 55,212-2 on expression of cell cycle regulator proteins. Western

Blot analyses demonstrate the changes in the expression of A) p27, Cdk4, and phosphorylated

retinoblastoma protein (pRb) in PC3 cells treated with WIN 55,212-2; B) Corresponding

densitometric analysis.

Page 110: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

96

0

50

100

150

200

250

300

350

400

450

17 19 21 24 26 28 31 33 35 38

Tum

or

Vo

lum

e (1

00m

m3)

Days Post Inoculation

Treatment Control

4.12 WIN 55,212-2 treatment reduces tumor growth in a mouse xenograft model

The anti-tumor effect of WIN 55,212-2 was tested in vivo using the PC3 xenograft model. Tumor

size and body weight were measured thrice weekly during the duration of the study. A significant

difference in the overall tumor growth rates between each group was observed over time

(p<0.05). Mice in the WIN 55,212-2 treatment group had significantly smaller tumors and slower

tumor growth rate compared to the control group (Figure 39, 41). Mice had minor changes in

body weight, which leveled off towards the end of the study (Figure 40). WIN 55,212-2 was well

tolerated with no toxicities as assessed histologically (data not shown).

Figure 39: WIN 55,212-2 significantly reduces tumor growth rate. Variation in tumor volume

(mm3) measured over time in 2 groups; Control (vehicle alone) and WIN 55,212-2 (5mg/kg body

weight). WIN 55,212-2 was administered on the days indicated on the horizontal axis. Tumor

growth was monitored over time. Error bars represent standard deviation (SD). The symbol *

denotes significance (p<0.05) relative to control.

* * * * *

Page 111: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

97

Figure 40: Lack of effect of WIN 55,212-2 treatment on animal weight. Average animal body

weight (grams) between control group and WIN 55-212-2 treatment group. Mice body weight

was measured thrice weekly for the duration of the study.

0

5

10

15

20

25

30

35

0 17 19 21 24 26 28 31 33 35 38

Wei

ght

(g)

Days Post Inoculation

Treatment Control

Page 112: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

98

Figure 41: Representative images of mice tumors before and after excision. A, C) Control

group; B, D) WIN 55,212-2 treatment group.

A

B

C

D

Page 113: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

99

4.13 Serum containing WIN 55,212-2 reduces PC3 cell proliferation

A mitogenicity experiment was conducted in which serum collected from control and WIN

55,212-2 treated mice was used to supplement DMEM/F12 media for PC3 cells. Using the MTS

cell proliferation assay, cells exposed to serum obtained from animals treated with WIN 55,212-

2 showed reductions in proliferation at 24 and 48 hours, but not at 72 hours. At 24 hours, cells

cultured in the serum from animals treated with WIN 55,212-2 displayed approximately 40%

reduction in proliferation, and a 55% reduction at 48 hours (p< 0.05) (Figure 42). A slight

increase in proliferation was seen at 72 hours.

Page 114: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

100

0.00

0.20

0.40

0.60

0.80

1.00

1.20

1.40

1.60

Control WINO

pti

cal D

ensi

ty (

490n

m)

Treatment Group

0.00

0.20

0.40

0.60

0.80

1.00

1.20

Control WIN

Op

tica

l Den

sity

(49

0nm

)

Treatment Group

0.00

0.10

0.20

0.30

0.40

0.50

0.60

0.70

0.80

0.90

1.00

Control WIN

Op

tica

l Den

sity

(49

0nm

)

Treatment Group

A)

B)

C)

Figure 42: Effect of WIN 55,212-2 containing serum on proliferation of PC3 cells. A) 24

hours; B) 48 hours; C) 72 hours. Cells were treated with mice serum from either control or WIN

55,212-2 group. Optical density was recorded at 490nm. The symbol * denotes significance

(p<0.05) relative to control.

*

Page 115: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

101

Table 3: Summary of In Vitro Results.

Assay Anandamide WIN 55,212-2 Anandamide +

AM630

WIN 55,212-2 +

AM630

Proliferation

Decrease in

LNCaP and

DU145

Decrease in

LNCaP, DU145,

and PC3

Increase in

LNCaP and

DU145

Increase in

LNCaP, DU145,

and PC3

Wound Healing Decrease in

DU145

Decrease in

DU145, and PC3 - -

Invasion Decrease in

DU145

Decrease in

DU145, and PC3 - -

Cell Cycle Analysis No Changes

Cell cycle arrest

in G1 phase in

DU145 and

G2/M phase in

PC3

- -

Annexin V Flow

Cytometry

LNCaP:

Decrease in

proliferating

cells, increase

in apoptotic

cells

Increase in

apoptotic PC3

and DU145 cells

- -

pRb Expression - Dose-dependent

decrease in PC3 - -

Cdk4 Expression - Dose-dependent

decrease in PC3 - -

Page 116: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

102

p27 Expression - Dose-dependent

increase in PC3

Mitogenicity -

Decrease in

proliferation at

24, 48 hours in

PC3

- -

Table 4: Summary of In Vivo Results

WIN 55,212-2

Animal Body Weight No changes relative to control

Tumor Size Decrease

Pathology No differences between groups (Data not

shown)

Page 117: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

103

Chapter 5 Discussion

Discussion

In addition to the well-known palliative effects of cannabinoids on cancer-associated symptoms,

a large body of evidence suggests these molecules may inhibit the growth of tumor cells in

culture and animal models by modulating key signalling pathways. The first evidence of these

anticancer effects was reported in lung cancer by Munson et al (Munson, Harris, Friedman,

Dewey, & Carchman, 1975) and since then, numerous studies have been carried out,

investigating the anti-tumor effects of cannabinoids in a variety of cancers.

Several cannabinoids, including plant-derived, endogenous, and synthetic cannabinoids, are

known to exert antiproliferative actions in a variety of cancer cells, such as breast, brain, skin,

thyroid and colorectal cancers (Guzmán, Sánchez, & Galve-Roperh, 2002). Additionally,

administration of cannabinoids to nude mice slows tumor growth rate in lung, glioma, thyroid

epithelioma, skin carcinoma and lymphoma xenograft models (Bifulco et al., 2001; Casanova et

al., 2003; McKallip et al., 2002). Despite evidence indicating the anti-cancer effect of

cannabinoids in other tumor types, there is very limited evidence for the use of cannabinoids,

anandamide and WIN 55,212-2 as a therapeutic agent in prostate cancer, and our study was

designed to address this gap in knowledge.

In this thesis, we have provided evidence supporting the hypothesis that cannabinoids,

anandamide and WIN 55,212-2, reduce proliferation, migration and invasion, and induce

apoptosis and cell cycle arrest in preclinical models of prostate cancer possibly through a CB2

receptor mediated pathway.

Initially, we demonstrated that treating various prostate cancer cell lines with anandamide or

WIN 55,212-2 caused a reduction in proliferation. A dose-dependent effect was seen upon

administration of AEA or WIN to prostate cancer cells, however, these effects were not seen in

PC3 cells treated with AEA. Furthermore, analysis by flow cytometry showed a decrease in

proliferation and an increase in apoptosis of LNCaP cells treated with AEA, however no

increases in apoptosis of DU145 cells. This may be due to inherent differences between the two

cells lines. It is possible that the apoptotic effects in LNCaP cells are dependent on AR or p53

Page 118: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

104

status, both of which are nonfunctional in DU145 cells. Our studies revealed an increase in

apoptosis in PC3 and DU145 cells treated with WIN, confirming results obtained through the

MTS cell proliferation assay. We were unable to demonstrate an induction of apoptosis in

LNCaP cells despite reports by Sarfaraz et al. This may be due to differences in experimental

approaches; we conducted our apoptosis studies using Annexin V, however Sarfaraz et al

completed a more detailed study exploring the expression of several apoptotic markers, including

Bax, Bcl-2, caspase 3,9, and PARP. It is possible that the assay used in our study is not sensitive

enough to detect the pro-apoptotic effect of WIN on LNCaP cells. Hence, conducting further

mechanistic studies may support the work published by Sarfaraz et al. Cell cycle analysis

revealed that WIN 55,212-2 caused cells to arrest in G1 and G2/M phase, while the percentage of

cells progressing to S phase was decreased. Studies combining AEA or WIN with the CB2

antagonist AM630 demonstrated a reversal in the cannabinoids’ anti-proliferative effects,

suggesting that alterations in cell proliferation may be occurring through a CB2 dependent

pathway. In PC3 cells, AM630 partially restored proliferation in the presence of WIN, relative to

control. These effects are possibly due to activation of the CB1 receptor, which may be counter-

acting the increase in proliferation observed through inhibition of CB2. Our migration and

invasion studies indicate a decrease in cell migration and invasion after treatment with AEA or

WIN for 24 hours in DU145 and PC3 cells. Mechanistic studies performed on key cell cycle

regulator proteins reveal that WIN may be exerting its effects through upregulation of the cyclin

dependent kinase inhibitor p27, downregulation of the tumor suppressor protein phosphorylated

retinoblastoma, and downregulation of the cell division protein kinase Cdk4.

Based on these in vitro findings, we went on to investigate the effects of WIN in a xenograft

mouse model. We found that animals treated with WIN over a period of three weeks

demonstrated significantly reduced tumor sizes. Moreover, administration was safe and well

tolerated, supporting its role as a safe and effective anti-cancer agent. Mice treated with WIN

demonstrated small, but non-significant reductions in weight loss. We did not observe

differences in food intake between the two groups, however the small, yet insignificant

reductions in body weight may be due to the influence of cannabinoid-induced changes in

metabolic activity. Future studies could explore changes in glycolysis through AMPK activation.

Subsequent mitogenicity studies using serum collected from mice showed significant reductions

in human prostate cancer cell proliferation.

Page 119: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

105

In this thesis, we have focused solely on the cannabinoid receptor 2, mainly due to its ability to

generate limited psychoactive side effects when stimulated. However, for the purpose of

gathering a better understanding of the mechanism through which cannabinoids exert their anti-

cancer effects in prostate cancer, it would be beneficial to explore a variety of the known

cannabinoid receptors discussed in literature, including the cannabinoid receptor 1, GRP55 and

TRPV1. A deeper understanding of role of these receptors could be achieved through the use of

antagonists or genetic silencing. This has been partially completed in prostate cancer, where

silencing of GPR55 was associated with reduced PC3 proliferative rates and was crucial for

anchorage-dependent and anchorage-independent cell growth though a mechanism involving

ERK (Pĩeiro, Maffucci, & Falasca, 2011). In glioblastoma xenograft mice models, silencing the

expression of GPR55 lead to significantly slower tumor growth rates compared to control

siRNA-treated mice (Andradas et al., 2011). With regard to TRPV1, studies have shown a

cannabidiol driven impairment in the invasion of human lung and cervical cancer cells through

activation of the MAPks p38 and p42/44, which was reversed by the TRPV1 antagonist

capsazepine (Ramer, Merkord, Rohde, & Hinz, 2010). These studies shed light on the unclear

aspects of the TRPV1 and GRP55 receptors, including their physio-pathological role and the

signalling pathways they are coupled with, and may help to understand their relevance in human

cancer.

Several studies could be conducted on a variety of promising cannabinoids, including

phytocannabinoids such as THC and CBD, synthetic cannabinoids and the endogenous

cannabinoid 2-AG. Majority of these compounds have shown promise in other areas of cancer;

however, a very limited number have been explored for therapeutic use in PCa. Synthetic

cannabinoids that bind to the CB2 receptor with higher affinity would be of greater efficacy for

its transition to clinical trials, as psychoactive effects would be mitigated or completely avoided.

Several of these cannabinoids alter varying pathways, thus their individual mechanisms of action

offer great knowledge into future pathways to target for the prevention and treatment of PCa. A

few of the pathways commonly discussed in the literature were mentioned in the molecular

targets section of this thesis (see section 1.7). Conducting more detailed mechanistic studies on

key proteins in each of the pathways related to endoplasmic reticulum stress, oxidative stress, as

well as RhoA GTPase, will allow for a more solid foundation into how these compounds are

involved in the anti-proliferative, anti-migratory, anti-invasive, and pro-apoptotic processes

Page 120: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

106

explored in this thesis. Additional experiments, including oxidative stress assays, calcium level

assays, ceramide kinase assays, immunostaining to examine structural changes in cytoskeleton,

in addition to the expression of autophagy-related markers would allow for a more in-depth

analysis of the processes involved in cannabinoid administration.

Targeting the cell cycle pathway is crucial in cancer therapy, as cancer development has been

associated with dysregulation of cell cycle machinery. Cell cycle arrest represents a survival

mechanism that provides the tumor cell with the opportunity to repair its damaged DNA or can

activate the apoptotic cascade, leading to cell death (Schwartz & Shah, 2005). Several

cannabinoids have been shown to target the cell cycle through inducing cell cycle arrest in G1

phase, allowing for a reduction in the percentage of cells in S phase (See Table 1). In this study

we have demonstrated that treatment with WIN 55,212-2 induces cell cycle arrest in PC3 cells

through a mechanism involving p27, Cdk4, and pRb. Our results indicate that WIN may be

regulated via this pathway and may provide a potential target for future therapy in the field of

prostate cancer. One approach to further validating these mechanistic studies are to inhibit the

various aspects of the signalling pathway using commercially available inhibitors. This would

validate my Western blot findings and add further credence to the mechanism of action.

With regard to the in vivo portion of this thesis, the bioavailability of WIN 55,212-2 in mice is

not well documented and very limited clinical studies have been conducted on the metabolism of

WIN in humans. In the first study discussing the metabolism of WIN 55,212-2 in humans, it was

reported that WIN is extensively metabolized in the liver, with a predicted human clearance rate

of 16mL/min/kg, suggesting a fast and nearly complete metabolism in vivo, as well as a short

half-life of the drug (Mardal, Gracia-Lor, Leibnitz, Castiglioni, & Meyer, 2016). In our study,

there was no indication of toxicity after intraperitoneal administration of 5mg/kg of WIN,

demonstrating that WIN is well tolerated.

Serum concentrations and metabolism of WIN were not detailed in this study. Experiments

analyzing the precursors or metabolites of WIN in media or mice serum would help us to better

understand concentrations and time points for optimal treatment. Thus, more in-depth analysis on

the bioavailability and pharmacokinetics of WIN 55,212-2 in vivo is warranted through serum

analysis of xenograft mice models.

Page 121: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

107

In summary, we have revealed that cannabinoids WIN and anandamide reduce the growth,

migration and invasion of prostate cancer cells. Furthermore, WIN induces cell cycle arrest in G1

and G2/M phase and alters expression levels of key proteins in the cell cycle pathway. The in

vivo studies revealed that intraperitoneal administration of 5mg/kg WIN 55,212-2 thrice weekly

in mice is well tolerated and significantly reduces the tumor growth rate, without significant

impact on body weight. Based on these findings, it would be important to conduct further studies

to assess the anti-cancer effects of WIN and anandamide and to determine their potential as a

treatment option for prostate cancer through future clinical trials.

Page 122: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

108

Chapter 6 Future Directions and Overall Conclusion

Future Directions and Overall Conclusion

Throughout the course of my studies, several questions arose. However, due to time constraints,

we were unable to address these questions or conduct the necessary experiments to investigate

them in greater detail. In the following section, some of the emerging areas of interest from this

thesis will be outlined.

6.1 Potential In Vivo Studies

Despite the promising anti-cancer properties of cannabinoids observed in vitro, there is quite a

gap in our understanding of the roles cannabinoids and the endocannabinoid system play in the

development of prostate cancer. To further explore this aspect, the use of transgenic model

systems that very closely resemble human prostate cancer development are necessary. Utilizing a

transgenic model will allow for the ability to commence treatment at different stages of disease

progression, including prior to cancer development, after PIN lesion development, or after the

development of metastasis. This will provide better insight as to optimal time point for maximum

treatment effectiveness and will allow us to determine the effect of cannabinoid treatment on

metastasis.

Another potential extension of this thesis is the completion of in vivo studies using cannabinoid

receptor knockout mice models. The development of CB1 and CB2 knockouts in prostate cancer

mouse models will provide a clearer insight into the role of CB1/2 in prostate cancer. This could

lead to the discovery of potential alterations in tumor microenvironment through changes in

immune cell populations, downstream signalling, or the promotion of inflammation. Thus far,

there are three lines of cannabinoid receptor 1 knockout mice and two lines of cannabinoid

receptor 2 knockout mice (N. E. Buckley, 2008). These knockouts have allowed researchers to

discover the role of the endocannabinoid system in vivo (Vincenzo Di Marzo et al., 2000), how

immunomodulation is affected (Nancy E. Buckley et al., 2000), and the involvement of the CB

receptors in immune cell function and development, autoimmune inflammation, apoptosis,

chemotaxis, and infection (Buckley et al 2008). Thus far, no prostate cancer mouse models

lacking either of these receptors have been developed. In colorectal cancer (CRC), azoxymethane

Page 123: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

109

(AOM)- and dextran sulfate sodium (DSS)-driven CRC mouse models have been developed in

which the CB1 and GPR55 receptors are knocked out. Using these models, Hasenoehrl et al

(2017) were able to demonstrate that GPR55 and CB1 play differential roles in colon

carcinogenesis where the former acts as an oncogene and the latter as a tumor suppressor. These

results help us understand the pathway in which cannabinoid receptor activation results in an

inhibitory effect on carcinogenesis and if translated to a prostate cancer model would provide

convincing evidence as to its relationship to prostate cancer.

6.2 Current and Potential Clinical Trials

Thus far, there are no clinical trials testing the use of cannabinoids as treatment for prostate

cancer. However, a few clinical trials have been launched testing the safety and efficacy of

cannabinoids in the treatment of cancers. The ongoing clinical trials involve the use of

dexanabinol, a synthetic cannabinoid which does not produce cannabis-like psychoactive effects,

and Sativex, an oromucosal spray containing a 1:1 ratio of CBD and THC.

In 2016, Phase I and II clinical trials were completed to assess the tolerability, safety and

pharmacodynamics of Sativex in combination with dose-intense temozolomide in patients with

recurrent glioblastoma. In part one of this study, 6 patients with recurrent glioblastoma

multiforme were treated with Sativex adjunct to dose-intense temozolomide to assess safety of

the combination. Part two involved 20 patients receiving either their individualized dose of

Sativex or placebo plus temozolomide. Results of this two-part safety and exploratory study was

recently published in the Journal of Clinical Oncology (Twelves et al 2017). There were no

Grade 3 or 4 toxicities associated with use of the drug, and patients in the Sativex treatment

group had a higher median survival and higher one-year survival rate compared to the

chemotherapy group.

In another clinical trial completed in 2016, varying doses of dexanabinol were used on patients

with solid tumors in order to determine the maximum safe dose and to further understand the

safety of the drug and measure any reductions in tumor size. In this Phase I clinical trial, 40

patients were assigned to 9 treatment arms where dosages of dexanabinol ranged from 2mg/kg to

36mg/kg. Results indicate the drug is well tolerated up to 22mg/kg, with minimal adverse events

reported. The patient’s progression free survival was increased to a maximum of 293 days in the

Page 124: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

110

group treated with 22mg/kg dexanabinol, and tumor development was delayed by the treatment

(ClinicalTrials.gov Identifier: NCT01489826).

Both of these studies have shown that cannabinoid treatment offers some efficacy in cancer

patients and confirms the safety and feasibility of individualized dosing. They set the basis for

future trials aimed at evaluating the antitumoral activity of cannabinoids.

Within the field of prostate cancer, little information is available on the pharmacokinetics,

metabolism and route of administration of cannabinoids both in animals and human, making the

transition towards clinical studies challenging. Future studies should explore the

pharmacokinetics of drug administration and investigate optimal drug dosage by administering

cannabinoids to patients through different routes and/or at a variety of dosages.

Additionally, one simple study that could be done to provide a proof of principle for a potential

pathway of action is to give patients the drug over a short period of time before surgery and look

for apoptosis or any changes in the expression of key proteins in the pathways of interest (i.e.

pRb, p27, Cdk4). This would confirm our in vitro findings and help us to determine whether or

not this proposed pathway is targeted by cannabinoids in humans, allowing for the potential to

examine this pathway as a target for future therapy of prostate cancer.

6.3 Overall Conclusion

In conclusion, this work provides novel evidence for the use of cannabinoids anandamide and

WIN 55,212-2 as a therapeutic for the treatment of prostate cancer. We initially reported that

treating various prostate cancer cells with anandamide and WIN 55,212-2 causes a reduction in

proliferation, migration, and invasion, and an increase in apoptosis. Cell cycle analysis revealed

that WIN 55,212-2 caused alterations in cell cycle distribution, whereby the proportion of cells in

G1 phase increased and the proportion of cells in S phase decreased. Mechanistic studies

revealed that WIN 55,212-2 exerts its anti-cancer effects through upregulation of the cell cycle

inhibitor protein, p27, downregulation of the tumor suppressor protein, phosphorylated

retinoblastoma protein, and downregulation of the cyclin dependent kinase protein, Cdk4.

Furthermore, blocking the activation of the CB2 receptor caused cell proliferation to increase,

suggesting anandamide and WIN 55,212-2’s anti-proliferative effects may be occurring through

a CB2-dependent manner.

Page 125: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

111

Based on our in vitro findings, we went on to investigate the effects of WIN 55,212-2 in a

xenograft model of prostate cancer. We found that administration of WIN 55,212-2 was well

tolerated by animals and tumor growth rate was significantly reduced. Mitogenicity studies

revealed decreases in cell growth following treatment with cannabinoid-containing mouse serum.

We strongly believe that the novel work presented in this thesis will further the understanding of

the therapeutic benefits of cannabinoids in prostate cancer. This information will provide a

framework for future studies and clinical trials that will help us to better understand the potential

benefit of cannabinoid use in prostate cancer and improve the way in which this disease is

managed and treated.

Page 126: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

112

References

Abdulkadir, S. A., Magee, J. A., Peters, T. J., Kaleem, Z., Naughton, C. K., Humphrey, P. A., &

Milbrandt, J. (2002). Conditional loss of Nkx3.1 in adult mice induces prostatic

intraepithelial neoplasia. Molecular and Cellular Biology, 22(5), 1495–503.

https://doi.org/10.1128/MCB.22.5.1495

Abel, E. L., & DiGiovanni, J. (2011). Multistage carcinogenesis. Current Cancer Research, 6,

27–51. https://doi.org/10.1007/978-1-61737-995-6_2

Adhami, V. M., Ahmad, N., & Mukhtar, H. (2003). Molecular targets for green tea in prostate

cancer prevention. The Journal of Nutrition, 133(7 Suppl), 2417S–2424S.

Adhami, V. M., Siddiqui, I. A., Ahmad, N., Gupta, S., & Mukhtar, H. (2004). Oral consumption

of green tea polyphenols inhibits insulin-like growth factor-I-induced signaling in an

autochthonous mouse model of prostate cancer. Cancer Research, 64(23), 8715–8722.

https://doi.org/10.1158/0008-5472.CAN-04-2840

Akopian, A. N., Ruparel, N. B., Patwardhan, A., & Hargreaves, K. M. (2008). Cannabinoids

Desensitize Capsaicin and Mustard Oil Responses in Sensory Neurons via TRPA1

Activation. Journal of Neuroscience, 28(5), 1064–1075.

https://doi.org/10.1523/JNEUROSCI.1565-06.2008

Alger, B. E., & Kim, J. (2011). Supply and demand for endocannabinoids. Trends in

Neurosciences. https://doi.org/10.1016/j.tins.2011.03.003

Andradas, C., Caffarel, M. M., Pérez-Gómez, E., Salazar, M., Lorente, M., Velasco, G., …

Sánchez, C. (2011). The orphan G protein-coupled receptor GPR55 promotes cancer cell

proliferation via ERK. Oncogene, 30(2), 245–252. https://doi.org/10.1038/onc.2010.402

Bambang, K. N., Karasu, T., Gebeh, A., Taylor, A. H., Marczylo, T. H., Lam, P., … Konje, J. C.

(2010). From Fertilisation to Implantation in Mammalian Pregnancy—Modulation of Early

Human Reproduction by the Endocannabinoid System. Pharmaceuticals, 3(9), 2910–2929.

https://doi.org/10.3390/ph3092910

Barber, N. J., Zhang, X., Zhu, G., Pramanik, R., Barber, J. A., Martin, F. L., … Muir, G. H.

(2006). Lycopene inhibits DNA synthesis in primary prostate epithelial cells in vitro and its

administration is associated with a reduced prostate-specific antigen velocity in a phase II

clinical study. Prostate Cancer and Prostatic Diseases, 9(4), 407–413.

https://doi.org/10.1038/sj.pcan.4500895

Ben Amar, M. (2006). Cannabinoids in medicine: A review of their therapeutic potential.

Journal of Ethnopharmacology. https://doi.org/10.1016/j.jep.2006.02.001

Bettuzzi, S., Brausi, M., Rizzi, F., Castagnetti, G., Peracchia, G., & Corti, A. (2006).

Chemoprevention of human prostate cancer by oral administration of green tea catechins in

volunteers with high-grade prostate intraepithelial neoplasia: A preliminary report from a

one-year proof-of-principle study. Cancer Research, 66(2), 1234–1240.

https://doi.org/10.1158/0008-5472.CAN-05-1145

Page 127: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

113

Bhatia-Gaur, R., Donjacour, A. A., Sciavolino, P. J., Kim, M., Desai, N., Young, P., … Shen, M.

M. (1999). Roles for Nkx3.1 in prostate development and cancer. Genes and Development,

13(8), 966–977. https://doi.org/10.1101/gad.13.8.966

Bifulco, M., Laezza, C., Portella, G., Vitale, M., Orlando, P., De Petrocellis, L., & Di Marzo, V.

(2001). Control by the endogenous cannabinoid system of ras oncogene-dependent tumor

growth. The FASEB Journal : Official Publication of the Federation of American Societies

for Experimental Biology, 15(14), 2745–2747. https://doi.org/10.1096/fj.01-0320fje

Bill-Axelson, A., Holmberg, L., Ruutu, M., Garmo, H., Stark, J. R., Busch, C., … Johansson, J.-

E. (2011). Radical prostatectomy versus watchful waiting in early prostate cancer. The New

England Journal of Medicine, 364(18), 1708–17. https://doi.org/10.1056/NEJMoa1011967

Bill-Axelson, A., Holmberg, L., Ruutu, M., Häggman, M., Andersson, S.-O., Bratell, S., …

Scandinavian Prostate Cancer Group Study No. 4. (2005). Radical prostatectomy versus

watchful waiting in early prostate cancer. The New England Journal of Medicine, 352(19),

1977–84. https://doi.org/10.1056/NEJMoa043739

Bono, A. V., Pagano, F., Montironi, R., Zattoni, F., Manganelli, A., Selvaggi, F. P., … Prayer-

Galletti, T. (2001). Effect of complete androgen blockade on pathologic stage and resection

margin status of prostate cancer: Progress pathology report of the Italian PROSIT study.

Urology, 57(1), 117–121. https://doi.org/10.1016/S0090-4295(00)00866-9

Bono, A. V. (2004). Overview of Current Treatment Strategies in Prostate Cancer. European

Urology, Supplements, 3(1), 2–7.

https://doi.org/http://dx.doi.org/10.1016/j.eursup.2003.12.002

Bowen, C., Bubendorf, L., Voeller, H. J., Slack, R., Willi, N., Sauter, G., … Gelmann, E. P.

(2000). Loss of NKX3.1 expression in human prostate cancers correlates with tumor

progression. Cancer Research, 60(21), 6111–6115. https://doi.org/10.1158/0008-5472.can-

06-0963

Brausi, M., Rizzi, F., & Bettuzzi, S. (2008). Chemoprevention of Human Prostate Cancer by

Green Tea Catechins: Two Years Later. A Follow-up Update. European Urology.

https://doi.org/10.1016/j.eururo.2008.03.100

Buckley, N. E. (2008). The peripheral cannabinoid receptor knockout mice: An update. British

Journal of Pharmacology. https://doi.org/10.1038/sj.bjp.0707527

Buckley, N. E., McCoy, K. L., Mezey, É., Bonner, T., Zimmer, A., Felder, C. C., … Zimmer, A.

(2000). Immunomodulation by cannabinoids is absent in mice deficient for the cannabinoid

CB2receptor. European Journal of Pharmacology, 396(2–3), 141–149.

https://doi.org/10.1016/S0014-2999(00)00211-9

Caffarel, M. M., Sarrió, D., Palacios, J., Guzmán, M., & Sánchez, C. (2006). Delta9-

tetrahydrocannabinol inhibits cell cycle progression in human breast cancer cells through

Cdc2 regulation. Cancer Research, 66(13), 6615–6621. https://doi.org/10.1158/0008-

5472.CAN-05-4566

Page 128: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

114

Cantley, L. C., & Neel, B. G. (1999). New insights into tumor suppression: PTEN suppresses

tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway. Proceedings

of the National Academy of Sciences, 96(8), 4240–4245.

https://doi.org/10.1073/pnas.96.8.4240

Carracedo, A., Lorente, M., Egia, A., Blázquez, C., García, S., Giroux, V., … Velasco, G.

(2006). The stress-regulated protein p8 mediates cannabinoid-induced apoptosis of tumor

cells. Cancer Cell, 9(4), 301–312. https://doi.org/10.1016/j.ccr.2006.03.005

Casanova, M. L., Blázquez, C., Martínez-Palacio, J., Villanueva, C., Fernández-Aceñero, M. J.,

Huffman, J. W., … Guzmán, M. (2003). Inhibition of skin tumor growth and angiogenesis

in vivo by activation of cannabinoid receptors. The Journal of Clinical Investigation,

111(1), 43–50. https://doi.org/10.1172/JCI16116

Castaneto, M. S., Gorelick, D. A., Desrosiers, N. A., Hartman, R. L., Pirard, S., & Huestis, M. A.

(2014). Synthetic cannabinoids: Epidemiology, pharmacodynamics, and clinical

implications. Drug and Alcohol Dependence.

https://doi.org/10.1016/j.drugalcdep.2014.08.005

Caterina, M. J., Schumacher, M. A., Tominaga, M., Rosen, T. A., Levine, J. D., & Julius, D.

(1997). The capsaicin receptor: A heat-activated ion channel in the pain pathway. Nature,

389(6653), 816–824. https://doi.org/10.1038/39807

Choan, E., Segal, R., Jonker, D., Malone, S., Reaume, N., Eapen, L., & Gallant, V. (2005). A

prospective clinical trial of green tea for hormone refractory prostate cancer: an evaluation

of the complementary/alternative therapy approach. Urologic Oncology, 23(2), 108–13.

https://doi.org/10.1016/j.urolonc.2004.10.008

Christie, M. J., & Vaughan, C. W. (2001). Neurobiology Cannabinoids act backwards. Nature,

410(6828), 527–530. https://doi.org/10.1038/35069167

Christopoulos, A., & Wilson, K. (2001). Interaction of anandamide with the M 1 and M 4

muscarinic acetylcholine receptors. Brain Research, 915(1), 70–78.

https://doi.org/10.1016/S0006-8993(01)02825-6

Chung, S. C., Hammarsten, P., Josefsson, A., Stattin, P., Granfors, T., Egevad, L., … Fowler, C.

J. (2009). A high cannabinoid CB1 receptor immunoreactivity is associated with disease

severity and outcome in prostate cancer. European Journal of Cancer, 45(1), 174–182.

https://doi.org/10.1016/j.ejca.2008.10.010

Clinical, N., Clinical, N., Guidelines, P., & Guidelines, P. (2009). NCCN Clinical Practice

Guidelines in OncologyTM. Cancer.

Colli, J. L., & Colli, A. (2006). International comparisons of prostate cancer mortality rates with

dietary practices and sunlight levels. Urologic Oncology: Seminars and Original

Investigations, 24(3), 184–194. https://doi.org/10.1016/j.urolonc.2005.05.023

Colvin, E. K., Weir, C., Ikin, R. J., & Hudson, A. L. (2014). SV40 TAg mouse models of cancer.

Seminars in Cell and Developmental Biology. https://doi.org/10.1016/j.semcdb.2014.02.004

Page 129: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

115

Compton, D. R., Gold, L. H., Ward, S. J., Balster, R. L., & Martin, B. R. (1992).

Aminoalkylindole analogs: cannabimimetic activity of a class of compounds structurally

distinct from delta 9-tetrahydrocannabinol. The Journal of Pharmacology and Experimental

Therapeutics, 263(3), 1118–26. Retrieved from

http://www.ncbi.nlm.nih.gov/pubmed/1335057

Cooperberg, M. R., Broering, J. M., & Carroll, P. R. (2010). Time trends and local variation in

primary treatment of localized prostate cancer. Journal of Clinical Oncology : Official

Journal of the American Society of Clinical Oncology, 28(7), 1117–23.

https://doi.org/10.1200/JCO.2009.26.0133

Cooperberg, M. R., Carroll, P. R., & Klotz, L. (2011). Active surveillance for prostate cancer:

progress and promise. Journal of Clinical Oncology : Official Journal of the American

Society of Clinical Oncology, 29(27), 3669–76. https://doi.org/10.1200/JCO.2011.34.9738

Cosma, G., Acampora, G., Brown, D., Rees, R. C., Khan, M., & Pockley, A. G. (2016).

Prediction of pathological stage in patients with prostate cancer: A neuro-fuzzy model.

PLoS ONE, 11(6). https://doi.org/10.1371/journal.pone.0155856

D’mbra, T. E., Estep, K. G., Bell, M. R., Eissenstat, M. A., Josef, K. A., Ward, S. J., … Daley,

G. T. (1992). Conformationally Restrained Analogues of Pravadoline: Nanomolar Potent,

Enantioselective, (Aminoalkyl)indole Agonists of the Cannabinoid Receptor. Journal of

Medicinal Chemistry, 35(1), 124–135. https://doi.org/10.1021/jm00079a016

Dall’Era, M. A., Albertsen, P. C., Bangma, C., Carroll, P. R., Carter, H. B., Cooperberg, M. R.,

… Soloway, M. S. (2012). Active surveillance for prostate cancer: A systematic review of

the literature. European Urology. https://doi.org/10.1016/j.eururo.2012.05.072

Dando, I., Donadelli, M., Costanzo, C., Dalla Pozza, E., D’Alessandro, A., Zolla, L., & Palmieri,

M. (2013). Cannabinoids inhibit energetic metabolism and induce AMPK-dependent

autophagy in pancreatic cancer cells. Cell Death and Disease, 4(6).

https://doi.org/10.1038/cddis.2013.151

De Petrocellis, L., Ligresti, A., Schiano Moriello, A., Iappelli, M., Verde, R., Stott, C. G., … Di

Marzo, V. (2013). Non-THC cannabinoids inhibit prostate carcinoma growth in vitro and in

vivo: Pro-apoptotic effects and underlying mechanisms. British Journal of Pharmacology,

168(1), 79–102. https://doi.org/10.1111/j.1476-5381.2012.02027.x

De Petrocellis, L., Starowicz, K., Moriello, A. S., Vivese, M., Orlando, P., & Di Marzo, V.

(2007). Regulation of transient receptor potential channels of melastatin type 8 (TRPM8):

Effect of cAMP, cannabinoid CB1 receptors and endovanilloids. Experimental Cell

Research, 313(9), 1911–1920. https://doi.org/10.1016/j.yexcr.2007.01.008

De Petrocellis, L., Vellani, V., Schiano-Moriello, A., Marini, P., Magherini, P. C., Orlando, P., &

Di Marzo, V. (2008). Plant-Derived Cannabinoids Modulate the Activity of Transient

Receptor Potential Channels of Ankyrin Type-1 and Melastatin Type-8. Journal of

Pharmacology and Experimental Therapeutics, 325(3), 1007–1015.

https://doi.org/10.1124/jpet.107.134809

Page 130: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

116

Devane, W. A., Dysarz, F. A., Johnson, M. R., Melvin, L. S., & Howlett, A. C. (1988).

Determination and characterization of a cannabinoid receptor in rat brain. Molecular

Pharmacology, 34(5), 605–613. https://doi.org/88/050605-09$02.OO/O

Devane, W. A., Hanus, L., Breuer, A., Pertwee, R. G., Stevenson, L. A., Griffin, G., …

Mechoulam, R. (1992). Isolation and structure of a brain constituent that binds to the

cannabinoid receptor. Science (New York, N.Y.), 258(5090), 1946–9.

https://doi.org/10.1126/science.1470919

Dhanasekaran, S. M., Dash, A., Yu, J., Maine, I. P., Laxman, B., Tomlins, S. a, … Chinnaiyan,

A. M. (2005). Molecular profiling of human prostate tissues: insights into gene expression

patterns of prostate development during puberty. The FASEB Journal : Official Publication

of the Federation of American Societies for Experimental Biology, 19(2), 243–245.

https://doi.org/10.1096/fj.04-2415fje

Di Cristofano, A., De Acetis, M., Koff, A., Cordon-Cardo, C., & P Pandolfi, P. (2001). Pten and

p27KIP1 cooperate in prostate cancer tumor suppression in the mouse. Nature Genetics,

27(2). https://doi.org/10.1038/84879

Di Cristofano, A., Pesce, B., Cordon-Cardo, C., & Pandolfi, P. P. (1998). Pten is essential for

embryonic development and tumour suppression. Nature Genetics, 19(4), 348–355.

https://doi.org/10.1038/1235

Di Marzo, V. (2008). Targeting the endocannabinoid system: To enhance or reduce? Nature

Reviews Drug Discovery. https://doi.org/10.1038/nrd2553

Di Marzo, V., Breivogel, C. S., Tao, Q., Bridgen, D. T., Razdan, R. K., Zimmer, A. M., …

Martin, B. R. (2000). Levels, metabolism, and pharmacological activity of anandamide in

CB1 cannabinoid receptor knockout mice: Evidence for non-CB1, non-CB2 receptor-

mediated actions of anandamide in mouse brain. Journal of Neurochemistry, 75(6), 2434–

2444. https://doi.org/10.1046/j.1471-4159.2000.0752434.x

Di Marzo, V., & Cristino, L. (2008). Why endocannabinoids are not all alike. Nature

Neuroscience. https://doi.org/10.1038/nn0208-124

Di Marzo, V., De Petrocellis, L., Fezza, F., Ligresti, A., & Bisogno, T. (2002). Anandamide

receptors. Prostaglandins, Leukotrienes and Essential Fatty Acids (PLEFA), 66(2–3), 377–

391. https://doi.org/10.1054/plef.2001.0349

Díaz-Laviada, I. (2011). The endocannabinoid system in prostate cancer. Nature Reviews

Urology, 8(10), 553–561. https://doi.org/10.1038/nrurol.2011.130

Drysdale, A. J., Ryan, D., Pertwee, R. G., & Platt, B. (2006). Cannabidiol-induced intracellular

Ca2+ elevations in hippocampal cells. Neuropharmacology, 50(5), 621–631.

https://doi.org/10.1016/j.neuropharm.2005.11.008

Page 131: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

117

Duffield-Lillico, A. J., Dalkin, B. L., Reid, M. E., Turnbull, B. W., Slate, E. H., Jacobs, E. T., …

Clark, L. C. (2003). Selenium supplementation, baseline plasma selenium status and

incidence of prostate cancer: An analysis of the complete treatment period of the Nutritional

Prevention of Cancer Trial. BJU International, 91(7), 608–612.

https://doi.org/10.1046/j.1464-410X.2003.04167.x

Ellwood-Yen, K., Graeber, T. G., Wongvipat, J., Iruela-Arispe, M. L., Zhang, J., Matusik, R., …

Sawyers, C. L. (2003). Myc-driven murine prostate cancer shares molecular features with

human prostate tumors. Cancer Cell, 4(3), 223–238. https://doi.org/10.1016/S1535-

6108(03)00197-1

Elmore, S. (2007). Apoptosis: A Review of Programmed Cell Death. Toxicologic Pathology.

https://doi.org/10.1080/01926230701320337

ElSohly, M. A., & Slade, D. (2005). Chemical constituents of marijuana: The complex mixture

of natural cannabinoids. In Life Sciences (Vol. 78, pp. 539–548).

https://doi.org/10.1016/j.lfs.2005.09.011

Endsley, M. P., Aggarwal, N., Isbell, M. A., Wheelock, C. E., Hammock, B. D., Falck, J. R., …

Nithipatikom, K. (2007). Diverse roles of 2-arachidonoylglycerol in invasion of prostate

carcinoma cells: Location, hydrolysis and 12-lipoxygenase metabolism. International

Journal of Cancer, 121(5), 984–991. https://doi.org/10.1002/ijc.22761

Endsley, M. P., Thill, R., Choudhry, I., Williams, C. L., Kajdacsy-Balla, A., Campbell, W. B., &

Nithipatikom, K. (2008). Expression and function of fatty acid amide hydrolase in prostate

cancer. International Journal of Cancer. Journal International Du Cancer, 123(6), 1318–

26. https://doi.org/10.1002/ijc.23674

Epstein, J. I., Zelefsky, M. J., Sjoberg, D. D., Nelson, J. B., Egevad, L., Magi-Galluzzi, C., …

Klein, E. A. (2016). A Contemporary Prostate Cancer Grading System: A Validated

Alternative to the Gleason Score. European Urology, 69(3), 428–435.

https://doi.org/10.1016/j.eururo.2015.06.046

Fan, F., Compton, D. R., Ward, S., Melvin, L., & Martin, B. R. (1994). Development of cross-

tolerance between delta 9-tetrahydrocannabinol, CP 55,940 and WIN 55,212. Journal of

Pharmacology and Experimental Therapeutics, 271(3), 1383–1390. Retrieved from

http://jpet.aspetjournals.org/content/271/3/1383.abstract

Ferlay, J., Soerjomataram, I., Ervik, M., Dikshit, R., Eser, S., Mathers, C., … Bray, F. (2013).

GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase.

No. 11 [Internet]. https://doi.org/10.1016/j.ucl.2013.01.011

Fizazi, K., Scher, H. I., Molina, A., Logothetis, C. J., Chi, K. N., Jones, R. J., … de Bono, J. S.

(2012). Abiraterone acetate for treatment of metastatic castration-resistant prostate cancer:

Final overall survival analysis of the COU-AA-301 randomised, double-blind, placebo-

controlled phase 3 study. The Lancet Oncology, 13, 983–992.

https://doi.org/http://dx.doi.org/10.1016/S1470-2045%2812%2970379-0

Page 132: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

118

Fong, E. L. S., Martinez, M., Yang, J., Mikos, A. G., Navone, N. M., Harrington, D. A., &

Farach-Carson, M. C. (2014). Hydrogel-based 3D model of patient-derived prostate

xenograft tumors suitable for drug screening. Molecular Pharmaceutics, 11(7), 2040–2050.

https://doi.org/10.1021/mp500085p

Fong, T. M., Shearman, L. P., Stribling, D. S., Shu, J., Lao, J., Huang, C. R. R., … Lanning, C.

L. (2009). Pharmacological efficacy and safety profile of taranabant in preclinical species.

Drug Development Research, 70(5), 349–362. https://doi.org/10.1002/ddr.20311

Fonseca, B. M., Costa, M. A., Almada, M., Correia-Da-Silva, G., & Teixeira, N. A. (2013).

Endogenous cannabinoids revisited: A biochemistry perspective. Prostaglandins and Other

Lipid Mediators. https://doi.org/10.1016/j.prostaglandins.2013.02.002

Fradet, Y., Klotz, L., Trachtenberg, J., & Zlotta, A. (2009). The burden of prostate cancer in

Canada. Journal of the Canadian Urological Association.

Galanti, G., Fisher, T., Kventsel, I., Shoham, J., Gallily, R., Mechoulam, R., … Toren, A. (2008).

Δ9-Tetrahydrocannabinol inhibits cell cycle progression by downregulation of E2F1 in

human glioblastoma multiforme cells. Acta Oncologica, 47(6), 1062–1070.

https://doi.org/10.1080/02841860701678787

Galiegue, S., Mary, S., Marchand, J., Dussossoy, D., Carriere, D., Carayon, P., … Casellas, P.

(1995). Expression of central and peripheral cannabinoid receptors in human immune

tissues and leukocyte subpopulations. Eur J Biochem, 232(1), 54–61.

https://doi.org/10.1111/j.1432-1033.1995.tb20780.x

Garber, K. (2009). From human to mouse and back: “Tumorgraft” models surge in popularity.

Journal of the National Cancer Institute. https://doi.org/10.1093/jnci/djn481

Gingrich, J. R., Barrios, R. J., Morton, R. A., Boyce, B. F., DeMayo, F. J., Finegold, M. J., …

Greenberg, N. M. (1996). Metastatic prostate cancer in a transgenic mouse. Cancer

Research, 56(18), 4096–4102.

Gordetsky, J., & Epstein, J. (2016). Grading of prostatic adenocarcinoma: current state and

prognostic implications. Diagnostic Pathology, 11(1), 25. https://doi.org/10.1186/s13000-

016-0478-2

Gratzke, C., Weinhold, P., Reich, O., Seitz, M., Schlenker, B., Stief, C. G., … Hedlund, P.

(2010). Transient Receptor Potential A1 and Cannabinoid Receptor Activity in Human

Normal and Hyperplastic Prostate: Relation to Nerves and Interstitial Cells. European

Urology, 57(5), 902–910. https://doi.org/10.1016/j.eururo.2009.08.019

Greenberg, N. M. (1996). Transgenic models for prostate cancer research. Urologic Oncology,

2(4), 119–122.

Gurel, B., Iwata, T., Koh, C. M., Jenkins, R. B., Lan, F., Van Dang, C., … De Marzo, A. M.

(2008). Nuclear MYC protein overexpression is an early alteration in human prostate

carcinogenesis. Modern Pathology, 21(9), 1156–1167.

https://doi.org/10.1038/modpathol.2008.111

Page 133: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

119

Guzman, M. (2003). Cannabinoids: potential anticancer agents. Nature reviews.Cancer, 3(10),

745–755. https://doi.org/10.1038/nrc1188

Guzmán, M., Sánchez, C., & Galve-Roperh, I. (2002). Cannabinoids and cell fate. Pharmacology

and Therapeutics, 95(2), 175–184. https://doi.org/10.1016/S0163-7258(02)00256-5

Hanahan, D., & Weinberg, R. A. (2000). The hallmarks of cancer. Cell, 100(1), 57–70.

https://doi.org/10.1007/s00262-010-0968-0

Hanahan, D., & Weinberg, R. A. (2011). Ha1. Hanahan D, Weinberg RA. Hallmarks of cancer:

The next generation. Vol. 144, Cell. 2011. p. 646–74. llmarks of cancer: The next

generation. Cell.

Hanus, L., Abu-Lafi, S., Fride, E., Breuer, A., Vogel, Z., Shalev, D. E., … Mechoulam, R.

(2001). 2-Arachidonyl glyceryl ether, an endogenous agonist of the cannabinoid CB1

receptor. Proceedings of the National Academy of Sciences, 98(7), 3662–3665.

https://doi.org/10.1073/pnas.061029898

Harbour, J. W., & Dean, D. C. (2000). Rb function in cell-cycle regulation and apoptosis. Nature

Cell Biology. https://doi.org/10.1038/35008695

Hasenoehrl, C., Feuersinger, D., Sturm, E. M., Bärnthaler, T., Heitzer, E., Graf, R., … Schicho,

R. (2017). G protein-coupled receptor GPR55 promotes colorectal cancer and has opposing

effects to cannabinoid receptor 1. International Journal of Cancer.

https://doi.org/10.1002/ijc.31030

Heidenreich, A., Bastian, P. J., Bellmunt, J., Bolla, M., Joniau, S., Van Der Kwast, T., … Mottet,

N. (2014). EAU guidelines on prostate cancer. Part II: Treatment of advanced, relapsing,

and castration-resistant prostate cancer. European Urology, 65(2), 467–479.

https://doi.org/10.1016/j.eururo.2013.11.002

Heidenreich, A., Bellmunt, J., Bolla, M., Joniau, S., Mason, M., Matveev, V., … Zattoni, F.

(2011). EAU guidelines on prostate cancer. Part 1: Screening, diagnosis, and treatment of

clinically localised disease. European Urology.

https://doi.org/10.1016/j.eururo.2010.10.039

Herrera, B., Carracedo, A., Diez-Zaera, M., Gómez del Pulgar, T., Guzmán, M., & Velasco, G.

(2006). The CB2 cannabinoid receptor signals apoptosis via ceramide-dependent activation

of the mitochondrial intrinsic pathway. Experimental Cell Research, 312(11), 2121–2131.

https://doi.org/10.1016/j.yexcr.2006.03.009

Holcomb, I. N., Young, J. M., Coleman, I. M., Salari, K., Grove, D. I., Li, H., … Trask, B. J.

(2009). Comparative analyses of chromosome alterations in soft-tissue metastases within

and across patients with castration-resistant prostate cancer. Cancer Research, 69(19),

7793–7802. https://doi.org/10.1158/0008-5472.CAN-08-3810

Hotte, S. J., & Saad, F. (2010). Current management of castrate-resistant prostate cancer.

Current Oncology, 17(SUPPL. 2). https://doi.org/10.3747/co.v17i0.718

Page 134: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

120

Howlett, A. C. (2005). Cannabinoid receptor signaling. Handbook of Experimental

Pharmacology, 168, 53–79. https://doi.org/10.1007/3-540-26573-2-2

Hsieh, C., Xie, Z., Yu, J., Martin, W. D., Datta, M. W., Wu, G., & Chung, L. W. K. (2007). Non-

invasive bioluminescent detection of prostate cancer growth and metastasis in a bigenic

transgenic mouse model. The Prostate, 67(7), 685–691. https://doi.org/10.1002/pros.20510

Hu, H., Jiang, C., Schuster, T., Li, G.-X., Daniel, P. T., & Lü, J. (2006). Inorganic selenium

sensitizes prostate cancer cells to TRAIL-induced apoptosis through superoxide/p53/Bax-

mediated activation of mitochondrial pathway. Molecular Cancer Therapeutics, 5(7), 1873–

82. https://doi.org/10.1158/1535-7163.MCT-06-0063

Israel, K., Sanders, B. G., & Kline, K. (1995). Rrr-Alpha-Tocopheryl Succinate Inhibits the

Proliferation of Human Prostatic Tumor-Cells with Defective Cell-Cycle Differentiation

Pathways. Nutrition and Cancer-an International Journal, 24(2), 161–169.

Izzo, A. A., & Camilleri, M. (2009). Cannabinoids in intestinal inflammation and cancer.

Pharmacological Research. https://doi.org/10.1016/j.phrs.2009.03.008

Johnson, J. L., Pillai, S., Pernazza, D., Sebti, S. M., Lawrence, N. J., & Chellappan, S. P. (2012).

Regulation of matrix metalloproteinase genes by E2F transcription factors: Rb-Raf-1

interaction as a novel target for metastatic disease. Cancer Research, 72(2), 516–526.

https://doi.org/10.1158/0008-5472.CAN-11-2647

Jordt, S. E., Bautista, D. M., Chuang, H. H., McKemy, D. D., Zygmunt, P. M., Högestätt, E. D.,

… Julius, D. (2004). Mustard oils and cannabinoids excite sensory nerve fibres through the

TRP channel ANKTM1. Nature, 427(6971), 260–265. https://doi.org/10.1038/nature02282

Kasper, S., Sheppard, P. C., Yan, Y., Pettigrew, N., Borowsky, A. D., Prins, G. S., … Matusik,

R. J. (1998). Development, progression, and androgen-dependence of prostate tumors in

probasin-large T antigen transgenic mice: a model for prostate cancer. Laboratory

Investigation; a Journal of Technical Methods and Pathology, 78(3), 319–333.

Kent, E. C., & Hussain, M. H. (2003). Neoadjuvant Therapy for Prostate Cancer: An

Oncologist’s Perspective. Reviews in Urology, 5 Suppl 3, S28-37. Retrieved from

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=1502344&tool=pmcentrez&ren

dertype=abstract

Kerbel, R. S. (2003). Human tumor xenografts as predictive preclinical models for anticancer

drug activity in humans: better than commonly perceived-but they can be improved. Cancer

Biology & Therapy. https://doi.org/213 [pii]

Khan, K. H., Blanco-Codesido, M., & Molife, L. R. (2014). Cancer therapeutics: Targeting the

apoptotic pathway. Critical Reviews in Oncology/Hematology.

https://doi.org/10.1016/j.critrevonc.2013.12.012

Khanapure, S. P., Garvey, D. S., Janero, D. R., & Letts, L. G. (2007). Eicosanoids in

inflammation: biosynthesis, pharmacology, and therapeutic frontiers. Current Topics in

Medicinal Chemistry, 7(3), 311–40. https://doi.org/10.2174/156802607779941314

Page 135: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

121

Khandrika, L., Kumar, B., Koul, S., Maroni, P., & Koul, H. K. (2009). Oxidative stress in

prostate cancer. Cancer Letters. https://doi.org/10.1016/j.canlet.2008.12.011

Kim, E. H., & Andriole, G. L. (2015). Prostate-specific antigen-based screening: controversy and

guidelines. BMC Medicine, 13(1), 61. https://doi.org/10.1186/s12916-015-0296-5

Kim, M. J., Cardiff, R. D., Desai, N., Banach-Petrosky, W. A., Parsons, R., Shen, M. M., &

Abate-Shen, C. (2002). Cooperativity of Nkx3.1 and Pten loss of function in a mouse model

of prostate carcinogenesis. Proceedings of the National Academy of Sciences of the United

States of America, 99(5), 2884–9. https://doi.org/10.1073/pnas.042688999

Kim, S. J., Uehara, H., Yazici, S., Busby, J. E., Nakamura, T., He, J. Q., … Fidler, I. J. (2006).

Targeting platelet-derived growth factor receptor on endothelial cells of multidrug-resistant

prostate cancer. J Natl Cancer Inst, 98(11), 783–793. https://doi.org/10.1093/jnci/djj211

Klein, R. D. (2005). The use of genetically engineered mouse models of prostate cancer for

nutrition and cancer chemoprevention research. Mutation Research, 576(1–2), 111–119.

https://doi.org/10.1016/j.mrfmmm.2005.02.012

Klink, J. C., Miocinovic, R., Galluzzi, C. M., & Klein, E. A. (2012). High-Grade prostatic

intraepithelial neoplasia. Korean Journal of Urology.

https://doi.org/10.4111/kju.2012.53.5.297

Klotz, L. (2005). Active surveillance for prostate cancer: For whom? Journal of Clinical

Oncology. https://doi.org/10.1200/JCO.2005.03.3134

Klotz, L., Vesprini, D., Sethukavalan, P., Jethava, V., Zhang, L., Jain, S., … Loblaw, A. (2015).

Long-term follow-up of a large active surveillance cohort of patients with prostate cancer.

Journal of Clinical Oncology, 33(3), 272–277. https://doi.org/10.1200/JCO.2014.55.1192

Koh, C. M., Bieberich, C. J., Dang, C. V., Nelson, W. G., Yegnasubramanian, S., & De Marzo,

A. M. (2010). MYC and prostate cancer. Genes and Cancer, 1(6), 617–628.

https://doi.org/10.1177/1947601910379132

KS, Z. (2010). Cell Migration: Signalling and Mechanisms. (Entschladen F, Ed.).

https://doi.org/10.1159/isbn.978-3-8055-9322-9

Kubota, T., Koshizuka, K., Koike, M., Uskokovic, M., Miyoshi, I., & Koeffler, H. P. (1998). 19-

nor-26,27-bishomo-vitamin D3 analogs: A unique class of potent inhibitors of proliferation

of prostate, breast, and hematopoietic cancer cells. Cancer Research, 58(15), 3370–3375.

Kumar, B., Koul, S., Khandrika, L., Meacham, R. B., & Koul, H. K. (2008). Oxidative stress is

inherent in prostate cancer cells and is required for aggressive phenotype. Cancer Research,

68(6), 1777–1785. https://doi.org/10.1158/0008-5472.CAN-07-5259

Laezza, C., Pisanti, S., Malfitano, A. M., & Bifulco, M. (2008). The anandamide analog, Met-F-

AEA, controls human breast cancer cell migration via the RHOA/RHO kinase signaling

pathway. Endocrine-Related Cancer, 15(4), 965–974. https://doi.org/10.1677/ERC-08-0030

Page 136: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

122

Laezza, C., Simona Pisanti, Crescenzi, E., & Bifulco, M. (2006). Anandamide inhibits Cdk2 and

activates Chk1 leading to cell cycle arrest in human breast cancer cells. FEBS Letters,

580(26), 6076–6082. https://doi.org/10.1016/j.febslet.2006.09.074

Lambrechts, A., Van Troys, M., & Ampe, C. (2004). The actin cytoskeleton in normal and

pathological cell motility. The International Journal of Biochemistry & Cell Biology,

36(10), 1890–909. https://doi.org/10.1016/j.biocel.2004.01.024

Lane, J. R., Beukers, M. W., Mulder-Krieger, T., & IJzerman, A. P. (2010). The

endocannabinoid 2-arachidonylglycerol is a negative allosteric modulator of the human A3

adenosine receptor. Biochemical Pharmacology, 79(1), 48–56.

https://doi.org/10.1016/j.bcp.2009.07.024

Lee, S. H., Jung, S. H., Shin, S., Kim, M. S., Baek, I. P., Lee, J. Y., … Chung, Y. J. (2016).

Genetic Progression of High Grade Prostatic Intraepithelial Neoplasia to Prostate Cancer.

European Urology, 69(5), 823–830. https://doi.org/10.1016/j.eururo.2015.10.031

Lee, S. O., Chun, J. Y., Nadiminty, N., Trump, D. L., Ip, C., Dong, Y., & Gao, A. C. (2006).

Monomethylated selenium inhibits growth of LNCaP human prostate cancer xenograft

accompanied by a decrease in the expression of androgen receptor and prostate-specific

antigen (PSA). Prostate, 66(10), 1070–1075. https://doi.org/10.1002/pros.20329

Ligresti, A., Moriello, A., Starowicz, K., Matias, I., Pisanti, S., De Petrocellis, L., … Di Marzo,

V. (2006). Antitumor activity of plant cannabinoids with emphasis on the effect of

cannabidiol on human breast carcinoma. J Pharmacol Exp Ther, 318, 1375–1387.

Ling, Y.-H., Liebes, L., Zou, Y., & Perez-Soler, R. (2003). Reactive oxygen species generation

and mitochondrial dysfunction in the apoptotic response to Bortezomib, a novel proteasome

inhibitor, in human H460 non-small cell lung cancer cells. The Journal of Biological

Chemistry, 278(36), 33714–33723. https://doi.org/10.1074/jbc.M302559200

Lippman, S. M., Klein, E. A., Goodman, P. J., Lucia, M. S., Thompson, I. M., Ford, L. G., …

Coltman, C. A. (2009). Effect of selenium and vitamin E on risk of prostate cancer and

other cancers: the Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA,

301(1), 39–51. https://doi.org/10.1001/jama.2008.864

Liu, J., Gao, B., Mirshahi, F., Sanyal, A. J., Khanolkar, A. D., Makriyannis, A., & Kunos, G.

(2000). Functional CB1 cannabinoid receptors in human vascular endothelial cells. The

Biochemical Journal, 346 Pt 3, 835–40. https://doi.org/10.1042/bj3460835

Locksley, R. M., Killeen, N., & Lenardo, M. J. (2001). The TNF and TNF receptor

superfamilies: Integrating mammalian biology. Cell. https://doi.org/10.1016/S0092-

8674(01)00237-9

Lombard, C., Nagarkatti, M., & Nagarkatti, P. (2007). CB2 cannabinoid receptor agonist, JWH-

015, triggers apoptosis in immune cells: potential role for CB2-selective ligands as

immunosuppressive agents. Clinical Immunology (Orlando, Fla.), 122(3), 259–70.

https://doi.org/10.1016/j.clim.2006.11.002

Page 137: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

123

Maccarrone, M., Lorenzon, T., Bari, M., Melino, G., & Finazzi-Agro, A. (2000). Anandamide

induces apoptosis in human cells via vanilloid receptors. Evidence for a protective role of

cannabinoid receptors. The Journal of Biological Chemistry, 275(41), 31938–45.

https://doi.org/10.1074/jbc.M005722200

Malhotra, J. D., & Kaufman, R. J. (2007). Endoplasmic Reticulum Stress and Oxidative Stress:

A Vicious Cycle or a Double-Edged Sword? Antioxidants & Redox Signaling, 9(12), 2277–

2294. https://doi.org/10.1089/ars.2007.1782

Mardal, M., Gracia-Lor, E., Leibnitz, S., Castiglioni, S., & Meyer, M. R. (2016). Toxicokinetics

of new psychoactive substances: plasma protein binding, metabolic stability, and human

phase I metabolism of the synthetic cannabinoid WIN 55,212-2 studied using in vitro tools

and LC-HR-MS/MS. Drug Testing and Analysis, 8(10), 1039–1048.

https://doi.org/10.1002/dta.1938

Martin, N. E., & D’Amico, A. V. (2014). Progress and controversies: Radiation therapy for

prostate cancer. CA: A Cancer Journal for Clinicians, 64(6), 389–407.

https://doi.org/10.3322/caac.21250

Massi, P., Vaccani, A., Bianchessi, S., Costa, B., Macchi, P., & Parolaro, D. (2006). The non-

psychoactive cannabidiol triggers caspase activation and oxidative stress in human glioma

cells. Cellular and Molecular Life Sciences, 63(17), 2057–2066.

https://doi.org/10.1007/s00018-006-6156-x

Masumori, N., Thomas, T. Z., Chaurand, P., Case, T., Paul, M., Kasper, S., … Matusik, R. J.

(2001). A probasin-large T antigen transgenic mouse line develops prostate adenocarcinoma

and neuroendocrine carcinoma with metastatic potential. Cancer Research, 61(5), 2239–

2249.

Matos, H. R., Capelozzi, V. L., Gomes, O. F., Mascio, P. D., & Medeiros, M. H. (2001).

Lycopene inhibits DNA damage and liver necrosis in rats treated with ferric

nitrilotriacetate. Archives of Biochemistry and Biophysics, 396(2), 171–7.

https://doi.org/10.1006/abbi.2001.2611

McKallip, R. J., Lombard, C., Fisher, M., Martin, B. R., Ryu, S., Grant, S., … Nagarkatti, M.

(2002). Targeting CB2 cannabinoid receptors as a novel therapy to treat malignant

lymphoblastic disease. Blood, 100(2), 627–634. https://doi.org/10.1182/blood-2002-01-

0098

McPartland, J. M., Matias, I., Di Marzo, V., & Glass, M. (2006). Evolutionary origins of the

endocannabinoid system. Gene, 370(1–2), 64–74.

https://doi.org/10.1016/j.gene.2005.11.004

Merrick, G. S., Butler, W. M., Galbreath, R. W., Lief, J. H., & Adamovich, E. (2003). Does

hormonal manipulation in conjunction with permanent interstitial brachytherapy, with or

without supplemental external beam irradiation, improve the biochemical outcome for men

with intermediate or high-risk prostate cancer? BJU International, 91(1), 23–29.

https://doi.org/10.1046/j.1464-410X.2003.04024.x

Page 138: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

124

Minelli, A., Bellezza, I., Conte, C., & Culig, Z. (2009). Oxidative stress-related aging: A role for

prostate cancer? Biochimica et Biophysica Acta - Reviews on Cancer.

https://doi.org/10.1016/j.bbcan.2008.11.001

Morgan, N. H., Stanford, I. M., & Woodhall, G. L. (2009). Functional CB2 type cannabinoid

receptors at CNS synapses. Neuropharmacology, 57(4), 356–368.

https://doi.org/10.1016/j.neuropharm.2009.07.017

Mori, A., Lehmann, S., O’Kelly, J., Kumagai, T., Desmond, J. C., Pervan, M., … Koeffler, H. P.

(2006). Capsaicin, a component of red peppers, inhibits the growth of androgen-

independent, p53 mutant prostate cancer cells. Cancer Research, 66(6), 3222–3229.

https://doi.org/10.1158/0008-5472.CAN-05-0087

Munro, S., Thomas, K. L., & Abu-Shaar, M. (1993). Molecular characterization of a peripheral

receptor for cannabinoids. Nature, 365(6441), 61–65. https://doi.org/10.1038/365061a0

Munson, a E., Harris, L. S., Friedman, M. a, Dewey, W. L., & Carchman, R. a. (1975).

Antineoplastic activity of cannabinoids. Journal of the National Cancer Institute, 55(3),

597–602. https://doi.org/10.1093/jnci/55.3.597

Navone, N. M., Olive, M., Ozen, M., Davis, R., Troncoso, P., Tu, S. M., … Logothetis, C. J.

(1997). Establishment of two human prostate cancer cell lines derived from a single bone

metastasis. Clinical Cancer Research : An Official Journal of the American Association for

Cancer Research, 3(12 Pt 1), 2493–500. Retrieved from

http://www.ncbi.nlm.nih.gov/pubmed/9815652

Nie, D., Guo, Y., Yang, D., Tang, Y., Chen, Y., Wang, M. T., … Honn, K. V. (2008).

Thromboxane A2 receptors in prostate carcinoma: expression and its role in regulating cell

motility via small GTPase Rho. Cancer Res., 68(1), 115–121. https://doi.org/10.1158/0008-

5472.CAN-07-1018

Nithipatikom, K., Endsley, M. P., Isbell, M. A., Falck, J. R., Iwamoto, Y., Hillard, C. J., &

Campbell, W. B. (2004). 2-Arachidonoylglycerol: A novel inhibitor of androgen-

independent prostate cancer cell invasion. Cancer Research, 64(24), 8826–8830.

https://doi.org/10.1158/0008-5472.CAN-04-3136

Nithipatikom, K., Gomez-Granados, A. D., Tang, A. T., Pfeiffer, A. W., Williams, C. L., &

Campbell, W. B. (2012). Cannabinoid receptor type 1 (CB1) activation inhibits small

GTPase RhoA activity and regulates motility of prostate carcinoma cells. Endocrinology,

153(1), 29–41. https://doi.org/10.1210/en.2011-1144

Olea-Herrero, N., Vara, D., Malagarie-Cazenave, S., & Díaz-Laviada, I. (2009). Inhibition of

human tumour prostate PC-3 cell growth by cannabinoids R()-Methanandamide and JWH-

015: Involvement of CB 2. British Journal of Cancer, 101(6), 940–950.

https://doi.org/10.1038/sj.bjc.6605248

Orellana-Serradell, O., Poblete, C. E., Sanchez, C., Castell??n, E. A., Gallegos, I., Huidobro, C.,

… Contreras, H. R. (2015). Proapoptotic effect of endocannabinoids in prostate cancer

cells. Oncology Reports, 33(4), 1599–1608. https://doi.org/10.3892/or.2015.3746

Page 139: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

125

Ortega, A., García-Hernández, V. M., Ruiz-García, E., Meneses-García, A., Herrera-Gómez, A.,

Aguilar-Ponce, J. L., … Del Angel, S. A. (2016). Comparing the effects of endogenous and

synthetic cannabinoid receptor agonists on survival of gastric cancer cells. Life Sciences,

165, 56–62. https://doi.org/10.1016/j.lfs.2016.09.010

Ortega, A., Rangel-López, E., Hidalgo-Miranda, A., Morales, A., Ruiz-García, E., Meneses-

García, A., … Del Angel, S. A. (2015). On the effects of CP 55-940 and other cannabinoid

receptor agonists in C6 and U373 cell lines. Toxicology in Vitro, 29(7).

https://doi.org/10.1016/j.tiv.2015.08.003

Paradies, G., Petrosillo, G., Pistolese, M., & Ruggiero, F. M. (2002). Reactive oxygen species

affect mitochondrial electron transport complex I activity through oxidative cardiolipin

damage. In Gene (Vol. 286, pp. 135–141). https://doi.org/10.1016/S0378-1119(01)00814-9

PArk, J., Xian, X., Choi, M., Park, H., Cho, Y., Lee, I., … Chung, I. (2011). Antiproliferative

mechanism of a cannabinoid agonist by cell cycle arrest in human gastric cancer cells. J

Cell Biochem, 112(4), 1192–1205.

Park, S. I., Kim, S. J., McCauley, L. K., & Gallick, G. E. (2010). Pre-Clinical Mouse Models of

Human Prostate Cancer and their Utility in Drug Discovery. Current Protocols in

Pharmacology / Editorial Board, S.J. Enna (Editor-in-Chief) ... [et Al.], 51, 14.15-14.15.27.

https://doi.org/10.1002/0471141755.ph1415s51

Park, S. I., Zhang, J., Phillips, K. a, Araujo, J. C., Najjar, A. M., Volgin, A. Y., … Gallick, G. E.

(2008). Targeting SRC family kinases inhibits growth and lymph node metastases of

prostate cancer in an orthotopic nude mouse model. Cancer Research, 68(9), 3323–33.

https://doi.org/10.1158/0008-5472.CAN-07-2997

Patel, K. D., Davison, J. S., Pittman, Q. J., & Sharkey, K. A. (2010). Cannabinoid CB(2)

receptors in health and disease. Curr Med Chem, 17(14), 1393–1410.

https://doi.org/BSP/CMC/E-Pub/ 092 [pii]

Patil, K. R., Goyal, S. N., Sharma, C., Patil, C. R., & Ojha, S. (2015). Phytocannabinoids for

Cancer Therapeutics: Recent Updates and Future Prospects. Current Medicinal Chemistry,

22(30), 3472–3501. https://doi.org/10.2174/0929867322666150716115057

Peehl, D. M. (2005). Primary cell cultures as models of prostate cancer development. Endocrine-

Related Cancer. https://doi.org/10.1677/erc.1.00795

Peehl, D. M., Krishnan, A. V, & Feldman, D. (2003). Pathways mediating the growth-inhibitory

actions of vitamin D in prostate cancer. The Journal of Nutrition, 133, 2461S–2469S.

Pellerito, O., Calvaruso, G., Portanova, P., De Blasio, A., Santulli, A., Vento, R., … Giuliano,

M. (2010). The Synthetic Cannabinoid WIN 55,212-2 Sensitizes Hepatocellular Carcinoma

Cells to Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL)-Induced

Apoptosis by Activating p8/CCAAT/Enhancer Binding Protein Homologous Protein

(CHOP)/Death Receptor . Molecular Pharmacology, 77(5), 854–863.

Page 140: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

126

Pertwee, R. G. (1999). Evidence for the presence of CB1 cannabinoid receptors on peripheral

neurones and for the existence of neuronal non-CB1 cannabinoid receptors. Life Sciences,

65(6–7), 597–605. https://doi.org/10.1016/S0024-3205(99)00282-9

Pertwee, R. G., Howlett, A. C., Abood, M. E., Alexander, S. P. H., Di Marzo, V., Elphick, M. R.,

… Ross, R. A. (2010). International Union of Basic and Clinical Pharmacology. LXXIX.

Cannabinoid Receptors and Their Ligands: Beyond CB1 and CB2. Pharmacological

Reviews, 62(4), 588–631. https://doi.org/10.1124/pr.110.003004

Pertwee, R. G., Thomas, A., Stevenson, L. A., Maor, Y., & Mechoulam, R. (2005). Evidence that

(-)-7-hydroxy-4′-dimethylheptyl-cannabidiol activates a non-CB1, non-CB2, non-TRPV1

target in the mouse vas deferens. Neuropharmacology, 48(8 SPEC. ISS.), 1139–1146.

https://doi.org/10.1016/j.neuropharm.2005.01.010

Peterson, J. K., & Houghton, P. J. (2004). Integrating pharmacology and in vivo cancer models

in preclinical and clinical drug development. European Journal of Cancer.

https://doi.org/10.1016/j.ejca.2004.01.003

Pĩeiro, R., Maffucci, T., & Falasca, M. (2011). The putative cannabinoid receptor GPR55 defines

a novel autocrine loop in cancer cell proliferation. Oncogene, 30(2), 142–152.

https://doi.org/10.1038/onc.2010.417

Pierorazio, P. M., Walsh, P. C., Partin, A. W., & Epstein, J. I. (2013). Prognostic Gleason grade

grouping: Data based on the modified Gleason scoring system. BJU International, 111(5),

753–760. https://doi.org/10.1111/j.1464-410X.2012.11611.x

Pillé, J. Y., Denoyelle, C., Varet, J., Bertrand, J. R., Soria, J., Opolon, P., … Li, H. (2005). Anti-

RhoA and Anti-RhoC siRNAs inhibit the proliferation and invasiveness of MDA-MB-231

breast cancer cells in vitro and in vivo. Molecular Therapy, 11(2), 267–274.

https://doi.org/10.1016/j.ymthe.2004.08.029

Pitot, H. C. (1993). The molecular biology of carcinogenesis. Cancer, 72(3 S), 962–970.

https://doi.org/10.1002/1097-0142(19930801)72:3+<962::AID-

CNCR2820721303>3.0.CO;2-H

Podsypanina, K., Ellenson, L. H., Nemes, A., Gu, J., Tamura, M., Yamada, K. M., … Parsons, R.

(1999). Mutation of Pten/Mmac1 in mice causes neoplasia in multiple organ systems.

Proceedings of the National Academy of Sciences, 96(4), 1563–1568.

https://doi.org/10.1073/pnas.96.4.1563

Qin, N., Neeper, M. P., Liu, Y., Hutchinson, T. L., Lubin, M. Lou, & Flores, C. M. (2008).

TRPV2 is activated by cannabidiol and mediates CGRP release in cultured rat dorsal root

ganglion neurons. The Journal of Neuroscience : The Official Journal of the Society for

Neuroscience, 28(24), 6231–6238. https://doi.org/10.1523/JNEUROSCI.0504-08.2008

Ramer, R., Merkord, J., Rohde, H., & Hinz, B. (2010). Cannabidiol inhibits cancer cell invasion

via upregulation of tissue inhibitor of matrix metalloproteinases-1. Biochemical

Pharmacology, 79(7), 955–966. https://doi.org/10.1016/j.bcp.2009.11.007

Page 141: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

127

Razdan, R. K. (1986). Structure-Activity Relationships in Cannabinoids. Pharmacology and

Experimantal Therapeutics, 38(2), 75–149.

Rea, D., Del Vecchio, V., Palma, G., Barbieri, A., Falco, M., Luciano, A., … Arra, C. (2016).

Mouse Models in Prostate Cancer Translational Research: From Xenograft to PDX. BioMed

Research International. https://doi.org/10.1155/2016/9750795

Ridley, A. J. (2001). Rho GTPases and cell migration. J Cell Sci, 114(Pt 15), 2713–2722.

https://doi.org/10.1083/jcb.150.4.807

Ridley, A. J. (2015). Rho GTPase signalling in cell migration. Current Opinion in Cell Biology.

https://doi.org/10.1016/j.ceb.2015.08.005

Roberto, D., Klotz, L. H., & Venkateswaran, V. (2017). Cannabinoids as an Anticancer Agent

for Prostate Cancer. Journal of Urology and Research, 4(3), 1090–1097.

Romero-Sandoval, A., & Eisenach, J. C. (2007). Spinal Cannabinoid Receptor Type 2 Activation

Reduces Hypersensitivity and Spinal Cord Glial Activation after Paw Incision.

Anesthesiology, 106(4), 787–794. https://doi.org/10.1097/01.anes.0000264765.33673.6c

Saad, F., & Hotte, S. J. (2010). Guidelines for the management of castrate-resistant prostate

cancer. Can Urol Assoc J, 44(6), 380–4.

Saelens, X., Festjens, N., Vande Walle, L., Van Gurp, M., Van Loo, G., & Vandenabeele, P.

(2004). Toxic proteins released from mitochondria in cell death. Oncogene.

https://doi.org/10.1038/sj.onc.1207523

Salazar, M., Carracedo, A., Salanueva, Í. J., Hernández-tiedra, S., Lorente, M., Egia, A., …

Velasco, G. (2009). Cannabinoid action induces autophagy- mediated cell death through

stimulation of ER stress in human glioma cells. The Journal of Clinical Investigation,

119(5), 1359–1372. https://doi.org/10.1172/JCI37948.of

Sampson, N., Neuwirt, H., Puhr, M., Klocker, H., & Eder, I. E. (2013). In vitro model systems to

study androgen receptor signaling in prostate cancer. Endocrine-Related Cancer.

https://doi.org/10.1530/ERC-12-0401

Sánchez, A. M., Sánchez, M. G., Malagarie-Cazenave, S., Olea, N., & Díaz-Laviada, I. (2006).

Induction of apoptosis in prostate tumor PC-3 cells and inhibition of xenograft prostate

tumor growth by the vanilloid capsaicin. Apoptosis, 11(1), 89–99.

https://doi.org/10.1007/s10495-005-3275-z

Sánchez, M. G., Ruiz-Llorente, L., Sánchez, A. M., & Díaz-Laviada, I. (2003). Activation of

phosphoinositide 3-kinase/PKB pathway by CB1and CB2cannabinoid receptors expressed

in prostate PC-3 cells. Involvement in Raf-1 stimulation and NGF induction. Cellular

Signalling, 15(9), 851–859. https://doi.org/10.1016/S0898-6568(03)00036-6

Page 142: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

128

Sarfaraz, S., Afaq, F., Adhami, V. M., Malik, A., & Mukhtar, H. (2006). Cannabinoid receptor

agonist-induced apoptosis of human prostate cancer cells LNCaP proceeds through

sustained activation of ERK1/2 leading to G1 cell cycle arrest. The Journal of Biological

Chemistry, 281(51), 39480–39491. https://doi.org/10.1074/jbc.M603495200

Sarfaraz, S., Afaq, F., Adhami, V. M., & Mukhtar, H. (2005). Cannabinoid receptor as a novel

target for the treatment of prostate cancer. Cancer Research, 65(5), 1635–1641.

https://doi.org/10.1158/0008-5472.CAN-04-3410

Sarker, K. P., & Maruyama, I. (2003). Anandamide induces cell death independently of

cannabinoid receptors or vanilloid receptor 1: possible involvement of lipid rafts. Cellular

and Molecular Life Sciences, 60(6), 1200–1208. https://doi.org/10.1007/s00018-003-3055-2

Sawzdargo, M., Nguyen, T., Lee, D. K., Lynch, K. R., Cheng, R., Heng, H. H. Q., … O’Dowd,

B. F. (1999). Identification and cloning of three novel human G protein-coupled receptor

genes GPR52, ΨGPR53 and GPR55: GPR55 is extensively expressed in human brain.

Molecular Brain Research, 64(2), 193–198. https://doi.org/10.1016/S0169-328X(98)00277-

0

Scher, H. I., Fizazi, K., Saad, F., Taplin, M.-E., Sternberg, C. N., Miller, K., … de Bono, J. S.

(2012). Increased Survival with Enzalutamide in Prostate Cancer after Chemotherapy. New

England Journal of Medicine, 367(13), 1187–1197.

https://doi.org/10.1056/NEJMoa1207506

Schröder, M., & Kaufman, R. J. (2005). THE MAMMALIAN UNFOLDED PROTEIN

RESPONSE. Annual Review of Biochemistry, 74(1), 739–789.

https://doi.org/10.1146/annurev.biochem.73.011303.074134

Schuel, H., Burkman, L. J., Lippes, J., Crickard, K., Forester, E., Piomelli, D., & Giuffrida, A.

(2002). N-Acylethanolamines in human reproductive fluids. In Chemistry and Physics of

Lipids (Vol. 121, pp. 211–227). https://doi.org/10.1016/S0009-3084(02)00158-5

Schwartz, G. K., & Shah, M. A. (2005). Targeting the cell cycle: a new approach to cancer

therapy. Journal of Clinical Oncology : Official Journal of the American Society of Clinical

Oncology, 23(36), 9408–21. https://doi.org/10.1200/JCO.2005.01.5594

Sharifi, N., Gulley, J. L., & Dahut, W. L. (2005). Androgen deprivation therapy for prostate

cancer. JAMA, 294(2), 238–44. https://doi.org/10.1001/jama.294.2.238

Sharma, M. K., Murumkar, P. R., Kanhed, A. M., Giridhar, R., & Yadav, M. R. (2014).

Prospective therapeutic agents for obesity: Molecular modification approaches of centrally

and peripherally acting selective cannabinoid 1 receptor antagonists. European Journal of

Medicinal Chemistry. https://doi.org/10.1016/j.ejmech.2014.04.011

Shrivastava, A., Kuzontkoski, P. M., Groopman, J. E., & Prasad, A. (2011). Cannabidiol Induces

Programmed Cell Death in Breast Cancer Cells by Coordinating the Cross-talk between

Apoptosis and Autophagy. Molecular Cancer Therapeutics, 10(7), 1161–1172.

https://doi.org/10.1158/1535-7163.MCT-10-1100

Page 143: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

129

Siddiqui, I. A., Sanna, V., Ahmad, N., Sechi, M., & Mukhtar, H. (2015). Resveratrol

nanoformulation for cancer prevention and therapy. Annals of the New York Academy of

Sciences, 1348(1), 20–31. https://doi.org/10.1111/nyas.12811

Sikka, S. C. (2003). Role of oxidative stress response elements and antioxidants in prostate

cancer pathobiology and chemoprevention--a mechanistic approach. Curr Med Chem,

10(24), 2679–2692. https://doi.org/10.2174/0929867033456341

Siolas, D., & Hannon, G. J. (2013). Patient-derived tumor xenografts: Transforming clinical

samples into mouse models. Cancer Research. https://doi.org/10.1158/0008-5472.CAN-13-

1069

Soloway, M. S., Pareek, K., Sharifi, R., Wajsman, Z., McLeod, D., Wood, D. P., & Puras-Baez,

A. (2002). Neoadjuvant androgen ablation before radical prostatectomy in cT2bNxMo

prostate cancer: 5-year results. The Journal of Urology, 167(1), 112–116.

https://doi.org/10.1097/00005392-200201000-00026

Soloway, M. S., Sharifi, R., Wajsman, Z., McLeod, D., Wood, D. P., & Puras-Baez, A. for the L.

D. N. P. C. S. G. (1995). Randomized Prospective Study Comparing Radical Prostatectomy

Alone Versus Radical Prostatectomy Preceded by Androgen Blockage in Clinical Stage B2

(T2bNxM0) Prostate Cancer. The Journal of Urology, 154(2), 424–428.

https://doi.org/10.1016/S0022-5347(01)67067-8

Somlyo, A. V., Bradshaw, D., Ramos, S., Murphy, C., Myers, C. E., & Somlyo, A. P. (2000).

Rho-Kinase Inhibitor Retards Migration and in Vivo Dissemination of Human Prostate

Cancer Cells. Biochemical and Biophysical Research Communications, 269(3), 652–659.

https://doi.org/10.1006/bbrc.2000.2343

Song, M. S., Salmena, L., & Pandolfi, P. P. (2012). The functions and regulation of the PTEN

tumour suppressor. Nature Reviews Molecular Cell Biology.

https://doi.org/10.1038/nrm3330

Stark, K., Dostalek, M., & Guengerich, F. P. (2008). Expression and purification of orphan

cytochrome P450 4X1 and oxidation of anandamide. FEBS Journal, 275(14), 3706–3717.

https://doi.org/10.1111/j.1742-4658.2008.06518.x

Stein, M. E., Boehmer, D., & Kuten, A. (2007). Radiation therapy in prostate cancer. Recent

Results in Cancer Research. Fortschritte Der Krebsforschung. Progrès Dans Les

Recherches Sur Le Cancer, 175, 179–99. Retrieved from

http://www.ncbi.nlm.nih.gov/pubmed/17432560

Stock, R. G., Cesaretti, J. A., & Stone, N. N. (2006). Disease-specific survival following the

brachytherapy management of prostate cancer. International Journal of Radiation Oncology

Biology Physics, 64(3), 810–816. https://doi.org/10.1016/j.ijrobp.2005.09.005

Storr, M. A., Keenan, C. M., Emmerdinger, D., Zhang, H., Yüce, B., Sibaev, A., … Sharkey, K.

A. (2008). Targeting endocannabinoid degradation protects against experimental colitis in

mice: Involvement of CB1 and CB2 receptors. Journal of Molecular Medicine, 86(8), 925–

936. https://doi.org/10.1007/s00109-008-0359-6

Page 144: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

130

Syed, D. N., Khan, N., Afaq, F., & Mukhtar, H. (2007). Chemoprevention of prostate cancer

through dietary agents: Progress and promise. Cancer Epidemiology Biomarkers and

Prevention. https://doi.org/10.1158/1055-9965.EPI-06-0942

Takeda, S., Okajima, S., Miyoshi, H., Yoshida, K., Okamoto, Y., Okada, T., … Aramaki, H.

(2012). Cannabidiolic acid, a major cannabinoid in fiber-type cannabis, is an inhibitor of

MDA-MB-231 breast cancer cell migration. Toxicology Letters, 214(3), 314–319.

https://doi.org/10.1016/j.toxlet.2012.08.029

Tanaka, M., Komuro, I., Inagaki, H., Jenkins, N. A., Copeland, N. G., & Izumo, S. (2000).

Nkx3.1, a murine homolog of Drosophila bagpipe, regulates epithelial ductal branching and

proliferation of the prostate and palatine glands. Developmental Dynamics, 219(2), 248–

260. https://doi.org/10.1002/1097-0177(2000)9999:9999<::AID-DVDY1054>3.3.CO;2-5

Tenke, P., Horti, J., Balint, P., & Kovacs, B. (2007). Prostate Cancer Screening. In Prostate

Cancer (Vol. 175, pp. 65–81). https://doi.org/10.1007/978-3-540-40901-4_5

Tentler, J. J., Tan, A. C., Weekes, C. D., Jimeno, A., Leong, S., Pitts, T. M., … Eckhardt, S. G.

(2012). Patient-derived tumour xenografts as models for oncology drug development.

Nature Reviews Clinical Oncology, 9(6), 338–350.

https://doi.org/10.1038/nrclinonc.2012.61

Tokanovic, S., Malone, D. T., & Ventura, S. (2007). Stimulation of epithelial CB1 receptors

inhibits contractions of the rat prostate gland. British Journal of Pharmacology, 150(2),

227–34. https://doi.org/10.1038/sj.bjp.0706952

Tu, W. H., Thomas, T. Z., Masumori, N., Bhowmick, N. A., Gorska, A. E., Shyr, Y., … Matusik,

R. J. (2003). The loss of TGF-beta signaling promotes prostate cancer metastasis. Neoplasia

(New York, N.Y.), 5(3), 267–77. https://doi.org/NO_DOI

Twelves, C., Short, S., & Wright Stephen. (2017). A two-part safety and exploratory efficacy

randomized double-blind, placebo-controlled study of a 1:1 ratio of the cannabinoids

cannabidiol and delta-9-tetrahydrocannabinol (CBD:THC) plus dose-intense temozolomide

in patients with recurrent glioblastoma m. Journal of Clinical Oncology, 35(15

supplementary), 2046.

Valkenburg, K. C., & Williams, B. O. (2011). Mouse Models of Prostate Cancer. Prostate

Cancer, 2011, 1–22. https://doi.org/10.1155/2011/895238

Velasco, G., Sánchez, C., & Guzmán, M. (2012). Towards the use of cannabinoids as antitumour

agents. Nature Reviews Cancer, 12(6), 436–444. https://doi.org/10.1038/nrc3247

Venier, N. A., Colquhoun, A. J., Sasaki, H., Kiss, A., Sugar, L., Adomat, H., … Venkateswaran,

V. (2015). Capsaicin: A novel radio-sensitizing agent for prostate cancer. Prostate, 75(2),

113–125. https://doi.org/10.1002/pros.22896

Page 145: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

131

Venier, N. A., Yamamoto, T., Sugar, L. M., Adomat, H., Fleshner, N. E., Klotz, L. H., &

Venkateswaran, V. (2015). Capsaicin reduces the metastatic burden in the transgenic

adenocarcinoma of the mouse prostate model. Prostate, 75(12), 1300–1311.

https://doi.org/10.1002/pros.23013

Venkatachalam, K., & Montell, C. (2007). TRP Channels. Annual Review of Biochemistry,

76(1), 387–417. https://doi.org/10.1146/annurev.biochem.75.103004.142819

Venkateswaran, V., Fleshner, N. E., Sugar, L. M., & Klotz, L. H. (2004). Antioxidants block

prostate cancer in Lady transgenic mice. Cancer Research, 64(16), 5891–5896.

https://doi.org/10.1158/0008-5472.CAN-04-0690

Venkateswaran, V., & Klotz, L. H. (2010). Diet and prostate cancer: mechanisms of action and

implications for chemoprevention. Nature Reviews Urology, 7(8), 442–453.

https://doi.org/10.1038/nrurol.2010.102

Verfaillie, T., Salazar, M., Velasco, G., & Agostinis, P. (2010). Linking ER Stress to Autophagy:

Potential Implications for Cancer Therapy. International Journal of Cell Biology, 2010,

930509. https://doi.org/10.1155/2010/930509

Voskoglou-Nomikos, T., Pater, J. L., & Seymour, L. (2003). Clinical predictive value of the in

vitro cell line, human xenograft, and mouse allograft preclinical cancer models. In Clinical

Cancer Research (Vol. 9, pp. 4227–4239).

Wang, D., Wang, H., Ning, W., Backlund, M. G., Dey, S. K., & DuBois, R. N. (2008). Loss of

cannabinoid receptor 1 accelerates intestinal tumor growth. Cancer Research, 68(15),

6468–6476. https://doi.org/10.1158/0008-5472.CAN-08-0896

Wang, J., Zhao, L. Y., Uyama, T., Tsuboi, K., Wu, X. X., Kakehi, Y., & Ueda, N. (2008).

Expression and secretion of N-acylethanolamine-hydrolysing acid amidase in human

prostate cancer cells. Journal of Biochemistry, 144(5), 685–690.

https://doi.org/10.1093/jb/mvn122

Wang, Z., Sun, X., Bao, Y., Mo, J., Du, H., Hu, J., & Zhang, X. (2017). E2F1 silencing inhibits

migration and invasion of osteosarcoma cells via regulating DDR1 expression.

International Journal of Oncology, 51(6), 1639–1650. https://doi.org/10.3892/ijo.2017.4165

Watanabe, H., Vriens, J., Prenen, J., Droogmans, G., Voets, T., & Nillus, B. (2003). Anandamide

and arachidonic acid use epoxyeicosatrienoic acids to activate TRPV4 channels. Nature,

424(6947), 434–438. https://doi.org/10.1038/nature01807

Wells, C. M., & Parsons, M. (2011). Cell Migration. https://doi.org/10.1007/978-1-61779-207-6

Whiteford, C. C., Bilke, S., Greer, B. T., Chen, Q., Braunschweig, T. A., Cenacchi, N., … Khan,

J. (2007). Credentialing preclinical pediatric xenograft models using gene expression and

tissue microarray analysis. Cancer Research, 67(1), 32–40. https://doi.org/10.1158/0008-

5472.CAN-06-0610

Page 146: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

132

Wong, M. C. S., Goggins, W. B., Wang, H. H. X., Fung, F. D. H., Leung, C., Wong, S. Y. S., …

Sung, J. J. Y. (2016). Global Incidence and Mortality for Prostate Cancer: Analysis of

Temporal Patterns and Trends in 36 Countries. European Urology, 70(5), 862–874.

https://doi.org/10.1016/j.eururo.2016.05.043

Yatani, R., Kusano, I., Shiraishi, T., Hayashi, T., & Stemmermann, G. N. (1989). Latent prostatic

carcinoma: pathological and epidemiological aspects. Japanese Journal of Clinical

Oncology, 19(4), 319–26. Retrieved from http://europepmc.org/abstract/med/2691730

Yoshimoto, M., Cutz, J. C., Nuin, P. A. S., Joshua, A. M., Bayani, J., Evans, A. J., … Squire, J.

A. (2006). Interphase FISH analysis of PTEN in histologic sections shows genomic

deletions in 68% of primary prostate cancer and 23% of high-grade prostatic intra-epithelial

neoplasias. Cancer Genetics and Cytogenetics, 169(2), 128–137.

https://doi.org/10.1016/j.cancergencyto.2006.04.003

Zhang, J., Wang, L., Zhang, Y., & Lu, J. (2013). Lobe-Specific Carcinogenesis in the Transgenic

Adenocarcinoma of Mouse Prostate (TRAMP) Mouse Model BT - (null), (Chapter 10).

Retrieved from http://www.intechopen.com/books/carcinogenesis/lobe-specific-

carcinogenesis-in-the-transgenic-adenocarcinoma-of-mouse-prostate-tramp-mouse-

model%5Cnpapers2://publication/doi/10.5772/54904

Zhou, C. K., Check, D. P., Lortet-Tieulent, J., Laversanne, M., Jemal, A., Ferlay, J., … Devesa,

S. S. (2016). Prostate cancer incidence in 43 populations worldwide: An analysis of time

trends overall and by age group. International Journal of Cancer, 138(6), 1388–1400.

https://doi.org/10.1002/ijc.29894

Page 147: The Use of Endogenous and Synthetic Cannabinoids in Prostate Cancer Therapy · 2018-11-15 · Cancer Therapy Domenica Roberto Master of Science Institute of Medical Science University

133

Appendix

Contributions

With the support and guidance of my supervisor Dr. Vasundara Venkateswaran and my co-

supervisor Dr. Laurence Klotz, I have gained the knowledge and experience to design and

perform experiments, and to analyze and interpret my results. They have contributed to my

successful publications and conference presentations and have ensured that my project remained

scientifically sound.

Dr. Geneve Awong and Courtney McIntosh provided me with the necessary training to complete

flow cytometry experiments for cell cycle analysis and apoptosis, and microscopy training for

wound healing experiments. Dr. Katerina Molnarova provided me with animal care training

needed to complete my in vivo studies. Dr. Roman Bass assisted with the in vivo component of

the project. Dr. Linda Sugar, pathologist at Sunnybrook Hospital, reviewed xenograft tumours

and liver samples obtained from the in vivo component of the project and contributed to

histopathological review of all animal tissues. Results were interpreted with the assistance of my

supervisor Dr. Vasundara Venkateswaran, and my co-supervisor Dr. Laurence Klotz.