210
The effects of hyperglycemia on glucose metabolism in epithelial ovarian cancer by Lisa Danielle Kellenberger A Thesis presented to The University of Guelph In partial fulfillment of the requirements for the degree of Doctor of Philosophy in Biomedical Science Guelph, Ontario, Canada © Lisa Kellenberger, February, 2016

The effects of hyperglycemia on glucose metabolism in

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: The effects of hyperglycemia on glucose metabolism in

The effects of hyperglycemia on glucose metabolism in epithelial ovarian cancer

by

Lisa Danielle Kellenberger

A Thesis presented to

The University of Guelph

In partial fulfillment of the requirements for the degree of

Doctor of Philosophy in

Biomedical Science

Guelph, Ontario, Canada

© Lisa Kellenberger, February, 2016

Page 2: The effects of hyperglycemia on glucose metabolism in

ABSTRACT

THE EFFECTS OF HYPERGLYCEMIA ON GLUCOSE METABOLISM IN EPITHELIAL OVARIAN CANCER

Lisa Danielle Kellenberger Advisor: University of Guelph, 2016 Dr. Jim Petrik

Glucose metabolism in cancer cells is uniquely adapted to maximize the production of

both ATP and the precursors needed to support cell proliferation and tissue growth.

Glycolysis, an anaerobic process normally limited by the presence of oxygen, proceeds

at an elevated rate in both aerobic and anaerobic conditions. Glycolysis consumes large

quantities of glucose while producing relatively small amounts of energy. The

inefficiency of this process is not detrimental to the cell when glucose is abundant.

However, evidence suggests that in cancer the normal concentration of circulating

glucose does not meet the energy demands of the tumour and is therefore a limiting

factor in cancer cell metabolism. Hyperglycemic conditions such as diabetes are

becoming common comorbidities in cancer patients and are associated with increased

risk and poorer prognosis of epithelial ovarian cancer (EOC). We hypothesise that

elevated blood glucose permits tumour metabolism to function at maximal capacity

thereby facilitating tumour growth and metastasis.

The goal of this thesis was to examine the metabolic response and glucose transport

kinetics of EOC in hyperglycemic environments. By inducing ovarian tumour growth in

Page 3: The effects of hyperglycemia on glucose metabolism in

mouse models of Type 1 and Type 2 diabetes, we found that accelerated disease

progression occurs in a glucose concentration-dependent manner.

In addition, for the first time we show that EOC cells express not only the passive

glucose transporters (GLUTs), but also a class of active glucose transporters, the

sodium-glucose symporters (SGLTs). Glucose uptake into EOC cells in hyperglycemia

is mediated by this sodium-dependent glucose transport. Surprisingly, SGLT2 appears

to be a tumour suppressor: knockdown of SGLT2 in ovarian cancer cells increases the

relative risk of death in tumour-bearing mice by more than 50 times.

PET scans measuring accumulation of the glucose analogue fluoro-deoxyglucose

(FDG) are a vital component of EOC treatment. However, FDG is a poor substrate for

SGLTs, suggesting that current imaging fails to detect a large fraction of glucose

transport into tumours. The use of SGLT-specific glucose analogues, particularly in

patients with metabolic dysfunction, presents an opportunity to enhance the power of

PET imaging and provide a more complete picture of tumour metabolism which may

have therapeutic benefits.

Page 4: The effects of hyperglycemia on glucose metabolism in

iv

Acknowledgements

Dr. Jim Petrik, Esteemed Professor, who always puts things in perspective. Thank you.

It’s been righteous.

Dr. Alison Holloway, a mentor, advocate, and voice of reason for me since I was 17.

My advisory committee: Dr. Jon Lamarre, Dr. Roger Moorehead and Dr. Lindsay

Robinson.

Maka Natsvlishvili, an expert and a wonderful teacher. Everyone at the Central Animal

Facility for their help, support and genuine interest in these projects.

Michelle Ross, who always listened and commiserated.

Dr. Jim Greenaway, the ultimate scientist, cynic, and skeptic.

Amanda Kerr, who shared the science happy-dance. Sam Russell, Jackie Dynes, and

Funk Nights. Simone ten Kortenaar, the dancer, whistler, and optimist. Kata Osz, an

artist, technician and general badass. Josh Antunes, for the techno and the drifting.

Dr. Jenny Bruin, who trusted me with her work.

Dr. Amy McPhedran, who always did more than her job.

Mom, Dad, Nonnie and Grandpa for their unconditional support. Mark and Kevin who

have been friends, foes, defenders, and great little brothers.

Kelsey, who promised to read my “book”.

Labmates, officemates, roommates.

The pool, the road, the gym.

The Science Gods (sometimes).

Generous funding from a CIHR Vanier Canada Graduate Scholarship.

Those damned mice.

Page 5: The effects of hyperglycemia on glucose metabolism in

v

Declaration of work performed

I declare that with the exception of the items listed below, all work reported in this thesis

was performed by me.

All tumour induction surgeries were performed with the assistance of Dr. Jim Petrik. Dr.

Jim Petrik and Dr. Jim Greenaway assisted with some of the tissue collection. Insulin

staining on pancreas sections in Figure 2A was done by Michelle Ross and serum

insulin quantification in Figure 8A was done by Nicole DeLong in the lab of Dr. Alison

Holloway at McMaster University. Intravenous injections of radiolabelled glucose

analogues (Chapter 3) were performed by Jennifer Randall of the University of Guelph

Central Animal Facility. Statistical analyses of mouse data in Chapter 3 were performed

with the help of Dr. Cate Dewey. Kata Osz stained the tumour tissue slides shown in

Figure 21B and C. The human ovarian tumour tissue microarray (Chapter 3) was

imaged at the Advanced Optical Microscopy Facility (AOMF) at the University Health

Network in Toronto.

Page 6: The effects of hyperglycemia on glucose metabolism in

vi

TABLE OF CONTENTS

Declaration of work performed .................................................................................... v

LIST OF TABLES ...........................................................................................................ix

LIST OF FIGURES.......................................................................................................... x

LIST OF ABBREVIATIONS .......................................................................................... xiii

CHAPTER 1: INTRODUCTION AND LITERATURE REVIEW ....................................... 1

Ovarian cancer ........................................................................................................... 1

Histological classifications ........................................................................................ 1

Screening and prevention ......................................................................................... 2

Tumour development ................................................................................................ 3

Diagnosis and clinical staging ................................................................................... 4

Treatment and disease management ....................................................................... 6

Current challenges .................................................................................................... 6

Glucose metabolism .................................................................................................. 7

Glycolysis ................................................................................................................. 7

Cellular Respiration .................................................................................................. 8

The citric acid cycle ............................................................................................... 8

The electron transport chain and oxidative phosphorylation .................................. 8

Anaerobic glycolysis (fermentation) .......................................................................... 9

Glucose metabolism in cancer ................................................................................. 9

The Warburg Effect ............................................................................................... 10

Mitochondria ........................................................................................................ 11

Hypoxia ............................................................................................................... 12

Tumour acidification ............................................................................................ 13

Speed of metabolism ........................................................................................... 13

Page 7: The effects of hyperglycemia on glucose metabolism in

vii

Metabolic intermediates ....................................................................................... 14

Primary genetic changes ..................................................................................... 15

Hexokinase 2 (HK2) ............................................................................................ 16

High interstitial fluid pressure .............................................................................. 16

Metabolic flexibility ............................................................................................... 17

Contributions of the tumour microenvironment ................................................ 18

The Crabtree Effect ............................................................................................. 18

Glucose transport .................................................................................................... 19

GLUTs .................................................................................................................... 19

SGLTs .................................................................................................................... 21

Positron emission tomography (PET) .................................................................... 24

Principle .................................................................................................................. 25

Limitations .............................................................................................................. 25

Use in gynecological cancers ................................................................................. 27

RATIONALE ................................................................................................................. 28

Hypothesis and objectives ...................................................................................... 28

CHAPTER 2: SYSTEMIC HYPERGLYCEMIA AND EOC PROGRESSION ................ 29

Introduction .............................................................................................................. 29

Materials and Methods ............................................................................................ 31

Results ...................................................................................................................... 39

Discussion ................................................................................................................ 58

CHAPTER 3: GLUCOSE TRANSPORT IN HYPERGLYCEMIC EOC ......................... 65

Introduction .............................................................................................................. 65

Materials and Methods ............................................................................................ 67

Discussion .............................................................................................................. 106

Page 8: The effects of hyperglycemia on glucose metabolism in

viii

CHAPTER 4: ANTIDIABETIC EFFECTS OF METFORMIN AS EOC THERAPY ...... 114

Introduction ............................................................................................................ 114

Materials and Methods .......................................................................................... 117

Results .................................................................................................................... 123

Discussion .............................................................................................................. 143

CHAPTER 5: GENERAL DISCUSSION ..................................................................... 149

Overview ................................................................................................................. 149

The metabolic syndrome, diabetes, and EOC ..................................................... 151

Coordinated metabolism in the tumour microenvironment ............................... 154

Intersection of tumour metabolism and systemic metabolism .......................... 155

Glucose transport and diagnostic potential ........................................................ 157

Study Limitations ................................................................................................... 158

Conclusions ........................................................................................................... 160

REFERENCES ............................................................................................................ 161

Appendix I: Supplementary figures ......................................................................... 184

Appendix II: Source of supplies and materials ....................................................... 188

Appendix III: Recipes for solutions ......................................................................... 192

Appendix IV: RT-PCR primer sets............................................................................ 196

Page 9: The effects of hyperglycemia on glucose metabolism in

ix

LIST OF TABLES

Table 1: Prevalence of ovarian tumours by histology ...................................................... 2

Table 2: Frequency of EOC diagnoses by FIGO stage ................................................... 5

Table 3: SGLT protein expression in human ovarian cancer ......................................... 81

Table 4: Cox proportional hazard regression model for ovarian cancer survival ......... 102

Table 5: Median survival times of mice with SGLT2 KD tumours ................................ 103

Page 10: The effects of hyperglycemia on glucose metabolism in

x

LIST OF FIGURES

Figure 1: SGLT secondary active symporters ............................................................... 22

Figure 2: Streptozotocin treatment models Type 1 diabetes ......................................... 41

Figure 3: Type 1 diabetes promotes tumour growth in proportion to blood glucose ...... 42

Figure 4: Chronic hyperglycemia limits ovarian tumour growth ..................................... 44

Figure 5: Acute and chronic hyperglycemia decrease survival in mice with EOC ......... 45

Figure 6: Tumour induction improves glucose tolerance in chronically diabetic mice ... 47

Figure 7: High blood glucose in Type 2 diabetic mice predicts larger tumours .............. 50

Figure 8: Tumours improve insulin sensitivity in Type 2 diabetic mice .......................... 51

Figure 9: EOC cells conditioned to hyperglycemic media accelerate scratch wound

closing .......................................................................................................... 54

Figure 10: Cells conditioned to hyperglycemia are sensitive to glucose deprivation ..... 55

Figure 11: ID8-25 cells, but not ID8-6 cells, increase glucose consumption in

hyperglycemia. ............................................................................................. 56

Figure 12: In high glucose media, cells conditioned to hyperglycemia have lower

metabolic viability than cells conditioned to physiological glucose ............... 57

Figure 13: GLUT and SGLT transporters are expressed in ID8 cells ............................ 79

Figure 14: SGLT2 is overexpressed in human serous ovarian cancer .......................... 80

Figure 15: SGLTs affect glucose transport in hyperglycemia in cells conditioned to

normal physiological glucose concentrations (ID8-6) ................................... 83

Figure 16: SGLTs affect glucose transport in hyperglycemia in cells conditioned to high

glucose concentrations (ID8-25) ................................................................... 84

Figure 17: SGLT inhibition does not affect cell metabolic viability ................................. 86

Page 11: The effects of hyperglycemia on glucose metabolism in

xi

Figure 18: Phlorizin and phloretin inhibit scratch wound healing in ID8-6 cells ............. 88

Figure 19: SGLT glucose transporters limit cell invasion in ID8-6 cells ......................... 90

Figure 20: Knockdown of SGLT2 in ID8 cells changes proliferative behaviour in culture

..................................................................................................................... 92

Figure 21: GLUT1 protein expression is higher the tumours of diabetic mice than in

those of WT mice ......................................................................................... 94

Figure 22: Both GLUT and SGLT transporters are responsible for functional glucose

uptake into tumours ...................................................................................... 96

Figure 23: SGLT2 KD impairs glucose uptake through active transport, but total glucose

uptake is maintained through compensation by GLUTs ............................... 97

Figure 24: Expression of GLUT1 is highly upregulated in tumours with reduced SGLT2

expression at 45 days PTI ............................................................................ 99

Figure 25: SGLT2 knockdown leads to significantly larger primary tumours at death . 101

Figure 26: SGLT2 KD in tumours significantly decreases overall survival................... 102

Figure 27: Secondary disease is much more severe in mice with SGLT2 KD tumours

................................................................................................................... 104

Figure 28: Poor overall survival is associated with severe secondary disease,

regardless of primary tumour weight, in mice bearing SGLT2 KD tumours 105

Figure 29: Cellular effects of metformin ....................................................................... 116

Figure 30: Metformin inhibits EOC cell viability in high glucose .................................. 125

Figure 31: Metformin reduces cell number in glucose-deprived cultures .................... 127

Figure 32: Metformin increases cellular glucose consumption .................................... 129

Page 12: The effects of hyperglycemia on glucose metabolism in

xii

Figure 33: Metformin-mediated increase in glucose consumption is independent of

glucose transporter expression .................................................................. 130

Figure 34: Small tumours normalize blood glucose in diabetic mice ........................... 133

Figure 35: Daily metformin treatment does not affect EOC outcomes in WT mice ...... 135

Figure 36: Metformin treatment in WT mice with tumours impairs glucose tolerance over

time. ........................................................................................................... 136

Figure 37: Daily metformin treatment does not affect EOC outcomes in Type 2 diabetic

mice ............................................................................................................ 139

Figure 38: The presence of a tumour or treatment with metformin both improve glucose

tolerance in diabetic mice ........................................................................... 140

Figure 39: Metformin treatment does not reduce the weight of established tumours .. 142

Figure 40: EOC metabolism is a product of cell genotype, the tumour microenvironment,

and systemic glucose conditions ................................................................ 151

Figure 41: Positive SGLT1 staining on human ovarian tumour tissue ......................... 184

Figure 42: Dapagliflozin treatment does not affect tumour weight ............................... 186

Page 13: The effects of hyperglycemia on glucose metabolism in

xiii

LIST OF ABBREVIATIONS ABAM antibiotic/antimycotic

AGE-RAGE advanced glycation end product receptor complex

AMG 14C-α-methyl-d-glucopyranoside

ANOVA analysis of variance

ATP adenosine triphosphate

BRCA breast cancer

CB cytochalasin B

DAB 3,3’-diaminobenzidine

DAPI 4’,6-diamidino-2-phenylindole

DMEM Dulbecco’s Modified Eagle Medium

DNA deoxyribonucleic acid

EOC epithelial ovarian cancer

ETC electron transport chain

FBG fasting blood glucose

FBS fetal bovine serum

FDG fluoro-deoxyglucose

FIGO International Federation of Gynecology and Obstetrics

G6P glucose-6-phosphate

GLOX glucose oxidase

GLUT facilitative glucose transporter

IPGTT intraperitoneal glucose tolerance test

HIF-1α hypoxia inducible factor alpha

HK hexokinase

HRP horseradish peroxidase

ID8 mouse surface epithelial ovarian cancer cells

IGF insulin-like growth factor

IHC immunohistochemistry

LDH lactate dehydrogenase

KD knockdown

Page 14: The effects of hyperglycemia on glucose metabolism in

xiv

KO genetic knockout

MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

MRI magnetic resonance imaging

OXPHOS oxidative phosphorylation

PBS phosphate buffered saline

PET positron emission tomography

PFK phosphofructokinase

PK pyruvate kinase

PPP pentose phosphate pathway

PT phloretin

PTI post tumour induction

PZ phlorizin

RIPA radioimmunoprecipitation assay

RT-PCR polymerase chain reaction

SDS PAGE sodium dodecyl sulfate polyacrylamide gel electrophoresis

SEER Surveillance Epidemiology and End Results (National Cancer Institute)

SGLT sodium-glucose transporter

STIC serous tubal intraepithelial carcinoma

STZ streptozotocin

T1DM Type one diabetes mellitus

T2DM Type two diabetes mellitus

TBST Tris-buffered saline with tween20

TCA the citric acid cycle

TZD thiazolidinedione

VDAC voltage-dependent anion channel

WB Western blot

WHO World Health Organization

WST-1 water-soluble tetrazolium salt 1

WT wild type

Page 15: The effects of hyperglycemia on glucose metabolism in

1

CHAPTER 1: INTRODUCTION AND LITERATURE REVIEW

Ovarian cancer

Currently there are approximately 17,000 Canadian women living with ovarian cancer.

Each year nearly 2,800 new cases are diagnosed and the disease claims the lives of

1,750 women, making it the most deadly gynecologic cancer and the fifth leading cause

of all cancer deaths in women.1,2 Ovarian cancer most commonly affects post-

menopausal women (median age at diagnosis is 62) and both incidence and mortality

increase with age. In the general population the lifetime risk of developing ovarian

cancer is 1.4% (1 in 71).2 BRCA gene mutation carriers are at a significantly increased

risk (BRCA1: 30-60%, BRCA2: 15-30%)3 and cases in these women appear ten years

earlier on average.4

Histological classifications

Ovarian tumours can have stromal, germinal, or epithelial origins. Epithelial tumours are

by far the most prevalent and make up close to 90% of ovarian cancers. The World

Health Organization (WHO) has identified eight histologic subtypes of epithelial ovarian

cancer (EOC): serous, endometrioid, mucinous, clear cell, transitional cell, squamous

cell, mixed epithelial and undifferentiated.5 Table 1 indicates the prevalence of ovarian

tumours based on histopathology.6

Page 16: The effects of hyperglycemia on glucose metabolism in

2

Table 1: Prevalence of ovarian tumours by histology

Origin* Tumour Malignancy* Histology

Epithelial 60%

Benign 60%

Borderline 10%

Malignant 30% 85-90%

High-grade serous 70%

Low-grade serous <5%

Mucinous 3%

Endometrioid 10%

Clear cell 5-10%

Germ cell 30% Malignant <5%

Sex-cord stromal

8% Malignant <5%

* Remaining percentages of disease have undefined pathologies

It is becoming clear that the histopathological subtypes of ovarian cancer are also

associated with particular mutations.7 One of the most significant advances in the past

decade has been the development of more descriptive classifications of ovarian cancer

subtypes that also include cells of origin and genetic signatures (reviewed in 6).

Screening and prevention

Disease screening and prevention have been extremely challenging in EOC. Up to 90%

of EOCs do not have an identified genetic component and there are no easily

identifiable precursor lesions. Traditionally, the most successful clinical tools have been

Page 17: The effects of hyperglycemia on glucose metabolism in

3

transvaginal ultrasound and identification of elevated levels of the serum marker CA-

125, however neither is sensitive or specific enough for screening.7 In women at high

risk for the disease, primarily BRCA mutation carriers, risk-reducing salpingo-

oophorectomy is the most protective strategy.8

Tumour development

For many years, epithelial ovarian cancer was suspected to arise from the continual

disruption of the ovarian surface epithelium during ovulation. According to this

“incessant ovulation” hypothesis, repeated post-ovulatory follicle repair9–12 and its

associated inflammation13 enhance the possibility of mutagenesis. Continual follicle

repair also increases the frequency of cortical inclusion cysts which are invaginations of

the surface epithelium that expose naïve epithelial cells to the hormone-rich, thus

potentially tumourigenic, ovarian stroma.14 This hypothesis supports many of the clinical

observations related to disease risk factors: in general, circumstances that increase the

number of ovulatory events are associated with increased risk of ovarian cancer, such

as nulliparity, history of pelvic inflammatory disease and endometriosis. Conversely,

fewer ovulatory events are generally associated with decreased risk of the disease,

including oral contraceptive use and multiple pregnancies.15

The understanding of ovarian tumour development has been complicated by the fact

that “epithelial ovarian cancer” has historically encompassed a histologically diverse

group of pelvic tumours.16 As a way to reconcile this diversity of tumours Kurman and

others used an integrated analysis of clinical, pathological and molecular genetics to

Page 18: The effects of hyperglycemia on glucose metabolism in

4

propose a two-pathway model that divides ovarian cancers into Type I, low-grade and

borderline lesions; and Type II, high-grade aggressive tumours.17 Characterization of

these pathways continues to improve as new genomic and microenvironmental data

become available.9

The past decade has seen a substantial paradigm shift in the understanding of the

cellular origins of EOC. Despite the fact that tumours were thought to arise from the

ovarian epithelium, both tubal ligation and hysterectomy were found to provide a 67%

reduction in the risk of developing ovarian cancer.15 In 2001, Piek and others noticed

that the fallopian tubes of women carrying BRCA mutations often had dysplastic regions

and occult cancers18 and shortly after, Medeiros and others implicated the fimbriae of

the fallopian tubes as sites development of early serous tumours.19 In one study 71% of

cases diagnosed as “serous ovarian carcinoma” showed lesions in the tubal epithelium

known as serous tubal intraepithelial carcinomas (STICs).20 It is now accepted that

many serous tumours originally considered as ovarian in origin actually arise from the

fimbriated ends of the fallopian tubes.21 Current mouse models provide direct evidence

that metastatic serous “ovarian” cancer can develop from primary fallopian tube tumours

that grow to the engulf the ovary.22

Diagnosis and clinical staging

Diagnosis of ovarian cancer is complicated by the fact that its symptoms are ambiguous

and often resemble gastrointestinal problems.23,24 The most significant barrier to

improving ovarian cancer outcomes is the inability to effectively identify the disease in

Page 19: The effects of hyperglycemia on glucose metabolism in

5

its early stages. The five-year survival rate drops precipitously once the disease has

spread beyond the ovary, yet the majority of cases are not detected until after this point.

At diagnosis, EOC is most commonly characterized as Stage III or IV using the

International Federation of Gynecology and Obstetrics (FIGO) staging system (Table

2).25 The most current FIGO staging criteria reflect the importance of histological

type/grading at the time of staging and the identification of STICs at the primary site.26

Table 2: Frequency of EOC diagnoses by FIGO stage

FIGO Stage Description Percent

at diagnosis* 5-year

survival

I Confined to ovary/fallopian tube and peritoneal fluid

15% 90%

II Extension/metastasis to other pelvic organs or peritoneum

19% 70%

III Spread along both pelvic and abdominal peritoneal surfaces 60%

39%

IV Distant metastases 17%

* Remaining percentages of disease have undefined stages

The spread of EOC is facilitated by the circulation of peritoneal fluid that carries

particularly aggressive cells from the primary tumour to other pelvic and abdominal

surfaces.10,27 Close to 40% of EOC patients present with malignant abdominal ascites

that plays a critical part in chemoresistant and recurrent disease.28

Page 20: The effects of hyperglycemia on glucose metabolism in

6

Treatment and disease management

Despite the fact that EOCs are so histologically diverse, the same treatment regime is

generally used on tumours of all subtypes: cytoreductive surgery followed by several

cycles of carboplatin and paclitaxel chemotherapy.29 The goal of the initial surgery is

histopathological diagnosis, staging, and maximum removal of tumour tissue.29

Minimum residual disease is associated with longer survival30 and it has been

suggested that a low volume of disease after surgery may be a more important

prognostic factor than an initial low stage.17,31 Recommended surgery may include total

hysterectomy, bilateral salpingo-oophorectomy, tumour debulking, and omentectomy.32

Because ovarian cancer usually presents as peritoneal disease, surgery often examines

structures adjacent to the ovaries including the fallopian tubes.33 Following surgery and

chemotherapy, the disease is monitored using medical imaging and/or serum markers

such as CA-125.7 Most women who succumb to the disease die of malignant bowel

obstruction from widespread metastatic disease and limited mortality is associated with

the primary ovarian tumour.29

Current challenges

Despite advances in treatment over the past forty years, death rates from ovarian

cancer have not changed appreciably between 2001 and 2012.25 In addition to late

detection and disease spread within the peritoneal cavity, treatment challenges include

drug resistance and cancer recurrence even after initial response to treatment. On

average, advanced disease recurs within 18 months of treatment.29 Patients are given

platinum-based chemotherapies until their tumours become resistant, at which point

Page 21: The effects of hyperglycemia on glucose metabolism in

7

doxorubicin or topotecan is recommended.34 The median survival of patients with

platinum-resistant, recurrent disease is about three years, and just one year from the

onset of platinum resistance.35 According to a recent international consortium, improving

patient outcomes will come from using distinct histological and genetic subtypes to:

stratify clinical trial design; identify new therapeutic targets; develop better experimental

models; and design tools for prevention and early detection.36

Glucose metabolism

In order to survive and replicate, the cells in ovarian tumours rely on the oxidation of

glucose as their primary source of ATP energy, using metabolic processes that can

occur both aerobically (oxidative phosphorylation, oxphos) and anaerobically (glucose

fermentation).

Glycolysis

The anaerobic process of glycolysis in the cell cytoplasm initiates both oxphos and

fermentation by metabolising a glucose molecule into two 3-carbon pyruvate molecules

and two molecules of ATP. The net result of glycolysis can be represented as the

following equation:

Glucose + 2 NAD+ + 2 ADP + 2 Pi 2 NADH + 2 pyruvate + 2 ATP + 2 H2O + 4 H+

The rate of pyruvate production is controlled at the levels of the regulatory enzymes

hexokinase (HK), phosphofructokinase (PFK), and pyruvate kinase (PK) in reactions

that are virtually irreversible and are allosterically regulated by their products. While the

Page 22: The effects of hyperglycemia on glucose metabolism in

8

efficiency of ATP formation in glycolysis is only about 3%, the glycolytic reactions also

supply the cell with various anabolic precursors. For example, in addition to continuing

through the glycolytic cycle the first product of glycolysis, glucose-6-phosphate (G6P),

can be further metabolised by G6P dehydrogenase to enter the pentose phosphate

pathway (PPP) and synthesize ribose-5-phosphate required for nucleotide

biosynthesis.37,38

Cellular Respiration

The citric acid cycle

When oxygen is present, pyruvate (the end product of glycolysis) is converted to acetyl

coenzyme A which is further metabolized through the enzymes of the citric acid cycle

(TCA), a series of reactions that take place in the mitochondria.

The net reaction of the TCA can be summarized as:

2 acetyl-CoA + 6 H2O + 2 ADP 4 CO2 + 16 H+ + 2 CoA + 2 ATP + 2 FADH2 + 6 NADH

While the TCA itself only produces two molecules of ATP, there are four dehydrogenase

enzymes in the cycle that reduce the electron carriers NAD+ to NADH and FAD+ to

FADH2. For every two electrons subsequently released from these carriers, up to three

molecules of ATP can be synthesized through the electron transport chain (ETC).

The electron transport chain and oxidative phosphorylation

Oxidative phosphorylation (oxphos) refers to the generation of ATP energy in the

mitochondria through the electron transport chain and the ATP synthetase enzyme. The

Page 23: The effects of hyperglycemia on glucose metabolism in

9

ETC is a series of four protein complexes located in the mitochondrial membrane,

where each consecutive protein has a higher affinity for electrons. As electrons are

released from the carriers NADH and FADH2 and move down the chain, the resulting

release of energy is used to create a proton gradient across the mitochondrial

membranes. When H+ ions are pumped from the inner mitochondrion to the outer

chamber of mitochondrion, they create a strong negative electrical potential in inner

mitochondrial matrix. Hydrogen atoms flow with this concentration gradient through ATP

synthetase to form ATP from ADP. The newly synthesized ATP diffuses through the

mitochondrial membranes to cytoplasm. In aerobic conditions, cells can produce up to

36 molecules of ATP per molecule of glucose from glycolysis and cellular respiration.

Anaerobic glycolysis (fermentation)

In the absence of oxygen, cells undergo lactic acid fermentation to regenerate the NAD+

required for glycolysis.

Glucose2 lactate + 2 H+

When oxygen becomes available again, this reaction is reversed and pyruvate + NADH

+ H+ are regenerated.

Glucose metabolism in cancer

Normal cell metabolism is under the influence of the “Pasteur effect” where oxygen

inhibits glycolysis through phosphofructokinase (PFK) regulation to facilitate the

mitochondrial oxidation of glucose. However, glucose metabolism in cancer cells does

Page 24: The effects of hyperglycemia on glucose metabolism in

10

not adhere to these same patterns of regulation. In 1924 the German researcher Otto

Warburg made a seminal observation of cancer cell metabolism: tumours produce

significantly more lactate than normal tissue, indicating an abnormally active glycolytic

pathway.39 The glycolytic rate in tumour cells can be more than 30 times higher than in

normal cells.40 Curiously, Warburg’s tumours appeared to have uncoupled glycolysis

from oxygen levels to maintain their highly glycolytic phenotype even in the presence of

adequate oxygen, thus demonstrating “aerobic glycolysis.”41 Although not all cancerous

cells have the same abnormalities in metabolism,42 the Warburg phenotype is so well

preserved across diverse malignancies that it is now considered an independent

hallmark of cancer.43

The Warburg Effect

Understanding the significance of the Warburg phenotype remains a challenge and it is

still not clear whether the effect is a “cause, correlate or facilitator” of a progressing

malignancy.44 Warburg originally postulated that the abnormal metabolism of cancer

cells is a result of defective mitochondria, writing that “the respiration of all cancer cells

is damaged”41 – despite the fact that many investigators, including Warburg himself,

found that cancer tissues maintain normal levels of respiration in addition to generating

high levels of lactic acid.39,45–47 Warburg believed that impaired respiration was the

ultimate cause of cancer, thus only cancer cells would show this property, even though

significant evidence showed that aerobic glycolysis is common to many rapidly

proliferating cell populations.48 Rather than damage to respiration, the Warburg effect

more accurately reflects an impairment of glycolytic control.49

Page 25: The effects of hyperglycemia on glucose metabolism in

11

The diversity of factors associated with the development of the Warburg effect suggests

that there is no single cause or consequence of abnormal metabolism in tumours.

Parallel, reciprocal changes to genetic and environmental factors drive the evolution of

the glycolytic switch.50,51 The structure and function of cancer cell mitochondria, the

hypoxic nature of tumours, lactate-driven acidification of the tumour microenvironment,

the capacity for metabolic turnover, production of biosynthetic intermediates, primary

genetic changes, and the interstitial fluid pressure of tumours are all associated with the

metabolic profile of cancer.

Mitochondria

Both structural and functional impairment of mitochondria are significant factors in the

altered metabolism of cancer cells.52,53 Although damages to cellular respiration are not

as complete or as universal as Warburg predicted, there is evidence that mitochondria

in some highly glycolytic tumour cells have an impaired ability to transport electrons

from pyruvate to molecular oxygen.54,55 While the mitochondria are not inactive, they

may act at a low capacity56 and cancerous cells often have fewer organelles and

reduced mitochondrial DNA (mtDNA) content.57 There are mutations and copy number

changes to the mtDNA of many cancers53 in addition to nuclear DNA mutations in

proteins involved in respiration.49 The function of TCA proteins located in the

mitochondria including succinate dehydrogenase, fumarate dehydrogenase, and

isocitrate dehydrogenase are also impaired in some cancers.49,58–60 Furthermore,

inhibition of the respiratory enzyme citrate synthase can activate an epithelial-

mesenchymal transition that increases metastasis and proliferation.61 Taken together,

Page 26: The effects of hyperglycemia on glucose metabolism in

12

mitochondrial dysfunction leads to a more highly glycolytic phenotype that has been

directly linked to tumourigenicity, while increasing oxidative phosphorylation can reduce

the growth of cancer in vivo and in vitro.62

Hypoxia

The Warburg Effect is often thought to arise as an adaptation to environmental

pressures. In this case, cancer cells are not believed to be inherently glycolytic, but

rather that the phenotype of aerobic glycolysis is secondary to intermittent hypoxia.63,64

Oxygen levels in the tumour microenvironment are significantly reduced as a result of

pathologic tumour vasculature. Solid tumours inefficiently and chaotically develop new

blood vessels to support their growing mass and consequently develop large areas of

poorly perfused tissue.43 In these areas a steep oxygen gradient develops between the

vasculature and the diffusion limit of oxygen, exposing many tumour cells to hypoxic

conditions.65 A cell with an increased capacity for glycolytic metabolism would have a

survival advantage in these transient hypoxic conditions. In support of this hypothesis,

short-term hypoxia has been found to lead to the irreversible oncogenic transformation

of embryonic mouse cells by promoting aerobic glycolysis that remains and supports

growth even once normal oxygen pressure is restored.41

The cellular response to low oxygen is mediated primarily through the hypoxia-inducible

factor (HIF) signaling cascade which regulates processes including glucose metabolism,

angiogenesis, apoptosis, proliferation and survival and pH regulation. Gregg Semenza’s

group in particular has made significant contributions to the understanding of HIF’s

Page 27: The effects of hyperglycemia on glucose metabolism in

13

effects on metabolism.66 Oncogenic stabilization of HIF1α induces the glycolytic

proteins HK2, PFK-1, aldolase, enolase, and PKM2.40,66 Thus, hypoxia leads to the

Warburg phenotype because glycolytic products build up and exceed the capacity of

aerobic respiration, the production of lactate acid increases as Warburg observed.

Tumour acidification

A number of studies have shown that the lactate produced by increased glycolysis

contributes to the aggressiveness of tumours. In order to prevent cellular acidification,

lactate is shuttled out of the cell leading to the acidification of the tumour

microenvironment.40 Acidity has been shown to promote proliferation,63 signal67 and

facilitate68 tumour angiogenesis, suppress the immune response to a developing

tumour,69 and increase mutation rate70,71 and resistance to chemotherapy.72 The pH of

tumour tissue is around 0.5 units lower than normal, so H+ ions from the tumour diffuse

and cause damage to surrounding normal tissue, thus facilitating invasion.73–75

Mathematical modeling shows that tumour acid production alone can explain both

benign (a result of autotoxicity) and invasive tumour growth76, though the Warburg effect

is not necessarily associated with metastatic potential.77

Speed of metabolism

Cancer cells produce about 10% more total ATP than normal cells to support their rapid

proliferation.49 When tissues are rapidly consuming energy, most of it is regenerated by

the high turnover of anaerobic glycolysis.38 Although this is a relatively inefficient

Page 28: The effects of hyperglycemia on glucose metabolism in

14

process, if the cell is not limited by glucose availability aerobic glycolysis can supply

ATP up to 100 times faster than oxphos.40 The production of lactate is necessary to

sustain a high rate of glycolysis. The coenzyme NAD+ is required for the GAPDH

reaction in the fifth glycolytic reaction. In oxphos NAD+ is regenerated by the electron

transport chain, a process that is too slow to sustain cancer cell metabolism. As a

consequence, tumour cells must regenerate NAD+ by converting pyruvate into lactate

through lactate dehydrogenase (LDH).

Metabolic intermediates

In order to proliferate, the cell not only requires ATP energy, but also sufficient

macromolecule biosynthesis and maintenance of redox balance.51 High rates of

glycolysis have been shown to increase the production of biosynthetic building blocks

including proteins, phospholipids, fat and nucleic acids.78,79 The ability of cells to

produce these precursors allows them to become autonomous for their ability to take up

nutrients, and thus reduce their dependence on the extracellular environment. Pyruvate

kinase activity in glycolysis appears to be central to the production of anabolic

precursors. Blocking the PKM2 isoform in cancer cells from converting

phosphoenolpyruvate to pyruvate causes accumulation of upstream products which are

then available substrates for the PPP and thus cell precursors for biosynthesis.80 Other

secreted molecules from glucose metabolism, like lactate and alanine, can be used as

precursors for hepatic gluconeogenesis which may provide further fuel for the tumour.81

Page 29: The effects of hyperglycemia on glucose metabolism in

15

Primary genetic changes

While the Warburg effect may arise secondary to factors in the tumour

microenvironment, there is also substantial evidence to suggest that it is a result of

mutations in non-metabolic tumour suppressors or oncogenes.50,82,83 As early as the

1960s, it was noticed that the treatment of cells with an oncolytic virus increases

glucose transport beyond what is expected for rapid growth alone, suggesting a switch

to a less efficient metabolic process.84,85 More recently, a study showed that a glycolytic

phenotype emerged from progressive transformation of primary human fibroblast cells

with different oncogenes.86 The p53 tumour suppressor, which is mutated in 60-80% of

all EOC cases,3 stimulates respiration and suppresses glycolysis. Loss of p53 function

induces a Warburg phenotype through elevated activity of the glycolytic enzyme

phosphofructokinase49 and enhanced flux through the pentose phosphate pathway.87,88

Several papers have implicated the Akt kinase pathway in cancer cells as the main

driver of Warburg effects as Akt oncogenes activate HK2 and PFK1/2, and promote

GLUT recruitment to the plasma membrane.89,90 Expression of the M2 fetal isoform of

pyruvate kinase (PK) rather than the PKM1 splice variant also plays an important role in

the glycolytic switch and results in tumorigenesis.91 Most of the research on cancer cell

metabolism and the Warburg effect has been done in solid tumours where the

microenvironment is thought to largely mediate the Warburg effect. However, the effect

also appears in hematologic malignancies: acute leukemia is dependent on glycolysis

for survival and abnormally high glucose uptake visualized by FdG-PET is used to

detect and stage Hodgkin’s disease, non-Hodgkin’s lymphomas and multiple

myeloma.92 Because the “metabolic switch” can occur independently of tumour stromal

Page 30: The effects of hyperglycemia on glucose metabolism in

16

tissue, it may be an inherent property of malignant cells rather than exclusively a

consequence of the microenvironment.

Hexokinase 2 (HK2)

Hexokinase 2 catalyses the formation of glucose to G6P, the first step of metabolism

through both PPP and glycolysis. HK2 has emerged as an essential mediator of the

Warburg Effect and, as the slowest step in the glycolytic pathway in some cancers,93 it

may be the most important step in determining glycolytic flux.94–96 HK2 expression has

been correlated with poor overall survival and its inhibition can restore oxidative

metabolism and reduce proliferation.97 The Pederson group has contributed much of the

research on the role of HK2 in cancer metabolism and describe hexokinase 2 as a

“facilitator and gatekeeper of malignancy.” 98,99 HK2 can translocate between the

mitochondria and cytosol in response to glucose. When it is bound to the mitochondrial

membrane by the voltage dependent anion channel (VDAC), it promotes glycolysis by

escaping allosteric inhibition by G6P.100 Pederson has proposed that the Warburg effect

results from overexpressed HK2, VDAC binding HK2 to the mitochondria, and

upregulation of the HK2 gene.99

High interstitial fluid pressure

Tumours have been described as “biological pressure pumps” due to their high

interstitial fluid pressure and microvascular pressure.101,102 This characteristic provides a

major challenge to the delivery of therapeutic agents and immune cells, while

Page 31: The effects of hyperglycemia on glucose metabolism in

17

simultaneously helping to propel potentially metastatic cells out of the tumour.103 The

Van’t Hoff equation predicts that CO2 from the aerobic metabolism of glucose lowers

intracellular osmotic pressure, while lactate produced from glycolysis increases

intracellular osmotic pressure. A “Warburg ratio” (lactate:CO2) of greater than 1.0

indicates that there is relatively more glycolysis than oxphos, and osmotic pressure will

rise, driving outward currents of interstitial fluid.103 Therefore the high interstitial fluid

pressure common to solid tumours may be a direct consequence of tumour metabolism.

Metabolic flexibility

Although tumours have been described as “glucose addicted” they are in fact quite

flexible in their fuel use and metabolize amino acids, fatty acids,104 lactate,51 ketones105

and particularly glutamine.106,107 Moreover, they are not committed to glycolysis. The

Warburg effect is often misunderstood as a preference for glycolysis over respiration

despite the fact that even in the most aggressive tumour cells tested, Warburg found

almost an equal contribution of fermentation and respiration.41 In some tumour cells

mitochondria are responsible for most of the ATP production.108 This flexibility means

that for cancer cells to “starve” both respiration and fermentation must be compromised.

In normal cells, impairing only one pathway is sufficient to kill.109 Cancer cells display

different balances of glycolysis, oxidative phosphorylation and glutaminolysis depending

on glucose availability and other environmental conditions.110–113

Page 32: The effects of hyperglycemia on glucose metabolism in

18

Contributions of the tumour microenvironment

In solid tumours, intercellular interactions are crucial for cancer progression. Stromal

cells including fibroblasts, immune cells and vascular endothelial cells make up the

heterogeneous tumour tissue and contribute to tumour growth and resistance to

therapeutics.114 Intratumoral heterogeneity of metabolism is emerging as an essential

determinant of carcinogenesis.115 Michael Lisanti’s group has described a “reverse

Warburg effect” in which cancer-associated fibroblasts (CAFs) are metabolically

reprogrammed to perform aerobic glycolysis. This transformation allows the fibroblasts

to enrich the microenvironment with lactate, pyruvate and ketone bodies which epithelial

cancer cells then take up and metabolise through oxidative phosphorylation to

synthesize high volumes of ATP.105,116,117 To facilitate this, cancer cells also induce

increased expression of the H+-coupled lactate export protein monocarboxylate

transporter 4 (MCT4) in stromal cells.117,118 Others have shown that a metabolic

symbiosis exists between cancer cells in oxygenated and hypoxic environments within

the tumour. The expression of the MCT1 importer in oxidative cancer cells allows them

to use lactate exported by glycolytic cancer cells as a primary fuel.119,120

The Crabtree Effect

Herbert Crabtree was a contemporary of Warburg who described a local, reversible

adaptation in tumours where high levels of glucose suppress oxygen consumption and

respiration and accelerate glycolysis. Crabtree confirmed Warburg’s observation that

tumours showed abnormally high levels of glycolysis and oxygen is ineffective at

Page 33: The effects of hyperglycemia on glucose metabolism in

19

reducing glycolysis. However, he also observed that glycolytic activity in vitro limited

respiration on average 12% in glucose vs non-glucose solution.121 Recent studies in

Barrett’s esophagus confirm that the Crabtree effect is important to tumour development

and is related advancement of pre-malignant lesions overt cancer.122

Glucose transport

A primary consequence of the Warburg Effect is that cells have abnormally high glucose

requirements. Glucose is a large, hydrophilic molecule that requires specific

transmembrane proteins to cross the cell’s lipid bilayer, and so the expression of

glucose transporters is a key mediator of aerobic glycolysis.123 There are two classes of

transporters: facilitative glucose transporters (GLUTs) and sodium/glucose

cotransporters (SGLTs).

GLUTs

The thirteen identified mammalian GLUT isoforms are grouped into three classes based

on sequence similarities. Class I GLUTS (1,2,3,4) have been well characterized and are

the most important in human disease.123 The GLUT proteins rely on a downhill

concentration gradient to move glucose and other hexoses across the lipid bilayer. The

magnitude and direction of glucose transport is dependent on both the number of

glucose transporters and the steepness of the gradient.123 The expression of transporter

isoforms is highly tissue specific. GLUT1 is expressed in erythrocytes but is also widely

distributed in tissues throughout the body. GLUT2 is specific to the liver, pancreas and

Page 34: The effects of hyperglycemia on glucose metabolism in

20

small intestine; GLUT3 to the brain; and GLUT4 to insulin-responsive tissues including

skeletal muscle, cardiac and adipose tissue.

GLUT expression is integral to many metabolic and energy-sensing pathways including

HIF, c-myc, Akt and AMPK. A pool of GLUTs is stored in vesicles in the cytoplasm and

are translocated to the cell membrane as needed, most notably through insulin signaling

pathways. GLUT transport works through a model of alternate conformation. Glucose

binds to sites either on inside or outside of cell and this binding changes the protein

conformation to open to the other side of membrane.38

GLUTs in cancer. GLUTs are essential to cancer metabolism (reviewed in 124) and their

function is well documented in the literature. Experiments that oncogenically transform

fibroblasts show that oncogenic transformation itself may trigger GLUT1 activity,40 one

of the main determinants of the Warburg effect.125

GLUT1 is highly expressed in ovarian cancer, where tumour status (benign, borderline,

or malignant) is correlated with the level of GLUT1 expression.126,127 Almost all invasive

epithelial carcinomas are positive for GLUT1, independent of stage, grade, or

histological subtype.126,128 Expression of GLUT-1 correlates with tumour proliferation

and microvessel density in EOC. In addition, patients with rapidly proliferating advanced

stage tumours overexpressing GLUT-1 have a lesser chance for optimal

cytoreduction.127 Antibodies to GLUT1 decrease proliferation, induce apoptosis in

nonsmall cell lung cancer and breast cancer cell lines, and appear to synergize with a

Page 35: The effects of hyperglycemia on glucose metabolism in

21

number of chemotherapeutics to enhance their apoptotic effects.79 Likewise, antisense

GLUT1 mRNA inhibits tumour growth.129

SGLTs

The sodium/glucose cotransporters work against the glucose concentration gradient

and are required to establish and maintain higher intracellular glucose

concentrations.130 The main actions of the SGLTs are in glucose reabsorption from the

proximal tubule of the kidney (primarily SGLT2) and the brush border of the small

intestine (primarily SGLT1). In the enterocyte, after being pumped into the cell, glucose

flows with its concentration gradient through GLUT2 on the basolateral side of the

enterocyte, thus moving glucose back into the blood stream (Figure 1).130 SGLTs are

also expressed in the heart, brain, liver, thyroid and skeletal muscle.131

Page 36: The effects of hyperglycemia on glucose metabolism in

22

Figure 1: SGLT secondary active symporters

SGLT in pathology. While there is a large body of research on the role of GLUTs in

cancer, the contributions of the second class of glucose transporters is virtually

unknown. Most of the research on SGLTs comes from outside of the cancer literature.

In the last decade, the action of SGLT2 has garnered attention as a therapeutic target in

Type II diabetes (reviewed in 132) as it increases renal glucosuria and adds to the effects

of other antidiabetic drugs to improve glucose tolerance.133 SGLTs also play essential

roles in mediating ischemic stress in the brain,134 protecting against the LPS-induced

Page 37: The effects of hyperglycemia on glucose metabolism in

23

apoptosis implicated in inflammatory bowel diseases (IBD) and bacterial enteritis,135 and

defending against cardiac disease.136

Isoforms and structure. SGLTs belong to the 12-member SLC5 gene family of hexose

transporters. SGLT1 and SGLT2 import sodium and glucose with 2:1 and 1:1

stoichiometry respectively and differ in the substrates they transport. SGLT1 is a high

affinity transporter that moves both glucose and galactose while SGLT2, specific for

glucose, transports at a high capacity but has a glucose affinity that is about four times

lower. SGLT3 appears to act as a glucose sensor rather than a functional transporter;

glucose binds similarly and produces an inward current of Na+, but no glucose

molecules are transported across the cell membrane.137–140

Regulation. SGLT1 upregulation is mediated by the cAMP-PKA signaling pathway and

takes several days to act.141 Over minutes, activation of cAMP pathway results in

recruitment of an intracellular reserve pool of transporters to the plasma membrane

where the number of cotransporters in the plasma membrane regulated by endo- and

exo- cytosis of vesicles.130 In cardiac tissue, SGLT1 expression and function are

regulated by insulin and leptin.136 SGLT expression and translocation to the cell

membrane is also regulated through AMPK activation.142–144 Protein-protein interaction

with extracellular domain of EGFR stabilizes SGLT-1 to prevent proteasomal

degradation.145 Because SGLT1 expression is dependent on EGFR expression, it has

been suggested that EGFR has a role in maintaining basal intracellular glucose level by

this mechanism.40,145

Page 38: The effects of hyperglycemia on glucose metabolism in

24

Mechanism of action. Using the electrochemical gradient maintained by the Na+/K+

ATPase pump, SGLTs are secondary active symporters that transport glucose into the

cell coupled with Na+. Glucose/sodium symport is followed by anions and water to

maintain electroneutral isotonic fluid absorption.146 In addition to Na+ driven cotransport,

glucose can enter the cell with protons although the affinity for sugar becomes orders of

magnitude lower. The SGLT protein transports with a 6-step model of internalization.

External Na+ binds, increasing transporter’s affinity for glucose. Glucose then binds,

creating a conformational change that expose Na+ and glucose to the inside of the cell.

Glucose and then sodium are released and the protein returns to its original

conformation. Transport is reversible depending on the concentration gradient of Na+,

but it is asymmetrical and favours the internalization of glucose.146

Actions other than glucose transport

SGLTs can act as uniporters to move sodium into the cell in the absence of glucose131

or glucose transport.147 In certain protein conformations, SGLT1 is also a water

cotransporter with an osmotic permeability similar to AQP0130,148,149 which has

therapeutic applications in oral rehydration therapy.150 These glucose-independent roles

may contribute to cancer cell function, particularly in maintaining osmotic balance and

cell membrane potential, both of which are altered in cancer cells.151

Positron emission tomography (PET)

Both glucose metabolism and transporter expression in cancer cells are clinically useful.

Positron emission tomography (PET) is an imaging technique that has become standard

Page 39: The effects of hyperglycemia on glucose metabolism in

25

in cancer diagnostics. PET takes advantage of the cancer cell’s metabolic requirements

by visualizing tumours based on their abnormally high uptake of glucose which may be

20-30 times higher than in normal cells.40 However, this imaging is not consistently

useful across a wide variety of tumours. In current practice only glucose taken up

through the GLUT transport channels is detectable130, suggesting that there is potential

to increase the strength of this imaging if SGLT-mediated transport is also visualized.

Principle

Patients are fasted overnight and given an intravenous injection of the radiolabelled

glucose analogue 18F-2-deoxy-D-glucose (FDG). FDG is taken up into cells through

GLUT transporters with similar kinetics as glucose and is phosphorylated by hexokinase

(HK) to FDG-6-phosphate, but no further.152 This property allows its accumulation in

tissue as a function of time and can be used to calculate transport and HK activity.130

Gamma rays emitted from the fluorine molecule are used to visualize the accumulation

of glucose in tissues on a PET scan.153 Tissues with high glucose transport and

hexokinase activity, like tumour cells, appear brighter on the scan. This technique is

usually combined with other imaging techniques like computerized tomography (CT) or

magnetic resonance imaging (MRI), to achieve better spatial resolution that improves

the power and utility of the technique.149,154 FDG-PET can be used as a quantitative

measurement using standardized uptake value (SUV) or ratio (SUR; uptake per dose).

Limitations

1. Tumour biology. Not all tumours have the same level of metabolic activity and up to

30% of tumours are FDG-PET-negative.155 Even within tumours up to a quarter of

Page 40: The effects of hyperglycemia on glucose metabolism in

26

FDG in a tumour can be from non-tumour tissue.156 Furthermore, while PET is good

at differentiating between live and dead cells, it cannot distinguish live cells with

lowered metabolism (quiescent cells).155 The technique requires that FDG

accumulates in tissues, so it is not useful in malignancies in tissues where de-

phosphorylation of FDG-6-phosphate allows glucose to diffuse out of cell.157 FDG

uptake is also at least partially dependent on blood flow which is often severely

compromised in tumours.158 In addition, the phase of menstrual cycle also affects

level of uptake in ovaries.159

2. Signal interference. Although patients are fasted to decrease circulating insulin

before a scan, insulin levels can affect FDG distribution, as can insulin-sensitizing

drugs such metformin.160 Chemotherapy or its associated inflammation can result in

false positive FDG uptake.161,162 Hyperglycemia can affect FDG uptake as normal

glucose provides greater competition for labelled glucose to enter the tumour.163 The

timing of imaging can have an effect as tissues have varying FDG uptake on

delayed images which can alter background levels of uptake.164 High glucose uptake

in pelvic organs, particularly the bladder, can obscure tumour metabolism in these

tissues.159

3. Sensitivity and resolution. The resolution of PET, even when combined with other

imaging modalities like CT, is poor below 0.8 cm3 and thus too low to detect early

tumours.165 It is especially poor at finding new, previously unknown metastases.166

Page 41: The effects of hyperglycemia on glucose metabolism in

27

There are numerous causes of false-negatives and false-positives that can make it

difficult to interpret scans.167

Use in gynecological cancers

Despite these limitations, FDG uptake is increased in both ovarian cell lines168 and

ovarian tumours,169 and FDG-PET reportedly has high specificity for ovarian lesions

larger than 5mm.159 Tumour morphology, background activity, limited spatial resolution

have been implicated where FDG-PET has performed poorly in clinical use.168

A 2009 report from Cancer Care Ontario concluded that PET adds limited value to

diagnosis of pelvic masses and does not recommend its use in the diagnosis of ovarian

cancer170,171, though in some cases the SUVmax of FDG-PET/CT can be useful for

differentiating benign or borderline from malignant tumours.170,172 FDG-PET can help

clinicians decide for or against surgery based on whether there is a localized mass or

widespread disease171 and it can help reduce the need for second-look surgeries

following first-line treatment.161 It is most useful in monitoring the response to treatment,

residual/refractory disease, and recurrence.161,166,173,174 In combination with rising serum

CA-125, FDG-PET/CT can also be sensitive and specific for early diagnosis of relapse

in otherwise asymptomatic patients.175

Page 42: The effects of hyperglycemia on glucose metabolism in

28

RATIONALE

Most tumours undergo a “glycolytic shift” as they become more aggressive. In contrast

to glucose metabolism through oxidative phosphorylation, glycolysis provides the cell

with rapid but inefficient ATP production. As a result, tumours consume exponentially

more glucose than normal tissue. Since circulating glucose is the primary source of fuel

for transformed cells, systemic metabolic control may be an important mediator of

epithelial ovarian cancer (EOC). As the prevalence of diabetes continues to rise, it will

be important to understand the effects of long-term comorbidities like hyperglycemia.

Hypothesis and objectives

We hypothesize that chronic hyperglycemia will accelerate the growth of ovarian

tumours by providing an abundance of fuel needed for rapid proliferation, and that

tumour metabolism may be mitigated by normalization of systemic blood glucose. The

following objectives were designed to test this hypothesis:

Objective 1. Determine the effects of hyperglycemia on EOC metabolism, growth and

survival in vitro and in a diabetic mouse model of EOC.

Objective 2. Examine the mechanisms of glucose uptake in physiologic and

hyperglycemic environments.

Objective 3. Assess the treatment potential of systemic glucose control to prolong

disease-free survival in vivo.

Page 43: The effects of hyperglycemia on glucose metabolism in

29

CHAPTER 2: SYSTEMIC HYPERGLYCEMIA AND EOC PROGRESSION

Introduction

Epithelial ovarian cancer (EOC) is the deadliest gynecologic cancer2 and despite

advances in treatment over the past thirty years, the overall survival rate remains below

40%.9 In the majority of cases, a diagnosis is not made until after the disease has

spread beyond the ovary and is much more challenging to treat. Although first-line

surgery and platinum-based chemotherapy can be effective, most late-stage cancers

recur within 16 to 18 months.29,176 There are limited options for second-line therapy and

most cancers will eventually become chemo-resistant.7

In addition to these challenges, comorbid diabetes affects 8-18% of cancer patients.177

Women with type 2 diabetes (T2DM) are at an increased risk of developing ovarian

cancer178 and EOC patients with pre-existing diabetes have a much poorer overall

survival rate than non-diabetic patients.179–182 T2DM is characterized by hyperglycemia

and hyperinsulinemia and is often related to obesity, which carries its own risk factors

including high body mass index (BMI), sex steroid metabolism, and inflammation.183,184

Despite substantial epidemiological evidence, the underlying mechanisms of the

cancer-diabetes association are still poorly understood.

Insulin increases the circulating, local and bioavailable growth factors including insulin-

like growth factor 1, leading many to believe that hyperinsulinemia is the primary

influence on cancer risk and progression in type 2 diabetics.181,185 The role of

Page 44: The effects of hyperglycemia on glucose metabolism in

30

hyperglycemia has been largely overlooked, despite its significant pro-tumorigenic

potential.186

Like most malignancies, EOC cells metabolize considerably more glucose than their

normal counterparts.187 Abnormally elevated glycolytic activity, even in aerobic

conditions, is characteristic of the glucose metabolism in most cancer cells,

accompanied by the conversion of pyruvate to lactate in large amounts (the Warburg

Effect).41,43 Evidence suggests that the quantity of glucose required to maintain this

metabolic phenotype can exceed the normal glucose content of blood.188 The rate of

glucose uptake also increases with increasing extracellular glucose,123 suggesting that

its supply is a rate-limiting factor for metabolism in cancerous cells.189

Increased glucose uptake has been shown to coincide with the transition from pre-

malignant lesions to invasive cancer and has been linked with overall tumour

aggressiveness.63,190 We hypothesized that the hyperglycemic conditions of diabetes

can remove growth restrictions imposed by limited glucose availability and lead to more

aggressive progression of epithelial ovarian cancer. In this study we investigated the in

vivo and in vitro effects of hyperglycemia on epithelial ovarian cancer cell growth.

Page 45: The effects of hyperglycemia on glucose metabolism in

31

Materials and Methods

Animals

Wildtype (WT) C57bl/6 mice were purchased from Charles River Laboratories

(Wilmington, MA, USA), and Akt2 null C57bl/6 mice (https://www.jax.org/strain/006966)

were purchased from Jackson Laboratories (Bar Harbour, ME, USA). An Akt2 null

mouse colony was established from breeder mice and animals were genotyped prior to

beginning experiments. Animals were housed at the Central Animal Facility at the

University of Guelph and all experiments were conducted according to Canadian

Council on Animal Care guidelines. Mice were kept on a 12h light:dark cycle and had

free access to food and water. For all experiments, adult mice were between 12 and 24

weeks old at the time of surgery.

Mouse model of epithelial ovarian cancer

Spontaneously transformed murine epithelial cells from C57Bl/6 mice (ID8) were diluted

in 5µl phosphate buffered saline (PBS) and injected under the ovarian bursa of WT or

Akt2 null mice. This orthotopic, syngeneic mouse model of EOC closely replicates

ovarian serous adenocarcinoma in women. By 90 days post-tumour induction, mice

develop large primary tumours, secondary peritoneal lesions, and significant abdominal

ascites.10 In survival experiments, mice were sacrificed when they became moribund

due to the accumulation of ascites fluid.

Page 46: The effects of hyperglycemia on glucose metabolism in

32

Type 1 Diabetes Mellitus (T1DM)

Streptozotocin ([2-deoxy-2-(3-(methyl-3-nitrosoureido)-D- glucopyranose]; STZ) is a

pancreatic β-cell toxin commonly used to model T1DM.191,192 Mice were treated with a

standard streptozotocin dosing schedule (Animal Models of Diabetic Complications

Consortium; AMDCC) two weeks prior to tumour induction surgery. Briefly, STZ (Sigma,

Oakville, ON) was dissolved in 50mM sodium citrate buffer (pH 4.5) made up fresh

daily. Mice received 200µl IP injections of 50mg/kg STZ or vehicle control for six

consecutive days. Blood glucose was measured ten days following the last injection and

mice were considered hyperglycemic if non-fasted glucose was above 300mg/dl

(16.7mM).

Type 2 Diabetes Mellitus (T2DM)

C57BL/6 mice homozygous for a mutant Akt2 allele have poor glucose tolerance and

reduced insulin sensitivity that result in hyperglycemia and hyperinsulinemia but are not

obese.193,194 Obesity often accompanies T2DM and is independently associated with

diabetes and cancer so this model allowed us to isolate the contributions of high

glucose and high insulin. Mice were genotyped using the following primers designed by

Jackson Laboratories: Akt2 wildtype reverse: TGCACAATCTGTCTTCATGCCAC; Akt2

common forward: ACCAACCCCCTTTCAGCACTTG; Akt2 mutant reverse:

TACACTTCATTCTCAGTATTGTTTTGC. Products are 110bp (wildtype), 277bp

(mutant), or 110bp and 277bp (heterozygote).

Page 47: The effects of hyperglycemia on glucose metabolism in

33

Food Consumption

Food consumption at three weeks post tumour induction (PTI) was calculated by taking

the initial weight of dry food and subtracting food weight after five days. This value was

divided by the number of mice in the cage and the number of days in the measurement

period to determine food consumed per mouse per day.

Intraperitoneal glucose tolerance tests

Mice were fasted for four hours prior to intraperitoneal glucose tolerance tests (IPGTT).

After recording baseline blood glucose, mice were given an intraperitoneal injection of

glucose (1 mg/g body weight in saline solution at a volume of 10µl/g body weight).

Blood glucose measurements were taken at 15, 30, 60, and 120 minutes after glucose

challenge using a handheld glucose monitor (Freestyle Lite, Abbott Laboratories).

Values that exceeded the measurement range of the glucose monitor were considered

27.8mM (maximum reading). Insulin levels were measured in trunk blood from fasted

animals at sacrifice using an ultra-sensitive mouse insulin ELISA kit (Crystal Chem,

Downers Grove, IL). Data are presented as the average integrated area under the curve

(iAUC) ± SEM where iAUC is the area under the curve relative to fasting blood glucose

(FBG) at time 0 (just before glucose injection). iAUC was calculated using the

trapezoidal method. The glucose:insulin ratio was used as an indication of insulin

sensitivity.195

Page 48: The effects of hyperglycemia on glucose metabolism in

34

Tissue collection and preparation

Mice were fasted for 4-6 hours prior to sacrifice by cervical dislocation. Following

euthanasia, trunk blood was collected and fasting blood glucose was measured

immediately using a handheld glucose monitor. The remaining blood was allowed to clot

then centrifuged to obtain serum samples and stored at -80oC. Ovarian tumours were

removed, weighed, and divided into sections for subsequent analysis. Sections were

either fixed in 10% neutral buffered formalin (Fisher Scientific, Whitby, ON) for wax

embedding and tissue sectioning, or flash frozen in liquid nitrogen and stored at -80oC

for protein extraction. Secondary disease was evaluated at time of sacrifice by

aspirating ascites and by visual assessment of the number of secondary lesions. Both

ascites volume and secondary lesions were scored qualitatively on a relative 4-point

scale with 0 indicating absence of secondary disease and 3 indicating the most

widespread disease. Mice with scores of 0 and 1 were considered “low” and scores of 2

and 3 were considered “high”.

Immunohistochemistry

In order to confirm the effects of streptozotocin, immunohistochemical detection of

insulin was performed on 5µm paraffin-embedded sections of pancreatic tissue from

STZ-treated mice. Sections were deparaffinized in xylene and rehydrated in graded

alcohol solutions. Endogenous peroxidase activity was quenched using 1% (vol/vol)

hydrogen peroxide and antigen retrieval was performed by immersing slides in 10mM

citrate buffer at 90°C for 12 minutes. Tissues were blocked in 5% (wt/vol) bovine serum

Page 49: The effects of hyperglycemia on glucose metabolism in

35

albumin in PBS for 10 minutes and slides were then incubated overnight with anti-

insulin primary antibody (Cell Signal Technologies, Danvers, MA) diluted in 0.01M PBS

(pH 7.5) containing 2% (wt/vol) BSA and 0.01% (wt/vol) sodium azide at 4°C in a

humidity chamber. All subsequent incubations were performed at room temperature.

Anti-rabbit biotinylated secondary antibody (Sigma, Oakville, ON) was diluted in the

same buffer and incubated for 2 hours. Tissues were then washed in PBS and

incubated with avidin and biotinylated horseradish peroxidase (ExtrAvidin, Sigma,

Oakville, ON) for 1 hour. Antibodies were visualized using 3’ 3’-diaminobenzidine

tetrahydrochloride (DAB) (Sigma, Oakville, ON) and tissue was counterstained with

Carazzi’s Hematoxylin, dehydrated, and mounted with Permount (Fisher Scientific,

Whitby, ON).

Cell Culture

Mouse-derived epithelial ovarian cancer cells (ID8; kindly donated by Drs. Paul

Terranova and Kathy Roby, Kansas State University, Kansas, USA), human ovarian

cancer cells (SKOV-3; ATCC, Manassas, VA, USA), and human normal ovarian surface

epithelium (NOSE; generously donated by Dr. Jinsong Liu, MD Anderson Cancer

Center, Houston, TX) were maintained in DMEM supplemented with 10% fetal bovine

serum (FBS), 1% antibiotic/antimycotic (Gibco BRL, Burlington, ON) and 2% L-

glutamine (Life Technologies, Burlington ON). All experimental treatments were

performed in serum-free DMEM.

Page 50: The effects of hyperglycemia on glucose metabolism in

36

ID8 and SKOV3 cells are typically cultured in DMEM that contains supra-physiological

amounts of glucose (25mM). We refer to these “diabetic” cells as ID8-25 and SKOV3-

25. To test the effects of hyperglycemia, we created a parallel line of “normal” cells

chronically cultured in physiological glucose (6mM) that we named ID8-6 and SKOV3-6.

The -6 lines were created by passaging the -25 cells in gradually reduced

concentrations of glucose over a period of three weeks. These lines were then

maintained in 6mM glucose. To account for any differences caused by higher passage

numbers during the glucose reduction, ID8-6 and ID8-25 cells were passage-matched in

all experiments.

Viability assay

The MTT assay measures cellular reduction of a tetrazolium salt to a coloured formazan

product in an NADH- and NADPH-dependent manner and reflects primarily glycolytic

activity.196 Cells were seeded at a density of 1500 cells/well (ID8), 3000 cells/well

(SKOV3), or 4000 cells/well (NOSE) in triplicate in 96 well plates and allowed to attach

overnight. Cells were then serum starved for 6 hours and treated with different

concentrations of glucose for 48 hours. Following treatment, cells were incubated with

MTT (5mg/ml; Sigma, Oakville, ON) for one hour at 37°C and lysed with MTT lysis

buffer overnight at 37°C. Absorbance was quantified in a microplate reader at 570nm.

Page 51: The effects of hyperglycemia on glucose metabolism in

37

Scratch wound assay

To examine the motility of cells in different concentrations of glucose, ID8-6 and ID8-25

cells were grown to confluency in 24 well plates and serum starved for 6 hours. A plastic

pipette tip was used to remove an area of cells down the centre of the dish. Wells were

gently washed with PBS to remove detached cells before treatment. Phase contrast

images were taken at three marked points along the scratch at t=0 and t=24 hours.

Wound sizes were determined using ImageScope software and are presented as the

fraction of the original scratch covered by cells.

Glucose consumption and cell number

Glucose concentration in the cell media was measured using the glucose oxidase

method and reagents from Pointe Scientific (Canton MI, USA). After 24 hours of

treatment, glucose concentration in the sample wells was subtracted from the glucose

concentration in blank wells to determine the amount of glucose consumed. Relative

cell number was approximated by the absorbance of the crystal violet nuclear stain; to

account for any differences in the rate of cell proliferation, glucose consumption was

divided by relative crystal violet absorbance (590nm) to determine per-cell glucose

uptake.

Page 52: The effects of hyperglycemia on glucose metabolism in

38

Statistical Analysis

Data were analyzed using GraphPad Prism statistical analysis software. Results are

reported as mean ± standard error of the mean (SEM). Significance threshold was set

at 0.05 and p values are listed in the figure legends. Comparisons between means were

measured by two-way ANOVA followed by a Bonferroni correction, or by unpaired two-

tailed Student’s t-test. The relationship between tumour weight and FBG or insulin was

considered significant if the slope of a linear regression was significantly different from

zero. Overall survival data are plotted on a Kaplan-Meier survival curve and differences

were determined by a log-rank (Mantel-Cox) test.

Page 53: The effects of hyperglycemia on glucose metabolism in

39

Results

Mice with higher systemic glucose develop more severe disease in a model of

epithelial ovarian cancer.

Fasting blood glucose is normally maintained within a narrow physiological range

through a highly dependent relationship with insulin. To determine the insulin-

independent effects of glucose on tumour growth, we used the beta-cell toxin

streptozotocin (STZ) to inhibit pancreatic insulin production and model Type 1 diabetes

(T1DM). Without the production of insulin, Type 1 diabetic mice are unable to import

glucose into cells and thus have significantly elevated fasting blood glucose (18.81 ±

1.93 mM vs 6.41 ± 0.27 mM in controls). As in T1DM, STZ-treated animals also have

lower body weights despite consuming more food than their untreated counterparts

(Figure 2C).

We treated mice with STZ two weeks prior to inducing ovarian tumours by sub-bursal

injection of syngeneic malignant cells (ID8). Blood glucose levels remained significantly

elevated until the end of the study at 90 days post-tumour induction (PTI) (Figure 3A).

Although diabetic patients are reported to have higher operative morbidity,197 we did not

find that STZ-treated mice suffered any adverse events related to hyperglycemia either

at surgery or during subsequent incision healing.

Mice were sacrificed at 30, 60 and 90 days after tumour initiation. STZ-treated animals

developed significantly larger tumours than control mice as early as 30 days PTI (Figure

3B) and tumour size was proportional to fasting blood glucose (Figure 3C).

Furthermore, STZ-treated hyperglycemic mice had a greater burden of secondary

Page 54: The effects of hyperglycemia on glucose metabolism in

40

disease than normoglycemic mice as measured by the number of secondary tumour

lesions disseminated throughout the peritoneal cavity (Figure 3E). The accumulation of

abdominal ascites (Figure 3D), another indicator of secondary disease, was unchanged

with STZ treatment.

Page 55: The effects of hyperglycemia on glucose metabolism in

41

Figure 2: Streptozotocin treatment models Type 1 diabetes

A) Representative insulin staining (brown) of pancreas sections from mice four weeks

after STZ treatment (two weeks post-tumour induction). Arrows in STZ image indicate

pancreatic islets. 100x magnification. B) Fasting blood glucose prior to tumour-induction

surgery. Bars are mean + SEM, n=8. ***p<0.001 vs mice without STZ treatment

(control). C) Food consumption at 3 weeks post tumour induction (PTI). Bars are mean

+ SEM, n≥3. ***p<0.0001 vs mice without STZ treatment (control). D) Body weights of

control and STZ treated animals. Values are mean ± SEM, n=4-16. *p<0.05.

Page 56: The effects of hyperglycemia on glucose metabolism in

42

Figure 3: Type 1 diabetes promotes tumour growth in proportion to blood glucose

A) Fasting blood glucose in tumour-bearing mice with or without STZ-induced

hyperglycemia. Values are mean ± SEM, n=4-16. *p<0.05. B) Mean tumour weights at

30, 60 and 90 days post tumour induction (PTI). Bars are mean + SEM n≥4. *p<0.05. C)

Relationship between fasting blood glucose and tumour weight at 60 days PTI. Each

point represents a single animal. Open circles represent control mice, closed circles

represent STZ-treated mice. n≥6. Slope is significantly non-zero by linear regression

(p<0.0001). D) Qualitatively assessed ascites volume and E) secondary lesions. Values

are proportion of mice with relatively “high” or “low” ascites volume or number of

secondary lesions. n=4.

Page 57: The effects of hyperglycemia on glucose metabolism in

43

Chronic glucose deficiency in mice interferes with ovarian tumour growth.

Type 1 Diabetes, formerly known as juvenile-onset diabetes, is commonly diagnosed in

youth and adolescents and leads to glucose-related tissue damage in patients as young

as eight years old.198 In addition to the acute hyperglycemia modeled by STZ treatment

two weeks prior to tumour induction, we wanted to assess the impact of type 1 diabetic

tissue damage on tumour development. To investigate the effects of long-term (chronic)

hyperglycemia, we treated mice with STZ 15 weeks before introducing ovarian tumours.

These mice had significantly higher fasting blood glucose (10.58 ± 0.59 mM vs 5.03 ±

0.29 mM in control mice) at surgery and the hyperglycemia was maintained until mice

became moribund (Figure 4D). Six mice were excluded from the study due to

complications of diabetes.

Surprisingly, we found that at death, hyperglycemic mice had smaller tumours (Figure

4A) than normoglycemic mice, in addition to more ascites (Figure 4B) and fewer

secondary lesions (Figure 4C). Furthermore, in four of the nine STZ-treated mice that

developed ascites, the abdominal fluid was pale and watery (not shown) unlike the

hemorrhagic ascites typical to this mouse model. In contrast to the effects of short-term

STZ treatment, there was an inverse relationship between fasting blood glucose at

surgery and tumour weight at death (Figure 4E). Fasting blood glucose at death was

unrelated to tumour weight (Figure 4F). Despite the development of smaller primary

tumours and fewer secondary lesions in these mice, both long-term and short-term

hyperglycemia led to decreased survival times compared to control mice (Figure 5).

Page 58: The effects of hyperglycemia on glucose metabolism in

44

Figure 4: Chronic hyperglycemia limits ovarian tumour growth

Mice were treated with STZ or vehicle control 15 weeks prior to tumour induction

surgery to model chronic Type 1 diabetes. A) Tumours were removed and weighed

when mice became moribund. Values are mean + SEM, n≥9. ***p<0.001 vs control.

Secondary disease was evaluated at time of death by B) ascites volume and C) number

of secondary lesions. Values are proportion of mice with “high” or “low” scores. n≥9. D)

Fasting blood glucose at time of surgery and when mice became moribund. Bars are

mean + SEM, n=9 control, n=19 STZ. Different letters indicate significant (p<0.05)

differences between bars. E) Relationship between fasting blood glucose at tumour

induction surgery and tumour weight at death. n=7 control, n=15 STZ. Each point

represents a single animal. Slope is significantly non-zero by linear regression

(p<0.001). F) Relationship between fasting blood glucose and tumour weight at death.

n=9 control, n=10 STZ. Slope is not significantly different from zero.

Page 59: The effects of hyperglycemia on glucose metabolism in

45

Figure 5: Acute and chronic hyperglycemia decrease survival in mice with EOC

Kaplan-Meier survival curve of acutely (dashed lines) and chronically (solid lines)

diabetic mice (Type 1 diabetes). There is no statistical difference in survival between

mice in acute or chronic groups. STZ-treated mice had a significantly poorer overall

survival than control (nondiabetic) mice. Log-rank (Mantel-Cox) between STZ and

controls, p<0.05. n≥8.

0 20 40 60 80 1000

50

100

Acute STZ

Acute control

Chronic control

Chronic STZ

Days PTI

Perc

en

t su

rviv

al

Page 60: The effects of hyperglycemia on glucose metabolism in

46

The development of ovarian tumours improves impaired glucose tolerance in

chronically hyperglycemic mice.

In addition to elevated fasting blood glucose, diabetes is associated with glucose

intolerance, or an impaired ability to clear glucose from the blood in response to a

glucose challenge. To better characterize the effects of systemic hyperglycemia on

EOC, we performed intraperitoneal glucose tolerance tests (IPGTT). After a period of

fasting, mice were injected with 1 mg/g body weight glucose in saline. Blood glucose

was then measured at 15, 30, 60 and 120 minutes after injection and the integrated

area under the plot of blood glucose was calculated in relation to the baseline glucose

level (integrated area under the curve, iAUC). One week after tumour induction, STZ-

treated mice had a significantly higher iAUC of blood glucose (Figure 6A and B) than

normoglycemic mice, indicating poorer glycemic control. Interestingly, by 55 days PTI

STZ-treated mice showed significant improvements in glucose tolerance that had

reduced iAUC values to normal (Figure 6C).

Page 61: The effects of hyperglycemia on glucose metabolism in

47

Figure 6: Tumour induction improves glucose tolerance in chronically diabetic mice

A) and B) Blood glucose is higher in STZ-treated mice (closed circles) than in control

mice (closed circles) at all timepoints (p<0.05) following an intraperitoneal glucose

challenge. Values are mean ± SEM, n≥9. C) Integrated area under the curve (iAUC) of

glucose tolerance graphs in A) and B). Different letters indicate significant differences

between bars (p<0.05).

Page 62: The effects of hyperglycemia on glucose metabolism in

48

Mice with Type 2 diabetes grow larger tumours and die earlier in a model of

epithelial ovarian cancer.

T2DM is a prevalent health concern that has been correlated with many cancers.199 In

contrast to T1DM which is a condition of insulin deficiency, Type 2 diabetes mellitus

(T2DM) is characterized by an impaired insulin response that promotes a compensatory

increase in production and leads to hyperinsulinemia. In the STZ model we

demonstrated the glucose alone can affect tumour size, but others have predicted that

the hyperinsulinemia, rather than hyperglycemia, drives cancer development.181,185 To

investigate the combined effects of insulin and glucose on EOC tumour growth, we used

a non-obese Akt2-/- (KO) mouse model of hyperglycemia (Figure 7C) and

hyperinsulinemia (Figure 8A). Sixty days after initiating ovarian tumours, KO mice had

larger tumours than Akt2+/+ (WT) mice, and mice heterozygous for Akt (+/-) had an

intermediate tumour weight (Figure 7A). A survival study also indicated more aggressive

cancer associated with T2DM as KO mice died significantly earlier than WT mice

(Figure 7B). Interestingly, growing tumours were associated with a drop in insulin

concentration in the blood (Figure 8C) and a lower glucose:insulin ratio which indicated

better insulin sensitivity.

Higher fasting blood glucose predicts larger tumours in Akt2 KO mice.

There was a significant positive correlation between the level of fasting blood glucose

(FBG) at 30 days post tumour induction (PTI) and tumour weight at 90 days PTI (Figure

7C). Blood glucose levels at the time of tumour collection had no correlation to tumour

weight (Figure 7D). Most surprisingly, we found that insulin levels were not associated

Page 63: The effects of hyperglycemia on glucose metabolism in

49

with tumour weight as many have predicted (Figure 8C). Furthermore, in contrast to the

normal relationship between insulin and glucose, we found no correlation of insulin with

fasting blood glucose at any experimental timepoint (Figure 8D).

Page 64: The effects of hyperglycemia on glucose metabolism in

50

Figure 7: High blood glucose in Type 2 diabetic mice predicts larger tumours

Ovarian tumours were induced in wildtype mice (WT) and mice homozygous (Akt2-/-) or

heterozygous (Akt2+/-) for Akt2. A) Mean tumour weight at 30, 60 and 90 days post

tumour induction (PTI). Bars are mean + SEM, n≥5. *p<0.05. B) Percent survival of

wildtype (solid line) and Akt2-/- (dashed line) tumour bearing mice. Time to death is

significantly shorter in Akt2-/- mice than in WT controls. p=0.0193, n=6. C) Mean fasting

blood glucose (FBG) measured at tumour induction surgery. Bars are mean + SEM,

n≥62. ***p<0.001. D) Relationship between FBG at 30 days PTI and tumour weight at

90 days PTI. Slope is significantly non-zero by linear regression (p=0.0093). E)

Relationship between FBG at 90 days and tumour weight at 90 days PTI. Linear

regression slope not significantly different from zero (p=0.067). Each point represents a

single animal. n=7 WT, n=6 Akt2-/-.

Page 65: The effects of hyperglycemia on glucose metabolism in

51

Figure 8: Tumours improve insulin sensitivity in Type 2 diabetic mice

A) Fasting blood insulin and B) glucose:insulin ratio at 30, 60 and 90 days post tumour

induction (PTI). Bars are mean + SEM, n≥6. Bars sharing the same letter are not

significantly different (p<0.05). C) Relationship between tumour weight and insulin

concentration at 90 days PTI and D) relationship between fasting blood glucose and

insulin concentration at 90 days PTI. Slope is not significantly different from zero. Each

point represents a single animal. n≥5.

Page 66: The effects of hyperglycemia on glucose metabolism in

52

Cells conditioned to high glucose media have altered glucose-dependent growth

kinetics.

In normal homeostatic conditions, fasting blood glucose in humans falls in the range of

3.9 - 5.5mM, while random (unfasted) samples are typically 4.4 - 6.6mM and under

7.8mM two hours after a meal.200 Many cancerous cell lines are cultured in media with

supra-physiological concentrations of glucose, including ID8 cells which are normally

grown in high glucose (25mM) DMEM. From these “diabetic” or “hyperglycemic” cells,

we derived a “normoglycemic” cell line, ID8-6, by reducing glucose concentrations in the

media over a period of three weeks and maintaining the cells in DMEM with 6mM

glucose. The chronic culture conditions of ID8 cells significantly altered their growth

kinetics. Although ID8-25 cells were more successful at closing a scratch wound than

ID8-6 cells (Figure 9) they also appeared to be more sensitive to glucose deprivation

(Figure 10) based on measurements of cell number.

We predicted that environmental glucose was a rate limiting factor in glucose

metabolism and that when more glucose was available, cells would respond by

increasing glucose uptake. We found that although this was true of ID8-25 cells, ID8-6

cells did not show the same response (Figure 11). Based on our hypothesis, we

predicted that cells would increase uptake in direct proportion to the total glucose

available. However, we found that all cells consumed proportionally less glucose in

hyperglycemic conditions (Figure 11A).

One of the hallmarks of cancer cells that drives their glucose consumption is the rate of

metabolism through the glycolytic and oxidative phosphorylation pathways. We

Page 67: The effects of hyperglycemia on glucose metabolism in

53

measured the metabolic viability of cells using the MTT assay and again found

differences between cells cultured in 6mM and those cultured in 25mM glucose. While

ID8-6 cell viability increased in a glucose-dependent fashion from 0mM to 25mM, ID8-

25 cell viability plateaued at physiological glucose levels (Figure 12A). To evaluate

whether these results were specific to our murine cell line, we established normal- and

high- glucose variants of the human ovarian cancer cell line SKOV3 (SKOV3-6 and

SKOV3-25). As in ID8 cells, SKOV3-6 cells were more metabolically active than

SKOV3-25 cells in hyperglycemia (Figure 12B).

Page 68: The effects of hyperglycemia on glucose metabolism in

54

Figure 9: EOC cells conditioned to hyperglycemic media accelerate scratch wound

closing

A) Images were analyzed to determine the fraction of the scratches covered by cells

after 24 hours. B) Representative images of cells after 24 hours at 10x magnification.

Dashed lines indicate the width of the initial scratch. Bars are mean + SEM, n=3.

**p<0.01, ***p<0.001.

Page 69: The effects of hyperglycemia on glucose metabolism in

55

Figure 10: Cells conditioned to hyperglycemia are sensitive to glucose deprivation

Cells were cultured in different glucose concentrations for 24 hours. Crystal violet

absorbance at 590 nm was used as a cell number proxy. Bars are mean + SEM relative

to normoglycemic (6mM glucose) conditions. Different letters represent significant

differences between ID8-25 cells (black bars) by one-way ANOVA (p<0.05). ID8-6 cells

(white bars) are not significantly different across glucose concentrations. Brackets

indicate differences between cell lines at the same glucose concentration. ID8-6 n=4;

ID8-25 n=9 *p<0.05.

0.0

0.5

1.0

1.5

2.0

2.5

2mM 6mM 16.7mM 25mM

a

aa

bID8-6

ID8-25

**

Glucose in media

Cells

rela

tiv

e

to n

orm

og

lycem

ic c

on

tro

l

Page 70: The effects of hyperglycemia on glucose metabolism in

56

Figure 11: ID8-25 cells, but not ID8-6 cells, increase glucose consumption in

hyperglycemia.

A) Fraction of glucose consumed relative to the initial glucose concentration in the cell

media. B) Total glucose consumed in mg/dL. C) Relative glucose consumption per cell.

Bars are mean + SEM, n≥5. Different letters represent significant differences between

bars (p<0.05).

Page 71: The effects of hyperglycemia on glucose metabolism in

57

Figure 12: In high glucose media, cells conditioned to hyperglycemia have lower

metabolic viability than cells conditioned to physiological glucose

MTT absorbance was used to determine the metabolic viability of cells. A) Viability of

mouse epithelial ovarian cells acclimatized to physiological (ID8-6, white bars) or

hyperglycemic (ID8-25, black bars) conditions. ID8-6, n≥8; ID8-25, n=13. B) Viability of

human ovarian cancer cells acclimatized to physiological (SKOV3-6, white bars) or

hyperglycemic (SKOV3-25, black bars) conditions. SKOV3-6, n=4; SKOV3-25, n=11.

Values are mean + SEM. Stars indicate differences vs 6mM (normoglycemic control).

Brackets indicate differences between cell lines at a single glucose concentration.

*p<0.05, **p<0.01, ***p<0.001.

Page 72: The effects of hyperglycemia on glucose metabolism in

58

Discussion

Type 2 diabetes (T2DM) is a large and growing health burden worldwide. In population

studies it is consistently associated with increased incidence and mortality in multiple

cancers.199 T2DM is characterized by insulin insensitivity which leads to a combination

of hyperglycemia and hyperinsulinemia. In this study we showed that high systemic

glucose, with or without insulin, accelerates ovarian tumour progression.

Tumour growth and survival in Type 1 diabetes

Insulin is a powerful mitogen and pro-tumorigenic factor and is generally considered a

driving force for cancer development in diabetics.201,202 Type 1 diabetes is characterized

by hyperglycemia but a deficiency in insulin which makes it a good model for isolating

the effects of high blood glucose from hyperinsulinemia.

Type 1 diabetes (T1DM) is an autoimmune disorder that results in the destruction of

insulin-producing beta cells in the pancreas. We induced T1DM in vivo by using the

beta-cell toxin streptozotocin (STZ). Two weeks after STZ treatment, we injected

transformed epithelial ovarian (ID8) cells under the ovarian bursa in c57bl/6 mice in an

orthotopic, syngeneic model of EOC developed in our lab. In this model, tumour

development at 30 days post-tumour induction (PTI) models Stage I human disease and

EOC progression at 60 and 90 days PTI are representative of Stages II and III. We

found that as early as 30 days PTI, STZ-treated mice had significantly larger tumours

than normoglycemic controls. By 60 days, STZ-treated mice showed more aggressive

Page 73: The effects of hyperglycemia on glucose metabolism in

59

secondary disease. Moreover, we found that the degree of dysglycemia directly

influenced the size of the primary tumour (Figure 3C). This is in agreement with the

glucose concentration-dependent increase in proliferation that has been shown in

cancer cells in vitro203 and human studies that have shown that there are proportional

increases in the risk of overall cancer mortality with concurrent increases in plasma

glucose.204 There are currently no other studies that have used a T1DM model of

diabetes to evaluate cancer progression.

Based on these intriguing results, we anticipate that a short-term STZ model will be a

powerful tool for investigating tumour cell metabolism. However, such an acute and

short-term hyperglycemia is probably not representative of a human diabetic condition.

In reality, diabetes is a chronic disease that causes significant damage to normal cells,

particularly those of the endothelium. It is reasonable to expect that these changes will

impact the development of a tumour, which relies on interactions with its local

environment. In order to more accurately model the physiological effects of chronic

hyperglycemia, we also treated mice with STZ 15 weeks prior to tumour induction.

These mice had milder, but still significant, hyperglycemia (10.58 ± 0.59 mM) at tumour

induction surgery than the mice with “acute” diabetes (18.81 ± 1.93 mM). We suspect

that this difference reflects a partial recovery of the beta-cell population in the time

between STZ treatment and tumour induction.205 Chronically elevated blood glucose

interfered with the growth of ovarian tumours, which were significantly smaller than

those in normoglycemic mice (Figure 4A). Because cancer cells rely on the recruitment

of other cell types to populate a tumour and facilitate its growth, these unanticipated

results could be a reflection of irreversible cellular damage caused by chronic

Page 74: The effects of hyperglycemia on glucose metabolism in

60

hyperglycemia which prevented somatic cells from interacting with cancer cells. Cancer

associated fibroblasts (CAF) in particular make up a large portion of tumour mass and

are significant contributors to cancer progression.206 Diabetes, however, is associated

with low numbers of fibroblasts and fibroblasts with impaired motility.207 If local

fibroblasts have sustained cellular damage due to hyperglycemia, it is possible that

cancer cells will be unable to efficiently recruit them into tumours. Similarly, vascular

damage in diabetes may inhibit the tumour’s ability to recruit new vessels which would

limit tumour growth and metastasis.43,208

In addition to metabolic influences of blood glucose on tumour growth, the pathological

stress of diabetes itself contributes to long-term mortality in cancer patients,209 affecting

overall survival even if disease-free survival is not significantly worse.179 Adding to this

evidence, we found that despite the differences in tumour size, STZ treatments at 2 and

15 weeks prior to surgery both similarly decreased survival compared to nondiabetic

mice. Comorbid hyperglycemia can accelerate time to death in cancer patients, and

may do so independently of primary tumour size. The discrepancy between survival and

tumour size in the T1DM STZ mice may also reflect differences in secondary disease

that our scoring system does not take into account, in particular immune response and

the tumour-promoting effects of chronic systemic inflammation.210 The ascites in the 15-

week STZ animals was less hemorrhagic than that in the control animals (data not

shown), which might indicate differences in composition or malignant potential which

could affect tumour spread within the peritoneal cavity.

Page 75: The effects of hyperglycemia on glucose metabolism in

61

Tumour growth and survival in Type 2 diabetes

In women, the hyperinsulinemia and hyperglycemia of T2DM is often associated with

obesity. Models of diet-induced T2DM have shown increased primary tumour growth

and more metastatic disease in orthotopic ovarian211 and breast201 cancers. However,

obesity can independently contribute to tumour growth. To separate the effects of

obesity from the influence of insulin and glucose in T2DM, we used non-obese Akt2

knockout mice on a c57bl/6 background that have an impaired ability to translocate

glucose transport proteins to the cell surface in response to insulin. As in the short-term

STZ-treated animals, we found that these mice grew larger primary tumours in

proportion to higher fasting blood glucose and died earlier than their wildtype

counterparts (Figure 7A and B).

Systemic glucose

Tumours have been shown to act as glucose sinks, inducing host hypoglycemia in a

tumour mass-dependent fashion,212,213 particularly in the areas closest to the

tumour.212,214 In some cancer patients with epithelial-derived malignancies, this property

of tumours leads a condition called non-islet cell tumour-induced hypoglycemia

(NICTH).215 In addition to testing whether circulating glucose can affect tumour growth,

we were interested in the reciprocal effect of the tumour on systemic glucose. We

showed that the growth of ovarian tumours improved glycemic control in T2DM by both

normalizing the fasting insulin level and by improving insulin sensitivity (Figure 8).

Intraperitoneal glucose tolerance tests in STZ-treated mice demonstrated that growing

Page 76: The effects of hyperglycemia on glucose metabolism in

62

tumours also improved glucose tolerance in T1DM (Figure 6), suggesting that the

tumour-mediated reduction in hyperglycemia is not entirely insulin-mediated.

Surprisingly, we found that many of our mice had substantial abdominal fat (data not

shown), despite the fact that Akt2 KO results in an 80-90% reduction of fat depots.194

We interpret this as further evidence that tumours caused significant disturbances in

global metabolic maintenance.

In vitro

ID8 cells are normally maintained in high glucose DMEM media (25mM glucose, ID8-

25). In order to compare EOC cell growth in normal and hyperglycemic conditions, we

created a parallel line of ID8 cells that were passaged in progressively lower

concentrations of glucose and eventually maintained at a physiological level (6mM, ID8-

6). These cells allowed us to model the effects of both glucose addition and glucose

deprivation. ID8-6 and ID8-25 cells showed markedly different responses to

environmental glucose. Subsequently, another group showed that breast cancer cells in

glucose concentrations above 5mM have significantly higher rates of proliferation,

clonogenicity, motility, upregulation/activation of pro-oncogenic signaling, and

reductions in apoptosis.216

In order to measure metabolic viability, we used the MTT assay which is sensitive to

NADH produced primarily through glycolysis. Although MTT activity is generally thought

to reflect mitochondrial succinate activity, only a small proportion of tetrazolium

reduction occurs this way.196

Page 77: The effects of hyperglycemia on glucose metabolism in

63

Although ID8-25 cells consumed more glucose in response to substrate availability, the

MTT assay showed no increase glycolysis compared to control (Figure 12). This

suggests that glycolytic metabolism may be saturated at normal glucose concentrations.

However, because the glucose uptake increases, the saturation point must occur

downstream of hexokinase (HK) which is necessary to keep glucose from diffusing back

out of the cell. An increase in glucose uptake with no change in glycolysis could suggest

less efficient metabolism. This observation could be an indication of the Crabtree Effect,

in which high glucose saturates enzymes responsible for oxidative phosphorylation.121 It

may also be reflective of a defect in oxidative phosphorylation in the ID8-25 cells that

could necessitate an increase in glycolysis, representative of the Warburg Effect. Both

the Crabtree and Warburg effects are associated with increased malignancy.217

Conversely, ID8-6 cells did not increase glucose uptake in response to substrate

availability, but they did show a relative increase in glycolytic flux in 25mM glucose

compared to 6mM glucose. These cells may have limited capacity to upregulate glucose

transporters, or HK could potentially be saturated in these conditions and prevent

further glucose uptake. However, the increase in glycolytic activity indicates that

enzymes lower down the glycolytic cascade (phosphofructokinase, pyruvate kinase) are

not saturated at normal glucose concentrations and are therefore able to increase

energy production. Although we did not see higher glucose consumption over 24 hours,

it is possible that over a longer period this increased glycolytic activity would result in

increased glucose uptake.

As mouse epithelial ovarian cells become more glycolytic they also become malignant

and aggressive218 which would suggest that in ID8-6 cells, a high glucose environment

Page 78: The effects of hyperglycemia on glucose metabolism in

64

would be associated with more aggressive disease than a lower glucose environment.

Glucose starvation, which we modeled in ID8-25 cells in 6mM glucose, has been shown

to induce a caspase-independent cell death similar to autophagy.219 Consequently, we

would expect that ID8-25 cells in 6mM would be susceptible to death. We found that

there were fewer ID8-25 cells in total in conditions of glucose deprivation (Figure 10),

but environmental glucose did not influence their ability to close a scratch wound

(Figure 9). Neither of these assays shows what processes are responsible for changes

in cell number, so it is unclear whether these data indicate differences in cell

proliferation or cell death. Also, although treating ID8-25 cells in 6mM glucose

represents glucose deprivation, the cells are not in hypoglycemic media and so may not

be under enough metabolic stress to induce cell death.

Both ovarian cancer and T2DM are insidious diseases that are often diagnosed well

after pathological changes have occurred.25,220 In patients presenting with both

illnesses, the combined systemic metabolic defects in diabetes and the cellular

metabolic transformations in ovarian cancer cells have had a long window of interaction.

The in vitro results from this study show that cancer cells developed in hyperglycemic

environments are metabolically distinct from those in normoglycemic environments.

These differences could indicate that certain cancer treatments will have different

outcomes in diabetic compared to nondiabetic patients.

Hyperglycemia is a potentially modifiable risk factor for poor overall survival in women

with EOC and the results of this study support the possibility that anti-hyperglycemic

drugs would be a beneficial adjuvant cancer therapy.

Page 79: The effects of hyperglycemia on glucose metabolism in

65

CHAPTER 3: GLUCOSE TRANSPORT IN HYPERGLYCEMIC EOC

Introduction

Epithelial ovarian cancer (EOC) is the fifth leading cause of cancer death among

women.1,2 EOC is a challenge both to detect and to treat, resulting in a survival rate that

has been virtually unchanged in the last 30 years.

In large population studies, T2DM has been associated with the risk and prognosis of

many different cancers.199 Previous studies in our lab have shown that ovarian tumour

growth is more aggressive in diabetic environments independent of insulin levels

(Chapter 2). Hyperglycemia also increases the ability of EOC cells to take up glucose,

but the expression, regulation and uptake kinetics of glucose transporters in these

conditions are unknown.

Glucose primarily enters cells by facilitated diffusion through glucose transporters

(GLUTs). The activation of GLUT genes is one of the earliest events in oncogenesis221

and high GLUT1 expression is often associated with poor prognosis and more

aggressive disease.222 Almost all invasive epithelial carcinomas are positive for GLUT1,

independent of stage, grade, or histological subtype.126,127 As a passive transporter,

GLUT activity is dependent on the direction and magnitude of the glucose concentration

gradient across the cell membrane. Glucose can also enter the cell against its

concentration gradient through a class of sodium-dependent secondary active

symporters (SGLTs). The most well studied isoforms, SGLT1 and SGLT2, are normally

expressed in the small intestine and kidney proximal tubule respectively. Few papers

Page 80: The effects of hyperglycemia on glucose metabolism in

66

have looked at SGLT expression in malignancies and only one published study has

demonstrated functional glucose uptake through the SGLTs in tumours.223

One of the hallmark characteristics of cancer is a glycolytic phenotype that necessitates

high glucose uptake to produce ATP and the biosynthetic precursors required for

proliferation. This elevated glucose uptake has been exploited in clinical imaging. Using

positron emission tomography (PET), the accumulation of the glucose analogue

18fluoro-deoxy-glucose (FDG) can differentiate tumour tissue from normal tissue. In

EOC, FGD-PET can be useful in monitoring response to treatment but it has poor utility

as a diagnostic tool. FDG is efficiently transported through GLUT transporters, but it is a

poor substrate for SGLTs.224 FDG is the only oncolytic tracer currently in clinical use. If

sodium-glucose transport is functionally important in glucose uptake into ovarian tumour

tissue, the use of an SGLT-transportable glucose analogue could substantially increase

the power of imaging.

Glucose transport through SGLTs has the potential to release cancer cells from the

constraints of extracellular glucose concentration and thus provide a level of self-

sufficiency for energy production. The goal of this study was to characterize the

expression and relative contributions of the GLUT and SGLT transporter families in the

glucose uptake and tumour growth of epithelial ovarian cancer, in both normoglycemic

and hyperglycemic environments.

Page 81: The effects of hyperglycemia on glucose metabolism in

67

Materials and Methods

Reverse transcription PCR

Reverse transcription (RT)-PCR was used to determine the mRNA expression of

glucose transporters SGLT1-3 and GLUT1-4 in ID8-25 cells. Total RNA was extracted

from cell lysates of serum-starved ID8-25 cells using the Qiagen RNA Extraction Kit

(RNeasy Mini kit, Qiagen, Mississauga, ON) and stored at -80°C prior to use.

RNA samples were reverse transcribed to cDNA using in 50µl reactions with 1µl

Oligo(dT) 12-18 primer, 1µl 10mM dNTP, 10µl of each RNA sample (65ºC for 5 minutes

in an MJ Research PTC-200 Thermo Cycler); four parts 5X First-Strand buffer, 2 parts

0.1M DTT (supplied with SuperScript II Reverse Transcriptase kit, Life Technologies

Inc., Burlington, ON) and one part RNasin in 7µl (42ºC for 2 minutes); 1µl of SuperScript

II Reverse Transcriptase. The reverse transcription reaction was then carried out at

42ºC for 50 minutes. Inactivation was induced by incubating the tubes at 70ºC for 15

minutes. Final cDNA products were stored at -20ºC.

Primer sequences were determined by Mus musculus gene sequences using NCBI

Primer-Blast. These primers, generated from Laboratory Services Molecular Biology

Lab (Guelph, ON), were stored at -20ºC. Sequences are provided in Appendix IV.

The cDNA was amplified in 50 µl reactions containing one part 10mM dNTP mix, five

parts 10x buffer and one part 1x Taq (New England BioLabs Ltd., Pickering, ON),

forward and reverse primers, 1µl of cDNA and nuclease-free water. The PCR reaction

95ºC for 30 seconds; appropriate annealing temperature (previously determined) for 30

Page 82: The effects of hyperglycemia on glucose metabolism in

68

seconds; 72ºC for 1 minute; for 35 cycles. PCR products were then incubated at 72ºC

for 10 minutes and stored at 4ºC until they were separated on a 1% agarose gel and

visualized with ethidium bromide using the FluorChem8800 gel documentation imaging

system.

Immunohistochemistry

In order to determine the protein expression of SGLT transporters in human EOC,

immunohistochemical detection of SGLT1 and SGLT2 was performed on a microarray

that included normal ovarian tissue, serous adenocarcinomas and metastatic tissue

cores (OV208 microarray; US Biomax, Rockville, MD). Sections were deparaffinized in

xylene and rehydrated in graded alcohol solutions. Endogenous peroxidase activity was

quenched using 1% (vol/vol) hydrogen peroxide and antigen retrieval was performed by

immersing slides in 10mM citrate buffer at 90°C for 12 minutes. Tissues were blocked in

5% (wt/vol) bovine serum albumin in PBS for 10 minutes and slides were then

incubated overnight with anti-SGLT1 or -SGLT2 primary antibody (Abcam, Cambridge

Science Park, Cambridge, MA) diluted in 0.01M PBS (pH 7.5) containing 2% (wt/vol)

BSA and 0.01% (wt/vol) sodium azide at 4°C in a humidity chamber. All subsequent

incubations were performed at room temperature. Anti-rabbit biotinylated secondary

antibody (Sigma, Oakville, ON) was diluted in the same buffer and incubated for 2

hours. Tissues were then washed in PBS and incubated with avidin and biotinylated

horseradish peroxidase (ExtrAvidin, Sigma, Oakville, ON) for 1 hour. Antibodies were

visualized using 3’ 3’-diaminobenzidine tetrahydrochloride (DAB) (Sigma, Oakville, ON),

tissue was counterstained with Carazzi’s Hematoxylin then dehydrated and mounted

Page 83: The effects of hyperglycemia on glucose metabolism in

69

with Permount (Fisher Scientific, Whitby, ON). Tissue cores were considered to have no

SGLT protein expression if total positive pixel counts were less than 2%.

Immunohistochemistry of SGLT2 (Abcam, Cambridge Science Park, Cambridge, MA)

and GLUT1 (Santa Cruz Biotechnologies, Santa Cruz, CA) in mouse tissue sections

was performed identically.

Cell Culture

Mouse-derived epithelial ovarian cancer cells (ID8; kindly donated by Drs. Paul

Terranova and Kathy Roby, Kansas State University, Kansas, USA), human ovarian

cancer cells (SKOV-3; ATCC, Manassas, VA, USA), and human normal ovarian surface

epithelium (NOSE; generously donated by Dr. Jinsong Liu, MD Anderson Cancer

Center, Houston, TX) were maintained in DMEM supplemented with 10% fetal bovine

serum (FBS), 1% antibiotic/antimycotic (Gibco BRL, Burlington, ON) and 2% L-

glutamine (Life Technologies, Burlington ON). All experimental treatments were

performed in serum-free DMEM.

ID8 and SKOV3 cells are typically cultured in DMEM that contains supra-physiological

amounts of glucose (25mM). These “diabetic” cells were referred to as ID8-25 and

SKOV3-25. To test the effects of hyperglycemia, we created a parallel line of “normal”

cells chronically cultured in physiological glucose (6mM) that we named ID8-6 and

SKOV3-6. The -6 lines were created by passaging the -25 cells in reducing

concentrations of glucose over a period of three weeks. These lines were then

maintained in 6mM glucose. To account for any differences caused by higher passage

Page 84: The effects of hyperglycemia on glucose metabolism in

70

numbers during the glucose reduction, ID8-6 and ID8-25 cells were passage-matched in

all experiments.

Drugs

Phlorizin (PZ; Sigma, Oakville, ON) competitively inhibits glucose transport through

sodium/glucose cotransporters (SGLTs). Cytochalasin B (CB; Sigma, Oakville, ON) and

phloretin (PT; Santa Cruz Biotechnologies, Santa Cruz, CA) inhibit glucose transport

through the passive glucose transporters (GLUTs). Cytochalasin B is a more potent

GLUT inhibitor than phloretin, however it has two binding sites, one of which affects cell

cytoskeleton by interfering with actin assembly. Thus, CB was not appropriate for

motility studies and was replaced by PT.225

Viability assay

The WST-1 assay measures extracellular reduction of a tetrazolium salt to a coloured

formazan product dependent on the net rate of NADH reduction. NADH produced in the

mitochondrial TCA cycle is the primary reductant for extracellular WST-1 reduction via

transmembrane electron transport.226 ID8 cells were seeded at a density of 1500

cells/well in triplicate in 96 well plates and allowed to attach overnight. Cells were then

serum starved for 6 hours and treated for 48 hours in serum-free conditions. Following

treatment, cells were incubated with treatment media containing water-soluble

tetrazolium salt (WST)-1 reagent (Roche Applied Science, Laval, QC) at a 1:10 dilution

Page 85: The effects of hyperglycemia on glucose metabolism in

71

at 37ºC for one hour. Absorbance was quantified 450nm with 630nm correction on a

microplate reader.

Scratch wound assay

To examine the motility of cells in different concentrations of glucose, ID8-6 and ID8-25

cells were grown to confluency in 24 well plates and serum starved for 6 hours. A plastic

pipette tip was used to remove an area of cells down the centre of the dish. Wells were

gently washed with PBS to remove detached cells before treatment. Phase contrast

images were taken at three marked points along the scratch at t=0 and t=24 hours.

Wound sizes were determined using ImageScope software and are presented as the

fraction of the original scratch covered by cells.

Glucose consumption and cell number

Glucose concentration in the cell media was measured using the glucose oxidase

method and reagents from Pointe Scientific (Canton MI, USA). After 24 hours of

treatment, glucose concentration in the sample wells was subtracted from the glucose

concentration in blank wells to determine the amount of glucose consumed. Relative

cell number was approximated by the absorbance of the crystal violet nuclear stain. To

account for any differences in the rate of cell proliferation, glucose consumption was

divided by relative crystal violet absorbance (590nm) to determine a per-cell uptake

comparison.

Page 86: The effects of hyperglycemia on glucose metabolism in

72

Invasion Assay (Boyden chamber)

In vitro cell invasive/migratory capacity was measured using a Boyden chamber assay

(Biocoat Matrigel invasion chambers; BD Biosciences, Franklin Lakes, NJ). ID8-6 and

ID8-25 cells were grown on a Matrigel-coated upper membrane (10,000 cells per well)

and treated with 100µM phlorizin or DMSO vehicle control, in serum-free DMEM

containing 6mM or 25mM glucose. The lower chamber contained drug-free media with

10% FBS to act as a chemoattractant. Cells were incubated at 37°C and allowed to

migrate for 20 hours. Cells were removed from the upper chamber using cotton swabs.

500µl of 5% glutaraldehyde PBS was added to the lower chamber and incubated for 10

minutes at room temperature. Migrated cells were stained by adding 500µl 0.5%

toluidine blue staining solution in the lower chamber and incubating for 15 minutes.

Manual counting of cells in three fields of view per membrane was used to quantify

number of invaded cells.

shRNA SGLT2 knockdown

ID8-6 and ID8-25 cells were cultured in 12-well plates in complete medium (6mM

glucose or 25mM glucose DMEM containing FBS). At 40% confluence, cells were

treated with 5µg/ml Polybrene (Santa Cruz Biotechnologies, Santa Cruz, CA) and

infected with either: a pool of three shRNA constructs, each encoding SGLT2-specific

19-25 nt (plus hairpin loop) shRNA; or with control shRNA particles (scrambled

sequence) (Santa Cruz Biotechnologies, Santa Cruz, CA) overnight. Polybrene media

was then replaced with normal media and incubated overnight. After 48 hours, cells

Page 87: The effects of hyperglycemia on glucose metabolism in

73

were selected with puromycin dihydrochloride (Santa Cruz Biotechnologies, Santa Cruz,

CA) for 12 weeks. Clonal knockdown colonies were grown from single puromycin-

resistant cells. Western blot analysis was used to evaluate SGLT2 protein knockdown.

Animals

Wildtype c57bl/6 mice were purchased from Charles River Laboratories (Wilmington,

MA, USA), c57bl/6 Akt2 null mice (https://www.jax.org/strain/006966) were purchased

from Jackson Laboratories (Bar Harbour, ME, USA). An Akt2 null mouse colony was

established from breeder mice and animals were genotyped prior to beginning

experiments. Animals were housed at the Central Animal Facility at the University of

Guelph and all experiments were conducted according to Canadian Council on Animal

Care guidelines. Mice were kept on a 12h light:dark cycle and had free access to food

and water. For all experiments, adult mice were between 12 and 24 weeks old at time of

surgery.

Mouse model of epithelial ovarian cancer

Spontaneously transformed murine epithelial cells from C57Bl/6 mice (ID8) were diluted

in 5µl phosphate buffered saline (PBS) and injected under the ovarian bursa of WT or

Akt2 null mice. This orthotopic, syngeneic mouse model of EOC closely replicates

ovarian serous adenocarcinoma in women. By 90 days post-tumour induction, mice

develop large primary tumours, secondary peritoneal lesions, and significant abdominal

ascites.10 In survival experiments, mice were sacrificed when they became moribund

due to the accumulation of ascites fluid.

Page 88: The effects of hyperglycemia on glucose metabolism in

74

Type 2 Diabetes Mellitus (T2DM)

C57BL/6 mice homozygous for a mutant Akt2 allele are non-obese animals with poor

glucose tolerance and reduced insulin sensitivity that result in hyperglycemia and

hyperinsulinemia.193,194 Mice were genotyped using the following primers designed by

Jackson Laboratories: Akt2 wildtype reverse: TGCACAATCTGTCTTCATGCCAC; Akt2

common forward: ACCAACCCCCTTTCAGCACTTG; Akt2 mutant reverse:

TACACTTCATTCTCAGTATTGTTTTGC. Products are 110bp (wildtype), 277bp

(mutant), or 110bp and 277bp (heterozygote).

Intraperitoneal glucose tolerance tests

Mice were fasted for four hours prior to intraperitoneal glucose tolerance tests (IPGTT).

After recording baseline blood glucose, mice were given an intraperitoneal injection of

glucose (1 mg/g body weight in saline solution at a volume of 10µl/g body weight).

Blood glucose measurements were taken at 15, 30, 60, and 120 minutes after glucose

challenge using a handheld glucose monitor (Freestyle Lite, Abbott Laboratories, Abbott

Park, IL). Values that exceeded the measurement range of the glucose monitor were

considered 27.8mM (maximum reading).

Plasma for insulin quantification was collected in heparinized capillary tubes during the

IPGTT or from trunk blood from fasted animals at sacrifice using an ultra-sensitive

mouse insulin ELISA kit (Crystal Chem, Downers Grove, IL). Data are presented as the

average integrated area under the curve (iAUC) ± SEM where iAUC is the area under

the curve relative to fasting blood glucose (FBG) at time 0 (immediately before glucose

Page 89: The effects of hyperglycemia on glucose metabolism in

75

injection). iAUC was calculated using the trapezoidal method. The glucose:insulin ratio

was used as an indication of insulin sensitivity.195

Tissue collection and preparation

Mice were fasted for 6 hours prior to sacrifice by cervical dislocation. Following

euthanasia, trunk blood was collected and fasting blood glucose was measured

immediately using a handheld glucometer. The remaining blood was allowed to clot

then centrifuged to obtain serum samples and stored at -80oC. Ovarian tumours were

removed, weighed, and divided into sections for subsequent analysis. Sections were

either fixed in 10% neutral buffered formalin (Fisher Scientific, Whitby, ON) for wax

embedding and tissue sectioning or flash frozen in liquid nitrogen and stored at -80oC

for protein extraction. Secondary disease was evaluated at time of sacrifice by ascites

volume and number of secondary lesions. Mice were scored on a 4-point scale with 0

indicating absence of secondary disease. Mice with scores of 0 and 1 were considered

“low” and scores of 2 and 3 were considered “high”.

Radioisotope uptake

Alpha-methyl-glucopyranoside (AMG) and 2-Deoxy-D-glucose (2-DG) are glucose

analogues that are preferentially transported by SGLTs and GLUTs respectively.224

Transporter-specific glucose uptake was determined using [3H] 2-DG and [14C] AMG

(Moravek Biochemicals, Brea, CA). Mice were given a single tail vein injection of 25µCi

Page 90: The effects of hyperglycemia on glucose metabolism in

76

of [3H]2-DG and 5µCi of [14C]AMG 1.5 hours before sacrifice. Volumes of blood and

ascites and sections of tumour, skeletal muscle, liver and ovary were collected. The

samples were incubated at 55°C in ScintiGest solubilisation fluid (Fisher Scientific,

Whitby, ON) until the tissue dissolved. Thirty percent H2O2 was added to decolorize

samples. An aliquot of scintillation cocktail (CytoScint; MP Biomedicals, Santa Ana, CA)

was added to each sample and the total radioactive disintegrations per minute (dpm)

were determined using a Beckman LS 6500 liquid scintillation counter (Beckman

Coulter, Fullerton, CA).

Western Blots

Protein was collected from tumour tissue or SGLT2 knockdown cells using RIPA lysis

buffer and quantified using the DC Protein Quantification kit (Bio-Rad, Mississauga,

ON). Total protein lysates (20-40 µg) were used for immunoblot analysis. Lysates were

separated by SDS-PAGE on 10% gels. Proteins were electro-transferred onto

nitrocellulose membranes (Amersham, Piscataway, NJ) then blocked for 1 hour at room

temperature in 5% (wt/vol) skim milk in TBS-T (TBS with 1%(vol/vol) Tween 20).

Primary antibodies (SGLT1 (1:500, Abcam, Cambridge Science Park, Cambridge, MA),

SGLT2 (1:500, Abcam, Cambridge Science Park, Cambridge, MA), GLUT1 (1:200,

Santa Cruz) and β-actin (1:8000, Cell Signal Technologies, Danvers, MA) were diluted

in TBS-T skim milk and incubated overnight at 4°C on a rocking platform. After washing

with TBS-T blots were incubated with peroxidase-conjugated anti-rabbit secondary

antibody (1:1000 dilution; Cell Signal Technologies, Danvers, MA) or anti-mouse IgG

(1:1000 dilution, Cell Signal Technologies, Danvers, MA) for one hour at room

Page 91: The effects of hyperglycemia on glucose metabolism in

77

temperature on a rocking platform. Reactive protein was detected with ECL

chemiluminescence (Perkin Elmer, Waltham, MA). Membranes were imaged on the Bio-

Rad ChemiDoc XRS+ and densitometry was performed using Bio-Rad Image Lab 5.1.

Protein values were expressed as a ratio with β-actin.

Statistical Analysis

In vivo data were analyzed using Statistix 7 analytical software. Results are reported as

mean ± standard error of the mean (SEM). Significance threshold was set at 0.05 and p

values are listed in the figure legends. Tumour weight at survival was analyzed using a

one-way ANOVA followed by Tukey’s post-hoc. The significance of metastastic

secondary disease was measured using a Wilcox Rank Sum test for metastasis by

knockdown (ID8-6 and genotype were not significant in this model). Differences in

ascites volume by group were significant (chi squared approximation p=0.0025) but

there were no differences in pairwise comparisons. Differences in ascites volume were

therefore determined using a Kruskal-Wallis one-way nonparametric ANOVA by

knockdown. Overall survival data are plotted on a Kaplan-Meier survival curve and

differences were determined by a proportional hazards regression. All other data were

analyzed using GraphPad Prism statistical analysis software. Comparisons between

means were measured by two-way ANOVA followed by a Bonferroni correction, or by

unpaired two-tailed Student’s t-test.

Page 92: The effects of hyperglycemia on glucose metabolism in

78

Results

Both passive and active glucose transporters are expressed in EOC

Using reverse transcription PCR we showed the expression of both passive (GLUT1,

GLUT3) and active (SGLT2) glucose transporters in ID8 murine epithelial ovarian

cancer (EOC) cells (Figure 13). This is the first demonstration that sodium-glucose

transporters are expressed in ovarian cancer.

To further characterize the expression of SGLT transporters, we immunostained human

tissue microarrays containing tissue samples from serous ovarian adenocarcinomas,

metastases, and normal ovarian tissue (Figure 14). Using Imagescope software to

quantify protein expression, we found that both SGLT1 and SGLT2 were present in

normal ovaries at low levels (3.1 ± 0.2 % and 23.8 ± 2.8 % respectively). In tumours,

SGLT1 was only expressed in about a third of samples, in which staining density was

only about 16%. SGLT2, however, was expressed in all samples with high density (67.5

± 2.9 % positive pixels). Both isoforms were overexpressed in tumours, though

expression in metastases was no different than in normal tissue. Expression did not

differ by stage, but low grade tumours were associated with higher SGLT expression.

Staining was diffuse throughout the cytoplasm and there was no clearly defined

expression at the cell membrane. Positive staining for SGLT1 is shown in the

supplementary figures (Figure 41, Appendix I).

Page 93: The effects of hyperglycemia on glucose metabolism in

79

Figure 13: GLUT and SGLT transporters are expressed in ID8 cells

Reverse transcription PCR for glucose transporters in mouse epithelial ovarian cancer

cells (ID8-25). A) Secondary active sodium glucose transporter 2 (SGLT-2) expressed.

Positive controls: mouse kidney. B) Facilitative glucose transporters (GLUT) GLUT-1

and GLUT-3 are expressed. Positive controls: GLUT-2, mouse liver and GLUT-4,

mouse skeletal muscle. NTC, no-template control.

Page 94: The effects of hyperglycemia on glucose metabolism in

80

Figure 14: SGLT2 is overexpressed in human serous ovarian cancer

Immunostaining for SGLT1 and SGLT2 on tissue microarrays of human serous

epithelial ovarian cancers. Staining was quantified using Imagescope software to

determine the number of positive and negative pixels within a tissue section. A)

Representative tissue cores of normal and malignant (FIGO Stage 3c, Grade 3) ovarian

Page 95: The effects of hyperglycemia on glucose metabolism in

81

tissue. Pixels intensity was categorized as negative, weak positive, positive, or strong

positive. Normal tissue (n=26), carcinomas (n=48), metastases (n=18). B) Staining by

tissue type C) Staining of serous adenocarcinomas by FIGO stage D) Staining of serous

adenocarcinomas by histological grade *p<0.05, **p<0.01, ***p<0.001 between total

positive pixel counts.

Table 3: SGLT protein expression in human ovarian cancer

SGLT1 SGLT2

Tissue expression Positive pixel density Tissue expression Positive pixel density

N Percent Mean ± SEM (%) N Percent Mean ± SEM (%)

Normal 18 100.0 3.1 ± 0.2 16 100.0 23.8 ± 3.8

Tumours 15 31.3 15.8 ± 4.0 48 100.0 67.5 ± 2.9

Grade 2 9 56.3 20.9 ± 5.9 16 100.0 78.4 ± 3.6

Grade 3 6 18.8 8.3 ± 2.4 32 100.0 62.1 ± 3.7

Stage 1

6 28.6 19.4 ± 8.4 15 100.0 68.4 ± 3.9

Stage 2 6 100.0 48.2 ± 11.9

Stage 3 8 33.3 13.1 ± 4.3 20 100.0 69.0 ± 4.5

Metastases 7 38.9 3.7 ± 0.6 18 100.0 37.2 ± 5.3

Page 96: The effects of hyperglycemia on glucose metabolism in

82

GLUT transporters are active in physiological glucose concentrations (6mM) and

SGLT transporters are active in hyperglycemic conditions (25mM)

In order to evaluate the relative contributions of GLUT and SGLT transporters, we used

class-specific glucose transport inhibitors. Phlorizin (PZ) selectively inhibits glucose

transport through the SGLTs, while phloretin (PT) and cytochalasin B (CB) inhibitor only

GLUT-mediated glucose transport. Based on the mRNA expression in ID8 cells, we can

infer that in ID8 cells, PZ is acting on SGLT2 and PT/CB are acting on GLUT1 and

GLUT3.

ID8 cells were differentiated into two subtypes through serial passaging. ID8-6 cells are

conditioned to normal physiological concentrations of glucose (6mM) and ID8-25 cells

are conditioned to supra-physiological glucose (25mM). In both cell lines, PZ inhibited

glucose consumption in high-glucose media compared to cells treated with a vehicle

control (Figures 15 and 16). PZ had no significant inhibitory effect on glucose uptake in

normal glucose concentrations. PT, on the other hand, significantly reduced glucose

uptake in 6mM but not 25mM glucose.

In ID8-25 cells the combination treatment with PZ and CB did not have additive

inhibitory effects on glucose uptake in either normal or high glucose (Figure 16).

Regardless of transporter inhibition, these cells consumed more glucose in high-glucose

media than in physiological glucose, even though glucose was plentiful. Even in 6mM

glucose media, the total consumption represented less than a third of the total glucose

available.

Page 97: The effects of hyperglycemia on glucose metabolism in

83

Figure 15: SGLTs affect glucose transport in hyperglycemia in cells conditioned to

normal physiological glucose concentrations (ID8-6)

Glucose consumption by ID8-6 cells in normoglycemic and hyperglycemic culture

conditions. Cells were treated with 50µM PT or 200µM PZ in 0.01% DMSO. A) Total

glucose consumed in mg/dL. B) Fraction of glucose consumed relative to the initial

glucose concentration. C) Relative glucose consumption per cell. Bars are mean +

SEM, n≥3. Bars sharing the same letter are not significantly different (p<0.05).

Page 98: The effects of hyperglycemia on glucose metabolism in

84

Figure 16: SGLTs affect glucose transport in hyperglycemia in cells conditioned to high

glucose concentrations (ID8-25)

A) Total glucose consumed in mg/dL. B) Fraction of glucose consumed relative to the

initial glucose concentration. C) Relative glucose consumption per cell. Drugs were

dissolved in 0.01% DMSO. Combination treatment was 50µM PZ and 0.5µM CB. Bars

are mean + SEM, n≥3. Different letters indicate significant differences between bars

(p<0.05)

Page 99: The effects of hyperglycemia on glucose metabolism in

85

Glucose concentration does not alter the effect of glucose inhibitors on viability.

The metabolic viability of cells was measured using the WST-1 assay (Figure 17).

Phlorizin (PZ, SGLT inhibitor) had no effect on cell viability, while phloretin (PT, GLUT

inhibitor) potently decreased viability in a concentration-dependent manner in both ID8-

6 and ID8-25 cells. The combination of PZ and CB on ID8-25 cells had no effect on cell

viability.

Page 100: The effects of hyperglycemia on glucose metabolism in

86

Figure 17: SGLT inhibition does not affect cell metabolic viability

A) and B) Viability of mouse epithelial ovarian cells acclimatized to physiological

conditions. ID8-6, n≥3; C)-E) Viability of mouse epithelial ovarian cells acclimatized to

hyperglycemic conditions. Combination treatment (PZ+CB) is 50uM PZ and 0.5uM CB.

ID8-25, n=3. *p<0.05, **p<0.01, ***p<0.001. Bars are mean + SEM. Different letters, or

brackets with stars, indicate significant differences between bars (p<0.05)

Page 101: The effects of hyperglycemia on glucose metabolism in

87

Inhibition of the SGLTs does not affect wound healing in hyperglycemia

In “non-diabetic” ID8-6 cells, the GLUT inhibitor phloretin (PT) inhibited wound healing a

scratch assay in both normal- and high-glucose media (Figure 18). The SGLT inhibitor

phlorizin (PZ), however, only inhibited wound healing in 6mM glucose. The combination

of phlorizin and phloretin did not have an additive effect on the fraction of the scratch

wound that was filled in over 24 hours.

Page 102: The effects of hyperglycemia on glucose metabolism in

88

Figure 18: Phlorizin and phloretin inhibit scratch wound healing in ID8-6 cells

Images were analyzed to determine the fraction of the scratches covered by cells after

24 hours. Representative images of cells after 24 hours at 4x magnification. Dashed

lines indicate the width of the initial scratch. A) ID8-6 cells in 6mM glucose. B) ID8-6

cells in 25mM glucose. Combo is 100µM PZ + 100µM PT. Bars are mean + SEM, n=3.

**p<0.01, ***p<0.001.

Page 103: The effects of hyperglycemia on glucose metabolism in

89

Phlorizin increases the invasive capacity of ID8-6 but not ID8-25 cells

Cells conditioned to high glucose (ID8-25) were more invasive than ID8-6 cells

regardless of available glucose (Figure 19A, a vs b). However, SGLT channel inhibition

with PZ equalized the number of migrated cells across all experimental groups by

increasing ID8-6 cell invasiveness and decreasing (6mM) or maintaining (25mM) ID8-25

invasiveness (Figure 19A, black bars). Relative to cells treated with DMSO vehicle

control, ID8-6 cells treated with PZ were significantly more invasive than ID8-25 cells

(Figure 19B).

Page 104: The effects of hyperglycemia on glucose metabolism in

90

Figure 19: SGLT glucose transporters limit cell invasion in ID8-6 cells

Cells were cultured in the upper chamber of trans-wells coated with Matrigel and

allowed to migrate towards the lower chamber using FBS as a chemoattractant. Purple

staining indicates cells that have migrated through the Matrigel membrane. Bars are

mean + SEM. Different letters indicate significant differences within DMSO control

treatments (white bars) p<0.05. Phlorizin treatments (black bars) were not significantly

Page 105: The effects of hyperglycemia on glucose metabolism in

91

different from one another. (p<0.05). Stars indicate significant differences between

DMSO and PZ treatments within each cell line/glucose condition. *p<0.05, ***p<0.001.

A) Number of cells migrated through Matrigel, with representative images. B) Migration

of PZ-treated cells relative to DMSO control.

Knockdown of SGLT2 opposes contact-dependent growth inhibition

The SGLT2, the sodium-glucose transporter isoform present in our cells, was stably

knocked down using shRNA (Figure 20A). Cells with lowered SGLT2 expression had an

altered pattern of growth in culture where individual cells appeared to migrate away

from a group of cells, rather than multiplying to expand an existing group of cells (Figure

20B). KD cells were also resistant to contact-dependent growth inhibition and continued

to grow on top of a fully confluent monolayer of cells, forming three-dimensional foci

with cords of stacked cells projecting outward (Figure 20C).

Page 106: The effects of hyperglycemia on glucose metabolism in

92

Figure 20: Knockdown of SGLT2 in ID8 cells changes proliferative behaviour in culture

SGLT2 expression in ID8-6 and ID8-25 cells was knocked down using shRNA

packaged in lentiviral particles. A) Representative Western blots showing protein

expression of SGLT2 in cells treated with empty vectors (left) and shRNA constructs

(KD, right). Protein knockdown was evaluated by densitometry. ID8-6 cells had an

apparently complete SGLT2 knockdown by this technique and ID8-25 cells protein

expression was reduced by 65%. B) Representative images of cell migration in cultures

of KD clones at 4x magnification. C) Representative images of cell foci formed at high

confluency by KD clones.

Page 107: The effects of hyperglycemia on glucose metabolism in

93

GLUT1 protein expression appears greater in diabetic mice than in WT mice

In both tumours with and without SGLT2 expression (ID8-25), GLUT1 expression

appears qualitatively greater in Akt2 KO mice than in normoglycemic WT mice (Figure

21A and B). In mice whose ovaries were injected with SGLT2 KD cells, SGLT2

expression did not differ by genotype, but in WT mice appeared considerably higher

than GLUT1 expression (Figure 21C).

Page 108: The effects of hyperglycemia on glucose metabolism in

94

Figure 21: GLUT1 protein expression is higher the tumours of diabetic mice than in

those of WT mice

Representative images of tumour tissue stained by immunohistochemistry. A) GLUT1

expression in from Akt2 KO mice appears higher than in WT mice. B) In mice whose

tumours were initiated with ID8-25 SGLT2 knockdown cells, GLUT1 expression appears

higher in Akt2 KO than in WT mice. C) SGLT2 expression in mice with SGLT2

knockdown cells does not appear to differ between genotypes. (Not quantified).

Page 109: The effects of hyperglycemia on glucose metabolism in

95

Glucose is transported into orthotopic ovarian tumours by both GLUT and SGLT.

In order to determine the functional expression of GLUT and SGLT transporters in

murine EOC tumours, we injected radioisotope-tagged glucose analogues into the tail

veins of wildtype (WT) and Akt2 -/- (KO) mice four months PTI. Alpha-methyl-

glucopyranoside (14C-AMG) is specifically transported through the SGLT transporter

family and deoxyglucose (3H-DG) is specifically transported through the GLUT

transporter family. Both GLUT and SGLT transporter families contribute to glucose

uptake in tumours in approximately equal amounts (Figure 22). All tissues took up the

same total glucose per gram (data not shown). Total glucose uptake per gram was

unchanged in SGLT2 KD cells and a significant increase in the amount of GLUT-

mediated transport completely compensated for the loss of SGLT2 expression (Figure

23).

Page 110: The effects of hyperglycemia on glucose metabolism in

96

Figure 22: Both GLUT and SGLT transporters are responsible for functional glucose

uptake into tumours

Bars show the relative uptake of each isotope. Stars indicate significant differences in

the fractions of 3H-DG (transported by GLUTs) and 14C-AMG (transported by SGLTs).

Different letters indicate differences in uptake between tissues. (p<0.05). N=4.

Page 111: The effects of hyperglycemia on glucose metabolism in

97

Figure 23: SGLT2 KD impairs glucose uptake through active transport, but total glucose

uptake is maintained through compensation by GLUTs

A) A 3H-DG : 14C-AMG ratio above 1.0 indicates that there was more GLUT-mediated

glucose transport than SGLT-mediated transport. There were no significant differences

between control WT and Akt2 KO ratios in SGLT2 control mice. B) Total glucose uptake

per gram of tissue (sum of disintegrations per minute of 3H-DG + 14C-AMG divided by

tissue weight). Control n=4; KD n=1. Bars represent mean + SEM (control) or values

from the single animal with an SGLT2 KD tumour.

Page 112: The effects of hyperglycemia on glucose metabolism in

98

45 days after tumour induction, GLUT1 protein expression is upregulated in

diabetic mice with SGLT2 KD tumours.

At 45 days PTI we found no significant differences in the weights of tumours in any

experimental condition (Figure 24A). Western blot analysis showed that in Akt2 KO

mice, GLUT1 expression was upregulated beyond accurate quantification. Tumours

from mice that were injected with SGLT2 KD ID8 cells had the same SGLT2 protein

expression as those injected with control ID8 cells (Figure 24B and C).

Page 113: The effects of hyperglycemia on glucose metabolism in

99

Figure 24: Expression of GLUT1 is highly upregulated in tumours with reduced SGLT2

expression at 45 days PTI

Transformed mouse ovarian epithelial cells (ID8-25) were injected under the ovarian

bursa of wildtype (WT) and Akt2-/- (KO) mice. Injected cells had normal (control) or

knocked down (SGLT2 KD) expression of SGLT2. A) Mean tumour weight at 45 days

Page 114: The effects of hyperglycemia on glucose metabolism in

100

post tumour induction (PTI). Bars are mean + SEM n=4. Differences in tumour weights

were non-significant. p=0.065 between control and SGLT2 KD in WT mice. B) Protein

expression of glucose transporters in 45-day tumours. Each lane represents protein

from an individual animal. C) Densitometry of Western blots relative to β-actin loading

control. Bars are mean + SEM n≥3. ***p<0.001.

SGLT2 KD increases tumour weight and decreases overall survival

SGLT knockdown significantly increased tumour weight at the time of death in a cell

line-dependent manner (Figure 25). Tumours initiated with ID8-6 cells were associated

with decreased tumour weight after controlling for the variation due to the SGLT2 KD. In

a univariate analysis, however, cell type (ID8-6 / ID8-25) had no effect. Knockdown of

SGLT2 in ID8-25 cells increased tumour weight in WT mice, while SGLT2 KD in ID8-6

cells caused significantly larger tumours in the diabetic Akt2 KO mice.

Mice whose tumours had knocked-down SGLT2 expression had a relative risk of dying

of nearly 25 times. When considered in a multivariate model, after controlling for

variation in ID8-6 and WT the relative risk of dying was more than 57 times higher in

mice with SGLT2-deficient tumours (Figure 26). ID8-6 cells or a WT genotype also

increased the relative risk of death by 5 and 3.5 times respectively, although their

effects were non-significant if SGLT2 KD mice were removed from the model.

Page 115: The effects of hyperglycemia on glucose metabolism in

101

Figure 25: SGLT2 knockdown leads to significantly larger primary tumours at death

Tumours were removed and weighed when mice became moribund. Representative

images of A) wildtype mice and B) Akt2-/- mice and tumours. (ID8-25 cells, SGLT2

control and SGLT2 KD.) Bars represent mean + SEM. Different letters represent

significant differences between all groups (p<0.05). n=5 per group. SGLT2 KD

(p<0.0001) increases tumour weight. ID8-6 decreases (p=0.0364) tumour weight after

controlling for the variation due to KD, but without KD its effects are not significant.

Page 116: The effects of hyperglycemia on glucose metabolism in

102

Figure 26: SGLT2 KD in tumours significantly decreases overall survival

Solid lines indicate tumours developed from ID8-25 cells, dashed lines indicate those

from ID8-6 cells. The experiment was terminated at 126 days and mice that were

sacrificed at 126 days were considered to have lived. N=5 per group. Survival of ID8-25

control mice overlaps survival of ID8-6 control mice.

Table 4: Cox proportional hazard regression model for ovarian cancer survival

Variable RR p 95% CI lower

95% CI upper

SGLT2 knockdown 24.48 0.0019 - - Univariate

SGLT2 knockdown 57.26 0.0002 79.628 103.3 Multivariate

ID8-6 (vs ID8-25) 5.29 0.0053 66.946 95.498 Multivariate

Akt2 WT (vs KO) 3.45 0.0319 74.61 103.79 Multivariate

0 50 100 1500

50

100 Control, WT

SGLT2 KD, WT

Control, KO

SGLT2 KD, KO

Days PTI

Perc

en

t su

rviv

al

Page 117: The effects of hyperglycemia on glucose metabolism in

103

Table 5: Median survival times of mice with SGLT2 KD tumours

Experimental Group Median survival (days PTI)

WT KD ID8-6 69

Akt2 KO KD ID8-6 86

WT KD ID8-25 111

WT Control ID8-25 112

Akt2 KO KD ID8-25 115.5

Akt2 KO Control ID8-6 126

Akt2 KO Control ID8-25 126

WT Control ID8-6 126

Tumours deficient in SGLT2 have more severe secondary disease

The number and dispersal of secondary lesions plus the volume of ascites in tumour-

bearing mice constitute our assessment of secondary disease. Knockdown mice had

higher rates of metastasis (p>0.00001), but differences were not significant by ID8-6 or

genotype (Figure 27). The severity of secondary disease is associated with overall

survival. Widespread metastatic lesions are more consistently associated with morbidity

than tumour size (Figure 28).

Page 118: The effects of hyperglycemia on glucose metabolism in

104

Figure 27: Secondary disease is much more severe in mice with SGLT2 KD tumours

A) Representative images of advanced peritoneal disease. Secondary lesions (B, C)

were scored on a 4-point scale with 0 indicating absence of secondary disease and 4

indicating more than 10 lesions spread throughout the peritoneal cavity. Tumours with

SGLT2 knockdown have higher rates of metastasis (p>0.00001). Neither cell type (ID8-

6/ID8-25) nor genotype (WT/Akt2-/-) significantly affected number of secondary lesions.

Page 119: The effects of hyperglycemia on glucose metabolism in

105

Figure 28: Poor overall survival is associated with severe secondary disease,

regardless of primary tumour weight, in mice bearing SGLT2 KD tumours

Data represent the cohort of mice (WT or Akt2 KO) injected with ID8-6 or ID8-25 SGLT2

KD cells. Coloured circles represent primary tumour weights at death. The secondary

lesion scores for animals that died on a particular day are marked by blue squares.

Shorter survival times were generally associated with small primary tumours but high

secondary scores which indicate multiple metastatic lesions spread throughout the

peritoneal cavity. In mice that survived longer, there appears to be more variability in

both tumour weight and secondary scores.

60 75 90 105 120 135

0

100

200

300

400

0

1

2

3

4

ID8-25 KO

Secondary lesions

ID8-6 KO

ID8-25 WT

ID8-6 WT

Primary tumours

Days PTI

Tu

mo

ur

weig

ht

(mg

) Seco

nd

ary

sco

re

Page 120: The effects of hyperglycemia on glucose metabolism in

106

Discussion

Glucose uptake in epithelial ovarian tumours occurs through the coordinated

actions of active (SGLT) and passive (GLUT) transporters

Glucose can enter cells through two classes of transporters. The GLUT proteins

facilitate transmembrane diffusion and are upregulated in most cancers.227 The sodium-

glucose transporters (SGLT) can move glucose against its concentration gradient by

using the inward sodium concentration gradient maintained by the Na+/K+ pump. Here

we demonstrated for the first time that members of both transporter families are

expressed in epithelial ovarian cancer (EOC). ID8 murine EOC cells express the mRNA

of SGLT2, GLUT1 and GLUT3, all of which are high capacity glucose transporters.

GLUT1 and GLUT3 have previously been reported in EOC, however in our subsequent

experiments we decided to focus on the contributions of the GLUT1 isoform because of

its stronger association with tumour development.126

Very few data exist on the expression of the SGLTs in human cancers. Where it has

been examined, SGLT isoforms vary among tumour types: preliminary studies have

shown that SGLT1 is expressed in breast145,228, prostate145,228, head and neck229,

colon230, laryngeal231, pancreatic232, oral233, and ovarian cancers234 while SGLT2

expression has been noted in lung,235 pancreatic223, and prostate223 cancers. We used a

tissue microarray of serous ovarian adenocarcinomas from women to examine the

protein expression of SGLT1 and SGLT2. SGLT1 protein was expressed at low levels in

about 30% of serous adenocarcinoma samples, while SGLT2 was strongly expressed in

all samples (Figure 14). Neither isoform was correlated with tumour stage, but lower

Page 121: The effects of hyperglycemia on glucose metabolism in

107

expression was associated with more poorly differentiated tumours. Subsequent to

these experiments, another group investigated SGLT expression in ovarian cancer.

Contrary to our findings, this large study found that SGLT1 was overexpressed and was

an independent biomarker for poor prognosis. The reason for the differences in SGLT1

expression in the two sets of samples is puzzling considering that the histochemical

methodology and antibody used were the same.234 It is unclear whether or not they

looked for SGLT2 expression.

In order to determine the relative contributions of each class of transporters, we used

class-specific inhibitors of GLUTs and SGLTs. Inhibition of the GLUT transporters with

cytochalasin B or phloretin reduced glucose uptake in cells cultured in a physiological

glucose concentration (6mM), but was ineffective in hyperglycemic conditions (25mM

glucose). Conversely, inhibition of the SGLT transporters with phlorizin had greater

effects in high glucose than in normal glucose. Rather surprisingly, the combination of

SGLT and GLUT inhibitors did not have an additive effect on glucose uptake. We

hypothesize that this is a result of their reciprocal effects in high and low glucose:

because the GLUT transporters had low activity in high glucose, competitively inhibiting

them in these conditions would have had little additional effect on overall glucose

consumption. The same would be true of inhibiting SGLT transporters physiological

concentrations of glucose.

In vivo, GLUT-mediated uptake compensated for a lack of SGLT2. Mice whose tumours

had reduced expression of SGLT2 took up relatively less of the SGLT-specific glucose

analogue 14C-AMG than those with SGLT2. However, compensation from the GLUT

transporters maintained similar total glucose uptake (Figure 23). Expression of the two

Page 122: The effects of hyperglycemia on glucose metabolism in

108

families of transporters appears to be inversely associated. In colon cancer cells SGLT1

is downregulated following oncogenic transformation while GLUT1 is upregulated.230

Similarly, in renal epithelial cells hypoxia increases GLUT1 expression but concurrently

decreases SGLT1 and SGLT2.236 The influence of oxygen levels on SGLT expression

suggests that they may be regulated by HIF-1α expression, whose pro-tumourigenic

effects could explain the increased SGLT expression after oncogenic transformation.

In tumours from diabetic mice at 45 days PTI, we saw significant upregulation of GLUT1

protein in the SGLT2 KD group which matched the pattern of radioactive glucose uptake

observed at 120 days (Figure 24). However, despite confirmation of SGLT2 knockdown

in the tumour-initiating cells, we did not see a decrease in SGLT2 protein expression in

tumours at 45 days by Western blot. In WT animals, tumours initiated with SGLT2

deficient cells did not show changes in the expression of either SGLT2 or GLUT1,

although again there was an increase functional activity of GLUT transporters at 120

days PTI. In addition, although GLUT1 appeared to be significantly upregulated in

SGLT2 KD cells by Western blot at 45 days PTI, immunohistochemical analysis showed

the opposite pattern in tumour samples taken at morbidity (Figure 21). This may be due

to the relatively small size of these tumours, as well as the fact that they were taken at

animal death. We were unable to collect enough sample to stain for SGLT2 expression

in tumours initiated with SGLT2-competent cells. These results indicate that transporter

expression is not directly associated with uptake kinetics, and suggest that SGLT2

expression in non-cancerous cells in the tumour may obscure the knockdown that was

clearly seen in isolated cells in vitro.

Page 123: The effects of hyperglycemia on glucose metabolism in

109

The active sodium-glucose symporter SGLT2 is protective against cancer death,

primary tumour growth, and metastatic secondary disease in an orthotopic

mouse model of EOC

The uptake and phosphorylation of glucose are the most important steps in determining

glycolytic flux.94–96 Hypothesising that inhibited SGLT transport would slow tumour

growth, we stably knocked down the expression of the SGLT2 isoform in ID8 cells using

shRNA. Although SGLT1 has the capacity to take up glucose when SGLT2 activity is

compromised,237 Western blot analysis suggested that this isoform had limited

expression in the ID8-cell tumours. Although there was efficient uptake of the lentiviral

particles containing the shRNA against SGLT2, achieving knockdown was very difficult

particularly in ID8-25 (cells conditioned to hyperglycemia). If SGLT2 is more important in

cells in high glucose (ID8-25), then KD cells would be less likely to survive even if they

are puromycin resistant. Because we selected single cells that appeared most active in

the presence of puromycin, it is possible that we selected for cells that had lesser

knockdown due to the random integration of the lentiviral construct into the genome of

the cell.

The results of this study strongly opposed our hypothesis and rather than improving the

outcome of EOC in a mouse model, SGLT2 KD actually increased the relative risk of

dying by 57 times. We can only speculate on the mechanism behind these surprising

results. Interestingly, SGLTs appear to be protective in many cases: increasing overall

survival231 or disease free survival232, and higher expression in better differentiated

tissues.229,230 We expected that the ability for SGLT2 to take up glucose independently

of its extracellular concentration would lead to more aggressive disease. However, it

Page 124: The effects of hyperglycemia on glucose metabolism in

110

could instead have relieved some of the selective pressure for mutations or non-genetic

actions that would facilitate metastasis. If SGLT2 KD provides a strong impetus for cells

to move away from the primary tumour to gain access to resources in a more abundant

and less competitive environment, we would expect to see more cell motility and a

greater spread of secondary disease. Results from the mouse model (Figure 27), cell

invasion assay (Figure 19) and the movement of individual cells in culture (Figure 20B)

all seem to support this premise. Secondary disease is a better predictor of death than

primary tumour weight. The impact of SGLT KD in EOC may also be related to its role

as an ion transporter. Transport through SGLTs can be driven by protons as well as

sodium.130 The dysregulated metabolism of cancer cells results in high extracellular H+

concentrations that may favour SGLT-mediated glucose uptake.

There is an extensive body of evidence indicating that GLUT1 overexpression is

associated with aggressive disease. Overexpression of GLUT1 in EOC predicts

response to chemotherapy and shorter disease-free survival238 and expression

progressively increases with stage and grade suggesting it may be related to malignant

transformation.239,240 These data indicate that SGLT2 knockdown may be promoting

EOC growth indirectly by increasing GLUT1, which would suggest that SGLT somehow

limits GLUT upregulation.

In our experiments GLUT1 compensation did not increase overall glucose uptake, but

simply maintained the levels seen in tumours with SGLT2-competent cells. However,

we saw profound effects on EOC outcome, so the contribution of SGLT2 is not

necessarily a glucose-related phenomenon. A similar situation has been noted in

neurons, where SGLT1 expression appears to be important even in cells that have an

Page 125: The effects of hyperglycemia on glucose metabolism in

111

abundance of GLUT3 and more than sufficient glucose uptake capacity.130 The glucose-

sensing or water- and ion- transporting roles of SGLT could be the significant effectors

preventing oncogenesis.

In the mouse model, the level of systemic glucose played a limited role in the impact of

SGLT2 KD on cancer progression. Neither genotype nor cell type had significant effects

on their own, which highlights the dominant effect of SGLT2 KD, but they did influence

survival in multivariate analyses. After controlling for SGLT2 KD and cell type,

normoglycemic (wildtype) mice were more 3.5 times more likely to die than

hyperglycemic and hyperinsulinemic (Akt2 KO) mice. This could be an indication that

diabetic conditions support tumour growth, particularly when glucose transport is

dysregulated. Mice whose tumours were induced by injection of ID8-6 cells were more

than five times more likely to die than those with ID8-25 tumours after controlling for

SGLT2 KD and genotype. Part of the effect of ID8-6 over ID8-25 could be attributed to

the difference in amount of SGLT knockdown (Figure 20A).

The studies presented here show compelling data that SGLT2 acts as a tumour

suppressor. Knockdown of SGLT2 in tumour cells increased tumour size, severity of

secondary disease, and substantially reduced survival times. In vitro observations

complemented this finding and collectively indicated a protective role for SGLT2 in cell

proliferation and migration, particularly in hyperglycemic conditions. Taken together,

SGLT2 appears to be most powerful on cell motility/invasion. There are several

examples of things that have limited effect on primary tumour growth but that promote

migration and increase the formation of metastases, including L-lactate.105

Page 126: The effects of hyperglycemia on glucose metabolism in

112

The overexpression of SGLT2 in the tissue microarray and the ability of its inhibition to

promote tumour growth seem at odds with one another. However, protein expression

does not necessarily reflect membrane expression or the kinetics of glucose uptake. As

yet there have been no functional studies of SGLT-mediated glucose uptake in human

ovarian tumours. Moreover, the ability of GLUTs to compensate for reductions in SGLT-

mediated glucose uptake make these results hard to interpret.

Based on these findings, we would expect the use of SGLT2 inhibitors to be associated

with an increase cancer or cancer-related death. Safety studies of SGLT2-inhibiting

diabetes drugs have not shown prohibitively high incidences of cancer, although it is

acknowledged that these data are not conclusive.241

FDG may underrepresent the total amount of glucose uptake into cancer cells

The high accumulation of the glucose analogue FDG is used to differentiate highly

glycolytic tumour tissue from normal tissue. Currently, FDG is the only FDA approved

oncolytic tracer and more than 90% of oncological PET imaging uses FDG.161 However,

FDG is a poor substrate for SGLTs. Because FDG is not transported through the SGLT

transporters, this suggests that tumours fulfilling metabolic needs through SGLTs will

have weaker PET results that may contribute to a false-negative diagnosis. Moreover,

the results of this study suggest that low or falling SGLT transport may indicate more

aggressive disease.

Page 127: The effects of hyperglycemia on glucose metabolism in

113

GLUTs and SGLTs appear to coordinate glucose transport into cancer cells and have

differential importance in normal and high glucose environments. GLUT was able to

compensate for the decreased 14C-AMG uptake in SGLT KD tumours by raising its

uptake of 3H-DG. It would be interesting to see if SGLT transport could compensate in

the same way if the GLUTs were compromised.

Conclusions

GLUT1 expression is historically considered the most important glucose transporter in

cancer cells and is correlated with many markers of advanced disease. For the first

time, we show that a second class of transporter, SGLT, is not only expressed, but also

makes significant functional contributions to EOC progression.

Page 128: The effects of hyperglycemia on glucose metabolism in

114

CHAPTER 4: ANTIDIABETIC EFFECTS OF METFORMIN AS EOC

THERAPY

Introduction

Metformin is a commonly prescribed and inexpensive first-line diabetes treatment that

has shown promise as a novel cancer therapeutic. In epithelial ovarian cancer (EOC),

however, its antineoplastic effects are inconsistent across different study populations

and are confounded by the underlying interaction between cancer and diabetes.242

Some studies have shown that metformin decreases cancer risk in patients with

T2DM,243,244 while others have found no association with the risk of ovarian cancer.245

Similarly, some studies have shown that metformin improves the survival for ovarian

cancer patients with concurrent diabetes181,246,247 while others have found that

metformin use among diabetic EOC patients does not affect progression-free survival or

overall survival.180,248

Metformin lowers glucose in an insulin-independent manner by inhibiting hepatic

gluconeogenesis, increasing peripheral glucose uptake in skeletal muscle and

decreasing renal and intestinal glucose reabsorption (reviewed in 249). These effects are

primarily mediated through the AMPK energy sensing pathway and its downstream

inhibition of mTOR.250,251 Increases in AMPK activation in response to a high AMP:ATP

ratio signal an energy deficit and promote a catabolic state through mechanisms such

as autophagy and the inhibition of anabolic proteins. Pre-clinical studies have shown

that the AMPK pathway also mediates anti-proliferative effect of metformin in EOC.252

Metformin has a further range of effects on cancer cells through AMPK-independent

Page 129: The effects of hyperglycemia on glucose metabolism in

115

mechanisms that inhibit mTOR signaling253 and cell proliferation, and promote apoptosis

and cell cycle arrest.254,255 Figure 29 illustrates some of metformin’s cellular effects.

Metformin appears to be particularly toxic to p53 negative cells in colon cancer256 and

can selectively kill cancer stem cells in breast cancer.257 However, the greatest potential

of metformin as a cancer treatment may be its synergistic effect with various

chemotherapeutics.258 Metformin potentiates the effects of doxorubicin,257

paclitaxel259,260 and cisplatin261–264 and can resensitize previously chemo-resistant cells.

Authors have postulated that the mechanisms behind this cooperation include the

additive affects of chemotherapy on activation of AMPK259,262, the ability of metformin to

selectively target cancer stem cells that are not affected by chemotherapy alone257,

further cytotoxicity261, or though inhibition of the MAPK/ERK pathway, leading to

apoptosis.264 Combination with chemotherapy may allow therapeutic effects to be

reached at much lower concentrations than required by chemotherapy as a single

agent.265 The NIH clinical trials database lists three phase II and III trials recruiting to

look at ovarian cancer and metformin + paclitaxel and/or carboplatin.

Although metformin is an antidiabetic drug and diabetes is related to poor cancer

outcomes,199 very little research has examined metformin’s antineoplastic effects

through this mechanism. We hypothesized that despite metformin’s direct cellular

effects on EOC, its primary action in diabetic cancer patients is through the reduction of

systemic fuel availability. The goals of this study were to investigate both the effects of

metformin on diabetic vs nondiabetic populations in a mouse model of EOC, and the

effects of hyperglycemia on metformin action in vitro.

Page 130: The effects of hyperglycemia on glucose metabolism in

116

Figure 29: Cellular effects of metformin

Page 131: The effects of hyperglycemia on glucose metabolism in

117

Materials and Methods

Cell Culture

Mouse-derived epithelial ovarian cancer cells (ID8; kindly donated by Drs. Paul

Terranova and Kathy Roby, Kansas State University, Kansas, USA) were maintained in

DMEM supplemented with 10% fetal bovine serum (FBS), 1% antibiotic/antimycotic

(Gibco BRL, Burlington, ON) and 2% L-glutamine (Life Technologies, Burlington ON).

All experimental treatments were performed in serum-free DMEM.

Drugs

Phlorizin (PZ; Sigma, Oakville, ON) competitively inhibits glucose transport through

sodium/glucose cotransporters (SGLTs). Cytochalasin B (CB; Sigma, Oakville, ON)

inhibits glucose transport through the passive glucose transporters (GLUTs). Metformin

hydrochloride was purchased from Sigma (Oakville, ON).

Viability assay

The MTT assay measures cellular reduction of a tetrazolium salt to a coloured formazan

product in an NADH- and NADPH-dependent manner and reflects primarily glycolytic

activity. Cells were seeded at a density of 1500 cells/well in triplicate in 96 well plates

and allowed to attach overnight. Cells were then serum starved for 6 hours and treated

for 48 h in serum-free conditions. Following treatment, cells were incubated with MTT

Page 132: The effects of hyperglycemia on glucose metabolism in

118

(5mg/ml; Sigma, Oakville, ON) for one hour at 37°C and lysed with MTT lysis buffer

overnight at 37°C. Absorbance was quantified in a microplate reader at 570nm.

Scratch wound assay

To examine the motility of cells in different concentrations of glucose, ID8-6 and ID8-25

cells were grown to confluency in 24 well plates and serum starved for 6 hours. A plastic

pipette tip was used to remove an area of cells down the centre of the dish. Wells were

gently washed with PBS to remove detached cells before treatment. Phase contrast

images were taken at three marked points along the scratch at t=0 and t=24 hours.

Wound sizes were determined using ImageScope software and are presented as the

fraction of the original scratch covered by cells.

Glucose consumption and cell number

Glucose concentration in the cell media was measured using the glucose oxidase

method and reagents from Pointe Scientific (Canton MI, USA). After 24 hours of

treatment, glucose concentration in the sample wells was subtracted from the glucose

concentration in blank wells to determine the amount of glucose consumed. Relative

cell number was approximated by the absorbance of the crystal violet nuclear stain. To

account for any differences in the rate of cell proliferation, glucose consumption was

divided by relative crystal violet absorbance (590nm) to determine a per-cell uptake

comparison.

Page 133: The effects of hyperglycemia on glucose metabolism in

119

Animals

Wildtype c57bl/6 mice were purchased from Charles River Laboratories (Wilmington,

MA, USA), and c57bl/6 Akt2 null mice (https://www.jax.org/strain/006966) were

purchased from Jackson Laboratories (Bar Harbour, ME, USA). An Akt2 null mouse

colony was established from breeder mice and animals were genotyped prior to

beginning experiments. Animals were housed at the Central Animal Facility at the

University of Guelph and all experiments were conducted according to Canadian

Council on Animal Care guidelines. Mice were kept on a 12h light:dark cycle and had

free access to food and water. For all experiments, adult mice were between 12 and 24

weeks old at time of surgery. Metformin hydrochloride (Sigma, Oakville, ON) was

dissolved in sterile PBS and administered as a daily IP injection at either 50mg/kg/day

or 150mg/kg/day.

Mouse model of epithelial ovarian cancer

Spontaneously transformed murine epithelial cells from C57Bl/6 mice (ID8) were diluted

in 5µl phosphate buffered saline (PBS) and injected under the ovarian bursa of WT or

Akt2 null mice. This orthotopic, syngeneic mouse model of EOC closely replicates

ovarian serous adenocarcinoma in women. By 90 days post-tumour induction, mice

develop large primary tumours, secondary peritoneal lesions, and significant abdominal

ascites.10 In survival experiments, mice were sacrificed when they became moribund

due to the accumulation of ascites fluid.

Page 134: The effects of hyperglycemia on glucose metabolism in

120

Type 2 Diabetes Mellitus (T2DM)

C57BL/6 mice homozygous for a mutant Akt2 allele have poor glucose tolerance and

reduced insulin sensitivity that result in hyperglycemia and hyperinsulinemia.193,194

Obesity often accompanies T2DM and is independently associated with diabetes and

cancer. Akt2-/- mice are not obese so this model isolates the contributions of high

glucose and high insulin. Mice were genotyped using the following primers designed by

Jackson Laboratories: Akt2 wildtype reverse: TGCACAATCTGTCTTCATGCCAC; Akt2

common forward: ACCAACCCCCTTTCAGCACTTG; Akt2 mutant reverse:

TACACTTCATTCTCAGTATTGTTTTGC. Products are 110bp (wildtype), 277bp

(mutant), or 110bp and 277bp (heterozygote).

Food Consumption

Weekly food consumption was calculated by taking the initial weight of dry food and

subtracting food weight after 5 days. This value was divided by the number of mice in

the cage and the number of days in the measurement period to determine food

consumed per mouse per day.

Intraperitoneal glucose tolerance tests

Mice were fasted for four hours prior to intraperitoneal glucose tolerance tests (IPGTT).

After recording baseline blood glucose, mice were given an intraperitoneal injection of

glucose (1 mg/g body weight in saline solution at a volume of 10µl/g body weight).

Blood glucose measurements were taken at 15, 30, 60, and 120 minutes after glucose

Page 135: The effects of hyperglycemia on glucose metabolism in

121

challenge using a handheld glucose monitor (Freestyle Lite, Abbott Laboratories, Abbott

Park, IL). Values that exceeded the measurement range of the glucose monitor were

considered 27.8mM (maximum reading possible). Data are presented as the average

integrated area under the curve (iAUC) ± SEM. The iAUC is the area under the curve

relative to FBG at time 0 and was calculated using the trapezoidal method.

Tissue collection and preparation

Mice were fasted for 6 hours prior to sacrifice by cervical dislocation. Following

euthanasia, trunk blood was collected and fasting blood glucose was measured

immediately using a handheld glucometer. The remaining blood was allowed to clot

then centrifuged to obtain serum samples and stored at -80oC. Ovarian tumours were

removed, weighed, and divided into sections for subsequent analysis. Sections were

either fixed in 10% neutral buffered formalin (Fisher Scientific, Whitby, ON) for wax

embedding and tissue sectioning or flash frozen in liquid nitrogen and stored at -80oC

for protein extraction. Secondary disease was evaluated at time of sacrifice by ascites

volume and number of secondary lesions. Mice were scored on a 4-point scale with 0

indicating absence of secondary disease. Mice with scores of 0 and 1 were considered

“low” and scores of 2 and 3 were considered “high”.

Statistical Analysis

Data were analyzed using GraphPad Prism statistical analysis software. Results are

reported as mean ± standard error of the mean (SEM). Significance threshold was set

Page 136: The effects of hyperglycemia on glucose metabolism in

122

at 0.05 and p values are listed in the figure legends. Comparisons between means were

measured by two-way ANOVA followed by a Bonferroni correction, or by unpaired two-

tailed Student’s t-test, unless otherwise noted. Overall survival data are plotted on a

Kaplan-Meier survival curve and differences were determined by a log-rank (Mantel-

Cox) test.

Page 137: The effects of hyperglycemia on glucose metabolism in

123

Results

In vitro

Metformin affects systemic metabolism in diabetic patients but has little risk of

hypoglycemia in nondiabetics. To determine if there were also differential effects at a

cellular level in EOC cells, we tested the effects of metformin in physiological glucose

concentrations (6mM) and hyperglycemic concentrations (25mM). The MTT assay was

used to measure the metabolic viability of ID8, SKOV3, and normal human ovarian

epithelium (NOSE) treated with metformin. The concentration of glucose did not affect

the metabolic response to metformin (Figure 29A).

In previous experiments we have found that the chronic culture conditions of cells can

affect their response to treatment media with different concentrations of glucose. We

compared the effects of metformin in 6mM glucose media on cells chronically cultured

in normoglycemic conditions (-6 cell lines) and those cultured in diabetic conditions (-25

cell lines). The metabolic viability of cells acclimatized to hyperglycemic conditions was

significantly more sensitive to the effects of metformin. ID8-6 cells showed no significant

response to metformin until 80mM and SKOV3-6 were unaffected until 40mM.

Interestingly, the metabolic response of SKOV3-6 cells was identical to that of normal

ovarian epithelium (Figure 29B).

In ovarian cancer cells acclimatized to hyperglycemia (ID8-25, SKOV3-25; Figure 31,

part A) there were dose-responsive declines in viability that were significant after 5mM

and 2.5mM metformin respectively. These findings are in line with other studies which

have also found no direct toxicity (necrosis, apoptosis) in this dose range indicating that

Page 138: The effects of hyperglycemia on glucose metabolism in

124

it is at a therapeutic level.266 The viability of NOSE cells was only reduced at metformin

concentrations greater than 40mM.

The MTT assay measures NADH formed primarily through glycolysis compared to

oxidative phosphorylation. These results imply that the glycolytic flux in “diabetic” cancer

cells is more sensitive to inhibition by metformin.

Page 139: The effects of hyperglycemia on glucose metabolism in

125

Figure 30: Metformin inhibits EOC cell viability in high glucose

MTT absorbance was used to determine cell viability. A) Viability of mouse epithelial

ovarian cancer, human ovarian cancer, and normal human ovarian epithelial cells (ID8,

SKOV3, NOSE) in media containing physiological glucose (6mM) or high glucose

(25mM). B) Viability of cells chronically cultured in physiological (red, 6mM glucose) or

hyperglycemic (blue, 25mM glucose) conditions. Left: murine, right: human. Black

dashed line indicates cells derived from normal epithelial and cultured in physiological

glucose. n=3. Values are mean ± SEM. Stars indicate differences between cell lines at a

single metformin concentration. *p<0.05, **p<0.01, ***p<0.001.

Page 140: The effects of hyperglycemia on glucose metabolism in

126

Although metformin had a greater effect on metabolic viability in high glucose, cell

number appeared to be more affected in conditions of glucose deprivation (Figure 30A).

Consistent with this, the ability of metformin-treated cells to heal a scratch wound was

impaired in low but not high glucose (Figure 30B).

Page 141: The effects of hyperglycemia on glucose metabolism in

127

Figure 31: Metformin reduces cell number in glucose-deprived cultures

A) ID8-25 cells were cultured in different glucose concentrations for 48 hours with or

without 50mM metformin. Crystal violet absorbance at 590 nm was used as an indicator

of cell number. Bars are mean + SEM. Stars indicate significant differences from crystal

violet absorbance in 6mM glucose. Brackets indicate differences between treatments at

the same glucose concentration. n=2; **p<0.01, ***p<0.001. B) Scratch wounds were

made by clearing an area of cells with a pipette tip. Images were analyzed to determine

the fraction of the scratches covered by cells after 48 hours of treatment with 5mM

metformin. Bars are mean + SEM, n=4. **p<0.01, ***p<0.001.

Page 142: The effects of hyperglycemia on glucose metabolism in

128

Metformin significantly increased cellular glucose consumption regardless of

environmental glucose availability (Figure 31). To determine whether glucose uptake

capacity contributed to this increase in glucose uptake, we used glucose transport

inhibitors to both the passive (GLUT; cytochalasin B) and active (SGLT; phlorizin)

glucose transporters. In low glucose, total glucose uptake was still enhanced compared

to control in the presence of glucose transport inhibitors. In high glucose, GLUT

inhibition abrogated this effect and glucose uptake was unchanged with cytochalasin B

treatment (Figure 32A). On a per-cell basis the increases in glucose consumption were

reduced, however a statistical trend remained (p<0.09) (Figure 32B).

Page 143: The effects of hyperglycemia on glucose metabolism in

129

Figure 32: Metformin increases cellular glucose consumption

Cells were treated with 5mM metformin in media containing normal (6mM) and high

(25mM) concentrations of glucose. A) Total glucose consumed in mg/dL B) Relative

glucose consumption per cell. C) Fraction of glucose consumed relative to the initial

glucose concentration in the cell media. Bars are mean + SEM, n≥5. Different letters

represent significant differences between bars (p<0.05).

Page 144: The effects of hyperglycemia on glucose metabolism in

130

Figure 33: Metformin-mediated increase in glucose consumption is independent of

glucose transporter expression

Cells were treated with 5mM metformin dissolved directly into culture medium, 50µM

phlorizin (PZ in 0.01% DMSO; SGLT inhibitor), or 0.5µM cytochalasin B (CB in 0.01%

DMSO; GLUT inhibitor) in media containing normal and high concentrations of glucose.

A) Total glucose consumed in mg/dL. B) Relative glucose consumption per cell. Bars

are mean + SEM, n≥4. *p<0.05, **p<0.01, ***p<0.001 between metformin and vehicle

control.

Page 145: The effects of hyperglycemia on glucose metabolism in

131

In previous work we have shown that EOC progresses faster in both type 1 and type 2

diabetic mice than in nondiabetic animals. We predicted that normalizing blood glucose

in diabetic animals would improve EOC outcome by reducing the amount of systemic

glucose available for the tumour to consume. Metformin has also been shown to have

beneficial effects in normoglycemic preclinical models through direct effects on cellular

signalling pathways. In order to assess the effects of metformin on EOC tumour growth

we used induced tumours in both wildtype (WT) mice and a genetic model of non-obese

mice with a T2DM hyperglycemic, hyperinsulinemic phenotype (Akt2 KO).

Three in vivo studies were performed using a mouse model of ovarian cancer

developed in our lab. Briefly, syngeneic ID8 cells were injected orthotopically beneath

the ovarian bursa of C57bl/6 mice. These cells develop into disease that closely

replicates human serous ovarian cancer. Tumour development over 30, 60 and 90 days

is representative of EOC Stages 1, 2 and 3 respectively.

In population studies, the use of metformin has been correlated with a lower risk of

developing cancer. The first study was designed to evaluate the ability of chronic

metformin administration to prevent tumour growth. WT and Akt2 KO mice received

daily IP injections of either metformin or PBS control at 50 mg/kg body weight between

two weeks after surgery until 90 days PTI. Metformin had no effect on fasting blood

glucose in WT mice. In KO mice, although FBG dropped significantly over time post-

tumour induction, the effects of metformin and PBS were identical (Figure 33B). At 90

days PTI, the weight of tumours in the metformin treated mice was no different than

those in PBS treated mice within each genotype (Figure 33A). In contrast to previous

experiments which showed that Akt2 KO mice grew significantly larger tumours than

Page 146: The effects of hyperglycemia on glucose metabolism in

132

WT mice 90 days PTI (Figure 7A), a one-way ANOVA of control and metformin in both

genotypes (comparison between Figure 33As) in this trial showed no significant

differences in tumour weight. Tumour initiation in this trial was weak and the lack of a

genotype-related effect was probably due to the very small size of these tumours.

Page 147: The effects of hyperglycemia on glucose metabolism in

133

Figure 34: Small tumours normalize blood glucose in diabetic mice

Tumours in wildtype and diabetic (Akt2 -/-) mice were treated with daily IP injections of

50mg/kg/day metformin (2 weeks PTI to 60 days PTI) then 150mg/kg/day (60 until 90

days PTI). A) Metformin treatment did not affect tumour. B) Fasting blood glucose. Bars

are mean + SEM, n=5. *p<0.05, **p<0.01, ***p<0.001.

Page 148: The effects of hyperglycemia on glucose metabolism in

134

The human dose is of metformin is approximately 35mg/kg/day orally.267 In our

experiments, the original dose of 50 mg/kg/day IP was chosen based on literature at the

time which used this dose in chronic administration.268 However, because this dose did

not affect the blood glucose in the Akt2 KO mice as we had expected, we increased the

daily dose of metformin to 150 mg/kg/day in a second study.263 In this second study, we

again began metformin treatment at two weeks PTI in order to evaluate the effects of

metformin on tumour growth prevention. Mice were sacrificed when they became

moribund. We found no differences in the length of survival or tumour weight at survival

in WT mice treated with either metformin or PBS control (Figure 34A and B).

Qualitatively there was a slight reduction in the number of secondary lesions, but our

scoring system for evaluating secondary disease did not allow us to determine statistical

significance from these data (Figure 34C).

We measured food consumption to determine whether the drug was affecting total

glucose intake and found that metformin had no effect (Figure 35A). In PBS treated

mice, the presence of the developing disease resulted in significant fluctuations in

fasting blood glucose. Metformin appeared to protect against this variability (Figure

35B). We performed IPGTTs in mice with and without tumours to determine whether

metformin alone could affect glucose tolerance. Metformin had no effect on the glucose

tolerance of mice with tumours, but surprisingly it significantly worsened glucose

tolerance in mice without tumours (Figure 35C and D).

Page 149: The effects of hyperglycemia on glucose metabolism in

135

Figure 35: Daily metformin treatment does not affect EOC outcomes in WT mice

A) Tumours were removed and weighed when mice became moribund. Values are

mean + SEM, control: n=9, metformin: n=6. There was no significant difference in

tumour weight at death between metformin- or control-treated mice. B) Kaplan-Meier

survival curve of WT mice with ovarian cancer that received 150mg/kg daily metformin

in PBS IP treatment from two weeks PTI until death. There is no statistical difference in

survival between groups by log-rank (Mantel-Cox) test. C) Secondary lesions and

ascites volume. Values are proportion of mice with “high” or “low” scores.

Page 150: The effects of hyperglycemia on glucose metabolism in

136

Figure 36: Metformin treatment in WT mice with tumours impairs glucose tolerance over

time.

WT mice bearing tumours were treated with daily metformin injections (150mg/kg)

beginning two weeks PTI. A) Food consumption at 2, 7 and 10 weeks PTI (0, 5 and 8

weeks of metformin treatment). Bars are mean + SEM, n=5. B) Fasting blood glucose

Page 151: The effects of hyperglycemia on glucose metabolism in

137

between surgery (0 weeks) and death (†). Bars are mean + SEM, n=5; *p<0.05 vs

baseline. Comparison of FBG from baseline is by two-way ANOVA with Dunnett’s post-

hoc test. C) Results of intraperitoneal glucose tolerance tests (IPGTT). ***p<0.001 vs

control at a specific time point. D) Integrated area under the curve (iAUC) of IPGTTs.

Glucose tolerance was impaired at 90 days PTI vs 60 days PTI in metformin-treated

mice with tumours. *p<0.05. Control: n=9, metformin: n=6.

Page 152: The effects of hyperglycemia on glucose metabolism in

138

In diabetic mice (Akt2 KO), there were no differences in tumour weight, length of

survival or severity of secondary disease (Figure 36). Mice in this trial died earlier than

expected (Figure 36B), likely due to tumour initiation with high passage cells that

accelerated disease progression. Food consumption decreased over time as EOC

progressed, but there were no differences between PBS and metformin treatments

(Figure 37A). In IPGTTs, metformin had no effect on glucose tolerance regardless of the

presence of a tumour. However, in all Akt2 KO groups, glucose tolerance improved with

time and the addition of metformin did not add to these effects (Figure 37D). Mice

without tumours that were treated with metformin showed the same improvement in

glucose tolerance as untreated mice with tumours. As in the wildtype mice, although

there were clear differences in glucose tolerance between groups, FBG was more

variable (Figure 37B).

Page 153: The effects of hyperglycemia on glucose metabolism in

139

Figure 37: Daily metformin treatment does not affect EOC outcomes in Type 2 diabetic

mice

Tumours were removed and weighed when mice became moribund. Values are mean +

SEM, control: n=14. There was no significant difference in tumour weight at death

between metformin- or control-treated mice. B) Kaplan-Meier survival curve of WT mice

with ovarian cancer that received 150mg/kg daily metformin in PBS IP treatment from

two weeks PTI until death. There is no statistical difference in survival between groups

by log-rank (Mantel-Cox) test. C) Secondary lesions and ascites volume. Values are

proportion of mice with “high” or “low” scores.

Page 154: The effects of hyperglycemia on glucose metabolism in

140

Figure 38: The presence of a tumour or treatment with metformin both improve glucose

tolerance in diabetic mice

Akt2 KO mice bearing tumours were treated with daily metformin injections (150mg/kg)

beginning two weeks PTI. A) Food consumption at 2, 4 and 7 weeks PTI (0, 2 and 5

weeks of metformin treatment). Bars are mean + SEM, n=5. B) Fasting blood glucose

Page 155: The effects of hyperglycemia on glucose metabolism in

141

between 3 weeks PTI (1 week of metformin treatment) and 8 weeks PTI (six weeks of

metformin treatment. Bars are mean + SEM, n=5; Brackets indicate significant

differences between bars. *p<0.05. C) Results of intraperitoneal glucose tolerance tests

(IPGTT). D) Integrated area under the curve (iAUC) of IPGTTs. In Akt2 KO diabetic

mice, glucose tolerance between 30 and 60 days PTI was improved by the presence of

tumours. Mice that underwent a sham surgery showed improved glucose tolerance with

metformin treatment *p<0.05. Control: n=9, metformin: n=6.

Page 156: The effects of hyperglycemia on glucose metabolism in

142

In the third study, we investigated whether metformin administered to WT mice with

established tumours (from 60d PTI to 90d PTI), could have an effect on disease

progression/regression. There were no differences in tumour size.

Figure 39: Metformin treatment does not reduce the weight of established tumours

Wildtype mice were allowed to grow tumours for 60 days before 30 days of daily

treatment with 150mg/kg/day metformin IP. Bars are mean tumour weight at 90 days

PTI + SEM, n=5.

Page 157: The effects of hyperglycemia on glucose metabolism in

143

Discussion

Preclinical evidence suggests that the glucose-lowering drug metformin is effective in

limiting EOC growth in vitro. The OCT1 transporter is necessary for metformin entry into

cells and is expressed in the human ovarian cancer cells lines SKOV3 and OVCAR3, as

well as in malignant cells in ovarian tumours.269 Metformin can inhibit proliferation,

migration, invasion, and adhesion in EOC monolayer culture as well as inhibit the

formation of tumour spheres, induce cell cycle arrest and increase apoptosis.260–263,270

We have previously shown that both Type 1 and Type 2 diabetes significantly

accelerate the growth of ovarian tumours in a mouse model of EOC. The goal of this

study was to determine if EOC progression can be prevented or reversed by lowering

blood glucose. Metformin is a widely prescribed antidiabetic drug which has recently

been shown to improve both the risk and outcome of a number of cancers. By inducing

ovarian cancer in both WT and diabetic (Akt2 KO) animals, we hoped to identify any

protective effects of metformin through its action in lowering systemic hyperglycemia as

well direct its effects on EOC cells.

Cellular effects in different glucose levels

The effects of metformin appear to be dependent on the level of environmental glucose.

Metformin reduced cell number in restricted glucose but not hyperglycemia (Figure 30A)

and it inhibited wound healing in normal but not high glucose (Figure 30B). These

findings are consistent with other studies that have shown high glucose impairs or even

reverses the anti-cancer effects of metformin.271,272 When treated with metformin, cells

Page 158: The effects of hyperglycemia on glucose metabolism in

144

in 5mM glucose show gene expression changes in pro-tumorigenic pathways; this effect

is abolished cells in high glucose (10mM or above).216 This is perhaps not surprising

because many of metformin’s effects are dependent on AMPK activation, which also

responds to the AMP:ATP ratio. A low AMP:ATP ratio indicates high energy availability

and would oppose the effects of metformin, meaning that the same drug dose in high

glucose would be less effective at promoting AMPK-mediated anti-neoplastic effects.

Several studies have suggested that increased metformin efficacy in low glucose is due

partly to downregulation of the glycolytic enzyme pyruvate kinase M2 (PKM2) in these

conditions.271,273,274

The effects of metformin on a systemic level are mostly concentrated in the liver and

skeletal muscle where AMPK activation increases glucose uptake and decreases

gluconeogenesis to reduce blood glucose. In vivo, we evaluated metformin’s effect on

tumour weight and overall survival. Although metformin has direct effects on cells, we

anticipated that when given systemically in hyperglycemic mice, metformin’s anti-cancer

effects would be mediated primarily through its ability to reduce blood glucose.

In our model, metformin was unable to oppose tumour growth in either normal or

hyperglycemic mice. The doses of metformin that we used (50mg/kg/day and

150mg/kg/day) did not reduce blood glucose and so we cannot draw conclusions on the

effect of lowering glucose on the progression of EOC. In a pilot study, we administered

up to 250 mg/kg/day IP in an effort to reduce blood glucose, however within four days of

this dosing three mice had died and we reduced the dose to 150mg/kg/day. However,

the lack of effect of metformin on tumour growth in normoglycemic mice suggests that it

has limited influence directly on cancer cells. This finding is in disagreement with a

Page 159: The effects of hyperglycemia on glucose metabolism in

145

number of studies of EOC that demonstrate metformin can reduce primary tumour

growth and proliferation, and the formation of metastases.260,262,263,266 Although the

metformin doses in these studies were similar to what we used, these studies were all

performed with subcutaneous xenografts in immunocompromised mice. Both the

ovarian microenvironment10 and immune-related inflammation43 significantly impact

tumour biology, which could explain why metformin treatments were ineffective in our

orthotopic model.

Litchfield and others used a similar ID8 orthotopic model and found that metformin

inhibited tumour growth in normoglycemic mice but not diet-induced hyperglycemic

mice.272 In their study, metformin treatment began three weeks prior to tumour induction

and the tumours that developed were much smaller than what we typically see (~40mg

in untreated normoglycemic mice and ~90mg in untreated hyperglycemic mice vs

~150mg and ~350mg in our mice). Taken together with our findings, these data suggest

that metformin’s effects against EOC are primarily preventative rather than reductive.

These same conclusions have been drawn in colorectal cancer where metformin has

been found to limit tumour initiation but have no effects on existing tumours.275

Alterations in angiogenesis may provide the mechanism for this limited tumour growth.

Metformin can decrease VEGF and/or microvessel density262,276 and in a breast cancer

model it has been shown to reduce tumour angiogenesis, but not affect tumour

volume.270

Page 160: The effects of hyperglycemia on glucose metabolism in

146

Metformin and reprogramming of cellular metabolism

Several studies contend that metformin's inhibitory effects on cancer progression are

cancer cell autonomous and AMPK-independent, instead acting through inhibition of

mitochondrial enzymes to decrease cellular respiration and promote a compensatory

increase in glucose uptake and glycolytic flux.277–280 We found that although short-term

changes in environmental glucose did not affect viability with metformin treatment

(Figure 29A), chronic culture in high glucose was associated with reduced viability, i.e. a

decrease in glycolytic flux (Figure 29B). A highly glycolytic phenotype is generally

related to more aggressively malignant cells, but forcing glycolysis also increases the

tumour’s demand for glucose and can make cells acutely sensitive to glucose

deprivation. The sensitivity of cells to metformin is dependent on their ability to cope

with energetic stress.280 In the presence of glucose, metformin can suppress the overall

efficiency of metabolism to inhibit the proliferation of human colon and thyroid

cancers.271 When glucose becomes a limiting factor for metabolism, this metformin-

induced metabolic inefficiency induces cell death.271,277,279 The higher rate of glucose

consumption we demonstrated with metformin treatment (Figure 31B) may suggest that

metformin increased glycolytic activity.

Dosing

Drug repositioning often means using different concentrations to elicit different

physiological responses. In diabetes, the effective dose for metformin has maximal

pharmacological effect on tissue-specific metabolism in the liver and skeletal muscle,

but minimal systemic toxicity. Using metformin in cancer treatment may require a

Page 161: The effects of hyperglycemia on glucose metabolism in

147

different dose and different dosing schedule that induces both pharmacological and

toxic effects.281 For example, metformin at a concentration that effectively upregulates

pAMPK may have no influence on decreasing mammary cancer severity in nondiabetic

rats and mice.282 There are also large differences in the drug concentrations that are

effective over different time periods. One group refers to the “metformin paradox” where

long term metformin treatment over weeks or months requires only micromolar

concentrations of metformin to elicit anti-cancer effects, whereas short term (12-48

hour) experiments need higher drug concentrations far beyond the human therapeutic

dose of 15-17 µM.216 It is possible that rather than having a dose-responsive effect

against cancer, there is a threshold concentration for drug effect that 150mg/kg/day did

not reach which could partially explain the lack of effect that we saw in our in vivo

models.

Conclusion

This study does not support a role for single-agent metformin treatment of EOC in either

physiological or diabetic environments. We were unable to evaluate its effect on tumour

growth through reduction in blood glucose, however others have shown that in diabetic

animals, combined treatment of both anti-tumour and anti-hyperglycemia therapies give

the best tumour-reductive outcome.213 Our in vitro findings support reports that high

environmental glucose inhibits metformin’s anti-cancer effects. Taken together with our

previous work that showed markedly worse EOC outcomes in diabetic mice, we

conclude that glycemic control is an important consideration in EOC management. In

diabetic mice the lower cell-level efficacy of metformin in hyperglycemia could be offset

Page 162: The effects of hyperglycemia on glucose metabolism in

148

by the fact that it reduces this hyperglycemia through actions on the liver and skeletal

muscle. If there is a role for metformin in the treatment of EOC, the variability of its

effects in preclinical models has shown that it will be essential to identify the subset of

patients likely to respond, which could include evaluation of systemic glucose

homeostasis, concurrent therapies, OCT1 transporter expression283 and oncogenic

mutations.284

Page 163: The effects of hyperglycemia on glucose metabolism in

149

CHAPTER 5: GENERAL DISCUSSION

Overview

Glucose is an important energy substrate, survival factor, and pro-angiogenic molecule.

This thesis examines the influence of high glucose availability on the growth of epithelial

ovarian tumours, and attempts to the regulation of systemic glucose can be exploited

therapeutically in cancer treatment.

Since beginning this work, a number of significant discoveries have altered our

understanding of cancer cell metabolism and tumour metabolism. In 2011, Hanahan

and Weinberg updated their seminal “Hallmarks of Cancer” to include “reprogramming

energy metabolism.”43 Hexokinase52,98,99 and the PKM2 fetal isoform of pyruvate

kinase91 have emerged as key drivers of the Warburg effect. The sodium glucose

transporter 1 (SGLT1) was found to be expressed in cancer cells and physically

stabilized by EGFR, suggesting a role for EGFR in glucose balance.145,228 Perhaps most

significantly, the meaning of abnormal “cancer metabolism” has expanded to include

both cellular and intercellular characteristics. A metabolic symbiosis has been

discovered between oxygenated and hypoxic cancer cells as they share lactate to

maximize energy production.119 In addition, observation of a “Reverse Warburg” effect

demonstrated an essential role for metabolically reprogrammed cancer-associated

fibroblasts.116 Disrupting the unique energy balance of both cancer cells and tumours

have direct anti-cancer consequences.

Advances in the efficacy of EOC treatment have come from stratifying patients by

disease type on histological and genetic levels. However, neither of these takes into

Page 164: The effects of hyperglycemia on glucose metabolism in

150

account the functional bioenergetic heterogeneity inherent at the levels of the cell,

tumour and body. As the results presented here demonstrate, tumours with the same

histologic and genetic characteristics can behave differently in different glucose

conditions.

As studies move toward the development of clinical applications, research will need to

consider another level of complexity—interaction with the systemic environment. The

results of this thesis demonstrate pro-tumorigenic roles for both long-term and short-

term exposure to systemic hyperglycemia and implicate common metabolic

comorbidities of EOC, particularly diabetes, as significant challenges to address. Can

systemic metabolic dysfunction mask cancer diagnosis or monitoring, particularly using

FDG-PET? Can it increase cancer aggressiveness and lead to poorer prognoses by

providing an unlimited fuel supply? Do standard treatments for diabetes, like insulin or

lactate infusions, have detrimental effects by enhancing tumour growth or interfering

with cancer treatments?

The key findings of this thesis are that:

1. Hyperglycemia promotes EOC independent of insulin;

2. Tumour development ameliorates dysfunctional systemic metabolism;

3. Knockdown of the active glucose transporter SGLT2 promotes EOC spread;

4. Secondary disease predicts death independent of primary tumour size; and

5. Anti-neoplastic effects of metformin are highly glucose-specific.

Page 165: The effects of hyperglycemia on glucose metabolism in

151

Figure 40: EOC metabolism is a product of cell genotype, the tumour microenvironment,

and systemic glucose conditions

The metabolic syndrome, diabetes, and EOC

The metabolic syndrome is defined by the pathologic triad of hyperglycemia and

hyperinsulinemia (T2DM), and obesity. It is a significant and growing population health

concern with as much as one third of the population affected. The syndrome has health

Page 166: The effects of hyperglycemia on glucose metabolism in

152

effects on multiple organs and is related to various vascular and microvascular

diseases.198 Its effect as both a risk factor and poor prognostic factor in cancers is

undeniable but poorly understood.199 In some studies, ovarian cancer has been linked

to the metabolic syndrome and each of its three conditions independently.178,179

The effects of mitogenic insulin signalling are thought to be the primary drivers of this

association. However, insulin-independent mechanisms may also contribute, particularly

through energy-sensing pathways and glucotoxic damage. Toxicities of excess glucose

could benefit tumour progression by facilitating cellular and genetic damage through the

development of an acidic environment, transient hypoxia, oxidative stress, and glycation

including the development of advanced glycation end products and their receptors

(AGE/RAGE).186,285 Taken together hyperglycemia provides ongoing genomic instability,

one of the hallmarks of cancer.43

Dysregulated systemic metabolism can also affect cancer cell metabolism. The aerobic

glycolysis in many cancer cells is associated with more aggressive malignancies in

vitro.48,121 Highly glycolytic cells demonstrate the Crabtree effect where glucose induces

a further shift away from oxidative energy production. Diabetes, particularly T1DM, is

associated with a “thrifty” genotype where cells are genetically or epigenetically

reprogrammed take up more glucose.286 It is possible that this is a risk factor for cellular

transformation or a promotional influence on tumour development.

The metabolic syndrome is primarily a consequence of lifestyle and thus understanding

its connections to cancer can potentially indicate modifiable risk factors. In ovarian

cancer, where the etiology is still poorly understood and treatment of advanced disease

Page 167: The effects of hyperglycemia on glucose metabolism in

153

is still largely ineffective over the long term, this would be a significant finding.

Moreover, researchers have suggested that the effects of diabetes can be extrapolated

to all cancer patients as the associated risks may occur within the normoglycemic and

insulinemic ranges.184

Diabetes treatment and cancer

Treatments for diabetes and cancer can interact in detrimental ways. Anti-

hyperglycemic treatments inducing elevated plasma insulin (sulfonylureas) seem to

increase cancer risk but insulin-sensitizers (metformin, thiazolidinediones) seem to

reduce cancer risk.242,287,288 Some groups have proposed that the increased risk of

cancer in diabetics is not attributable to insulin itself, but rather to the effects of insulin

insensitivity on the dysregulation of AMPK signalling or the action of insulin-like growth

factor-1 (IGF-1)-cholesterol synthesis pathway.202

On the other hand, patients with diabetes tend to receive less aggressive cancer

treatment.289 Diabetes is associated with higher infection rates177 and compared with

their nondiabetic counterparts, cancer patients with pre-existing diabetes are

approximately 50% more likely to die after surgery.290 This high operative morbidity may

limit the number of diabetic patients having comprehensive surgical staging179 and

explain why diabetic patients are more often diagnosed with higher tumour stages.289

Page 168: The effects of hyperglycemia on glucose metabolism in

154

Coordinated metabolism in the tumour microenvironment

Transformed cells are one part of a complex and heterogeneous tumour

microenvironment that plays an essential role in cancer progression.114 Our lab has

shown that the interaction of transformed ovarian epithelial cells with the ovarian stroma

is crucial to the development of highly aggressive tumours in the mouse model

described in this thesis.10 The microenvironment, particularly its oxygen levels, also

influences metabolism.121 While the Warburg effect describes genetic adaptations of

tumour cells, the Crabtree effect (inhibition of oxphos and promotion of glycolysis in high

glucose) describes reversible, short term adaptations to changes in energy and oxygen

balance which may occur at protein or epigenetic levels. In the 1960s, the Crabtree

effect was considered characteristic of cancer cells and part of the malignant

transformation.291 Although the Crabtree effect is not often mentioned in contemporary

literature, in addition to the Warburg effect it is important to consider this as part of the

microenvironment-metabolism interaction.217,292

The reverse Warburg effect demonstrates a central role of reprogrammed cancer-

associated fibroblasts (CAFs) in tumour metabolism.116 Similarly, lactate is shared in a

metabolic symbiosis between oxic and hypoxic cancer cells within a tumour.119 The in

vitro studies in this thesis show that increased environmental glucose can influence

cells to increase their glucose uptake. However, in vivo tumours are poorly perfused

tissues. Interestingly, the reverse Warburg effect shows that tumours can enrich their

own microenvironment and thus gain some autonomy from systemic circulation and the

need for tumour vascularization or angiogenesis.105 This could have implications for the

effectiveness of antiangiogenic therapies or chemotherapy delivery to the tumour. In

Page 169: The effects of hyperglycemia on glucose metabolism in

155

hyperglycemic conditions, tumours are also able to increase glucose uptake by

promoting glucose transfer across the capillary walls into the IF, which occurs without

the need for glucose transport proteins.214 Glucose can accumulate within the tumour

tissue to potentially create a locally hyperglycemic environment, which could explain the

ability of passive GLUTs to compensate for lack of active SGLT2-mediated glucose

transport (Chapter 3).

Intersection of tumour metabolism and systemic metabolism

Just as cellular growth occurs within the tumour microenvironment, tumour growth

occurs within the larger ecosystem of the host which varies in its own systemic

metabolism. In this thesis we hypothesized that high blood glucose levels would

promote tumour growth by providing an abundance of energetic fuel. However, the

relationship between systemic and tumour glucose control is reciprocal and cancer itself

can drive changes in systemic metabolism. We saw that tumours in our animal model

actually improved the diabetes of both STZ and Akt2 KO mice. In T1DM mice lacking

insulin, the presence of a tumour improved glucose tolerance (Figure 6). In

hyperinsulinemic Akt2 KO mice, tumours improved insulin sensitivity (Figure 8A and B).

In fact, tumours appeared to be as effective as metformin at normalizing glucose

tolerance in T2DM mice (Figure 37D).

The high glucose demands of tumour tissue might suggest that it acts as a glucose sink

and improvements in systemic glucose would be due to a large tumour mass. However,

the tumours in the chronically diabetic STZ mice with improved glucose tolerance were

Page 170: The effects of hyperglycemia on glucose metabolism in

156

smaller than tumours in control mice (Figure 4A). This could indicate that the tumour is

influencing systemic glucose indirectly through secreted factors in the blood.

Hypoglycemia has been noted in many cancer patients and is thought to be due to a

combination of cachexia, renal and hepatic impairment, excessive glucose consumption

or decreased glucose production293 which can be resolved with removal of the

tumour.294 However, the body is able to compensate for much of the tumour’s effect, in

some cases overcompensating,295–297 by increasing glycogenolysis and

gluconeogenesis from non-carbohydrate sources212,298 This compensation at the site of

the liver could lead to relative hyperglycemia in the hepatic veins and might contribute to

the fact that the liver is such a common site of metastasis (Figure 27A). Similarly, we

saw that the intestines were a major site of metastasis. Aside from being well perfused,

metastatic cells in this location may co-opt dietary glucose as it is absorbed.

High blood glucose, or an associated hyperglycemic peritoneal fluid, could promote the

survival of circulating tumour cells with metastatic potential. If high blood glucose is

cancer-promoting, then it might be expected that haematological malignancies would be

most acutely accelerated in diabetic conditions, however there is no published data

looking at these effects.

Diabetes has similar influences on the body as cancer: cachexia, heightened

gluconeogenesis concomitant with an increase in liver mass, and dysregulation of

insulin and glucagon levels. At a cellular level diabetes induces similar host

compensation: impaired oxphos leads to increased glucose tolerance, reduced fat

mass, and increased insulin sensitivity, which oppose the progression of the metabolic

Page 171: The effects of hyperglycemia on glucose metabolism in

157

syndrome.299 As a comorbidity with EOC, diabetes may accelerate these cancer-driven

changes in the host ecosystem.

Glucose transport and diagnostic potential

These studies demonstrated that glucose enters cancer cells through both GLUT and

SGLT. It is possible that PET scans using FDG are only accounting for a portion of

glucose uptake, underestimating the number and metabolic activity of malignant lesions.

A first generation SGLT-specific tracer, alpha-methyl-4-deoxy-4-[18F]fluoro-D-

glucopyranoside (Me4FDG) has been developed.130 Because SGLT-mediated glucose

transport is reliant on the extracellular sodium concentration, using multi-modal uptake

imaging combining FDG + Me4FDG + Na+ could add power to PET scanning in

diagnosis and treatment monitoring.300–303

Implications for EOC treatment

The best available treatment tools against ovarian cancer are surgery304, imaging and

monitoring blood levels of CA-125305–309, and platinum-based chemotherapies310, all of

which are confounded by hyperglycemia.

In addition to the dysregulation of systemic metabolism caused by the tumour itself,

many standard cancer treatments induce hyperglycemia. Total parenteral nutrition, tube

feeding, steroids and glucocorticoids are often used in oncology and have been shown

to result in hyperglycemia.177,311 Intravenous solutions that contain lactate (Ringer’s,

Page 172: The effects of hyperglycemia on glucose metabolism in

158

Hartmann’s) are also often given to cancer patients and, given the tumour’s ability to

use lactate as a fuel, may in fact be detrimental.105 Even in previously diabetic patients,

hyperglycemic treatment varies widely. In many cases, comprehensive glucose

management may not be included as part of an overall treatment plan for the cancer,312

partly because it can be particularly difficult to both aggressively monitor blood glucose

and maintain quality of life during cancer treatment.313 Since blood glucose can often be

managed effectively through diet, nutrition could become an important part of EOC

treatment. Specific low-carbohydrate diets have been suggested as a means to starve

cancer cells of glucose, and studies using ketogenic diets have shown potential anti-

cancer benefits.314 However, ketones have recently been shown to actually fuel cancer

in part because of their contributions to the “reverse Warburg” effect. The ability of

tumours to use ketones as fuel has even been suggested to partly explain the diabetes-

cancer connection.105 Taken together, any modifications to systemic fuel availability

should be taken into consideration as potentially confounding factors when treating

ovarian cancer patients.

Study Limitations

One of the most significant limitations of cancer metabolic research in vitro is that cell

culture conditions are hyperglycemic and hyperoxic,64 both of which directly oppose the

usual conditions of the tumour microenvironment. The phenotypic differences between

ID8-6 and ID8-25 cells clearly demonstrated that high glucose medium influences cell

robustness and metabolism.

Page 173: The effects of hyperglycemia on glucose metabolism in

159

Through the use of both Type 1 and Type 2 diabetic animal models, we were able to

separate some of the effects of insulin from those of glucose. However, the reductive

approach we took did not consider other contributors to glucose regulation, particularly

other hormonal factors.315 Furthermore, we did not consider the effects of inflammation,

immunity and angiogenesis which are inextricably linked to both metabolism and tumour

growth.

Limitations of animal models

The Akt2 KO model was originally used because of its non-obese type 2 diabetic

phenotype, however it appears that under some conditions, Akt2 KO can also influence

tumour development, potentially confounding our results.316 In addition, we were unable

to reduce systemic blood glucose with metformin in these mice and therefore could not

directly test the hypothesis that normalizing blood glucose could reduce tumour growth

in diabetic environments. This lack of impact on blood glucose could be due to opposing

effects of the Akt2 KO and metformin. Akt2 plays an important role in the exocytosis of

intracellular GLUT4 to the cell membrane to facilitate insulin-dependent glucose

uptake.317 Eliminating Akt2 expression in mice results in insulin insensitivity and

consequently hyperglycemia. On the other hand, metformin achieves most of its

antidiabetic effects by increasing glucose uptake into skeletal muscle. Metformin

decreases the endocytosis of GLUT4 transporters from the cell membrane back into the

cytosol.318 The inactivation of Akt2-mediated GLUT4 trafficking antagonizes metformin’s

ability to maintain transporter expression and increase glucose consumption in cells,

which would limit its ability to reduce blood glucose in Akt2 KO mice.

Page 174: The effects of hyperglycemia on glucose metabolism in

160

Another limitation related to in vivo work was the difficulty in characterizing secondary

disease, which we found to be an essential measure of morbidity. In chronically diabetic

mice, we saw significant reductions in overall survival compared to normoglycemic

animals together with significantly smaller tumours which suggested a large contribution

of secondary disease. In addition, mice with SGLT2-deficient tumours presented with

many metastatic lesions that were strongly associated with poor survival. In women

metastases lead to death because of their physical affect vital organs, but the

secondary lesions in our model were too small to have this same influence. “Secondary

disease” includes humoral and intraperitoneal factors that are not accounted for in our

scoring method.

Conclusions

Collectively, the data presented in this thesis indicate that altered tumour metabolism is

much broader than the Warburg effect. The effects of systemic metabolic comorbidities

such as diabetes can confound EOC diagnosis, treatment and overall survival through

fundamental changes in tumour biology.209 Antidiabetic pharmaceuticals have great

potential as adjuvants to cancer treatment and because altered glucose homeostasis is

common in cancer patients, these therapies have wide applicability to normoglycemic

patients. Moreover, the identification of SGLT2 as a powerful tumour suppressor opens

exciting possibilities for improved screening and drug targeting.

Page 175: The effects of hyperglycemia on glucose metabolism in

161

REFERENCES

1. Navaneelan, T. Trends in the incidence and mortality of female reproductive system cancers. Statistics Canada, catalogue no. 82–624–X (2015).

2. Canadian Cancer Society. Canadian Cancer Statistics Special topic : Predictions of the future burden of cancer in Canada. 1–151 (2015).

3. Bast, R. C., Hennessy, B. & Mills, G. B. The biology of ovarian cancer: new opportunities for translation. Nat. Rev. Cancer 9, 415–28 (2009).

4. Boyd, J. & Rubin, S. C. Hereditary ovarian cancer: molecular genetics and clinical implications. Gynecol. Oncol. 64, 196–206 (1997).

5. Kurman, R. J., Carcangiu, M. & Herrington, C. World Health Organisation Classification of Tumours of the Female Reproductive Organs. International Agency for Research on Cancer 4th ed., (2014).

6. Devouassoux-Shisheboran, M. & Genestie, C. Pathobiology of ovarian carcinomas. Chin. J. Cancer 34, 50–5 (2015).

7. Karst, A. M. & Drapkin, R. Ovarian cancer pathogenesis: a model in evolution. J. Oncol. 2010, 932371 (2010).

8. Kauff, N. D. et al. Risk-reducing salpingo-oophorectomy in women with a BRCA1 or BRCA2 mutation. N. Engl. J. Med. 346, 1609–15 (2002).

9. Landen, C. N., Birrer, M. J. & Sood, A. K. Early events in the pathogenesis of epithelial ovarian cancer. J. Clin. Oncol. 26, 995–1005 (2008).

10. Greenaway, J., Moorehead, R., Shaw, P. & Petrik, J. Epithelial-stromal interaction increases cell proliferation, survival and tumorigenicity in a mouse model of human epithelial ovarian cancer. Gynecol. Oncol. 108, 385–94 (2008).

11. Risch, H. A. Hormonal etiology of epithelial ovarian cancer, with a hypothesis concerning the role of androgens and progesterone. J. Natl. Cancer Inst. 90, 1774–86 (1998).

12. Choi, J.-H., Wong, A. S. T., Huang, H.-F. & Leung, P. C. K. Gonadotropins and ovarian cancer. Endocr. Rev. 28, 440–61 (2007).

13. Ness, R. B. & Cottreau, C. Possible role of ovarian epithelial inflammation in ovarian cancer. J. Natl. Cancer Inst. 91, 1459–67 (1999).

14. Auersperg, N., Wong, a S., Choi, K. C., Kang, S. K. & Leung, P. C. Ovarian surface epithelium: biology, endocrinology, and pathology. Endocr. Rev. 22, 255–88 (2001).

Page 176: The effects of hyperglycemia on glucose metabolism in

162

15. Holschneider, C. H. & Berek, J. S. Ovarian cancer: epidemiology, biology, and prognostic factors. Semin. Surg. Oncol. 19, 3–10 (2000).

16. Dubeau, L. & Drapkin, R. Coming into focus: the nonovarian origins of ovarian cancer. Ann. Oncol. 24, viii28–viii35 (2013).

17. Kurman, R. J. & Shih, I.-M. Pathogenesis of ovarian cancer: lessons from morphology and molecular biology and their clinical implications. Int. J. Gynecol. Pathol. 27, 151–60 (2008).

18. Piek, J. M. et al. Dysplastic changes in prophylactically removed Fallopian tubes of women predisposed to developing ovarian cancer. J. Pathol. 195, 451–6 (2001).

19. Medeiros, F. et al. The tubal fimbria is a preferred site for early adenocarcinoma in women with familial ovarian cancer syndrome. Am. J. Surg. Pathol. 30, 230–6 (2006).

20. Kindelberger, D. W. et al. Intraepithelial Carcinoma of the Fimbria and Pelvic Serous Carcinoma: Evidence for a Causal Relationship. Am. J. Surg. Pathol. 31, 161–169 (2007).

21. Dubeau, L. The cell of origin of ovarian epithelial tumours. Lancet Oncol. 9, 1191–1197 (2008).

22. Kim, J. et al. High-grade serous ovarian cancer arises from fallopian tube in a mouse model. Proceedings of the National Academy of Sciences 109, 3921–3926 (2012).

23. Bankhead, C. R. et al. Identifying symptoms of ovarian cancer: a qualitative and quantitative study. BJOG 115, 1008–14 (2008).

24. Goff, B. A., Mandel, L. S., Melancon, C. H. & Muntz, H. G. Frequency of symptoms of ovarian cancer in women presenting to primary care clinics. JAMA 291, 2705–12 (2004).

25. National Cancer Institute. Surveillance Epidemiology and End Results (SEER). (2012).

26. Prat, J. FIGO Committee on Gynecologic Oncology. Staging classification for cancer of the ovary, fallopian tube, and peritoneum. Int. J. Gynecol. Obstet. 124, 1–5 (2014).

27. Hynninen, J. et al. FDG PET/CT in staging of advanced epithelial ovarian cancer: Frequency of supradiaphragmatic lymph node metastasis challenges the traditional pattern of disease spread. Gynecol. Oncol. 126, 64–68 (2012).

28. Ahmed, N. & Stenvers, K. L. Getting to know ovarian cancer ascites: opportunities

Page 177: The effects of hyperglycemia on glucose metabolism in

163

for targeted therapy-based translational research. Front. Oncol. 3, 256 (2013).

29. Jayson, G. C., Kohn, E. C., Kitchener, H. C. & Ledermann, J. A. Ovarian cancer. Lancet 384, 1376–1388 (2014).

30. du Bois, A. et al. Role of surgical outcome as prognostic factor in advanced epithelial ovarian cancer: a combined exploratory analysis of 3 prospectively randomized phase 3 multicenter trials: by the Arbeitsgemeinschaft Gynaekologische Onkologie Studiengruppe Ovarialkarzin. Cancer 115, 1234–44 (2009).

31. Kurman, R. J., Visvanathan, K., Roden, R., Wu, T. C. & Shih, I.-M. Early detection and treatment of ovarian cancer: shifting from early stage to minimal volume of disease based on a new model of carcinogenesis. Am. J. Obstet. Gynecol. 198, 351–6 (2008).

32. King, M.-C., Marks, J. H. & Mandell, J. B. Breast and ovarian cancer risks due to inherited mutations in BRCA1 and BRCA2. Science 302, 643–6 (2003).

33. Kyriazi, S., Kaye, S. B. & deSouza, N. M. Imaging ovarian cancer and peritoneal metastases--current and emerging techniques. Nat. Rev. Clin. Oncol. 7, 381–93 (2010).

34. Gordon, A. N. et al. Recurrent epithelial ovarian carcinoma: a randomized phase III study of pegylated liposomal doxorubicin versus topotecan. J. Clin. Oncol. 19, 3312–22 (2001).

35. Griffiths, R. W. et al. Outcomes After Multiple Lines of Chemotherapy for Platinum-Resistant Epithelial Cancers of the Ovary, Peritoneum, and Fallopian Tube. Int. J. Gynecol. Cancer 21, 58–65 (2011).

36. Vaughan, S. et al. Rethinking ovarian cancer: recommendations for improving outcomes. Nat. Rev. Cancer 11, 719–25 (2011).

37. Guyton, A. C. & Hall, J. E. Guyton and Hall Textbook of Medical Physiology. (Saunders Elsevier, 2011).

38. Voet, D. & Voet, J. G. Biochemistry. (John Wiley & Sons, Inc, 2004).

39. Warburg, O., K, P. & E, N. Ueber den stoffwechsel der tumoren. Biochem Z 152, 319–344 (1924).

40. Ganapathy, V., Thangaraju, M. & Prasad, P. D. Nutrient transporters in cancer: Relevance to Warburg hypothesis and beyond. Pharmacol. Ther. 121, 29–40 (2009).

41. Warburg, O. On the Origin of Cancer Cells. Science (80-. ). 123, 309–314 (1956).

Page 178: The effects of hyperglycemia on glucose metabolism in

164

42. Funes, J. M. et al. Transformation of human mesenchymal stem cells increases their dependency on oxidative phosphorylation for energy production. Proc. Natl. Acad. Sci. U. S. A. 104, 6223–8 (2007).

43. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–74 (2011).

44. Kim, J. & Dang, C. V. Cancer’s molecular sweet tooth and the Warburg effect. Cancer Res. 66, 8927–30 (2006).

45. Chance, B. & Hess, B. On the control of metabolism in ascites tumor cell suspensions. Ann. N. Y. Acad. Sci. 63, 1008–16 (1956).

46. Chance, B. & Castor, L. N. Some Patterns of the Respiratory Pigments of Ascites Tumors of Mice. Science 116, 200–2 (1952).

47. Weinhouse, S. On respiratory impairment in cancer cells. Science 124, 267–9 (1956).

48. Greiner, E. F., Guppy, M. & Brand, K. Glucose is essential for proliferation and the glycolytic enzyme induction that provokes a transition to glycolytic energy production. J. Biol. Chem. 269, 31484–90 (1994).

49. Koppenol, W. H., Bounds, P. L. & Dang, C. V. Otto Warburg’s contributions to current concepts of cancer metabolism. Nat. Rev. Cancer 11, 325–337 (2011).

50. Kroemer, G. & Pouyssegur, J. Tumor cell metabolism: cancer’s Achilles' heel. Cancer Cell 13, 472–82 (2008).

51. Cairns, R. A., Harris, I. S. & Mak, T. W. Regulation of cancer cell metabolism. Nature reviews. Cancer 11, 85–95 (2011).

52. Pedersen, P. L. The cancer cell’s ‘power plants’ as promising therapeutic targets: an overview. J. Bioenerg. Biomembr. 39, 1–12 (2007).

53. Modica-Napolitano, J. S., Kulawiec, M. & Singh, K. K. Mitochondria and human cancer. Curr. Mol. Med. 7, 121–31 (2007).

54. Eboli, M. L., Paradies, G., Galeotti, T. & Papa, S. Pyruvate transport in tumour-cell mitochondria. Biochim. Biophys. Acta 460, 183–7 (1977).

55. Pelicano, H. et al. Mitochondrial respiration defects in cancer cells cause activation of Akt survival pathway through a redox-mediated mechanism. J. Cell Biol. 175, 913–23 (2006).

56. Bellance, N. et al. Bioenergetics of lung tumors: Alteration of mitochondrial biogenesis and respiratory capacity. Int. J. Biochem. Cell Biol. 41, 2566–2577 (2009).

Page 179: The effects of hyperglycemia on glucose metabolism in

165

57. Desouki, M. M., Kulawiec, M., Bansal, S., Das, G. M. & Singh, K. K. Cross talk between mitochondria and superoxide generating NADPH oxidase in breast and ovarian tumors. Cancer Biol. Ther. 4, 1367–73 (2005).

58. Pollard, P. J. Accumulation of Krebs cycle intermediates and over-expression of HIF1 in tumours which result from germline FH and SDH mutations. Hum. Mol. Genet. 14, 2231–2239 (2005).

59. Dang, L. et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 465, 966 (2010).

60. Gottlieb, E. & Tomlinson, I. P. M. Mitochondrial tumour suppressors: a genetic and biochemical update. Nat. Rev. Cancer 5, 857–66 (2005).

61. Lin, C.-C. et al. Loss of the respiratory enzyme citrate synthase directly links the Warburg effect to tumor malignancy. Sci. Rep. 2, 785 (2012).

62. Schulz, T. J. et al. Induction of oxidative metabolism by mitochondrial frataxin inhibits cancer growth: Otto Warburg revisited. J. Biol. Chem. 281, 977–81 (2006).

63. Gatenby, R. a & Gillies, R. J. Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer 4, 891–9 (2004).

64. Zu, X. L. & Guppy, M. Cancer metabolism: facts, fantasy, and fiction☆. Biochem.

Biophys. Res. Commun. 313, 459–465 (2004).

65. Hockel, M. & Vaupel, P. Tumor Hypoxia: Definitions and Current Clinical, Biologic, and Molecular Aspects. JNCI J. Natl. Cancer Inst. 93, 266–276 (2001).

66. Semenza, G. L. Targeting HIF-1 for cancer therapy. Nat. Rev. Cancer 3, 721–732 (2003).

67. Goodwin, M. L., Gladden, L. B., Nijsten, M. W. N. & Jones, K. B. Lactate and Cancer: Revisiting the Warburg Effect in an Era of Lactate Shuttling. Front. Nutr. 1, 2014–2016 (2015).

68. Walenta, S. & Mueller-Klieser, W. F. Lactate: mirror and motor of tumor malignancy. Semin. Radiat. Oncol. 14, 267–74 (2004).

69. Choi, S. Y. C., Collins, C. C., Gout, P. W. & Wang, Y. Cancer-generated lactic acid: a regulatory, immunosuppressive metabolite? J. Pathol. 230, 350–5 (2013).

70. Morita, T., Nagaki, T., Fukuda, I. & Okumura, K. Clastogenicity of low pH to various cultured mammalian cells. Mutat. Res. 268, 297–305 (1992).

71. Yuan, J., Narayanan, L., Rockwell, S. & Glazer, P. M. Diminished DNA repair and elevated mutagenesis in mammalian cells exposed to hypoxia and low pH. Cancer Res. 60, 4372–6 (2000).

Page 180: The effects of hyperglycemia on glucose metabolism in

166

72. Raghunand, N., Mahoney, B., van Sluis, R., Baggett, B. & Gillies, R. J. Acute metabolic alkalosis enhances response of C3H mouse mammary tumors to the weak base mitoxantrone. Neoplasia 3, 227–35

73. Martínez-Zaguilán, R. et al. Acidic pH enhances the invasive behavior of human melanoma cells. Clin. Exp. Metastasis 14, 176–86 (1996).

74. Gatenby, R. A. & Gawlinski, E. T. The glycolytic phenotype in carcinogenesis and tumor invasion: insights through mathematical models. Cancer Res. 63, 3847–54 (2003).

75. Gatenby, R. A. & Gawlinski, E. T. A reaction-diffusion model of cancer invasion. Cancer Res. 56, 5745–53 (1996).

76. Smallbone, K., Gavaghan, D. J., Gatenby, R. A. & Maini, P. K. The role of acidity in solid tumour growth and invasion. J. Theor. Biol. 235, 476–84 (2005).

77. Xu, H. N. et al. Is Higher Lactate an Indicator of Tumor Metastatic Risk? A Pilot MRS Study Using Hyperpolarized (13)C-Pyruvate. Acad. Radiol. 21, 223–31 (2014).

78. Arora, K. K. & Pedersen, P. L. Functional significance of mitochondrial bound hexokinase in tumor cell metabolism. Evidence for preferential phosphorylation of glucose by intramitochondrially generated ATP. J. Biol. Chem. 263, 17422–8 (1988).

79. Rastogi, S., Banerjee, S., Chellappan, S. & Simon, G. R. Glut-1 antibodies induce growth arrest and apoptosis in human cancer cell lines. Cancer Lett. 257, 244–251 (2007).

80. Friedrich, M. J. Researchers aim to stop tumor growth by shutting off cancer’s fuel supply. JAMA 303, 1021–2 (2010).

81. Deberardinis, R. J., Sayed, N., Ditsworth, D. & Thompson, C. B. Brick by brick: metabolism and tumor cell growth. Curr. Opin. Genet. Dev. 18, 54–61 (2008).

82. Jang, M., Kim, S. S. & Lee, J. Cancer cell metabolism: implications for therapeutic targets. Exp. Mol. Med. 45, e45 (2013).

83. Ward, P. S. & Thompson, C. B. Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell 21, 297–308 (2012).

84. Venuta, S. & Rubin, H. Effects of glucose starvation on normal and rous sarcoma virus-transformed chick cells. J. Natl. Cancer Inst. 54, 395–400 (1975).

85. Hatanaka, M., Huebner, R. J. & Gilden, R. V. Alterations in the characteristics of sugar uptake by mouse cells transformed by murine sarcoma viruses. J. Natl. Cancer Inst. 43, 1091–6 (1969).

Page 181: The effects of hyperglycemia on glucose metabolism in

167

86. Ramanathan, A., Wang, C. & Schreiber, S. L. Perturbational profiling of a cell-line model of tumorigenesis by using metabolic measurements. Proc. Natl. Acad. Sci. U. S. A. 102, 5992–7 (2005).

87. Jiang, P. et al. p53 regulates biosynthesis through direct inactivation of glucose-6-phosphate dehydrogenase. Nat. Cell Biol. 13, 310–6 (2011).

88. Goldstein, I. et al. P53 Promotes the Expression of Gluconeogenesis-Related Genes and Enhances Hepatic Glucose Production. Cancer Metab. 1, 9 (2013).

89. Elstrom, R. L. et al. Akt stimulates aerobic glycolysis in cancer cells. Cancer Res. 64, 3892–9 (2004).

90. Robey, R. B. & Hay, N. Is Akt the ‘Warburg kinase’?-Akt-energy metabolism interactions and oncogenesis. Semin. Cancer Biol. 19, 25–31 (2009).

91. Christofk, H. R. et al. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature 452, 230–3 (2008).

92. Shanmugam, M., McBrayer, S. K. & Rosen, S. T. Targeting the Warburg effect in hematological malignancies: from PET to therapy. Curr. Opin. Oncol. 21, 531–6 (2009).

93. Schulz, J. et al. Regulation of anaerobic glycolysis in Ehrlich ascites tumour cells. Acta Biol. Med. Ger. 36, 1379–91 (1977).

94. Mathupala, S. P., Rempel, A. & Pedersen, P. L. Aberrant glycolytic metabolism of cancer cells: a remarkable coordination of genetic, transcriptional, post-translational, and mutational events that lead to a critical role for type II hexokinase. J. Bioenerg. Biomembr. 29, 339–43 (1997).

95. Rivenzon-Segal, D., Boldin-Adamsky, S., Seger, D., Seger, R. & Degani, H. Glycolysis and glucose transporter 1 as markers of response to hormonal therapy in breast cancer. Int. J. Cancer 107, 177–82 (2003).

96. Artemov, D., Bhujwalla, Z. M., Pilatus, U. & Glickson, J. D. Two-compartment model for determination of glycolytic rates of solid tumors by in vivo 13C NMR spectroscopy. NMR Biomed. 11, 395–404 (1998).

97. Wolf, A. et al. Hexokinase 2 is a key mediator of aerobic glycolysis and promotes tumor growth in human glioblastoma multiforme. J. Exp. Med. 208, 313–26 (2011).

98. Mathupala, S. P., Ko, Y. H. & Pedersen, P. L. Hexokinase-2 bound to mitochondria: cancer’s stygian link to the ‘Warburg Effect’ and a pivotal target for effective therapy. Semin. Cancer Biol. 19, 17–24 (2009).

99. Pedersen, P. L. Warburg, me and Hexokinase 2: Multiple discoveries of key

Page 182: The effects of hyperglycemia on glucose metabolism in

168

molecular events underlying one of cancers’ most common phenotypes, the ‘Warburg Effect’, i.e., elevated glycolysis in the presence of oxygen. J. Bioenerg. Biomembr. 39, 211–22 (2007).

100. John, S., Weiss, J. N. & Ribalet, B. Subcellular localization of hexokinases I and II directs the metabolic fate of glucose. PLoS One 6, e17674 (2011).

101. Netti, P. A., Baxter, L. T., Boucher, Y., Skalak, R. & Jain, R. K. Time-dependent behavior of interstitial fluid pressure in solid tumors: implications for drug delivery. Cancer Res. 55, 5451–8 (1995).

102. Boucher, Y. & Jain, R. K. Microvascular pressure is the principal driving force for interstitial hypertension in solid tumors: implications for vascular collapse. Cancer Res. 52, 5110–4 (1992).

103. Rutz, H. P. A biophysical basis of enhanced interstitial fluid pressure in tumors. Med. Hypotheses 53, 526–529 (1999).

104. Samudio, I. et al. Pharmacologic inhibition of fatty acid oxidation sensitizes human leukemia cells to apoptosis induction. J. Clin. Invest. 120, 142–56 (2010).

105. Bonuccelli, G. et al. Ketones and lactate ‘fuel’ tumor growth and metastasis: Evidence that epithelial cancer cells use oxidative mitochondrial metabolism. Cell Cycle 9, 3506–3514 (2010).

106. Wise, D. R. & Thompson, C. B. Glutamine addiction: a new therapeutic target in cancer. Trends Biochem. Sci. 35, 427–33 (2010).

107. Buzzai, M. et al. The glucose dependence of Akt-transformed cells can be reversed by pharmacologic activation of fatty acid beta-oxidation. Oncogene 24, 4165–73 (2005).

108. Rodríguez-Enríquez, S. et al. Oxidative phosphorylation is impaired by prolonged hypoxia in breast and possibly in cervix carcinoma. Int. J. Biochem. Cell Biol. 42, 1744–1751 (2010).

109. Warburg, O., Wind, F. & Negelein, E. THE METABOLISM OF TUMORS IN THE BODY. J. Gen. Physiol. 8, 519–30 (1927).

110. Jose, C., Bellance, N. & Rossignol, R. Choosing between glycolysis and oxidative phosphorylation: A tumor’s dilemma? Biochim. Biophys. Acta 1807, 552–61 (2011).

111. Anderson, A. S., Roberts, P. C., Frisard, M. I., Hulver, M. W. & Schmelz, E. M. Ovarian tumor-initiating cells display a flexible metabolism. Exp. Cell Res. 328, 44–57 (2014).

112. Griguer, C. E., Oliva, C. R. & Gillespie, G. Y. Glucose metabolism heterogeneity

Page 183: The effects of hyperglycemia on glucose metabolism in

169

in human and mouse malignant glioma cell lines. J. Neurooncol. 74, 123–33 (2005).

113. Berridge, M. V, Herst, P. M. & Tan, A. S. Metabolic flexibility and cell hierarchy in metastatic cancer. Mitochondrion 10, 584–8 (2010).

114. Bissell, M. J. & Radisky, D. Putting tumours in context. Nat. Rev. Cancer 1, 46–54 (2001).

115. Yoshida, G. J. Metabolic reprogramming: the emerging concept and associated therapeutic strategies. J. Exp. Clin. Cancer Res. 34, 111 (2015).

116. Pavlides, S. et al. The reverse Warburg effect: Aerobic glycolysis in cancer associated fibroblasts and the tumor stroma. Cell Cycle 8, 3984–4001 (2009).

117. Martinez-Outschoorn, U. E., Lisanti, M. P. & Sotgia, F. Catabolic Cancer-Associated Fibroblasts (CAFs) Transfer Energy and Biomass to Anabolic Cancer Cells, Fueling Tumor Growth. Semin. Cancer Biol. (2014).

118. Fiaschi, T. et al. Reciprocal metabolic reprogramming through lactate shuttle coordinately influences tumor-stroma interplay. Cancer Res. 72, 5130–40 (2012).

119. Sonveaux, P. et al. Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice. J. Clin. Invest. 118, 3930–42 (2008).

120. Semenza, G. L. Tumor metabolism: cancer cells give and take lactate. J. Clin. Invest. 118, 3835–7 (2008).

121. Crabtree, H. G. Observations on the carbohydrate metabolism of tumours. Biochem. J. 23, 536–45 (1929).

122. Suchorolski, M. T., Paulson, T. G., Sanchez, C. A., Hockenbery, D. & Reid, B. J. Warburg and Crabtree effects in premalignant Barrett’s esophagus cell lines with active mitochondria. PLoS One 8, e56884 (2013).

123. Bouché, C., Serdy, S., Kahn, C. R. & Goldfine, A. B. The cellular fate of glucose and its relevance in type 2 diabetes. Endocr. Rev. 25, 807–830 (2004).

124. Szablewski, L. Expression of glucose transporters in cancers. Biochim. Biophys. Acta 1835, 164–9 (2013).

125. Yamamoto, T. et al. Over-expression of facilitative glucose transporter genes in human cancer. Biochem. Biophys. Res. Commun. 170, 223–30 (1990).

126. Rudlowski, C. et al. GLUT1 mRNA and protein expression in ovarian borderline tumors and cancer. Oncology 66, 404–410 (2004).

127. Semaan, A. et al. Expression of GLUT-1 in epithelial ovarian carcinoma:

Page 184: The effects of hyperglycemia on glucose metabolism in

170

correlation with tumor cell proliferation, angiogenesis, survival and ability to predict optimal cytoreduction. Gynecol. Oncol. 121, 181–6 (2011).

128. Kalir, T. et al. Immunohistochemical staining of GLUT1 in benign, borderline, and malignant ovarian epithelia. Cancer 94, 1078–1082 (2002).

129. Noguchi, Y. et al. Suppression of facilitative glucose transporter 1 mRNA can suppress tumor growth. Cancer Lett. 154, 175–82 (2000).

130. Wright, E. M., Loo, D. D. F. & Hirayama, B. A. Biology of human sodium glucose transporters. Physiol. Rev. 91, 733–794 (2011).

131. Wright, E. M., Loo, D. D. F., Hirayama, B. a & Turk, E. Surprising versatility of Na+-glucose cotransporters: SLC5. Physiology (Bethesda). 19, 370–376 (2004).

132. Narendran, P. & Saeed, M. Dapagliflozin for the treatment of type 2 diabetes: a review of the literature. Drug Des. Devel. Ther. 8, 2493 (2014).

133. Tahara, A., Takasu, T., Yokono, M., Imamura, M. & Kurosaki, E. Effects of the combination of SGLT2 selective inhibitor ipragliflozin and various antidiabetic drugs in type 2 diabetic mice. Arch. Pharm. Res. (2015). doi:10.1007/s12272-015-0621-8

134. Yu, A. S. et al. Regional distribution of SGLT activity in rat brain in vivo. Am. J. Physiol. Cell Physiol. 304, C240–7 (2013).

135. Yu, L. C. H., Flynn, A. N., Turner, J. R. & Buret, A. G. SGLT-1-mediated glucose uptake protects intestinal epithelial cells against LPS-induced apoptosis and barrier defects: a novel cellular rescue mechanism? FASEB J. 19, 1822–35 (2005).

136. Banerjee, S. K., McGaffin, K. R., Pastor-Soler, N. M. & Ahmad, F. SGLT1 is a novel cardiac glucose transporter that is perturbed in disease states. Cardiovasc. Res. 84, 111–118 (2009).

137. Barcelona, S., Menegaz, D. & Diez-Sampedro, A. Mouse SGLT3a generates proton-activated currents but does not transport sugar. AJP Cell Physiol. 302, C1073–C1082 (2012).

138. Balon, T. W. SGLT and GLUT: are they teammates? Focus on ‘Mouse SGLT3a generates proton-activated currents but does not transport sugar’. Am. J. Physiol. Cell Physiol. 302, C1071–2 (2012).

139. Diez-Sampedro, A. et al. A glucose sensor hiding in a family of transporters. Proc. Natl. Acad. Sci. 100, 11753–11758 (2003).

140. Bianchi, L. & Díez-Sampedro, A. A single amino acid change converts the sugar sensor SGLT3 into a sugar transporter. PLoS One 5, (2010).

Page 185: The effects of hyperglycemia on glucose metabolism in

171

141. Wright, E. M., Hirsch, J. R., Loo, D. D. & Zampighi, G. a. Regulation of Na+/glucose cotransporters. J. Exp. Biol. 200, 287–93 (1997).

142. Sakar, Y. et al. Metformin-induced regulation of the intestinal d-glucose transporters. J. Physiol. Pharmacol. 61, 301–307 (2010).

143. Sopjani, M. et al. Regulation of Na+-coupled glucose carrier SGLT1 by AMP-activated protein kinase. Mol. Membr. Biol. 27, 137–44 (2010).

144. Yamazaki, Y., Ogihara, S., Harada, S. & Tokuyama, S. Activation of cerebral sodium-glucose transporter type 1 function mediated by post-ischemic hyperglycemia exacerbates the development of cerebral ischemia. Neuroscience 310, 674–85 (2015).

145. Weihua, Z. et al. Survival of cancer cells is maintained by EGFR independent of its kinase activity. Cancer Cell 13, 385–93 (2008).

146. Wright, E. M., Hirayama, B. A. & Loo, D. F. Active sugar transport in health and disease. J. Intern. Med. 261, 32–43 (2007).

147. Kothinti, R. K., Blodgett, A. B., North, P. E., Roman, R. J. & Tabatabai, N. M. A novel SGLT is expressed in the human kidney. Eur. J. Pharmacol. 690, 77–83 (2012).

148. Sasseville, L. J., Cuervo, J. E., Lapointe, J.-Y. & Noskov, S. Y. The structural pathway for water permeation through sodium-glucose cotransporters. Biophys. J. 101, 1887–95 (2011).

149. Panayotova-Heiermann, M. & Wright, E. M. Mapping the urea channel through the rabbit Na(+)-glucose cotransporter SGLT1. J. Physiol. 535, 419–25 (2001).

150. Hirschhorn, N. & Greenough, W. B. Progress in oral rehydration therapy. Sci. Am. 264, 50–6 (1991).

151. Yang, M. & Brackenbury, W. J. Membrane potential and cancer progression. Front. Physiol. 4, 185 (2013).

152. Blodgett, T. M., Meltzer, C. C. & Townsend, D. W. PET/CT: form and function. Radiology 242, 360–85 (2007).

153. Pimlott, S. L. & Sutherland, A. Molecular tracers for the PET and SPECT imaging of disease. Chem. Soc. Rev. 40, 149–162 (2011).

154. Kitajima, K., Ebina, Y. & Sugimura, K. Present and future role of FDG-PET/CT imaging in the management of gynecologic malignancies. Jpn J Radiol 32, 313–323 (2014).

155. Gambhir, S. S. Molecular imaging of cancer with positron emission tomography.

Page 186: The effects of hyperglycemia on glucose metabolism in

172

Nat. Rev. Cancer 2, 683–93 (2002).

156. Kubota, R. et al. Intratumoral distribution of fluorine-18-fluorodeoxyglucose in vivo: high accumulation in macrophages and granulation tissues studied by microautoradiography. J. Nucl. Med. 33, 1972–80 (1992).

157. Som, P. et al. A fluorinated glucose analog, 2-fluoro-2-deoxy-D-glucose (F-18): nontoxic tracer for rapid tumor detection. J. Nucl. Med. 21, 670–675 (1980).

158. Smith, T. a. The rate-limiting step for tumor [18F]fluoro-2-deoxy-D-glucose (FDG) incorporation. Nucl. Med. Biol. 28, 1–4 (2001).

159. Prakash, P., Cronin, C. G. & Blake, M. A. Role of PET/CT in Ovarian Cancer. American Roentgen Ray Society W464–W470 (2009). at <http://www.ajronline.org/doi/pdf/10.2214/AJR.09.3843>

160. Steenkamp, D. W., McDonnell, M. E. & Meibom, S. Metformin may be associated with false-negative cancer detection in the gastrointestinal tract on PET/CT. Endocr. Pract. 20, 1079–83 (2014).

161. Zhu, A., Lee, D. & Shim, H. Metabolic positron emission tomography imaging in cancer detection and therapy response. Semin. Oncol. 38, 55–69 (2011).

162. Ryu, J. S., Choi, N. C., Fischman, A. J., Lynch, T. J. & Mathisen, D. J. FDG-PET in staging and restaging non-small cell lung cancer after neoadjuvant chemoradiotherapy: correlation with histopathology. Lung Cancer 35, 179–87 (2002).

163. Alavi, A. et al. Positron emission tomography imaging in nonmalignant thoracic disorders. Semin. Nucl. Med. 32, 293–321 (2002).

164. Cheng, G. et al. Dynamic changes of FDG uptake and clearance in normal tissues. Mol. Imaging Biol. 15, 345–52 (2013).

165. Kostakoglu, L., Agress, H. & Goldsmith, S. J. Clinical role of FDG PET in evaluation of cancer patients. Radiographics 23, 315–40; quiz 533 (2003).

166. Gaeta, C. M., Sher, A. C., Kohan, A., Rubbert, C. & Avril, N. Recurrent and metastatic breast cancer PET, PET/CT, PET/MRI: FDG and new biomarkers. Q J Nucl Med Mol Imaging (2013).

167. Chang, J. M. et al. False positive and false negative FDG-PET scans in various thoracic diseases. Korean J. Radiol. 7, 57–69

168. Lutz, a M., Ray, P., Willmann, J. K., Drescher, C. & Gambhir, S. S. 2-deoxy-2-[F-18]fluoro-D-glucose accumulation in ovarian carcinoma cell lines. Mol. Imaging Biol. 9, 260–6 (2007).

Page 187: The effects of hyperglycemia on glucose metabolism in

173

169. Rohren, E. M., Turkington, T. G. & Coleman, R. E. Clinical applications of PET in oncology. Radiology 231, 305–32 (2004).

170. Schwarz, J. K., Grigsby, P. W., Dehdashti, F. & Delbeke, D. The role of 18F-FDG PET in assessing therapy response in cancer of the cervix and ovaries. J. Nucl. Med. 50 Suppl 1, 64S–73S (2009).

171. Prefontaine, M. & Walker-Dilks, C. PET Recommendation Report 7 IN REVIEW PET Imaging in Ovarian Cancer PET Imaging in Ovarian Cancer : Recommendations. (2009).

172. Tanizaki, Y. et al. Diagnostic Value of Preoperative SUVmax on FDG-PET/CT for the Detection of Ovarian Cancer. Int. J. Gynecol. Cancer (2014).

173. Son, H. et al. Role of FDG PET/CT in staging of recurrent ovarian cancer. Radiographics 31, 569–83 (2011).

174. Gouhar, G. K., Siam, S., Sadek, S. M. & Ahmed, R. A. Prospective assessment of 18F-FDG PET/CT in detection of recurrent ovarian cancer. Egypt. J. Radiol. Nucl. Med. 44, 913–922 (2013).

175. Ghosh, J. et al. Role of FDG PET-CT in asymptomatic epithelial ovarian cancer with rising serum CA-125: a pilot study. Natl. Med. J. India 26, 327–31 (2013).

176. Berek, J. S., Crum, C. & Friedlander, M. Cancer of the ovary, fallopian tube, and peritoneum. Int. J. Gynecol. Obstet. 119, S118–S129 (2012).

177. Psarakis, H. M. Clinical Challenges in Caring for Patients With Diabetes and Cancer. Diabetes Spectr. 19, 157–162 (2006).

178. Lee, J.-Y. et al. Diabetes mellitus and ovarian cancer risk: a systematic review and meta-analysis of observational studies. Int. J. Gynecol. Cancer 23, 402–12 (2013).

179. Bakhru, A., Buckanovich, R. J. & Griggs, J. J. The impact of diabetes on survival in women with ovarian cancer. Gynecol. Oncol. 121, 106–11 (2011).

180. Shah, M. M. et al. Diabetes mellitus and ovarian cancer: More complex than just increasing risk. Gynecol. Oncol. 135, 273–277 (2014).

181. Gallagher, E. J. & LeRoith, D. Diabetes, antihyperglycemic medications and cancer risk: smoke or fire? Curr. Opin. Endocrinol. Diabetes. Obes. 20, 485–94 (2013).

182. Lamkin, D. M. et al. Glucose as a prognostic factor in ovarian carcinoma. Cancer 115, 1021–7 (2009).

183. Leitzmann, M. F. et al. Body mass index and risk of ovarian cancer. Cancer 115,

Page 188: The effects of hyperglycemia on glucose metabolism in

174

812–22 (2009).

184. Grote, V. A., Becker, S. & Kaaks, R. Diabetes mellitus type 2 - an independent risk factor for cancer? Exp. Clin. Endocrinol. diabetes Off. J. Ger. Soc. Endocrinol. Ger. Diabetes Assoc. 118, 4–8 (2010).

185. Giovannucci, E. et al. Diabetes and cancer: a consensus report. in Diabetes care 33, 1674–1685 (2010).

186. Kellenberger, L. D. et al. The role of dysregulated glucose metabolism in epithelial ovarian cancer. J. Oncol. 2010, 514310 (2010).

187. Zhang, X. D. et al. Effect of 2-deoxy-D-glucose on various malignant cell lines in vitro. Anticancer Res. 26, 3561–6

188. Tayek, J. A. A review of cancer cachexia and abnormal glucose metabolism in humans with cancer. J. Am. Coll. Nutr. 11, 445–56 (1992).

189. Rodríguez-Enríquez, S., Marín-Hernández, A., Gallardo-Pérez, J. C. & Moreno-Sánchez, R. Kinetics of transport and phosphorylation of glucose in cancer cells. J. Cell. Physiol. 221, 552–559 (2009).

190. Younes, M., Lechago, L. V, Somoano, J. R., Mosharaf, M. & Lechago, J. Wide expression of the human erythrocyte glucose transporter Glut1 in human cancers. Cancer Res. 56, 1164–7 (1996).

191. Van Belle, T. L. L., Taylor, P. & von Herrath, M. G. G. Mouse Models for Type 1 Diabetes. Drug Discov. Today Dis. Model. 6, 41–45 (2009).

192. King, A. J. F. The use of animal models in diabetes research. Br. J. Pharmacol. 166, 877–894 (2012).

193. Dummler, B. et al. Life with a single isoform of Akt: mice lacking Akt2 and Akt3 are viable but display impaired glucose homeostasis and growth deficiencies. Mol. Cell. Biol. 26, 8042–51 (2006).

194. Garofalo, R. S. et al. Severe diabetes, age-dependent loss of adipose tissue, and mild growth deficiency in mice lacking Akt2/PKB beta. J. Clin. Invest. 112, 197–208 (2003).

195. Vuguin, P., Saenger, P. & Dimartino-Nardi, J. Fasting Glucose Insulin Ratio: A Useful Measure of Insulin Resistance in Girls with Premature Adrenarche. J. Clin. Endocrinol. Metab. 86, 4618–4621 (2001).

196. Berridge, M. V, Tan, A. N. S., Mccoy, K. D. & Wang, R. U. I. The Biochemical and Cellular Basis of Cell Proliferation Assays That Use Tetrazolium Salts. 4–9 (1996).

Page 189: The effects of hyperglycemia on glucose metabolism in

175

197. Bower, W. F. et al. Overt diabetes mellitus adversely affects surgical outcomes of noncardiovascular patients. Surgery 147, 670–5 (2010).

198. White, N. H. Long-term Outcomes in Youths with Diabetes Mellitus. Pediatr. Clin. North Am. 62, 889–909 (2015).

199. Zelenko, Z. & Gallagher, E. J. Diabetes and cancer. Endocrinology and Metabolism Clinics of North America 43, 167–185 (2014).

200. American Diabetes Association. Standards of medical care in diabetes. Diabetes Care 37, (Suppl 1): S14–S80 (2014).

201. Phoenix, K. N., Vumbaca, F., Fox, M. M., Evans, R. & Claffey, K. P. Dietary energy availability affects primary and metastatic breast cancer and metformin efficacy. Breast Cancer Res. Treat. 123, 333–44 (2010).

202. Yang, X.-L. & Chan, J. C. Diabetes, insulin and cancer risk. World J. Diabetes 3, 60–4 (2012).

203. Han, L. et al. High Glucose Promotes Pancreatic Cancer Cell Proliferation via the Induction of EGF Expression and Transactivation of EGFR. PLoS One 6, e27074 (2011).

204. Parekh, N., Lin, Y., Hayes, R. B., Albu, J. B. & Lu-Yao, G. L. Longitudinal associations of blood markers of insulin and glucose metabolism and cancer mortality in the third National Health and Nutrition Examination Survey. Cancer Causes Control 21, 631–42 (2010).

205. Plesner, A., Ten Holder, J. T. & Verchere, C. B. Islet remodeling in female mice with spontaneous autoimmune and streptozotocin-induced diabetes. PLoS One 9, (2014).

206. Karagiannis, G. S. et al. Cancer-associated fibroblasts drive the progression of metastasis through both paracrine and mechanical pressure on cancer tissue. Mol. Cancer Res. 10, 1403–18 (2012).

207. Lerman, O. Z., Galiano, R. D., Armour, M., Levine, J. P. & Gurtner, G. C. Cellular dysfunction in the diabetic fibroblast: impairment in migration, vascular endothelial growth factor production, and response to hypoxia. Am. J. Pathol. 162, 303–12 (2003).

208. Nerlich, A. G., Hagedorn, H. G., Böheim, M. & Schleicher, E. D. Patients with diabetes-induced microangiopathy show a reduced frequency of carcinomas. In Vivo 12, 667–70

209. Renehan, A. G. et al. Diabetes and cancer (2): Evaluating the impact of diabetes on mortality In patients with cancer. Diabetologia 55, 1619–1632 (2012).

Page 190: The effects of hyperglycemia on glucose metabolism in

176

210. Kerr, A. D. The Role of Chronic , Systemic Inflammation in the Progression of Epithelial Ovarian Cancer. (2012).

211. Al-Wahab, Z. et al. Metformin prevents aggressive ovarian cancer growth driven by high-energy diet: similarity with calorie restriction. Oncotarget 6, 10908–10923 (2015).

212. Shapot, V. S. & Blinov, V. a. Blood glucose levels and gluconeogenesis in animals bearing transplantable tumors. Cancer Res. 34, 1827–32 (1974).

213. Pavelić, K. et al. Growth and treatment of Ehrlich tumor in mice with alloxan-induced diabetes. Cancer Res. 39, 1807–13 (1979).

214. Gullino, P. M., Grantham, F. H. & Courtney, a H. Glucose consumption by transplanted tumors in vivo. Cancer Res. 27, 1031–40 (1967).

215. de Groot, J. W. B. et al. Non-islet cell tumour-induced hypoglycaemia: a review of the literature including two new cases. Endocr. Relat. Cancer 14, 979–93 (2007).

216. Wahdan-Alaswad, R. et al. Glucose promotes breast cancer aggression and reduces metformin efficacy. Cell Cycle 12, 3759–69 (2013).

217. Diaz-Ruiz, R., Rigoulet, M. & Devin, A. The Warburg and Crabtree effects: On the origin of cancer cell energy metabolism and of yeast glucose repression. Biochim. Biophys. Acta 1807, 568–76 (2011).

218. Anderson, A. S. et al. Metabolic changes during ovarian cancer progression as targets for sphingosine treatment. Exp. Cell Res. 319, 1431–42 (2013).

219. Kueck, A. et al. Resveratrol inhibits glucose metabolism in human ovarian cancer cells. Gynecol. Oncol. 107, 450–7 (2007).

220. Harris, M. I., Klein, R., Welborn, T. A. & Knuiman, M. W. Onset of NIDDM occurs at least 4-7 yr before clinical diagnosis. Diabetes Care 15, 815–9 (1992).

221. Shibata, K. et al. Placental leucine aminopeptidase (P-LAP) and glucose transporter 4 (GLUT4) expression in benign, borderline, and malignant ovarian epithelia. Gynecol. Oncol. 98, 11–8 (2005).

222. Cho, H., Lee, Y. S., Kim, J., Chung, J.-Y. & Kim, J.-H. Overexpression of Glucose Transporter-1 (GLUT-1) Predicts Poor Prognosis in Epithelial Ovarian Cancer. Cancer Invest. 31, 607–15 (2013).

223. Scafoglio, C. et al. Functional expression of sodium-glucose transporters in cancer. Proc. Natl. Acad. Sci. 112, E4111–E4119 (2015).

224. Bormans, G. M. et al. Synthesis and biologic evaluation of (11)c-methyl-d-glucoside, a tracer of the sodium-dependent glucose transporters. J. Nucl. Med.

Page 191: The effects of hyperglycemia on glucose metabolism in

177

44, 1075–1081 (2003).

225. Lin, S., Lin, D. C. & Flanagan, M. D. Specificity of the effects of cytochalasin B on transport and motile processes. Proc. Natl. Acad. Sci. U. S. A. 75, 329–33 (1978).

226. Berridge, M. V, Herst, P. M. & Tan, A. S. Tetrazolium dyes as tools in cell biology: new insights into their cellular reduction. Biotechnol. Annu. Rev. 11, 127–52 (2005).

227. Macheda, M. L., Rogers, S. & Best, J. D. Molecular and cellular regulation of glucose transporter (GLUT) proteins in cancer. J. Cell. Physiol. 202, 654–662 (2005).

228. Blessing, A. et al. Sodium/Glucose Co-transporter 1 Expression Increases in Human Diseased Prostate. J. Cancer Sci. Ther. 4(9), 302–312 (2012).

229. Helmke, B. M. et al. Expression of SGLT-1 in preneoplastic and neoplastic lesions of the head and neck. Oral Oncol. 40, 28–35 (2004).

230. Baron-Delage, S. et al. Deregulation of hexose transporter expression in Caco-2 cells by ras and polyoma middle T oncogenes. Am. J. Physiol. 270, G314–23 (1996).

231. de Miguel-Luken, M.-J. et al. MAP17 (PDZKIP1) as a novel prognostic biomarker for laryngeal cancer. Oncotarget 6, 12625–36 (2015).

232. Casneuf, V. F. et al. Expression of SGLT1, Bcl-2 and p53 in primary pancreatic cancer related to survival. Cancer Invest. 26, 852–859 (2008).

233. Hanabata, Y., Nakajima, Y., Morita, K.-I., Kayamori, K. & Omura, K. Coexpression of SGLT1 and EGFR is associated with tumor differentiation in oral squamous cell carcinoma. Odontology 100, 1–8 (2011).

234. Lai, B. et al. Overexpression of SGLT1 is correlated with tumor development and poor prognosis of ovarian carcinoma. Arch. Gynecol. Obstet. 285, 1455–61 (2012).

235. Ishikawa, N., Oguri, T., Isobe, T., Fujitaka, K. & Kohno, N. SGLT gene expression in primary lung cancers and their metastatic lesions. Jpn. J. Cancer Res. 92, 874–9 (2001).

236. Zapata-Morales, J. R., Galicia-Cruz, O. G., Franco, M. & Morales, F. M. Hypoxia-inducible factor-1α(HIF-1α) protein diminishes sodium glucose transport 1 (SGLT1) and SGLT2 protein expression In renal epithelial tubular cells (LLC-PK1) under hypoxia. J. Biol. Chem. 289, 346–357 (2014).

237. Vallon, V. et al. Knockout of Na-glucose transporter SGLT2 attenuates hyperglycemia and glomerular hyperfiltration but not kidney growth or injury in

Page 192: The effects of hyperglycemia on glucose metabolism in

178

diabetes mellitus. Am. J. Physiol. Renal Physiol. 304, F156–67 (2013).

238. Cantuaria, G. et al. GLUT-1 expression in ovarian carcinoma: association with survival and response to chemotherapy. Cancer 92, 1144–50 (2001).

239. Cantuaria, G. et al. Expression of GLUT-1 glucose transporter in borderline and malignant epithelial tumors of the ovary. Gynecol. Oncol. 79, 33–7 (2000).

240. Shin, S. J. et al. Ciglitazone enhances ovarian cancer cell death via inhibition of glucose transporter-1. Eur. J. Pharmacol. 743, 17–23 (2014).

241. Lin, H.-W. & Tseng, C.-H. A Review on the Relationship between SGLT2 Inhibitors and Cancer. Int. J. Endocrinol. 2014, 719578 (2014).

242. Bowker, S. L., Yasui, Y., Veugelers, P. & Johnson, J. a. Glucose-lowering agents and cancer mortality rates in type 2 diabetes: assessing effects of time-varying exposure. Diabetologia 53, 1631–7 (2010).

243. Evans, J. M. M., Donnelly, L. a, Emslie-Smith, A. M., Alessi, D. R. & Morris, A. D. Metformin and reduced risk of cancer in diabetic patients. BMJ 330, 1304–5 (2005).

244. Dilokthornsakul, P. et al. The effects of metformin on ovarian cancer: a systematic review. Int. J. Gynecol. Cancer 23, 1544–51 (2013).

245. Franciosi, M. et al. Metformin therapy and risk of cancer in patients with type 2 diabetes: systematic review. PLoS One 8, e71583 (2013).

246. Zhang, Z.-J. & Li, S. The prognostic value of metformin for cancer patients with concurrent diabetes: a systematic review and meta-analysis. Diabetes. Obes. Metab. 16(8), 707–710 (2014).

247. Kumar, S. et al. Metformin intake is associated with better survival in ovarian cancer: a case-control study. Cancer 119, 555–62 (2013).

248. Romero, I. L. et al. Relationship of type II diabetes and metformin use to ovarian cancer progression, survival, and chemosensitivity. Obstet. Gynecol. 119, 61–7 (2012).

249. Hur, K. Y. & Lee, M.-S. New mechanisms of metformin action: Focusing on mitochondria and the gut. J. Diabetes Investig. 6, 600–9 (2015).

250. Zakikhani, M., Dowling, R., Fantus, I. G., Sonenberg, N. & Pollak, M. Metformin is an AMP kinase-dependent growth inhibitor for breast cancer cells. Cancer Res. 66, 10269–73 (2006).

251. Dowling, R. J. O., Zakikhani, M., Fantus, I. G., Pollak, M. & Sonenberg, N. Metformin inhibits mammalian target of rapamycin-dependent translation initiation

Page 193: The effects of hyperglycemia on glucose metabolism in

179

in breast cancer cells. Cancer Res. 67, 10804–12 (2007).

252. Rattan, R., Giri, S., Hartmann, L. C. & Shridhar, V. Metformin attenuates ovarian cancer cell growth in an AMP-kinase dispensable manner. J. Cell. Mol. Med. 15, 166–78 (2011).

253. Kalender, A. et al. Metformin, independent of AMPK, inhibits mTORC1 in a rag GTPase-dependent manner. Cell Metab. 11, 390–401 (2010).

254. Ben Sahra, I. et al. The antidiabetic drug metformin exerts an antitumoral effect in vitro and in vivo through a decrease of cyclin D1 level. Oncogene 27, 3576–86 (2008).

255. Yasmeen, A. et al. Induction of apoptosis by metformin in epithelial ovarian cancer: involvement of the Bcl-2 family proteins. Gynecol. Oncol. 121, 492–8 (2011).

256. Buzzai, M. et al. Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth. Cancer Res. 67, 6745–52 (2007).

257. Hirsch, H. a., Iliopoulos, D., Tsichlis, P. N. & Struhl, K. Metformin selectively targets cancer stem cells, and acts together with chemotherapy to block tumor growth and prolong remission. Cancer Res. 69, 8832–8833 (2009).

258. Patel, S., Kumar, L. & Singh, N. Metformin and epithelial ovarian cancer therapeutics. Cell. Oncol. (Dordr). 38, 365–75 (2015).

259. Rocha, G. Z. et al. Metformin Amplifies Chemotherapy-Induced AMPK Activation and Antitumoral Growth. Clin. Cancer Res. 17, 3993–4005 (2011).

260. Lengyel, E. et al. Metformin inhibits ovarian cancer growth and increases sensitivity to paclitaxel in mouse models. Am. J. Obstet. Gynecol. 212, 479.e1–479.e10 (2015).

261. Gotlieb, W. H. et al. In vitro metformin anti-neoplastic activity in epithelial ovarian cancer. Gynecol. Oncol. 110, 246–50 (2008).

262. Rattan, R., Graham, R. P., Maguire, J. L., Giri, S. & Shridhar, V. Metformin suppresses ovarian cancer growth and metastasis with enhancement of cisplatin cytotoxicity in vivo. Neoplasia 13, 483–491 (2011).

263. Shank, J. J. et al. Metformin targets ovarian cancer stem cells in vitro and in vivo. Gynecol. Oncol. 127, 390–7 (2012).

264. Xie, Y. et al. Metformin combined with p38 MAPK inhibitor improves cisplatin

sensitivity in cisplatin‑resistant ovarian cancer. Mol. Med. Rep. 10, 2346–50 (2014).

Page 194: The effects of hyperglycemia on glucose metabolism in

180

265. Iliopoulos, D., Hirsch, H. A. & Struhl, K. Metformin decreases the dose of chemotherapy for prolonging tumor remission in mouse xenografts involving multiple cancer cell types. Cancer Res. 71, 3196–201 (2011).

266. Wu, B. et al. Metformin inhibits the development and metastasis of ovarian cancer. Oncol. Rep. 28, 903–8 (2012).

267. Kinaan, M., Ding, H. & Triggle, C. R. Metformin: An Old Drug for the Treatment of Diabetes but a New Drug for the Protection of the Endothelium. Med. Princ. Pract. 24, 401–15 (2015).

268. Hawley, S. A., Gadalla, A. E., Olsen, G. S. & Hardie, D. G. The antidiabetic drug metformin activates the AMP-activated protein kinase cascade via an adenine nucleotide-independent mechanism. Diabetes 51, 2420–5 (2002).

269. Segal, E. D. et al. Relevance of the OCT1 transporter to the antineoplastic effect of biguanides. Biochem. Biophys. Res. Commun. 414, 694–9 (2011).

270. Gao, S. et al. Attenuating tumour angiogenesis: a preventive role of metformin against breast cancer. Biomed Res. Int. 2015, 592523 (2015).

271. Bikas, A. D. et al. Glucose-deprivation increases thyroid cancer cell sensitivity to metformin. Endocr. Relat. Cancer 22, 919–32 (2015).

272. Litchfield, L. M. et al. Hyperglycemia-induced metabolic compensation inhibits metformin sensitivity in ovarian cancer. Oncotarget 6, 23548–23560 (2015).

273. Silvestri, A. et al. Metformin Induces Apoptosis and Downregulates Pyruvate Kinase M2 in Breast Cancer Cells Only When Grown in Nutrient-Poor Conditions. PLoS One 10, e0136250 (2015).

274. Birsoy, K. et al. Metabolic determinants of cancer cell sensitivity to glucose limitation and biguanides. Nature 508, 108–112 (2014).

275. Sui, X. et al. Use of metformin alone is not associated with survival outcomes of colorectal cancer cell but AMPK activator AICAR sensitizes anticancer effect of 5-fluorouracil through AMPK activation. PLoS One 9, e97781 (2014).

276. Yilmaz, B. et al. Metformin regresses endometriotic implants in rats by improving implant levels of superoxide dismutase, vascular endothelial growth factor, tissue inhibitor of metalloproteinase-2, and matrix metalloproteinase-9. Am. J. Obstet. Gynecol. 202, 368.e1–8 (2010).

277. Scotland, S. et al. Mitochondrial energetic and AKT status mediate metabolic effects and apoptosis of metformin in human leukemic cells. Leukemia 27, 2129–38 (2013).

278. Dykens, J. a et al. Biguanide-induced mitochondrial dysfunction yields increased

Page 195: The effects of hyperglycemia on glucose metabolism in

181

lactate production and cytotoxicity of aerobically-poised HepG2 cells and human hepatocytes in vitro. Toxicol. Appl. Pharmacol. 233, 203–10 (2008).

279. Wheaton, W. W. et al. Metformin inhibits mitochondrial complex I of cancer cells to reduce tumorigenesis. Elife 3, e02242 (2014).

280. Andrzejewski, S., Gravel, S.-P., Pollak, M. & St-Pierre, J. Metformin directly acts on mitochondria to alter cellular bioenergetics. Cancer Metab. 2, 12 (2014).

281. Menendez, J. A. et al. Oncobiguanides: Paracelsus’ law and nonconventional routes for administering diabetobiguanides for cancer treatment. Oncotarget 5, 2344–8 (2014).

282. Thompson, M. D. et al. Lack of effect of metformin on mammary carcinogenesis in nondiabetic rat and mouse models. Cancer Prev. Res. (Phila). 8, 231–9 (2015).

283. Shu, Y. et al. Effect of genetic variation in the organic cation transporter 1 (OCT1) on metformin action. J. Clin. … 117, 1422–31 (2007).

284. Ma, Y. et al. K‑ras gene mutation as a predictor of cancer cell responsiveness to metformin. Mol. Med. Rep. 8, 763–768 (2013).

285. Aleksandrovski, Y. A. Molecular mechanisms of the cross-impact of pathological processes in combined diabetes and cancer. Research and clinical aspects. Biochem. Biokhimiia 67, 1329–1346 (2002).

286. Ross, S. a & Milner, J. a. Epigenetic modulation and cancer: effect of metabolic syndrome? Am. J. Clin. Nutr. 86, s872–7 (2007).

287. Simon, D. & Balkau, B. Diabetes mellitus, hyperglycaemia and cancer. Diabetes Metab. 36, 182–191 (2010).

288. Bodmer, M., Becker, C., Meier, C., Jick, S. S. & Meier, C. R. Use of metformin and the risk of ovarian cancer: a case-control analysis. Gynecol. Oncol. 123, 200–4 (2011).

289. Van De Poll-Franse, L. V et al. Less aggressive treatment and worse overall survival in cancer patients with diabetes: a large population based analysis. Int. J. cancer 120, 1986–1992 (2007).

290. Barone, B. B. et al. Postoperative mortality in cancer patients with preexisting diabetes: systematic review and meta-analysis. Diabetes Care 33, 931–9 (2010).

291. Rogozan, I. O. N. Relations Between Tumor and Metastasis. 978–984 (1965).

292. Vadlakonda, L., Dash, A., Pasupuleti, M., Anil Kumar, K. & Reddanna, P. Did we get pasteur, warburg, and crabtree on a right note? Front. Oncol. 3, 186 (2013).

Page 196: The effects of hyperglycemia on glucose metabolism in

182

293. Singh, R. et al. Non-hyperinsulinemic hypoglycemia in a patient with a gastrointestinal stromal tumor. Eur. J. Intern. Med. 17, 127–9 (2006).

294. Marks, L. J., Steinke, J., Podolsky, S. & Egdahl, R. H. Hypoglycemia associated with neoplasia. Ann. N. Y. Acad. Sci. 230, 147–60 (1974).

295. Bishop, J. S. & Marks, P. A. Studies on carbohydrate metabolism in patients with neoplastic disease. II. Response to insulin administration. J. Clin. Invest. 38, 668–72 (1959).

296. Jasani, B., Donaldson, L. J., Ratcliffe, J. G. & Sokhi, G. S. Mechanism of impaired glucose tolerance in patients with neoplasia. Br. J. Cancer 38, 287–92 (1978).

297. Norton, J. A., Maher, M., Wesley, R., White, D. & Brennan, M. F. Glucose intolerance in sarcoma patients. Cancer 54, 3022–3027 (1984).

298. Denton, I. C., Kerlan, R. A. & McGraw, R. Brain tumor with hyperosmolar hyperglycemic nonketotic diabetic coma. JAMA 218, 256–7 (1971).

299. Pospisilik, J. A. et al. Targeted deletion of AIF decreases mitochondrial oxidative phosphorylation and protects from obesity and diabetes. Cell 131, 476–91 (2007).

300. Jacobs, M. A. et al. Multiparametric magnetic resonance imaging, spectroscopy and multinuclear (23Na) imaging monitoring of preoperative chemotherapy for locally advanced breast cancer. Acad. Radiol. 17, 1477–85 (2010).

301. Ouwerkerk, R. Sodium magnetic resonance imaging: from research to clinical use. J. Am. Coll. Radiol. 4, 739–41 (2007).

302. Ouwerkerk, R. et al. Elevated tissue sodium concentration in malignant breast lesions detected with non-invasive 23Na MRI. Breast Cancer Res. Treat. 106, 151–160 (2007).

303. Jacobs, M. A. et al. Monitoring of neoadjuvant chemotherapy using multiparametric, 23Na sodium MR, and multimodality (PET/CT/MRI) imaging in locally advanced breast cancer. Breast Cancer Res. Treat. 128, 119–126 (2011).

304. Ferriss, J. S. et al. Does significant medical comorbidity negate the benefit of up-front cytoreduction in advanced ovarian cancer? Int. J. Gynecol. Cancer 22, 762–9 (2012).

305. Menzin, A. W., Kobrin, S., Pollak, E., Goodman, D. B. & Rubin, S. C. The effect of renal function on serum levels of CA 125. Gynecol. Oncol. 58, 375–7 (1995).

306. Turgutalp, K. et al. Serum levels of cancer biomarkers in diabetic and non-diabetic proteinuric patients: a preliminary study. Clin. Chem. Lab. Med. 51, 889–95 (2013).

Page 197: The effects of hyperglycemia on glucose metabolism in

183

307. Esteghamati, A. et al. The inverse relation of CA-125 to diabetes, metabolic syndrome, and associated clinical variables. Metab. Syndr. Relat. Disord. 11, 256–61 (2013).

308. Breborowicz, A., Breborowicz, M., Pyda, M., Połubinska, A. & Oreopoulos, D. Limitations of CA125 as an index of peritoneal mesothelial cell mass. Nephron. Clin. Pract. 100, c46–51 (2005).

309. Joo, N.-S., Kim, K.-N. & Kim, K. S. Serum CA125 concentration has inverse correlation with metabolic syndrome. J. Korean Med. Sci. 26, 1328–32 (2011).

310. da Silva Faria, M. C. et al. Effect of diabetes on biodistribution, nephrotoxicity and antitumor activity of cisplatin in mice. Chem. Biol. Interact. 229, 119–31 (2015).

311. Harris, D. et al. Glucocorticoid-induced hyperglycemia is prevalent and unpredictable for patients undergoing cancer therapy: an observational cohort study. Current Oncology 20, e532–e538 (2013).

312. Balding, L., Philips, T. M. & Fleming, J. S. The management of diabetes in cancer patients receiving palliative care in a hospital setting. Palliat. Med. 24, S129 (2010).

313. Poulson, J. The management of diabetes in patients with advanced cancer. J. Pain Symptom Manage. 13, 339–46 (1997).

314. Stanley, I. a, Ribeiro, S. M., Giménez-Cassina, A., Norberg, E. & Danial, N. N. Changing appetites: the adaptive advantages of fuel choice. Trends Cell Biol. 24, 118–27 (2014).

315. Aronoff, S. L., Berkowitz, K., Shreiner, B. & Want, L. Glucose Metabolism and Regulation: Beyond Insulin and Glucagon. Diabetes Spectr. 17, 183–190 (2004).

316. Linnerth-Petrik, N. M., Santry, L. A., Petrik, J. J. & Wootton, S. K. Opposing functions of Akt isoforms in lung tumor initiation and progression. PLoS One 9, e94595 (2014).

317. Cho, H. et al. Insulin resistance and a diabetes mellitus-like syndrome in mice lacking the protein kinase Akt2 (PKB beta). Science 292, 1728–31 (2001).

318. Yang, J. & Holman, G. D. Long-term metformin treatment stimulates cardiomyocyte glucose transport through an AMP-activated protein kinase-dependent reduction in GLUT4 endocytosis. Endocrinology 147, 2728–36 (2006).

319. Cefalu, W. T. Paradoxical insights into whole body metabolic adaptations following SGLT2 inhibition. J. Clin. Invest. 124, 485–7 (2014).

320. Merovci, A. et al. Dapagliflozin improves muscle insulin sensitivity but enhances endogenous glucose production. J. Clin. Invest. 124, 509–14 (2014).

Page 198: The effects of hyperglycemia on glucose metabolism in

184

Appendix I: Supplementary figures

Figure 41: Positive SGLT1 staining on human ovarian tumour tissue

Page 199: The effects of hyperglycemia on glucose metabolism in

185

Dapagliflozin treatment does not affect the weight of established tumours

We hypothesized that reducing systemic blood glucose in diabetic mice would moderate

the growth of epithelial ovarian tumours by limiting fuel availability. We were unable to

reduce fasting blood glucose in metformin-treated mice (Chapter 4), so we attempted to

do so using the SGLT2 inhibitor dapagliflozin, a member of the thiazolidinedione (TZD)

class of drugs that is based on the structure of phlorizin (PZ). TZDs have recently been

developed as a promising new treatment in Type 2 diabetes (T2DM): SGLT2 increases

glucose reuptake into the renal circulation, and blocking this reuptake significantly

reduces blood glucose in an insulin-independent manner.

Dapagliflozin (Cayman Chemical, Ann Arbor, MI) was formulated in polyethylene

glycol/water/ethanol (45:45:10, v/v/v) at a concentration of 5 mg/ml. Mice received a

daily dose of 1 mg/kg dapagliflozin by oral gavage for 45 days (80 days PTI to 125 days

PTI).

Page 200: The effects of hyperglycemia on glucose metabolism in

186

Figure 42: Dapagliflozin treatment does not affect tumour weight

A) Fasting blood glucose after two weeks (PTI day 95) and six weeks (PTI day 125) of

dapagliflozin treatment. B) Tumour weights from mice sacrificed at PTI day 125. Bars

are mean + SEM. Brackets with stars indicate differences between bars by two-way

ANOVA with Bonferroni corrections. N=5 mice per group. *p<0.05, **p<0.01.

Page 201: The effects of hyperglycemia on glucose metabolism in

187

In mice 125 days post-tumour induction, daily dapagliflozin treatment for approximately

six weeks (45 days) did not result in a reduction in blood glucose. After two weeks of

treatment, WT animals had a paradoxical increase in blood glucose that may represent

a systemic compensation for dapagliflozin-induced glucosuria (Figure 41A).

Dapagliflozin has been associated with an increase in endogenous glucose production

that is independent of the decrease in plasma glucose concentration319 and can reduce

the glucose-lowering effects by half.320 When mice were sacrificed at 125 days PTI, the

differences in FBG were abrogated. It is possible that by this point in EOC development,

tumours had already altered glucose metabolism (Figures 6 and 8), reducing the

efficacy of dapagliflozin to reduce systemic glucose.

Dapagliflozin treatment was also unable to affect tumour weight (Figure 41B), although

the small size of the tumours may have masked any statistical differences. These data

indicate that dapagliflozin cannot reduce the weight of established tumours, however

they do not address whether or not glucose reduction at tumour initiation can limit

tumour growth.

Page 202: The effects of hyperglycemia on glucose metabolism in

188

Appendix II: Source of supplies and materials

[14C] α-methyl-D-glucopyranoside Moravek Biochemicals, Brea, CA

[3H] 2-deoxy-D-glucose Moravek Biochemicals, Brea, CA

Acrylamide Bio-Rad Laboratories, Mississauga, ON

Agarose Life Technologies Inc., Burlington, ON

Ammonium persulfate (APS) Fisher Scientific, Whitby, ON

Antibiotic-antimycotic (ABAM) Life Technologies Inc., Burlington, ON

Anti-GLUT1 antibody Santa Cruz Biotechnologies, Santa Cruz, CA

Anti-mouse secondary antibody Cell Signal Technologies, Danvers, MA

Anti-rabbit secondary antibody Cell Signal Technologies, Danvers, MA

Anti-SGLT1 antibody Abcam, Cambridge, MA

Anti-SGLT2 antibody Abcam, Cambridge, MA

Anti-β-actin antibody Cell Signal Technologies, Danvers, MA

Aprotinin Sigma, Oakville, ON

Biocoat Matrigel invasion chambers BD Biosciences, Franklin Lakes, NJ

Biotinylated anti-rabbit secondary antibody Sigma, Oakville, ON

Bovine serum albumin (BSA) Sigma, Oakville, ON

Buffered formalin (10%) Fisher Scientific, Whitby, ON

Citric acid Sigma, Oakville, ON

Corning cryovials Fisher Scientific, Whitby, ON

Costar culture dishes Corning Life Sciences, Lowell, MA

Crystal Violet Sigma, Oakville, ON

Cytochalasin B Sigma, Oakville, ON

CytoScint liquid scintillation cocktail MP Biomedicals, Santa Ana, CA

Dapagliflozin Cayman Chemical, Ann Arbor, MI

DC Protein Quantification kit Bio-Rad Laboratories, Mississauga, ON

Deoxyribonucleoside triphosphate (dNTP) Life Technologies Inc., Burlington, ON

Diaminobenzidine tetrahydrochloride (DAB) Sigma, Oakville, ON

Dithiothreitol (DTT) Life Technologies Inc., Burlington, ON

Page 203: The effects of hyperglycemia on glucose metabolism in

189

DMSO Sigma, Oakville, ON

DNA ladder (100bp) Life Technologies Inc., Burlington, ON

Dulbecco's Modified Eagle Medium (DMEM) Life Technologies Inc., Burlington, ON

Enhanced chemiluminescence (ECL) Perkin Elmer, Waltham, MA

Ethanol Greenfield Ethanol Inc., Brampton, ON

Ethidium Bromide Bio-Rad Laboratories, Mississauga, ON

Ethylenediaminetetraacetic acid (EDTA) Fisher Scientific, Whitby, ON

Extravidin peroxidase Sigma, Oakville, ON

Fetal bovine serum (FBS) Life Technologies Inc., Burlington, ON

Glass coverslips Fisher Scientific, Whitby, ON

Glucose Fisher Scientific, Whitby, ON

Glycerol Fisher Scientific, Whitby, ON

Hematoxylin Fisher Scientific, Whitby, ON

Hydrogen peroxide Sigma, Oakville, ON

Leupeptin Sigma, Oakville, ON

L-glutamine Life Technologies Inc., Burlington, ON

Metformin hydrochloride Sigma, Oakville, ON

Methanol Fisher Scientific, Whitby, ON

MTT Sigma, Oakville, ON

Nitrocellulose membranes Amersham, Piscataway, NJ

Nuclease-free water Applied Biosystems, Foster City, CA

Pepstatin A Sigma, Oakville, ON

Permount Fisher Scientific, Whitby, ON

Phenylmethanesulfony fluoride (PMSF) Roche Applied Science, Laval, QC

Phloretin Santa Cruz Biotechnologies, Santa Cruz, CA

Phlorizin Sigma, Oakville, ON

Phosphate buffered saline Life Technologies Inc., Burlington, ON

Polybrene Santa Cruz Biotechnologies, Santa Cruz, CA

Potassium chloride Fisher Scientific, Whitby, ON

Page 204: The effects of hyperglycemia on glucose metabolism in

190

Potassium iodide Fisher Scientific, Whitby, ON

Potassium phosphate monobasic Fisher Scientific, Whitby, ON

puromycin dihydrochloride Santa Cruz Biotechnologies, Santa Cruz, CA

Rnasin Fisher Scientific, Whitby, ON

Rneasy Mini Kit Qiagen, Toronto, ON

ScintiGest tissue solubilizer Fisher Scientific, Whitby, ON

SGLT2 shRNA lentiviral particles Santa Cruz Biotechnologies, Santa Cruz, CA

Skim milk powder Fisher Scientific, Whitby, ON

SKOV-3 cell line ATCC, Manassas, VA

Sodium azide Fisher Scientific, Whitby, ON

Sodium chloride Fisher Scientific, Whitby, ON

Sodium citrate dihydrate Fisher Scientific, Whitby, ON

Sodium dodecyl sulfate (SDS) Sigma, Oakville, ON

Sodium fluoride (NaF) Fisher Scientific, Whitby, ON

Sodium orthovanadate (NaV) Sigma, Oakville, ON

Streptozotocin Sigma, Oakville, ON

Sucrose Sigma, Oakville, ON

Superfrost Plus glass slides Fisher Scientific, Whitby, ON

SuperScript II Reverse Transcriptase kit Life Technologies Inc., Burlington, ON

Taq DNA polymerase New England BioLabs, Pickering, ON

Tetramethylethylenediamine (TEMED) Sigma, Oakville, ON

Tissue microarray (human ovarian cancer) US Biomax, Rockville, MD

Tris base Fisher Scientific, Whitby, ON

Tris Hydrochloride Fisher Scientific, Whitby, ON

Triton X-100 Sigma, Oakville, ON

Trypsin-EDTA (10x) Invitrogen (Life Technologies)

Turbo DNA-free kit Applied Biosystems, Foster City, CA

Tween-20 Fisher Scientific, Whitby, ON

Vacutainer Serum Separator Tube Fisher Scientific, Whitby, ON

Page 205: The effects of hyperglycemia on glucose metabolism in

191

Western Blot apparatus Bio-Rad

WST-1 Roche Applied Science, Laval, QC

Xylene Fisher Scientific, Whitby, ON

Page 206: The effects of hyperglycemia on glucose metabolism in

192

Appendix III: Recipes for solutions

50 mM sodium citrate buffer (pH 4.5 for streptozotocin)

Sodium citrate 0.441 g in

RO H2O up to 15mL

20x Phosphate Buffered Saline (Stock PBS for IHC)

Sodium chloride 16.0g or

Potassium chloride 0.4g

Sodium potassium dibasic anhydrous 2.3g

Potassium phosphate monobasic (pH 7.4) 0.4g

Immunohistochemistry Blocking Solution

5% BSA 10g

Sodium azide 0.2g

1x PBS 200mL

Citrate Buffer

Stock A – Citrate Buffer

Citric Acid 2.1g

RO H2O 100mL

Stock B – Citrate Buffer

Sodium citrate dihydrate 14.7g

RO H2O 500mL

Page 207: The effects of hyperglycemia on glucose metabolism in

193

Working Citrate Buffer (pH 6.0)

Stock A 18mL

Stock B 82mL

RO H2O up to 1L

Sodium Citrate with Tween 20 (pH 6.0)

Sodium citrate dihydrate 2.94g

RO H2O up to 1L

Tween 20 0.5mL

Carazzi’s Hematoxylin

Hematoxylin 0.25g

Glycerol 50mL

Aluminum potassium sulfate (APS) 12.5g

Potassium iodide 0.05g

RO H2O 200mL

RIPA Lysis Buffer

10mM Tris HCl 0.788g

RO H2O (pH adjusted to 7.6) up to 495mL

5mM Ethylenediaminetetraacetic acid (EDTA) 0.7306g

50mM Sodium chloride 1.461g

30mM Tetrasodium pyrophosphate 3.988g

1% Triton X-100 5mL

Page 208: The effects of hyperglycemia on glucose metabolism in

194

Protease inhibitors per mL RIPA (added fresh before each use)

Aprotinin (2.5mg/mL) 2µL

Phenylmethanesulfonyl fluoride (PMSF; 0.871g/mL) 20µL

Sodium Orthovanadate (NaV; 0.1mM) 2µL

Sodium fluoride (NaF; 50mM) 50µL

Pepstatin A (1mg/mL) 1µL

Leupeptin (2mg/mL) 1µL

Reducing Buffer (3x) (Western blot)

10% SDS 2mL

Glycerol 1mL

1M Tris HCl 0.5mL

pH adjusted to 6.8

Bromeophenol Blue 10mg

RO H2O up to 9mL

DTT 1:7 ratio

5x Running Buffer (Tris-Glycine Buffer; Western blot)

Tris base 15.1g

Glycine 72.1g

10% SDS 10mL

RO H2O up to 1L

Transfer Buffer (Western blot)

Tris base 7.0g

Glycine 3.02g

Methanol (added fresh before use) 200mL

RO H2O up to 1L

Page 209: The effects of hyperglycemia on glucose metabolism in

195

10x Tris buffered saline (TBS; pH 7.6)

Tris base 24.2g

Sodium chloride 80.0g

RO H2O up to 1L

Tween 20 1 ml

50x Tris-acetate-EDTA (TAE) Buffer (PCR)

Tris base 121g

Glacial acetic acid 28.6g

0.5M EDTA 50mL

RO H2O up to 500mL

Page 210: The effects of hyperglycemia on glucose metabolism in

196

Appendix IV: RT-PCR primer sets

Primer set and accession number

Sequence (3’-5’) Annealing

temperature Product size (bp)

SGLT1

(slc5a1)

nm_019810

Forward TCTTCGTCATCAGCGTCATC

55°C 191

Reverse GGGGGCTTCTGTGTCCATTTC

SGLT2

(slc5a2)

nm_133254

Forward ATGATGACCGTGGCTGTGT

55°C 156

Reverse TTCCTGCCCTGTTCCTTTTC

SGLT3

(slc5a3)

nm_017391

Forward TGAGAGACCCAACAGATGAAGA

55°C 165

Reverse TCAAGAAGCCAGCCATTAGAG

GLUT1

(slc2a1)

nm_011400

Forward CAGATGATGCGGGAGAAGAA

60°C 236

Reverse ACAACAAACAGCGACACCAC

GLUT2

(slc2a2)

nm_031197

Forward TCCTGGTCTTCACCCTGTTT

60°C 189

Reverse GGTCGGTTCCTCGGTTTTAG

GLUT3

(slc2a3)

nm_011401

Forward GAACAAAAGACCCCCTCCTC

60°C 209

Reverse AACCTCTCTCCAGCACCAAA

GLUT4

(slc2a4)

nm_009204

Forward CCTGCTTGGCTTCTTCATCT

60°C 171

Reverse TTTGCCCCTCAGTCATTCTC