+ All Categories
Home > Documents > IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of...

IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of...

Date post: 22-Jul-2020
Category:
Upload: others
View: 2 times
Download: 0 times
Share this document with a friend
85
IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC LYMPHOCYTIC LEUKEMIA by Stephanie Yee Ping Tung A thesis submitted in conformity with the requirements for the degree of Master of Science Graduate Department of Medical Biophysics University of Toronto Copyright by Stephanie Yee Ping Tung 2013
Transcript
Page 1: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

IMPROVING GLUCOCORTICOID THERAPY IN

CHRONIC LYMPHOCYTIC LEUKEMIA

by

Stephanie Yee Ping Tung

A thesis submitted in conformity with the requirements

for the degree of Master of Science

Graduate Department of Medical Biophysics

University of Toronto

Copyright by Stephanie Yee Ping Tung 2013

Page 2: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

ii

Improving glucocorticoid therapy in Chronic Lymphocytic Leukemia

Stephanie Yee Ping Tung

Master of Science Graduate Department of Medical Biophysics

University of Toronto 2013

Abstract Glucocorticoids (GCs) are commonly used in the clinic as a treatment for Chronic

Lymphocytic Leukemia (CLL). The exact mechanism of GC action remains unclear and

patients eventually develop resistance to this group of agents. Our findings show that GC-

cytotoxicity in circulating CLL cells is caused by bioenergetic restriction resulting from

the down-regulation of a key glycolytic enzyme, pyruvate kinase, muscle isozyme 2

(PKM2). Conversely, GCs were shown to promote fatty acid oxidation instead by up-

regulating the expression of peroxisome proliferator activated receptor α (PPARα).

These findings establish PPARα and fatty acid oxidation as novel mediators of GC

resistance in CLL. Our findings also demonstrate that GCs enhance the cytotoxic effects

of membrane-damaging agents such as ionophores and complement-mediated

cytotoxicity. A clinically relevant agent known to intercalate in the cell membrane,

Danazol was also found to have activity against CLL and can be combined safely with

GCs for enhanced treatment efficacy.

Page 3: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

iii

Table of Contents Page

Abstract ii Table of Contents iii List of Figures iv List of Publications v List of Abbreviations vi CHAPTER 1: Introduction 1 1.1 Chronic Lymphocytic Leukemia (CLL) 2 1.2 CLL clinical staging 3 1.3 CLL therapy 4 1.4 Glucocorticoids (GCs) 6 1.5 Clinical applications of GCs 8 1.6 Metabolic effects of GCs 10 1.7 Resistance to GC therapy 11 1.8 Fatty acid oxidation 12 1.9 Danazol 15 1.9 Hypothesis 16 1.10 Thesis objectives and organization 16 CHAPTER 2: PPAR-alpha and fatty acid oxidation mediate glucocorticoid 17 resistance in Chronic Lymphocytic Leukemia 2.1 Abstract 18 2.2 Introduction 19 2.3 Materials and methods 20 2.4 Results 24 2.5 Discussion 44 CHAPTER 3: A role for Danazol in Chronic Lymphocytic Leukemia 46 3.1 Introduction 47 3.2 Results 48 3.3 Discussion 54 CHAPTER 4: Discussion and Future Perspectives 55 4.1 GC treatment for proliferating CLL cells 56 4.2 Role of glutaminolysis in GC-mediated cytotoxicity of proliferating CLL cells 58 4.3 Rationale for combinatorial GC treatment of CLL disease 59 4.4 Mechanism of GC-mediated down-regulation of PKM2 59 4.5 Involvement of miR-17 in GC-induced cell death 60 4.6 Concluding remarks 62 LIST OF REFERENCES 63

Page 4: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

iv

List of Figures

Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25

Figure 2 Effects of DEX on mitochondrial membrane potential, 28 intracellular metabolites, and PKM2 expression Figure 3 Enhancement of cytotoxicity from membrane damage by DEX 31 Figure 4 Effect of DEX on PPARα expression 34 Figure 5 Effect of fatty acid oxidation substrates on DEX-mediated 37 cell death Figure 6 Effect of PPARα and fatty acid oxidation inhibitors on 40 DEX-mediated cytotoxicity in vitro and in vivo Figure 7 Supplementary figure 42 Figure 8 Effect of Danazol on CLL cells in vivo and in vitro 49 Figure 9 Effect of Danazol and glucocorticoids on CLL cells 52 in vivo and in vitro

Page 5: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

v

List of Publications

1. Tung, S., Spaner, D.E. A role for Danazol in chronic lymphocytic leukemia. Leukemia. 2012;7:1684-1686.

2. Spaner, D.E., Lee, E. Shi, Y., Wen, F., Li, Y., Tung, S., McCaw, L., Wong, K.,

Gary-Gouy, H., Dalloul, A., Ceddia, R., Gorzcynski, R. PPAR-alpha is a therapeutic target for chronic lymphocytic leukemia. Leukemia. 2012; [19 November Epub ahead of print]

Page 6: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

vi

List of Abbreviations γc

null 2-DG 2-ME 3’-UTR 7-AAD ACM ADP ALL AP-1 ATM ATP Bcl-2 cAMP CBP CCND1 CCND3 cDNA cES CHK6 CLL CPT1 CR CREB Dan Dex/DEX DiOC6(3) DNA DON DSS ERK FACS FAO FBS FCR FFA FISH GC GLS GLUT1 GR GRE HDGCs

IL-2Rγ complete null mutation 2-deoxy-D-glucose 2-mercaptoethanol 3’-untranslated region 7-amino-actinomycin D Adipocyte conditioned media Adenosine diphosphate Acute lymphoblastic leukemia Activating protein 1 Ataxia telangiectasia mutated Adenosine triphosphate B-cell lymphoma 2 Cyclic adenosine monophoshate CREB binding protein Cyclin D1 Cyclin D3 Complementary DNA Embryonic stem cell media Cyclin-dependent kinase 6 Chronic lymphocytic leukemia Carnitine palmitoyl transferase 1 Complete remission cAMP response element-binding Danazol Dexamethasone 3,3’-dihexyloxacarbocyanine iodide Deoxyribonucleic acid 6-diazo-5-oxo-L-norleucine 4,4-dimethyl-4-silapentane-1-sulfonic acid Extracellular-signal-regulated kinase Fluorescence activated cell sorting Fatty acid oxidation Fetal bovine serum Fludarabine-cyclophosphamide-rituximab Free fatty acid Fluorescence in situ hybridization Glucocorticoid Glutaminase Glucose transporter 1 Glucocorticoid receptor Glucocorticoid receptor element High dose glucocorticoids

Page 7: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

viii

HDL HDMP HPA HPRT HSP IgVH I-κB IL-2 IL-6 ip LDL LDT LPL MAG-L MDM2 MEM-α MFI miR MM mRNA mTOR NEFA NF-κB NLS NMR NOD-SCID OS PBS PC PDK4 PEP P-GR PI3K PKB PKM2 PPARα PPARδ Propionyl-CoA Q-PCR

High-density-lipoproteins High dose methylprednisolone Hypothalamic-pituitary-adrenal Hypoxanthineguanine phosphoribosyl transferase Heat shock protein Immunoglobulin heavy-chain variable gene Inhibitor of nuclear factor kappa B Interleukin-2 Interleukin-6 Intraperitoneal Low-density-lipoprotein Lymphocyte doubling time Lipoprotein lipase Monoacylglycerol lipase Murine double minute 2 Minimum essential media α Median fluorescence intensity MicroRNA Multiple myeloma Messenger RNA Mammalian target of rapamycin Non-essential fatty acid Nuclear factor kappa B Nuclear localization signal Nuclear magnetic resonance Non-obese diabetic/Severe combined immunodeficient Overall survival Phosphate buffered saline Proliferation center Pyruvate dehydrogenase kinase 4 Phosphoenolpyruvate Phosphorylated-glucocorticoid receptor Phosphoinositol-3-kinase Protein kinase B Pyruvate kinase muscle isozyme 2 Peroxisome proliferator activated receptor α Peroxisome proliferator activated receptor δ Propionyl coenzyme A Quantitative polymerase chain reaction

Page 8: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

ix

RPMI Roswell Park Memorial Institute medium

RNA Ribonucleic acidSDS-PAGE Sodium-dodecyl sulfate

polyacrylamide gel electrophoresis SRC1 Steroid receptor co-activator-1STAT3 Signal transducer and activator of

transcription 3 STAT5 Signal transducer and activator of

transcription 5 SWI/SNF SWItch/Sucrose NonFermentable TAGs Triacylglycerides TBS-T Tris-buffered saline plus 0.05%

Tween-20 TCA Tricarboxylic acid TLR Toll-like receptor TNF-α Tumor necrosis factor α VLDL Very-low-density-lipoprotein WBC White blood cell ZAP70 70-kDa-zeta-associated protein

Page 9: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

1

1

CHAPTER 1:

Introduction

Page 10: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

2

2

Glucocorticoids (GCs) are one of the oldest agents that have been utilized for the

clinical treatment of a variety of immunological diseases such as autoimmune diseases

and hematological malignancies. The cytotoxic effects of GCs against lymphocytes have

been studied extensively and yet the exact mechanism of action remains uncertain.

Cytotoxicity appears to result from numerous genomic, non-genomic and tissue-specific

effects of GCs. B cell leukemias such as chronic lymphocytic leukemia (CLL) are

commonly treated with GCs as a single agent or in combination with other treatment

modalities including immunotherapy and chemotherapy. GCs have been shown to be an

important element of highly effective regimens for specific subsets of CLL patients

including high-risk patients and patients who develop resistance to first-line

chemotherapy (Thornton et al., 1999; Thornton et al., 2003; Xu et al., 2010).

Understanding the precise mechanism of action of GCs against CLL cells may

allow for the development of strategies to further enhance the efficacy of these drugs.

The limiting factor of this treatment is the eventual development of GC resistance.

Various mechanisms of resistance have been suggested however few have presented

promising targets for the development of clinically-relevant agents. Results presented in

this thesis comprise of investigations into the mechanism of action and resistance of GCs

in circulating CLL cells. Furthermore, data on a new agent, Danazol, with clinical

activity against CLL as a treatment option in combination with GCs will be presented.

1.1 Chronic Lymphocytic Leukemia (CLL) CLL is the most common form of leukemia in North America with an incidence of 1.5 in

100,000 people and is typically found in patients over the age of 40 (Gribben et al.,

2011). The diagnosis of CLL requires a circulating B-cell count of at least 5,000 B

cells/µL as outlined in the International Workshop on CLL guidelines (Hallek et al.,

2008). A distinguishing characteristic of CLL is its unique immunophenotype, which is

the co-expression of the cell surface markers CD5, CD19, CD20 and CD23. However, in

comparison to normal B cells, CLL cells’ expression levels of surface immunoglobulin,

CD20 and CD79b are low (Ginaldi et al., 1998).

CLL cells are thought to arise from CD5+ B cells that have been transformed by

genetic damages sustained at sites of inflammation (Landgren et al., 2007; Klein et al.,

Page 11: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

3

3

2010). These CLL cells divide in proliferation centers (PCs) located in secondary

lymphoid organs such as the bone marrow, spleen and lymph nodes, and eventually enter

the blood stream (Ponzoni et al., 2011). CLL cells divide in PCs in response to

inflammatory microenvironmental factors, such as antigens, chemokines, cytokines and

Toll-like receptor (TLR)-ligands (Chiorazzi et al., 2005). Our lab (Tomic et al., 2011)

and others (Chiorazzi et al., 2005) have shown that responses to these immunoreceptor-

mediated signals are altered to support cell proliferation to promote malignancy of B

cells.

1.2 CLL clinical staging While the clinical course of CLL is highly variable, clinical staging can provide

important prognostic information and guide treatment decisions. Traditionally there are

two widely accepted staging methods, the Rai system (Rai et al., 1975) and the Binet

system (Binet et al., 1981), which both rely on physical examinations and standard

laboratory testing without the need for imaging modalities.

Rai classification defines low-risk disease as patients who exhibit lymphocytosis

(increase in the number of lymphocytes) with leukemia cells in the blood and/or marrow

(formerly Rai stage 0). Intermediate-risk disease is categorized as patients with

lymphocytosis, enlarged nodes in any site and splenomegaly and/or hepatomegaly

(formerly Rai stage I or II). Finally, high-risk disease is defined as patients with disease-

related anemia (formerly Rai stage III) or thrombocytopenia (formerly Rai stage IV)

(Hallek et al., 2008). Binet staging system uses letters A to C to stage CLL disease

according to how many areas are involved as indicated by the presence of enlarged lymph

nodes (>1cm in diameter) and whether there are signs of anemia (low red blood cell

count) or thrombocytopenia (low platelet count) (Hallek et al., 2008).

It is becoming clear that the molecular profile of CLL provides important insights

into disease pathogenesis and prognosis. Currently, fluorescence in situ hybridization

(FISH) is used to categorize CLL into 5 main sub-types based on cytogenetic

abnormalities (Döhner et al., 2000). According to Döhner et al., cytogenetic

abnormalities are detected in 80% of CLL patients. Normal cytogenetic profiles are

detected in around 20% of patients. Deletions in chromosome 13q (13q- deletions) occur

Page 12: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

4

4

in roughly 55% of patients and are associated with a loss of microRNAs (miR-15 and

miR-16) (Calin et al., 2002). Deletions of chromosome 11q (11q- deletions) and trisomy

12 (T12) can be found in 18% and 16% of patients respectively. Chromosome 11q

contains the locus for the ataxixa telangiectasia mutated (ATM) gene, which encodes for

a kinase involved in p53-mediated DNA damage repair (Schaffner et al., 1999). T12

results in the increased expression of genes on chromosome 12, including MDM2, which

is an inhibitor of p53 (Watanabe et al., 1994). The most uncommon abnormality is the

deletion of chromosome 17p (17p- deletions), which encompasses the p53 locus and

occurs in only 10% of patients. p53 is an important mediator of the cellular response to

DNA damage. CLL patients with T12, 11q- or 17p- deletions tend to demonstrate more

aggressive forms of disease, which can be attributed to a malfunction in the p53 DNA

damage pathway (el Rouby et al., 1993; de Viron et al., 2012). 17p- deletion patients tend

to have inferior prognosis and greater resistance to standard chemotherapy. In

comparison, CLL patients with only 13q- deletions tend to have a better prognosis, with

some patients never requiring treatment (Döhner et al., 2000). These cytogenetic changes

can evolve over time and it is important to reassess these markers appropriately to

determine treatment options.

Other common prognostic biomarkers include the mutational status of the

immunoglobulin heavy-chain variable gene (IgVH), use of IgVH and expression of 70-

kDa-zeta-associated protein (ZAP70) and CD38 (Gribben et al., 2011). In addition, there

are also serum markers that can predict survival or progression-free survival in CLL

patients, such as CD23, thymidine kinase and β2-microglobulin (Hallek et al., 2008).

1.3 CLL therapy Upon initial diagnosis of CLL, treatment may not always be necessary. This practice is

due to evidence that has shown that there is no benefit to early or immediate therapy at

least until there is clinical indication that warrants it. For early-stage, asymptomatic CLL

patients (Rai 0, Binet A) who are categorized as low risk, a wait-and-watch approach is

used until the patient starts developing symptoms (Gribben et al., 2011). Patients are

categorized with active disease if they fulfill one of the following criteria: progressive

bone marrow failure (development or worsening of anemia and/or thrombocytopenia),

Page 13: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

5

5

splenomegaly, lymphadenopathy, progressive lymphocytosis (lymphocyte doubling time

(LDT) < 6 months), autoimmune anemia and/or thrombocytopenia that is poorly

responsive to corticosteroids, or the development of constitutional symptoms such as

weight loss, extreme fatigue, high fevers or night sweats (Hallek et al., 2008). For

patients who are symptomatic with active or advanced CLL disease, treatment needs to

be started immediately. Currently, the standard treatment for CLL is combination

chemoimmunotherapy, which consists of fludarabine, cyclophosphamide and rituximab

(FCR) (Gribben et al., 2011). Fludarabine is a purine analog that interferes with DNA

synthesis and has largely replaced the alkylating agent, chlorambucil as the initial, single

agent treatment for patients with symptomatic CLL (Rai et al., 2000). Cyclophosphamide

is an alkylating agent and rituximab is a monoclonal antibody against CD20. FCR

treatment is associated with higher complete remission (CR) rates and improved overall

survival (OS) compared to single agent treatments or FC treatment (Hallek et al., 2010;

Keating et al., 2005; Tam et al., 2008). Even with initial treatment of FCR, some patients

fail to respond and are deemed to have primary resistance. Other patients who do respond

to FCR treatment eventually experience disease relapse and become resistant to

fludarabine-based regimens, which portends extremely poor prognosis (Zenz et al.,

2012).

Complications also arise when the patient’s prognosis is categorized as high-risk,

particularly CLL patients with 17p- deletions. There are debates as to whether starting

these patients on treatment earlier in their disease course is beneficial however there is

still no conclusive evidence to support it (Gribben et al., 2011). In addition, the response

rate of 17p- deletion patients to front line chemotherapy and chemoimmunotherapy is

very poor therefore it is questionable whether FCR is a suitable treatment option

(Hillmen, 2011).

For high-risk 17p- deletion patients and fludarabine-refractory patients, there are

limited treatment options available. In these cases, non-chemotherapeutic agents play an

important role in CLL therapy. Some of the non-chemotherapeutic agents utilized for

treatment include monoclonal antibodies, like Alemtuzumab (anti-CD52 antibody) and

Ofatumumab (fully humanized anti-CD20 antibody), cellular immunotherapy,

immunomodulatory agents (Lenalidomide) and high dose glucocorticoids (HDGCs)

Page 14: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

6

6

(Faderi et al., 2009). Finally, allogeneic stem cell transplantation provides a potential

cure for CLL however this treatment is only suitable for a very small subset of patients

(younger, physically fit patients with high-risk or refractory CLL disease) and there is the

prevalent risk of mortality (Toze et al., 2012).

1.4 Glucocorticoids (GCs) Glucocorticoids (GCs) are naturally occurring steroid hormones produced in the

adrenal cortex under the control of the hypothalamic-pituitary-adrenal (HPA) axis in

response to stress (Smith et al., 2010). GCs have a variety of physiological functions in

the body, including alteration of metabolism, regulation of immune responses, cell

growth and proliferation, development, and reproduction (Kfir-Erenfeld et al., 2010).

Generally, GCs mediate their functions via signaling through the GC receptor (GR)

pathway.

The GR is a member of the steroid/thyroid hormone receptor superfamily that is

ubiquitously expressed, ligand-dependent transcription factor. The gene expressing the

GR is found on chromosome 5 (5q31) in humans and contains nine exons (Theriault et

al., 1989). There are three characteristic domains found in the GR protein: the N-terminal

domain, internal DNA binding domain and the C-terminal domain.

The N-terminal domain contains a transactivation domain that is involved in

transcriptional activation of target genes (Hollenberg et al., 1988). The internal DNA

binding domain contains two zinc finger domains that are highly conserved as a result of

its function for binding to glucocorticoid response element (GRE) sequences of target

genes such as nuclear factor-κB (NF-κB) (Tao et al., 2001). This domain also contains a

nuclear localization sequence (NLS1) (Picard et al., 1987). Finally, the C-terminal

domain houses the ligand-binding domain, which is required for binding heat shock

proteins (HSPs) and for GR dimerization (Giguere et al., 1986). It also contains a second

nuclear localization signal (NLS2) (Picard et al., 1987).

The GR is primarily expressed in the cytoplasm however a cell membrane-

associated receptor has been described as well (Gametchu et al., 1993). As a result of

alternative splicing, there are multiple isoforms of the GR protein that are expressed

differentially in the various tissue types, GRα, GRβ, GRγ, GR-A and GR-P (Hollenberg

Page 15: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

7

7

et al., 1985; Moalli et al., 1993; Bamberger et al., 1995; Rivers et al., 1999; Zhou et al.,

2005). The GRγ isoform is largely expressed in lymphocytes (Rivers et al., 1999).

The unliganded GR protein exists in the cytoplasm as a multi-protein

heterocomplex containing chaperones like HSP90, stabilizing proteins like p23 and

various other molecules (Pratt et al., 1997; Cheung et al., 2000). GCs bind to the inactive

form of the GR in the cytoplasm, which initiates a conformational change in the receptor

that releases it from its chaperones (Elbi et al., 2004). The GR then undergoes multiple

phosphorylation events and subsequent homodimerization (Frankfurt et al., 2004). The

ligand bound, dimerized receptor translocates into the nucleus via the interaction between

its NLS and importins in the nuclear membrane (Freedman et al., 2004).

Once in the nucleus, the dimerized GR can exert its transcriptional effects by

binding to GC response elements (GREs) found in the promoter regions of target genes to

induce either transactivation or transrepression. The consensus GRE sequence consists of

two hexamer half-sites separated by three random nucleotides (Smith et al., 2010). The

most commonly found hexamer half-site sequence is TGTTCT. The number and location

of the GREs within the promoter region of the target genes impacts the intensity of the

transcriptional response (Freedman et al., 1993).

Upon binding to the GRE, the GR can recruit basal transcriptional machinery and

transcriptional co-activators to the start site; such co-activators include cyclic adenosine

monophosphate (cAMP) response element-binding (CREB)-binding protein (CBP),

steroid receptor co-activator-1 (SRC1), p300 and SWI/SNF (Adcock, 2001; Wallberg et

al., 2000). These co-activators induce histone acetylation allowing for transactivation of

target genes. The GR can also induce transrepression of target genes by directly binding

negative GREs within the promoter region (Sakai et al., 1988). On the other hand, GR

can also regulate transcription via direct protein-protein interaction with other nuclear

transcription factors such as NF-κB, AP-1, STAT5 and STAT3, thus affecting the

transcription of genes controlled by these factors (McKay et al., 2000; Ray et al., 1994;

Stocklin et al., 1996; Yang-Yen et al., 1990; Zhang et al., 1997).

One main mechanism of GC action is mediated through its inhibition of NF-κB.

NF-κB is a heterodimeric transcription factor that mediates transcription of cytokines,

cytokine receptors, chemotactic proteins and adhesion molecules, all of which are

Page 16: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

8

8

involved in cell survival, inflammation and various other cellular processes. There are a

few hypotheses that have been suggested regarding the mechanism of GC-mediated NF-

κB inhibition. The first theory is that the GR can directly interact with subunits of NF-κB

(p65) thus inhibiting its activation (Adcock et al., 1999; Liden et al., 1997; Nissen et al.,

2000). Secondly, the GR is also capable of inducing the expression of I-κB, a negative

regulator that sequesters NF-κB in the cytoplasm to prevent nuclear translocation

(Auphan et al., 1995; Scheinman et al., 1995). GCs are also capable of stimulating the

activity of phosphatases involved in the regulation of NF-κB (Shipp et al., 2010). The

variability with which GRs can mediate their transcriptional effects could account for the

cell-specific effects of GCs that have been observed over the years.

1.5 Clinical application of GCs For the purposes of therapeutic usage, synthetic GCs, such as dexamethasone (DEX),

prednisolone and prednisone have been manufactured to be far more potent than

endogenous GCs like cortisol. These synthetic GCs are widely used in the clinic as

immunosuppressive drugs for inflammatory and autoimmune disease, and are also used

for the treatment of various hematological malignancies due to their well-known ability

to induce apoptosis in lymphocytes (Dougherty et al., 1943; Pearson et al., 1949).

Apoptosis is a mode of programmed cell death that regulates the elimination of

cells that are no longer required, have developed improperly or have been genetically

damaged. Apoptosis is a unique process whereby dead cells are broken down into

apoptotic bodies that are removed by phagocytosis without releasing their cytotoxic

cellular contents to the surrounding microenvironment thereby avoiding an inflammatory

response (Sankari et al., 2012). The mechanism for GC-mediated apoptosis in

lymphocytes is postulated to be a combination of both genomic (GR regulation of gene

transcription) and non-genomic (mitochondrial translocation of GR and activation of

signal transduction pathways) effects, however the exact mechanism is unclear and may

vary depending on cell-type (Kfir-Erenfeld et al., 2010).

The first reported use of GCs in CLL treatment dates back to 1949 (Pearson et al.,

1949) and since then, synthetic GCs (especially prednisone and prenisolone) have been

Page 17: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

9

9

commonly used as single agents or in combination with other chemotherapeutic agents.

However, standard-doses of GCs with chemotherapy have not been shown to be

beneficial to the survival of CLL patients (Catovsky et al., 1988; Keating et al., 1998;

O’Brien et al., 1993) and therefore were confined to palliative uses, treating autoimmune

complications and improving cytopenias (Hamblin, 2006; Zent et al., 2010). The first use

of HDGCs in CLL was reported in 1995, whereby refractory CLL patients were treated

with high-dose methylprednisolone (HDMP). HDGC treatment is defined as a 1g/m2 dose

of GCs administered for 5 days (Bosanquet et al., 1995).

Since then, multiple studies have shown that HDGCs alone or in combination

with chemotherapeutic agents can generate better responses for refractory and 17p-

deletion CLL patients (Thornton et al., 1999; Thornton et al., 2003). Overall, HDGCs

were well tolerated with some minor side effects. The rationale for utilizing HDGCs in

the treatment of 17p- deletion patients relates to the p53-independent mechanism through

which GCs induce apoptosis (Johnston et al., 1997). Interestingly, recently published

findings indicate that HDMP treatment can induce complete remission (CR) in

fludarabine-refractory CLL patients (Xu et al., 2010).

Another successful avenue for HDGC therapy in CLL is in combination with

immunotherapy. Combining HDGCs with monoclonal antibodies is logical due to their

different mechanisms of action. It has been shown that the chimeric anti-CD20 antibody,

Rituximab and DEX have synergistic antiproliferative and apoptotic effects in vitro (Rose

et al., 2011). This combination was tested in vivo and proven to generate better response

rates than seen with either agent alone in both fludarabine-refractory and 17p- deletion

patients (Bowen et al., 2007; Dungarwalla et al., 2008; Castro et al., 2008). Another anti-

CD20 antibody clinically available is Ofatumumab, a fully humanized monoclonal

antibody that targets a different epitope on the CD20 molecule than Rituximab (Du et al.,

2009). Preclinical studies have shown that Ofatumumab is more efficient than Rituximab

in inducing complement-induced cell death, which is a major mechanism of action of

monoclonal antibodies (Beum et al., 2008). In vivo, Ofatumumab has been found to be

active against CLL as a single agent and in combination with HDGCs in both

fludarabine-refractory and 17p- deletion patients (Castro et al., 2010; Teichman et al.,

2011; Spaner et al., 2011 Wierda et al., 2010). There have also been successful reports of

Page 18: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

10

10

combining Alemtuzumab (anti-CD52 antibody) with HDGCs for treatment of high-risk

17p- deletion patients (Pettitt et al., 2006; Pettitt et al., 2012).

Unfortunately, a major problem for HDGC therapy is the eventual development of

therapeutic resistance. Another issue with HDGC therapy is the development of adverse

side effects such as hypertension, protein catabolism, cushingoid features, adrenal

suppression, myopathy, diabetes, obesity, osteoporosis and cataract (Kfir-Erenfeld et al.,

2010). However, it is important to note that a majority of these side effects are either

avoidable or treatable if patients are monitored carefully.

1.6 Metabolic effects of GCs Traditionally, GCs are thought to modulate metabolic processes in the body to conserve

glucose during stress or starvation periods (Vegiopoulos et al., 2007). To do this, glucose

uptake is inhibited in muscle and adipose tissue and fat is broken down in adipose tissues

thereby releasing fatty acids for the production of energy (Vegiopoulos et al., 2007). The

notion that GCs’ effects on glucose metabolism could be responsible in part for their

therapeutic effects on malignant lymphocytes has seldom been addressed.

GC-induced apoptosis is known to induce a loss of mitochondrial membrane

potential in human leukemic cells, both established cell lines and primary cells (Eberhart

et al., 2011; Tiefenthaler et al., 2001). The synthetic GC, DEX was found to induce

changes in mitochondrial membrane properties, reduce expression of mitochondrial

transporters of substrates and proteins, which lead to repressed mitochondrial respiratory

activity and lower cellular adenotriphosphate (ATP) levels, which contributed to

apoptosis (Eberhart et al., 2011). Similarly, HDGCs were shown to interfere with the

sodium and potassium transport processes across the plasma membrane therefore leading

to a restriction of cellular ATP supply in activated lymphocytes (Buttergereit et al.,

2000). This bioenergetic restriction was deemed responsible for the therapeutic effects of

HDGCs in activated lymphocytes, such as those found in autoimmune diseases

(Buttergereit et al., 2000). Interestingly, autoimmunity is a common complication in CLL

disease, which is also conventionally treated using HDGCs (Hamblin et al., 2006; Zent et

al., 2010).

Page 19: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

11

11

Recently, it has also been shown that an alternative form of cell death, autophagy

was responsible for GC-mediated cell death in ALL and CLL cells as opposed to the

traditional apoptotic pathway (Grandér et al., 2009). Autophagy is a form of cell death

induced when cells undergo starvation and resort to degrading cellular components using

lysosomal machinery to maintain cellular energy levels (Banergi et al., 2012). This

mechanism of GC-mediated cell death ties in with the energy restriction effects that were

established earlier both with GCs and HDGCs (Tiefenthaler et al., 2001; Eberhart et al.,

2011; Buttergereit et al., 2000).

More recently, it was discovered that the synthetic GC, DEX has a profound

effect on glucose metabolism and uptake in lymphoid and leukemic cells, very similar to

the role they play in skeletal muscle tissue (Buentke et al., 2011). It was shown that DEX

managed to reduce glucose consumption, utilization and uptake in ALL cells (Buentke et

al., 2011). The mechanism of DEX-mediated decrease in glucose uptake was via

decreased expression levels of plasma membrane-associated glucose transporter 1

(GLUT1) (Buentke et al., 2011). More importantly, the inhibition of glucose uptake was

found to correlate with DEX-mediated cell death and lowering glucose concentrations

resulted in increased DEX-mediated cell death (Buentke et al., 2011). All this

information fuels the emerging idea that the metabolic effects of GCs could be linked to

their cytotoxic effects in lymphocytes.

1.7 Resistance to GC therapy The major issue with GC treatment in the clinic is the development of resistance, which

stands in the way of achieving CR in hematological malignancies. Thus far, various

mechanisms of GC resistance have been proposed that mostly involve the GR signaling

pathway and apoptotic machinery, such as altered expression of GR isoforms, GC-

induced alterations in GR expression, mutations in the GR, aberrant expression of anti-

apoptotic proteins such as Bcl-2, failure to induce expression of pro-apoptotic proteins,

like Bim, and interactions with the kinome (Kfir-Erenfeld et al., 2010; Melarangi et al.,

2012; Smith et al., 2010). However, mutations of and alterations to the expression of the

GR are not thought be important in GC resistance in leukemia (Haarman et al., 2003;

Page 20: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

12

12

Tissing et al., 2005). In addition, few mechanisms have offered promising targets for

drug development to overcome GC resistance.

Recent discoveries have found links between glucose metabolism and GC-

resistance in leukemia. B-ALL cells are known to acquire altered glucose metabolism,

higher expression of GLUT1 and sensitivity to the glycolysis inhibitor, 2-deoxy-D-

glucose (2-DG) (Boag et al., 2006). It was demonstrated that GC resistance in precursor

B-ALL is associated with increased expression of genes involved in glucose metabolism

(Holleman et al., 2004) and the glycolytic pathway was confirmed as a modulator GC

resistance in ALL cells (Hulleman et al., 2009). There have also been claims that

modulation of glucose levels influences the effectiveness of GC treatment in ALL

(Buentke et al., 2011). A key factor involved in the regulation of glycolysis is the

serine/threonine kinase, Akt, which is known to regulate glucose uptake and induce

expression of glucose transporters, GLUT1 and GLUT3. Akt is a pro-survival factor that

is part of the mammalian target of rapamycin (mTOR) pathway (Schultze et al., 2012).

Phosphorylation events in the mTOR pathway are also known to regulate glycolysis.

Evidence of the mTOR inhibitor, rapamycin treatment overcoming GC resistance in ALL

cell lines implicates a role for the Akt/mTOR pathway in GC resistance (Wei et al., 2006;

Zhang et al., 2012).

It has also been shown that there is an association between the upregulation of

glycolytic signaling pathways and GC-resistance in T-ALL cell lines (Beesley et al.,

2009). Similar findings have also been discovered in multiple myeloma (MM) whereby

responses to DEX were determined to be glucose-sensitive (Friday et al., 2011). All

together, these findings present strong evidence for the role of metabolism, specifically

glycolysis, in GC-resistance in leukemia.

1.8 Fatty acid oxidation Free fatty acids (FFAs) are a major source of fuel for oxidative metabolism aside from

glucose and glutamine, particularly in cardiac and skeletal muscles. They also play

additional roles in membrane function and structure such as prostaglandin synthesis

(Macfarlane et al., 2008). Fatty acids are carboxylic acids typically categorized based on

Page 21: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

13

13

two aspects of the molecule: the length of the aliphatic chain and the presence of carbon-

carbon double bonds (saturated or unsaturated).

FFAs need to be complexed to albumin or esterified with glycerol to

triacylglycerides (TAGs) and packaged into lipoprotein particles for transport in the

circulation (Spector, 1975). In vivo, the majority of fatty acids are derived from dietary

sources and then stored as TAGs in adipose tissue. When required, TAGs packaged in

lipoprotein or chylomicrons are released into the circulation from the liver and the gut.

These TAGs are then hydrolyzed by lipoprotein lipase (LPL) located on the luminal

surface of the capillary endothelium (Linder et al., 1976), which then allows the FFAs to

be taken up into the cell either via a passive or facilitated process (Garfinkel et al., 1976;

Kalant and Cianflone, 2004). There are two pathways that can process FFAs inside the

cell, the first is β-oxidation in the mitochondria and the second is re-esterification to

TAGs (Macfarlane et al., 2008).

Mitochondrial β-oxidation is the process whereby FFAs are broken down to

generate acetyl-CoA for the production of cellular energy (Eaton et al., 1996). The first

step in β-oxidation is the activation of the FFAs catalyzed by the enzyme, long chain

acyl-CoA synthetase (McGarry and Brown, 1997). The FFA reacts with ATP and

Coenzyme A to generate fatty acyl-CoA ester and AMP. The activated fatty acid is then

transported into the mitochondria via carnitine palmitoyl transferase 1 (CPT1) (Bremer,

1983; McGarry and Brown, 1997). Once inside the mitochondria, two carbon units

(acetyl-CoA) are released with every cycle of β-oxidation (Eaton et al., 1996). This

continues until the entire chain is converted into acetyl-CoA. These acetyl-CoA

molecules can then enter the tricarboxylic acid (TCA) cycle, which produces ATP. This

process is typical for even numbered saturated fatty acids, however the final product is a

propionyl-CoA molecule for odd numbered saturated fatty acids. The propionyl-CoA

molecule can also enter the TCA cycle but it needs to be converted into succinyl-CoA

beforehand (Raph et al., 2010).

The Warburg hypothesis has largely been used to describe the aberrant

metabolism that fuels the growth of most cancer cells. The hypothesis proposes that

cancer cells originate from non-neoplastic cells that acquire a permanent respiratory

defect that bypasses the Pasteur effect (the inhibition of anaerobic fermentation by

Page 22: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

14

14

oxygen). These cancer cells mostly produced energy via a high rate of glycolysis

followed by lactic fermentation in the cytosol, regardless of the presence or absence of

oxygen (Warburg, 1956). In the presence of oxygen, normal non-proliferating cells

typically undergo a low rate of glycolysis followed by pyruvate oxidation in the

mitochondria to maintain efficient energy production.

An alternative hypothesis was proposed that suggested that the increased

dependence of cancer cells on glycolysis stemmed from their inability to synthesize ATP

in response to the mitochondrial proton gradient (Lynen, 1951; Ronzoni and Ehrenfest,

1936). Following work demonstrated that mitochondrial uncoupling (inability to

synthesize ATP in response to mitochondrial proton gradient) results in a shift in

metabolism to the use of non-glucose carbon sources, such as fatty acids, to maintain

mitochondrial function (Belfroy et al., 2008; Pecquer et al., 2008; Rossmeisl et al., 2000;

Samudio et al., 2008).

Interestingly, there is a substantial amount of evidence that implicates fatty acid

oxidation (FAO) as a metabolic hallmark of cancer. Increased FAO has been shown to

promote chemoresistance in tumour cells (Harper et al., 2002). The expression of various

genes involved in lipid metabolism was found to be increased in tumour cells (Hirsch et

al., 2010). An enzyme that releases FFAs from lipid stores, monoacylglycerol lipase

(MAG-L) was found to promote cancer pathogenesis (Nomura et al., 2010) Other studies

have also shown that pharmacologic inhibition of FAO is capable of sensitizing leukemic

cells to apoptosis (Samudio et al., 2010). Recently, the expression of FAO genes such as

carnitine palmitoyltransferase 1C (CPT1C) (Zaugg et al., 2011) and peroxisome

proliferator activated receptor α (PPARα) (Spaner et al., 2012) were found to confer a

survival advantage to tumour cells under conditions of metabolic stress. Given the

metabolic role GCs play in stimulating FAO, it is worthwhile to consider the contribution

of FAO to GC resistance in tumour cells.

Page 23: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

15

15

1.9 Danazol Danazol is a derivative of the synthetic androgen, ethisterone (Dmowski et al., 1971).

Danazol was initially created for the purposes of treating women with endometriosis, an

estrogen-dependent proliferative disease with extrauterine endometrium-like tissue

formation (Friedlander, 1973). The main mechanism by which Danazol prevents

endometriosis is by inhibiting endogenous estrogen production and acting like a

progesterone-like steroid hormone (Barbieri, 1977).

Danazol was subsequently found to be effective in treating various autoimmune

and hematologic disorders such as hereditary angioedema, autoimmune hemolytic anemia

and immune thrombocytopenic purpura (Ahn et al., 1983; Ahn et al., 1985). There has

also been evidence to suggest that Danazol has activity against myelodysplatic

syndromes, which are disorders of the stem cells in the bone marrow (Fontana et al.,

2011; Sadek et al., 2000). The mechanism of action of Danazol in immune cells still

remains a subject of investigation. However, various mechanisms have been suggested

such as decreased production of inflammatory cytokines, plasma cell membrane

intercalation and decreased expression of serum soluble CD23 from the membranes of

activated B cells (Horstman et al., 1995; Matalliotakis et al., 2000; Odukoya et al., 1995;

Tanaka et al., 2009). The therapeutic activity Danazol has against B-lymphocytes

suggests that it could be utilized in the treatment of hematological malignancies, such as

CLL.

Page 24: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

16

16

1.10 Hypothesis Glucocorticoid (GC) treatment of chronic lymphocytic leukemia (CLL) can be improved

by targeting metabolic aspects of resistance.

1.11 Thesis objectives and organization 1. In an effort to improve GC treatment, the relationship between the metabolic and

cytotoxic effects of GCs against CLL cells was investigated. In addition,

metabolic strategies to overcome GC resistance were studied. The results of these

studies are presented in Chapter 2.

2. The role of a clinically relevant agent, Danazol, was also studied as a potential

combination option with GC treatment of CLL. The results of these studies are

presented in Chapter 3.

Page 25: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

17

17

CHAPTER 2:

PPAR-alpha and fatty acid oxidation mediate glucocorticoid-

resistance in Chronic Lymphocytic Leukemia

Stephanie Tung,1,2 Yonghong Shi,1 Karry Wong,5,6 Reg Gorczynski,5,6 Robert

Laister,7 Mark Minden,2,4,7 and David E. Spaner,1,2,3,4,5

1Division of Molecular and Cellular Biology, Sunnybrook Research Institute, Toronto, Canada, M4N 3M5; 2Department of Medical Biophysics, University of Toronto, Toronto, Canada, M5G 2M9; 3Sunnybrook Odette Cancer Center, Toronto, Canada, M4N 3M5; 4Department of Medicine, University of Toronto, Toronto, Canada, M5G 2C4; 5Department of Immunology, University of Toronto, Toronto, Canada, M5S 1A8; 6Transplant Research Division, Toronto General Hospital, Toronto, Canada M5G 2C4; 7Ontario Cancer Institute / Princess Margaret Hospital, Toronto, Canada M5G 2M9

Page 26: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

18

18

2.1 Abstract Glucocorticoids (GCs) are an important treatment for Chronic Lymphocytic

Leukemia (CLL). However, resistance to GCs inevitably develops and the mechanisms of

action of GCs in CLL are not well understood. The synthetic GC dexamethasone (DEX)

was found to decrease pyruvate kinase M2 (PKM2) expression and metabolic activity of

CLL cells, indicated by loss of mitochondrial membrane potential and decreased levels of

pyruvate and its metabolites. This metabolic restriction was associated with decreased

size and resulted in the tumor cell death that was increased by concomitant damage to

plasma membranes. However, the nuclear receptor Peroxisome proliferator activated

receptor (PPAR)-alpha, which regulates fatty acid oxidation, was also increased by DEX

and adipocyte-derived lipids, lipoproteins, and propionic acid protected CLL cells from

DEX-mediated death. PPARalpha and fatty acid oxidation enzyme inhibitors increased

DEX-mediated killing of CLL cells in vitro and clearance of CLL xenografts in vivo.

These findings suggest that GCs prevent tumor cells from generating the energy they

need to repair membrane damage, fatty acid oxidation is a mechanism of resistance to

GC-mediated cytotoxicity, and PPARalpha-inhibition is a strategy to improve the

therapeutic efficacy of GCs.

Page 27: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

19

19

2.2 Introduction The outcome of high-risk CLL patients whose tumor cells harbor deletions in the

region of chromosome 17 that encodes p53 or who relapse shortly after completing first-

line therapy with fludarabine, cyclophosphamide, and rituximab (FCR) is poor (Zenz et

al., 2012). Despite the advent of newer agents (Wiestner et al., 2012), high-dose synthetic

glucocorticoids (GCs) such as methylprednisolone or dexamethasone (DEX), with or

without monoclonal antibodies such as Ofatumumab, remain among the most effective

treatments for these patients (Spaner, 2011). Unfortunately, GC-based regimens are not

curative and resistance inevitably develops with extended use. Considering their long

clinical history (Hench et al., 1949), knowledge of how GCs exert their therapeutic

effects is surprisingly incomplete. A better understanding of the mechanism of action and

nature of resistance are needed to improve the efficacy of GC therapy for CLL patients.

GCs are steroid hormones made by the adrenal cortex that affect a variety of cell

functions. They inhibit glucose metabolism and promote fatty acid oxidation during

starvation and also have potent immunosuppressive properties (Sapolsky et al., 2000).

GCs bind to glucocorticoid receptors (GRs), which are members of the nuclear receptor

family of ligand-dependent transcription factors. Activation by ligands causes GRs to

become phosphorylated and translocate to the nucleus where they bind GC-response

elements (GREs) and mediate gene transcription. Ligand-activated GRs transactivate

genes such as IκB that inhibit signaling pathways (Flammer et al., 2011) and also

transrepress signaling through direct binding to kinases (Glass et al., 2010). GCs cause

CLL cells to undergo apoptosis (Kfir-Erenfeld et al., 2010) but the processes that lead

from GR-phosphorylation to cell death are not fully understood. The studies here were

designed to provide more insight into the mechanisms of action of GCs in CLL.

Page 28: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

20

20

2.3 Materials and Methods Antibodies and reagents.

7-aminoactinomycin D (7-AAD) was obtained from Pharmingen (San Francisco, CA).

Fatty acid free bovine serum albumin, 2-mercaptoethanol (2-ME), Mifepristone, 3,3’-

dihexyloxacarbocyanine iodide (DiOC6(3)), propionic acid, ionomycin, GW6471

(PPARα-antagonist), and β-actin antibodies were from Sigma (St. Louis, MO). RPMI-

1640, lipid-rich bovine albumin (AlbuMAX®II), and Mitotracker® Red and Green were

from Invitrogen (Carlsbad, CA). Clinical grade dexamethasone sodium phosphate

(Omega, Montreal, Quebec) was purchased from the hospital pharmacy. Ofatumumab

was obtained from GlaxoSmithKline (London, UK). Low-Tox®-M Rabbit Complement

was purchased from Cedarlane (Burlington, ON, Canada). Antibodies against phospho-

(Ser211)glucocorticoid receptor and pyruvate kinase muscle isozyme (PKM2) were from

Cell Signaling Technology (Beverly, MA), as were secondary horseradish peroxidase-

conjugated anti-rabbit and anti-mouse antibodies (Cat. Nos. 7074 and 7076, respectively).

PPARα antibodies were from Cayman Chemical Co. (Ann Arbor, MI). High-, low- and

very-low-density lipoproteins were from EMD Chemicals (San Diego, CA). MK886 and

Compound A (PPARα-antagonists) were generous gifts from Inception Sciences (San

Diego, CA) and CVT-4325 (GS449794, β-oxidation inhibitor) was a generous gift from

Gilead Sciences (Foster City, CA).

CLL cell purification.

CLL cells were isolated as previously described by negative selection from the blood of

consenting patients (Tomic et al., 2011), diagnosed by a persistent monoclonal expansion

of CD19+CD5+IgMlo lymphocytes and untreated for at least 3 months. Patient

characteristics are described in Supplementary Table 1. Protocols were approved by the

Sunnybrook Ethics Review Board.

Cell culture.

Unless otherwise specified, purified CLL cells were cultured at a concentration of 1 x 106

cells/mL in RPMI-1640 medium supplemented with Transferrin and 0.02% AlbuMAX II

in 6- or 24-well plates (BD Labware) at 37°C in 5% CO2 for the times indicated in the

figure legends.

Page 29: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

21

21

Preparation of adipocytes and adipocyte conditioned medium (ACM).

OP-9 cells were maintained in OP-9 propagation medium consisting of MEM-α

(Minimum Essential Media α), 20% FBS (Fetal Bovine Serum; BioWhittaker,

Walkersville, MD), 100 U/ml penicillin, and 100 mg/ml streptomycin (Wisent USA). The

cells were re-plated every 3 days upon growing to confluence. Adipocytes were obtained

by the method of Wollins et al. (Wollins et al., 2006). OP-9 cells were grown to

confluence in propagation media which was then replaced by stem cell medium (cES)

(Garcia-Gonzalo et al., 2008), consisting of Advanced-DMEM-F12, 15% KnockOut™

Serum Replacement, 1% Glutamax-1 (all from Invitrogen), 1% MEM nonessential amino

acids (Wisent USA), and 0.1µM 2-ME. Within 3-4 days, more than 90% of the OP-9

cells become filled with large lipid droplets that were visible in a light microscope

(Fig.10C). To obtain ACM, OP-9 adipocytes were washed and incubated with fresh cES

for 3 days. ACM was collected and heat-inactivated at 60°C for 30 min.

Flow cytometry.

Cell viability was measured by washing cells in phosphate-buffered saline (PBS) and

then incubating with 3.5 µL of 7-AAD for 10 min. Mitochondrial membrane potential

was measured by staining with 0.2 nM of 3,3’-dihexyloxacarbocyanine iodide

(DiOC6(3)) for 15 min at room temperature followed by washing with PBS.

Mitochondrial mass was measured by staining with 2 nM of MitoTracker Green FM for

15 min at 37°C followed by washing with PBS. Ten thousand viable counts were

analyzed with a FACScan flow cytometer using Cellquest software (Becton Dickinson).

Standardization of the flow cytometer was performed before each experiment using

SpheroParticles (Spherotech Inc., Chicago, IL, USA).

Immunoblotting.

Protein extraction and immunoblotting were performed as previously described (Tomic et

al., 2011). Proteins were resolved in 10% sodium dodecyl sulphate-polyacrylamide gel

electrophoresis (SDS-PAGE) and transferred onto Immobilon-P transfer membranes

(Millipore Corp., Billerica, MA, USA). Western blot analysis was performed according

to the manufacturers’ protocols for each antibody. Chemiluminescent signals were

created with SupersignalWest Pico Luminal Enhancer and Stable Peroxide Solution

(Pierce, Rockford, IL, USA) and detected with a Syngene InGenius system (Syngene,

Page 30: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

22

22

Cambridge, UK). For additional signal, blots were stripped for 60 min at 37°C in Restore

Western Blot Stripping Buffer (Pierce) and washed twice in TBS-T (Tris-buffered saline

plus 0.05% Tween-20) at room temperature and then re-probed as required. Densitometry

was performed using Image J software. The densitometry value for each patient sample

was normalized against the value obtained for β-actin to obtain the intensities for P-GR,

PKM2 and PPARα reported in the figures.

Real-time PCR.

RNA was prepared with the RNeasy mini kit (Qiagen, Valencia, CA, USA), and cDNA

was synthesized from 2 µg of RNA using Superscript III reverse transcriptase (Life

Technologies, Invitrogen), according to the manufacturer’s instructions. PKM2, PPARα,

PPARδ, pyruvate dehydrogenase kinase 4 (PDK4), and hypoxanthineguanine

phosphoribosyl transferase (HPRT) transcripts were amplified with the following

primers: PKM2 forward, GTCGAAGCCCCATAGTGAAG; reverse,

ATGTCCTTCTCCGACACAGC, PPARα forward, CTGGAAGCTTTGGCTTTACG;

reverse, ACCAGCTTGAGTCGAATCGT, PPARδ forward,

CTCTATCGTCAACAAGGACG; reverse, GTCTTCTTGATCCGCTGCAT, PDK4

forward, CATACTCCACTGCACCAACG; reverse, CCTGCTTGGGATACACCAGT,

and HPRT forward, GAGGATTTGGAAAGGGTGTT; reverse

ACAATAGCTCTTCAGTCTGA. Polymerase chain reactions were carried out in a DNA

engine Opticon System (MJ Research, Waltham, MA, USA) and cycled 34 times after

initial denaturation (95°C, 15 min) with the following parameters: denaturation at 94°C

for 20 sec, annealing of primers at 58°C for 20 sec, and extension at 72°C for 20 sec.

mRNA abundance was evaluated by a standard amplification curve relating initial copy

number to cycle number. Copy numbers were determined from two independent cDNA

preparations for each sample. The final result was expressed as the relative fold change of

the target gene to HPRT.

Sample preparation for NMR.

To measure intracellular metabolite levels, 5x 107 CLL cells were pelleted, resuspended

in 1 mL of ice cold PBS, transferred into a microfuge tube and pelleted again. Pellets

were extracted with methanol 3 times by adding 1 mL of -80°C, 80% methanol,

vortexing, incubating on ice for 30 min, vortexing again, and then centrifuging for 30 min

Page 31: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

23

23

at 10000 rpm at 4°C. The supernatants were collected into 2 mL microfuge tubes and the

solvent was removed with a centrifugal evaporator (SpeedVac) at room temperature.

Metabolite extracts were stored at -80C until NMR analysis. 1H NMR.

The extracts were taken up in 120µL of NMR buffer (50mM Na2HPO4, 0.1% NaN3 in

D2O pH 7.0; uncorrected). DSS (0.5mM) was added as an internal reference standard and

the sample was transferred to a 3mm NMR tube. All NMR spectra were run on a Bruker

Avance II 800MHz spectrometer equipped with a triple resonance cryoprobe. One

dimensional (1D) 1H NMR spectra were collected at 298K with 128 scans using a 90°

proton pulse and recycle delay of 2 sec. A total of 32K points were collected over a

spectral width of 12.8kHz (16ppm). Metabolites were identified and quantified using the

Chenomx 7.1 NMR software suite (Chenomx Inc., Edmonton, AB). Metabolite

concentrations are reported as mean values ± standard deviation.

CLL xenograft model.

NOD-SCIDγcnull mice were sublethally irradiated (245 Rads) and then injected

intraperitoneally (ip) with 2.5 x108 thawed CLL splenocytes that had been obtained at the

time of therapeutic splenectomy. Subsequently mice were injected bi-weekly ip with 700

µl of plasma pooled from 8-10 patients with white cell counts greater than 100x106

cells/ml. Plasma injections continued until engraftment was established in the spleen and

the peritoneal cavities (6 weeks). Single cell suspensions from spleens and peritoneal

cavities (PCs) were counted in a hemocytometer and analyzed by multi-color FACS to

assess engraftment of CLL cells, as before. (Spaner et al., 2012; Wong et al., 2012)

Statistical analysis.

The Student’s t-test and paired t-tests were used to determine p-values.

Page 32: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

24

24

2.4 Results Dexamethasone induces cell death and decreases the size of circulating CLL cells

DEX (30 µM) killed purified CLL cells within 48 h in serum-free culture conditions

involving RPMI-1640 media with 0.02% lipid-rich albumin. An example (Fig.1A) and

results for 52 patient samples (Fig.1B) are shown. The dose of DEX was chosen to

approximate plasma levels that result when patients are treated with high-dose GCs

(Spaner et al., 2012). Cell death was prevented by the GR antagonist Mifepristone

(Agarwai et al., 1996) (Fig.4A, bottom panel), suggesting it was a direct, transcriptional

effect of DEX.

The number of CLL cells killed by DEX exhibited inter-patient variability

(Fig.1B), which could not be correlated with the clinical characteristics of the patients. In

all cases studied, the GR was phosphorylated by DEX, suggesting this variability was not

due to differences in activation of the GR (Fig.1C). Despite the fact that many DEX-

treated cells remained alive, as measured by their ability to exclude the DNA-dye 7-

AAD, in all cases they decreased in size, as measured by the forward scatter parameter of

flow cytometry (Fig.1D).

Page 33: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

25

25

Page 34: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

26

26

Figure 1. Effect of Dexamethasone on circulating CLL cells. A. CLL cells were

purified and cultured for 48 h with or without dexamethasone (DEX) (30 µM) in serum

free media (RPMI-1640 with Transferrin and 0.02% Albumax) in the presence or absence

of the GR antagonist Mifepristone (1 µM). After 48 h, percentages of viable 7AAD- cells

that exclude 7AAD were determined by flow cytometry and shown in the right upper

corners of the dot-plots. Mifepristone prevented DEX-induced cell death. B. Results for

52 different CLL patient samples are shown. Each line represents percentages of 7AAD+

cells after 48 h in the presence or absence of DEX. C. Circulating CLL cells from the

indicated patients were cultured for 4 h with or without DEX. Levels of Ser211

phosphorylated glucocorticoid receptor (P-GR) were measured by immunoblotting with

β-actin as a loading control and quantified by densitometry. D. Summary of forward

scatter median measurements at 48 h by flow cytometry indicating that DEX significantly

decreased the size of circulating CLL cells. Paired t-tests were used to determine p-

values. **, p<0.01

Page 35: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

27

27

Dexamethasone decreases mitochondrial membrane potential and intracellular

metabolites

Since size reflects metabolic activity (DeBerardinis et al., 2008), the smaller size

suggested that DEX might be acting to restrict the metabolic activity of CLL cells.

Cellular metabolism is supported by mitochondria and requires the generation and

maintenance of a mitochondrial membrane potential that can be measured by flow

cytometry with the cell-permeable, green-fluorescent dye 3,3’-dihexyloxacarbocyanine

iodide (DiOC6(3)). As early as 4 h after DEX treatment, CLL cells exhibited a significant

decrease in mitochondrial membrane potential (Fig.2A, left) that was sustained for at

least 18 h (Fig.5A, right). The decrease in membrane potential could not be attributed to

decreased mitochondrial mass (Supplementary Fig.7A) but could be prevented by

Mifepristone (see Fig.5F), suggesting it was a transcriptional effect of DEX.

Levels of small-molecule intracellular metabolites were then measured by nuclear

magnetic resonance (NMR) spectroscopy. Consistent with restricted metabolism, a

number of metabolites were decreased by DEX. Intracellular glutamate, alanine, and

lactate levels were significantly lower (Fig.2B). Pyruvate, succinate, and glutamine levels

were also decreased while acetate was relatively preserved (Fig.2B). Pyruvate is a

product of glycolysis and glutaminolysis that gives rise to lactate and alanine. Glutamate

is a product of glutaminolysis that provides Tricarboxylic Acid (TCA) cycle

intermediates, such as succinate. (DeBerardinis et al., 2008) Acetate is a product of fatty

acid oxidation (Lin et al., 1996). Note that these changes in metabolite levels were

measured at 18 h, prior to any loss of membrane integrity associated with cell death

(Fig.2B) and could not be attributed simply to cytotoxicity.

Dexamethasone downregulates pyruvate kinase expression

The decrease in pyruvate, lactate, and alanine levels suggested that genes associated with

pyruvate generation were decreased by DEX. Pyruvate kinase muscle isozyme-2 (PKM2)

catalyzes the last step of the glycolytic pathway and transfers a phosphate group from

phosphoenolpyruvate to adenosine diphosphate to make pyruvate and ATP (Tamada et

al., 2012). DEX-treated CLL cells down-regulated PKM2 mRNA and protein levels

within 4 h (Fig.2C,D).

Page 36: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

28

28

Page 37: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

29

29

Figure 2. Effect of DEX on mitochondrial membrane potential, intracellular

metabolites, and PKM2 expression. A. Cells from 8 different CLL patients were

cultured with or without DEX. At the indicated times, the cells were stained with

DiOC6(3) to measure mitochondrial membrane potential. Median fluorescence intensities

(MFIs) are shown for each sample. B. 5 x 107 cells from 5 different CLL patients were

cultured for 18 h in the presence or absence of DEX and then analyzed by 1D 1H NMR

spectroscopy. The averages and standard errors for the 5 samples are shown. Glutamate,

alanine and lactate levels were significantly lowered by DEX, while acetate levels were

relatively preserved. C. CLL cells from 9 patients were cultured with or without DEX.

After 4 h, PKM2 mRNA transcripts (relative to HPRT transcripts) were measured by

quantitative PCR. D. After 18 h of culture, phospho-GR and PKM2 expression were

measured in DEX-treated cells from 4 different patients by immunoblotting and

quantified by densitometry, with β-actin used as a loading control. PKM2 mRNA and

protein were both decreased by DEX. Student’s t-tests and paired t-tests were used to

determine p-values. **, p<0.01; *, p<0.05. NMR spectroscopy data courtesy of Rob

Laister.

Page 38: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

30

30

Concomitant membrane damage enhances DEX-mediated cytotoxicity

The above results suggested that DEX restricted cellular metabolism in CLL cells in part

by down-regulating PKM2. However, increased metabolic activity is needed to generate

the substrates and energy required to repair cellular damage. Accordingly, DEX may

inhibit the ability of CLL cells to repair membrane damage and subsequently increase

killing by membrane damaging agents. To address this hypothesis, CLL cells were

treated with Ofatumumab and/or complement in the presence or absence of DEX. In the

absence of complement, Ofatumumab did not change basal killing by DEX in serum-free

conditions (Fig.3A). Ofatumumab binds to CD20 and causes complement to deposit on

the cell surface, forming the membrane attack complex and resulting in membrane

damage and cell lysis (Rose et al., 2002). Death of CLL cells from complement damage

was increased significantly by DEX (Fig.3A).

Ionomycin is a calcium ionophore that creates pores in plasma membranes,

causing electrochemical gradient disturbances that require significant energy to repair.

(Krauss et al., 2001) Consistent with the hypothesis that DEX restricts the metabolic

activity needed to repair such damage, ionomycin-mediated cell death was increased

significantly by DEX (Fig.3B). The serum-free conditions used in these studies would

also be expected to damage plasma membranes through detergent-like effects of the fatty

acids bound to lipid-rich albumin (Glatz et al., 2010; Holzer et al., 2011). Removing this

source of membrane disruption should then decrease killing of CLL cells by DEX.

Consistent with this idea, DEX-mediated cell death was significantly reduced when

Albumax was replaced by fatty acid free albumin (Fig.3C). Note that the decrease in cell

size was still observed (not shown).

Page 39: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

31

31

Page 40: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

32

32

Figure 3. Enhancement of cytotoxicity from membrane damage by DEX. A. Purified

CLL cells were cultured with or without DEX, the monoclonal CD20 antibody

Ofatumumab (7 µg/ml), or rabbit complement (1:40 final dilution). Cells were stained

with 7AAD after 48 h and analyzed by flow cytometry. Specific death is the difference

between the percentages of 7AAD+ cells in control samples and treated samples. The

averages and standard errors of the results for the indicated numbers of samples are

shown. B. Similar experiments were carried out in the presence or absence of the

indicated doses of ionomycin. C. Purified CLL cells from 52 different patients were

treated with DEX in RPMI-1640, Transferrin, and 0.02% lipid-rich Albumax or with

RPMI-1640, Transferrin, and 0.02% fatty acid free (FAF) albumin to remove free fatty

acids that could potentially damage cell membranes in culture. Specific death was

determined after 48 h and was significantly lower in FAF albumin. **, p<0.01; *, p<0.05

Page 41: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

33

33

DEX increases PPARα expression

The results in Figure 2 suggested that the capacity of CLL cells to generate pyruvate was

compromised by DEX. However, the relatively preserved acetate levels (Fig.2B)

suggested that genes associated with fatty acid oxidation might be increased in DEX-

treated CLL cells. PPARα and PPARδ are nuclear receptors involved in the regulation of

fatty acid oxidation (Harmon et al., 2011; Spaner et al., 2012). DEX up-regulated PPARA

and PPARD mRNA and PPARα protein expression (Fig.4A,B,D). The changes in

PPARA mRNA occurred later than the down-regulation of PKM2.

Among the genes that are regulated by PPARα and PPARδ is pyruvate

dehydrogenase kinase 4 (PDK4). PDK4 phosphorylates and inactivates pyruvate

dehydrogenase (PDH), preventing pyruvate from being oxidized in mitochondria and

promoting fatty acid oxidation (Schulze et al., 2011). Consistent with the increase in

PPARα and PPARδ, PDK4 mRNA expression was also increased by DEX (Fig.4C).

Page 42: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

34

34

Figure 4. Effect of DEX on PPARα expression. CLL cells from the indicated patient

samples were treated with or without DEX for 18 h. PPARA (A), PPARD (B), and PDK4

(C) transcripts (relative to HPRT) were then measured by quantitative PCR. D.

Expression of phospho-GR and PPARα proteins were also measured in 4 patients by

immunoblotting, with β-actin used as a loading control. PPARα protein and mRNA were

both increased by DEX at this time.

Page 43: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

35

35

Adipocyte-derived lipids, lipoproteins and propionic acid reverse the effects of DEX

The decreased expression of PKM2 coupled with increased expression of PPARα,

PPARδ, and PDK4 suggested that DEX might cause CLL cells to depend primarily on

fatty acid oxidation as a metabolic strategy. Accordingly, they might be able to use fatty

acids to generate energy, support mitochondrial activity, and resist the cytotoxic effects

of GCs. To investigate this possibility, cultures of DEX-treated cells in serum-free

conditions were supplemented with various forms of fatty acid oxidation substrates.

Adipocytes are a major source of fuel for fat-burning tissues and are often found

in lymphoid microenvironments where CLL cells reside in vivo (Pond et al., 2005). To

test whether adipocyte-derived lipids could confer resistance to DEX, CLL cells were co-

cultured with OP-9-derived adipocytes (Wollins et al., 2006). OP-9 is a mesenchymal

stem cell line that differentiates rapidly into adipocytes in serum-free conditions

(Supplementary Fig.7C). CLL cells were highly resistant to DEX in the presence of

adipocytes, compared to CLL cells in serum-free conditions alone (Fig.5A).

Soluble factors from adipocytes were mainly responsible for the enhanced

survival of DEX-treated CLL cells since conditioned media from OP-9-derived

adipocytes also prevented DEX-mediated killing of CLL cells (Fig.5B). Much of the

protective effect survived heat inactivation, suggesting it was due to lipid factors.

Lipoproteins transport lipids to tissues that use fatty acid oxidation to generate

energy. Very-low density lipoproteins (VLDL) and low-density lipoproteins (LDL)

increased the survival of DEX-treated cells (Fig.5C) while high-density lipoproteins

(HDL), with lower triglyceride content, had little effect (not shown). Addition of long

chain (>12 carbon) fatty acids increased the death of DEX-treated CLL cells, perhaps

because of additional detergent-like effects in serum-free culture, as described above (not

shown). However, short-chain fatty acids and ketone bodies also failed to increase the

survival of DEX-treated CLL cells, despite their capacity to be used as fuel by fat-

burning tissues without causing membrane damage (not shown). Given that an intact

TCA cycle is required to oxidize fatty acids in mitochondria, even-numbered short-chain

fatty acids may not be able to be oxidized in DEX-treated cells because of the impaired

TCA cycle (Fig.5). However, odd-numbered fatty acids can be used for both anaplerosis

(i.e. to restore depleted TCA cycle intermediates) and as a fuel source. (Brunengraber et

Page 44: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

36

36

al., 2006) Propionic acid is a short chain 3-carbon fatty acid that is first converted to

propionyl coenzyme A (propionyl-CoA) and then to succinyl-CoA, an intermediate of the

TCA cycle. Supplementation with proprionic acid maintained the mitochondrial potential

of DEX-treated cells, as measured by DiOC6(3) staining, and increased their viability

(Fig.5D,E,F).

Note that supplementation with glucose or pyruvate did not increase survival of

DEX-treated CLL cells (Supplementary Fig.10E), consistent with a reduced ability to

oxidize glucose (Fig.2,4). Supplementation with glutaminolysis metabolites, including

glutamine, glutamate, or alpha-ketoglutarate also failed to rescue DEX-treated CLL cells

(Supplementary Fig.7D).

Page 45: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

37

37

Page 46: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

38

38

Figure 5. Effect of fatty acid oxidation substrates on DEX-mediated death. A.

Adipocytes and adipocyte conditioned media (ACM) were prepared as described in the

materials and methods. 6 x 106 CLL cells were co-cultured with OP-9-derived adipocytes

(A) or in ACM (with or without heat-inactivation) (B) in the presence or absence of DEX

for 48 h. Specific death was then determined by staining with 7AAD and flow cytometric

analysis. C. CLL cells from 10 different patients were cultured with or without DEX in

the presence or absence of low-density lipoproteins (LDLs) or very-low-density

lipoproteins (VLDLs) (1:200 and 1:100 final concentrations, respectively). The averages

and standard errors of the results for each treatment are shown. D. CLL cells from 15

individual patients were cultured with or without DEX in the presence or absence of

propionic acid (1 mM), an odd-numbered fatty acid that provides both acetyl-CoA and

succinate to support the TCA cycle. Percentages of viable 7AAD- cells were measured 48

h later by flow cytometry. Specific death is the difference between the percentages of

7AAD+ cells in control and DEX-treated samples. E. Mitochondrial membrane potentials

of DEX-treated CLL cells supplied with propionic acid were determined after 18 h by

staining with DiOC6(3) and flow cytometric analysis. As a control, DEX-treated CLL

cells were also treated with the GR antagonist Mifepristone (1 µM). An example is

shown. F. Summary of DiOC6(3) MFI measurements for 8 different patient samples,

indicating that propionic acid and Mifepristone both restore mitochondrial membrane

potential in DEX-treated CLL cells. **, p<0.01; *, p<0.5

Page 47: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

39

39

PPARα and fatty acid oxidation mediate GC resistance

The findings that PPARα was upregulated by DEX (Fig.4) and fatty acid oxidation

substrates prevented DEX-mediated death (Fig.5) suggested that PPARα and fatty acid

oxidation might mediate resistance of CLL cells to GCs. A previously described CLL

cell-line model was then used to determine the effect of increased PPARα-expression on

sensitivity to DEX (Spaner et al., 2012). CD5+ Daudi cells were transfected with human

PPARA and a clone was obtained with high PPARα expression (Fig.6A) (Spaner et al.,

2012). DEX activated the GR in both the PPARα-expressing cell line and its empty

vector control, as determined by increased phospho-GR levels (Fig.6A). However the

PPARα-expressing cell line was found to be resistant to DEX treatment in comparison to

the empty vector control (Fig.6B).

Conversely, circulating CLL cells treated with the small molecule PPARα-

inhibitors MK886 (Spaner et al., 2012), GW6471 (Xu et al., 2002) or Compound A

(Shearer et al., 2007) became more sensitive to DEX. (Fig.6C) In addition, the fatty acid

oxidation inhibitor CVT-4325 (Elzein et al., 2004) also enhanced killing of CLL cells by

DEX (Fig.6C, bottom right panel).

PPARα antagonists enhance clearance of CLL cells by DEX in vivo

The ability of MK886 to improve the efficacy of DEX against CLL cells in vivo was then

studied in a xenograft model involving transfer of spleen cells from patients who have

undergone therapeutic splenectomies into NOD-SCIDγcnull mice along with injections of

CLL plasma to support tumor engraftment (Spaner et al., 2012; Wong et al., 2012).

Human T and CLL-B cells are found mainly in spleens and peritoneal cavities of tumor-

bearing mice by 6 weeks after adoptive transfer (Wong et al., 2012). Groups of

reconstituted mice were then given 4 daily ip injections of either saline, MK886 at 10

mg/kg, which was shown previously to clear CLL cells from peritoneal cavities but not

spleens (Spaner et al., 2012), DEX at 4.6 mg/kg (the dose in the high-dose GC regimen

for CLL (Spaner et al., 2011)) or both MK886 and DEX. DEX alone did not clear CLL

cells from spleen. However, the combination of MK886 and DEX decreased splenic

tumor burdens better than either agent alone (Fig.6D).

Page 48: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

40

40

Page 49: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

41

41

Figure 6. Effect of PPARα and fatty acid oxidation inhibitors on DEX-mediated

cytotoxicity in vitro and in vivo. A. CD5+ Daudi cells engineered to over-express

PPARα and vector-control cells were treated with DEX for 48 h. Expression of PPARα

and the activated, phosphorylated GR were then determined by immunoblotting and

quantified by densitometry, using β-actin as a loading control. B. Percentages of viable

Daudi cells that excluded 7AAD were determined by flow cytometry after 48 h. Despite

strong activation of GR, DEX-treated PPARαhi cells were resistant to DEX. C. CLL cells

were cultured in the presence or absence of DEX with or without the indicated

concentrations of the PPARα antagonists MK886, GW6471, or Compound A and the

fatty acid oxidation inhibitor CVT-4325 (GS449794). After 48 h, specific death was

determined by the difference of the percentages of viable 7AAD- cells in control and

treated cultures as measured by flow cytometry. PPARα and fatty acid oxidation

inhibitors significantly increased specific death of DEX-treated CLL cells. Averages and

standard errors of the results for each inhibitor are shown. D. NOD-SCIDγcnull mice

engrafted 6 weeks earlier with CLL splenocytes were treated with 4 consecutive

injections of MK886 (10 mg/kg), DEX (4.6 mg/kg) or both MK886 and DEX. Four days

after the last injection, human CD5+CD19+ CLL cells were measured in spleen cell

suspensions by flow cytometry. **, p<0.01; *, p<0.05. In vivo CLL xenograft data

courtesy of Karrie Wong.

Page 50: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

42

42

Page 51: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

43

43

Supplementary Figure 7. A. Summary of MitoTracker Green MFI measurements of

control and DEX-treated CLL cells performed at 4 and 18 h, indicating no significant

difference in mitochondrial mass between DEX-treated and control samples. B. Viability

of DEX-treated CLL cells used for intracellular metabolite measurements by NMR

spectroscopy. Viability is reported as percentage of 7AAD- cells measured by flow

cytometry. C. Pictures (11x) of OP9 cells taken after 5 days in differentiation media.

Adipocytes are characterized by the presence of large intracellular lipid droplets. D, E.

DEX-treated CLL cells were supplemented with various concentrations of L-glutamine

(D) or glucose (14mM) (E). The results show that these fuel substrates could not reverse

the cytotoxic effects of DEX.

Page 52: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

44

44

2.5 Discussion The mechanism of action of high-dose GCs in CLL is not clear. GCs alter

signaling processes important for tumor growth by increasing the expression of

regulatory molecules such as I-κB and phosphatases (Sapolsky et al., 2000; Flammer et

al., 2011; Glass et al., 2010) or directly altering the expression of apoptotic genes (Kfir-

Erenfeld et al., 2010). It is widely known that many of the complications of GC therapy,

such as diabetes and hyperlipidemia, are metabolic in nature and that a major function of

endogenous GCs is to turn off glucose usage while at the same time to turn on fatty acid

oxidation to allow cells to survive an overnight fast (Sapolsky et al., 2000). Despite these

well-known facts, the possibility that the therapeutic activity of GCs in CLL is due to

metabolic effects is not widely appreciated.

The findings in this paper suggest: 1. GCs alter the expression of metabolic genes

(Fig.2C, 4) and restrict metabolism in CLL cells (Fig.2). 2. GCs may prevent tumor cells

from being able to activate the bioenergetic programs needed to respond to membrane

damage (Fig.3). 3. GCs increase the dependence of CLL cells on fatty acid oxidation by

altering their expression of PPARα and PDK4 (Fig.4). Use of fatty acids to support

metabolism can alleviate restrictions imposed by GCs and thereby constitutes a resistance

mechanism to GC-mediated cytotoxicity (Fig.5). 5. PPARα inhibitors may offer a

strategy to improve the therapeutic efficacy of GCs in CLL (Fig.6).

It has been shown that GCs cause leukemia cells to undergo autophagy in some

conditions, which is a response to energy deprivation (Grander et al., 2009). However,

we could not find evidence for autophagy, such as lipidation of LC3, in the serum-free

conditions employed here (not shown). Previous studies have also shown that GCs inhibit

glycolysis in acute leukemia cells by restricting glucose uptake and consumption

(Buentke et al., 2011). However, the effect of GCs on pyruvate kinase expression (Fig.2)

have not been previously reported and may possibly be unique to CLL cells. Regardless,

the concept that GCs restrict the metabolic activity of leukemia cells is appealing in that

it helps explain why GCs can enhance the therapeutic effects of cytotoxic drugs in

combination chemotherapy regimens.

The mechanism(s) whereby GCs down-regulate pyruvate kinase and increase

PPARα expression are not clear. The effects appear to be due to transcriptional

Page 53: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

45

45

regulation by GCs because they can be prevented by Mifepristone (Fig.1,5). GCs may act

directly at the promoters of metabolic genes to positively or negatively regulate their

expression and may also regulate the expression of microRNAs (miRs) that control

metabolic gene expression (Harada et al., 2012).

Fatty acid oxidation has been recently described as a mechanism used by cancer

cells to generate energy and survive under conditions of metabolic stress (Spaner et al.,

2012; Samudio et al., 2010; Schafer et al., 2009; Zaugg et al., 2011; Carracedo et al.,

2012). The results reported here are in keeping with these observations and suggest that

PPARα-mediated fatty acid oxidation also allows CLL cells to survive the metabolic

stress imposed by GCs. Concomitant administration of GCs and PPARα-antagonists such

as MK886, which has been used before in humans (Spaner et al., 2012), appears feasible

(Fig.6) and constitutes a novel strategy to improve the therapeutic efficacy of GC therapy

in high-risk CLL patients.

Page 54: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

46

46

CHAPTER 3:

A role for Danazol in Chronic Lymphocytic Leukemia

Stephanie Tung1,2, David E. Spaner1,2,3,4

1Division of Molecular and Cellular Biology, Sunnybrook Research Institute, Toronto, Canada, M4N 3M5; 2Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada, M5G 2M9; 3Sunnybrook Odette Cancer Center, Toronto, Canada, M4N 3M5, 4Department of Medicine, University of Toronto, Toronto, Ontario, Canada, M5G 2C4.

Published in Leukemia. 2012. 26(7):1684-1686

© 2012 Nature Publishing Group, a division of Macmillan Publishers Limited

Page 55: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

47

47

3.1 Introduction The clinical management of chronic lymphocytic leukemia (CLL) patients with

fludarabine-resistant disease or tumor cells with 17p deletions remains problematic

(Gribben et al., 2011). High-dose glucocorticoids (HDGCs) are among the most effective

treatment approaches for such patients, as well as elderly patients who cannot tolerate

conventional chemotherapy. However, HDGCs are not without significant side effects

and are only palliative in nature (Castro et al., 2009). Identification of non-toxic agents

that could improve the therapeutic efficacy of HDGCs would help significantly in

managing these patients. We report a case that suggests the weak androgen Danazol

(Fontana et al., 2011) may be such an agent.

Page 56: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

48

48

3.2 Results Clinical activity and in vitro toxicity of Danazol against CLL

A 61 year old female with slowly progressive Rai Stage II CLL, diagnosed 6 years earlier

and characterized by the 11q deletion with high CD38 expression (44%), was being

considered for first-line chemotherapy on the basis of increased fatigue and lymphocyte

doubling time (LDT) (Fig.8A). The patient also had episodal angioedema from an

acquired C1 esterase inhibitor deficiency related to CLL (Cicardi et al., 2003). Prior to a

dental procedure, her allergist prescribed Danazol 300 mg PO BID for one week to raise

C1-inhibitor levels in order to prevent possible airway obstruction from oral angioedema

(Morvacallo et al., 2010). Coincidentally, she was seen in the CLL clinic a week later

where her white blood cell (WBC) count had dropped from 76 to 42 x 106 cells/ml

(Fig.8A) and she reported improvements in subjective symptoms. Another dental

procedure was scheduled for the following month and the patient consented to give blood

before and after the prophylactic course of Danazol. Remarkably, the WBC count, which

had increased in the time off Danazol, decreased significantly again in response to

Danazol, although it increased again in the following weeks (Fig.8A). Similar declines in

circulating WBC counts have been observed in 2 male patients treated with Danazol (400

mg PO BID) for 2 months (not shown).

This apparent clinical activity of Danazol prompted laboratory studies of its effects

on CLL cells. Danazol was found to be toxic to CLL cells above 3 µM (Fig.8B) although

it was generally less toxic than the synthetic glucocorticoid Dexamethasone (Dex)

(Fig.8C). Danazol killed both male and female CLL cells although the former appeared

somewhat more sensitive (Fig.8D).

Page 57: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

49

49

Page 58: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

50

50

Figure 8. Effect of Danazol on CLL cells in vivo and in vitro. A. WBC counts over 12

months for the patient described in the text. One week of Danazol caused a drop in the

WBC count which was confirmed by the response to a second treatment. B. CLL cells

obtained from consenting patients who were untreated for at least 3 months, and with

local REB approval, were purified by negative selection12 and cultured in serum-free

media in different concentrations of Danazol (Sigma, St. Louis, MO). Exclusion of

7AAD by flow cytometry was used to measure cell viability after 48 h. Danazol-specific

death is the difference in %7AAD+ cells between Danazol-treated cells and control cells.

Subsequent experiments used 10 µM of Danazol. C. Example of flow cytometric

analysis. Numbers in the dot-plots represent percentages of viable 7AAD- cells. D.

Danazol-specific death after 48 h for 6 female and 5 male patient samples. The P value

for the difference between sample means was calculated using a two sample t-test.

Page 59: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

51

51

Danazol and Glucocorticoids have enhanced efficacy in CLL

We considered that Danazol might have off-target effects on the glucocorticoid receptor

(GR) to explain its cytotoxic activity against CLL cells. Indeed, Danazol induced

phosphorylation of the GR (indicating activation of the receptor) in CLL cells although it

was weaker than Dex (Fig. 9A). However Danazol-mediated killing was not blocked by

the GR antagonist Mifepristone (Johanssen et al., 2007), in contrast to Dex (not shown).

Consistent with a GR-independent mechanism, Danazol had additive effects on Dex-

induced cytotoxicity (Fig.9B).

We then examined the effects of Danazol specifically on CLL cells with 17p

deletions (Fig.9C, bottom left panel). At a dose of 10 µM, Danazol remained capable of

killing such cells, especially in combination with Dex. Compared to CLL cells without

this cytotogenetic abnormality, 17p- tumor cells appeared less sensitive to Danazol, with

or without Dex, although the differences were not statistically significant (Fig.9C, right

panel). Note the results for Fig. 9C were obtained with cryopreserved cells and the higher

levels of spontaneous death following thawing account for the lower specific death levels

in comparison with Fig. 9B which were obtained with fresh tumor cells.

While Danazol and low doses of glucocorticoids could potentially compete for the

GR, the possibility of combining Danazol with HDGCs is supported by the example of an

85 year old female with symptomatic Rai Stage IV CLL and trisomy 12 tumor cells.

Fludarabine- and alkylator-based regimens were felt to be contraindicated because of the

patient’s congestive heart failure and she was treated with HDGCs (1500 mg Prednisone

PO x 5 days) but suffered side effects of fluid retention and profound fatigue. Based on

previous use of Danazol as a steroid-sparing agent in autoimmune disorders, the

prednisone dose was lowered to 1000 mg PO daily for 4 days and Danazol (200 mg po

BID) was added to the next 2 treatment cycles. The treatments were better tolerated and

the clinical response was maintained and possibly even enhanced (Fig.9D).

Page 60: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

52

52

Page 61: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

53

53

Figure 9. Effect of Danazol and Glucocorticoids on CLL cells in vitro and in vivo. A.

Purified CLL cells from 3 different patients were cultured alone, with Dexamethasone

(Dex) (Sigma) (30 µM), Danazol (Dan) (10 µM) or both. After 4 h, cell lysates were

collected (Tomic et al., 2011) and examined by immunoblotting with antibodies to the

phosphorylated glucorticoid receptor (pGR) (Cell Signaling, Beverley, MA), using β-

actin as a loading control. The results indicate that Danazol can weakly phosphorylate

and activate the GR in CLL cells. Similar results were seen with 4 other samples. B.

Summary of cell viability results, measured with 7AAD-staining and flow cytometry

after 48 h of culture, for 13 different patient samples. C. Cryopreserved cells from 11

additional patients, with (n=5) or without (n=6) 17p deletions determined by fluorescence

in situ hybridization (FISH), were thawed, rested overnight, and examined for cell

viability following treatment with Dex, Danazol, or both. Specific deaths for individual

patient samples are shown in the left panels and summarized in the right graph. P-values

for the differences between sample means were calculated using a single factor ANOVA

test. * p<.01; ** p<.02. D. Changes in the WBC count of the patient described in the text,

indicating that addition of Danazol to HDGCs can potentially enhance efficacy without

increased toxicity.

Page 62: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

54

54

3.3 Discussion These observations suggest that Danazol has clinical activity in CLL and can be used

safely with HDGCs. Danazol has been prescribed for almost 40 years (Fontana et al.,

2011) and has a well-known toxicity profile. It causes virilization in young women with

endometriosis, which is not necessarily a problem for older male or post-menopausal

female CLL patients. Danazol is otherwise relatively safe but patients must be monitored

for thrombosis and lipid and liver abnormalities (Shephed et al., 1995). Along with its use

in autoimmune disorders (Cicardi et al., 2003; Boruchoy et al., 2007), Danazol is thought

to have activity in myelodysplastic syndromes (Fontana et al., 2011) and can prevent

Interferon-induced thrombocytopenia (Alvarez et al., 2011). Over 2500 papers on

Danazol are listed in PubMED but, to our knowledge, its activity in CLL has not been

reported before.

The mechanism of action of Danazol in CLL is not clear. Blood levels of CD23

were lowered by Danazol in endometriosis (Matalliotakis et al., 2000), suggesting a direct

effect on activated B cells which are considered the normal counterparts of CLL cells

(Tomic et al., 2011). However, we could not detect an effect of Danazol on CD23

expression by CLL cells in vitro (not shown). Danazol has been reported to decrease the

production of inflammatory cytokines, including IL-6 and TNF-α, which may limit their

ability to cause proliferation of CLL cells in vivo (Tanaka et al., 2009). Danazol can also

intercalate into cell membranes (Horstman et al., 1995) which may dysrupt growth-

promoting signaling processes and account for a mechanism of action apparently

independent of glucocorticoid, estrogen, and androgen receptors (Fig.8D).

The observations reported in this letter suggest clinical trials to determine if the

anti-CLL effect of Danazol can be exploited to improve the clinical efficacy of HDGCs.

Conventional oral doses of Danazol have been reported to achieve erythrocyte membrane

levels that approach the µM levels that appear to be needed for cytotoxicity (Fig.8,9).

Initial clinical studies will focus on determining if optimal dosing of Danazol in CLL

patients can be achieved through the oral route (Horstman et al., 1995) or if intravenous

administration will be required.

Page 63: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

55

55

CHAPTER 4:

Discussion and Future Perspectives

Page 64: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

56

56

In order to improve GC treatment for CLL, a better understanding of its

mechanisms of action and resistance is required. The findings of this thesis demonstrate a

novel mechanism of GC action that is based on its metabolic-altering effects (Chapter 2).

GC cytotoxicity is mediated by changes to the metabolic programs of circulating CLL

cells, which allow the CLL cells to switch from glucose oxidation to fatty acid oxidation

in order to resist cytotoxicity. In order to explore new avenues to overcome GC

resistance, fatty acid oxidation inhibitors were studied in combination with GCs to assess

their efficacy against CLL cells in vitro and in vivo. In addition, a clinically relevant

agent, Danazol was also investigated for its clinical activity against CLL cells as a single

agent and in combination with GCs (Chapter 3). In this final chapter, I will discuss the

importance and relevance of these findings will be presented and outline the future

directions of research in this area will be discussed.

4.1 GC treatment for proliferating CLL cells The relationship between the metabolic and cytotoxic effects of GCs in lymphocytes still

remains an incomplete picture. The findings of this thesis suggest that the metabolic state

of CLL cells plays an important role in GC sensitivity. I have found that in circulating

CLL cells, GC treatment causes bioenergetic restriction that is responsible for mediating

cell death (Chapter 2). As previously mentioned (Chapter 1), CLL disease exists in two

compartments in the body: proliferation centers (PCs) in lymphoid organs and the

circulation. Generally, non-dividing circulating CLL cells can be effectively cleared by

conventional cytotoxic treatments. The clearance of these circulating cells results in a

drastic drop in the WBC count however this does not cure the disease.

Circulating CLL cells are known to originate from PCs in lymphoid organs. In PCs,

CLL cells are driven to proliferate by external signals provided by the microenvironment

(Herishanu et al., 2011). These signals can originate from T cells and stromal cells found

in PCs that can produce soluble factors like IL-2 (Tomic et al., 2006) and activating

ligands like fibroblast growth factors (Xia et al., 2012) respectively. Since CLL cells

from PCs are the source of CLL relapse following conventional treatment and GC

therapy, future studies would focus on understanding and targeting the metabolic impact

of GCs on proliferating CLL cells in order to improve GC treatment.

Page 65: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

57

57

The need to eliminate tumour cells in PCs requires novel treatments to be evaluated

in this compartment. However, due to the inaccessible nature of these proliferating cells

in vivo modeling PCs of CLL disease in vitro is required. In general, such models include

culturing circulating CLL cells with antigen mimetics, cytokines, costimulatory

molecules and other cells that are encountered in vivo in PCs (Cols et al., 2012; Ding et

al., 2010). Our group has shown that IL-2 along with a TLR-7/8 agonist, Resiquimod can

induce CLL cells to proliferate in vitro (Tomic et al., 2006). We have also developed a

stromal cell-line from a patient’s spleen that supports proliferation by secreting soluble

factors that activate signaling pathways like STAT3. Based on this information, culturing

CLL cells in conditioned media from this stromal cell-line along with IL-2 and

Resiquimod could function as an in vitro PC model.

Our findings show that GCs invariably induce atrophy in circulating CLL cells,

however the proliferating CD5+ Daudi Vector leukemic cell-line (Chapter 2) was largely

unaffected. The resistance of proliferating tumour cells may explain why GCs are mostly

palliative to high-risk CLL patients. Pro-survival and growth-stimulating signaling

pathways are turned on in proliferating CLL cells in vitro such as phosphoinositol-3-

kinase (PI3K), Akt/protein kinase B (PKB), extracellular signal-regulated kinases (ERK)

and nuclear factor-κB (NFκB) (Tomic et al., 2006). Activation of these signaling

pathways results in increased metabolic activity to sustain the induced proliferation. This

increased metabolic activity could be sufficient to resist the energy restriction mediated

by GC treatment and to maintain the TCA cycle. Congruently, GC resistance in B-acute

lymphoblastic leukemia (B-ALL) is associated with increased glycolysis (Holleman et

al., 2004; Hulleman et al., 2009) and treatment with the glycolysis inhibitor, 2-deoxy-D-

glucose (2-DG) conferred GC sensitivity (Boag et al., 2006).

Using the in vitro PCs model, the sensitivity of proliferating CLL cells to GC

treatment can be assessed. It is expected that the activating signals provided to the CLL

cells will render the cells resistant to GC treatment due to their enhanced metabolic

activity. Therefore, treating the proliferating CLL cells with glycolysis inhibitors (2-DG)

is anticipated to overcome GC resistance. Other promising targets for inhibition include

the signaling pathways that up-regulate glycolysis, including PI3K and Akt.

Page 66: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

58

58

4.2 Role of glutaminolysis in GC-mediated cytotoxicity Based on the findings in this thesis, I have established that bioenergetic restriction is a

major mechanism of GC-mediated cytotoxicity in circulating CLL cells (Chapter 2). I

have shown that the primary lesion involves the down-regulation of a crucial glycolytic

enzyme, PKM2. The down-regulation of PKM2 and in turn of glucose metabolism in the

mitochondria leads to a compensatory up-regulation of fatty acid oxidation gene

programs including, PPARα, PPARδ and PDK4. This thesis has demonstrated the key

role PPARα-mediated fatty acid oxidation plays in GC resistance and established the

major role metabolism plays in GC-mediated cytotoxicity in malignant lymphocytes.

Another major source of cellular energy is glutamine. Glutamine is the most

abundant amino acid in the human circulation and can contribute to the TCA cycle via

glutaminolysis (Newsholme et al., 1985). Glutaminolysis is a series of biochemical

processes that degrades glutamine to substrates that can support the TCA cycle.

Glutamine is converted to glutamate, which is then converted to α-ketoglutarate, an

important anaplerotic compound that maintains the TCA cycle in the mitochondria (Daye

et al., 2012).

Glutaminolysis occurs in proliferating cells like lymphocytes and tumour cells

(Newsholme et al., 1985). Most tumour cells rely on glutaminolysis in addition to

glycolysis to fulfill their high-energy requirements for growth (Maruzek and Eigenbrodt,

2003). Theoretically, the upregulation of glutaminolysis gene programs could allow

proliferating leukemic cells to utilize alternate fuels to overcome the GC-mediated energy

restriction. The contribution of glutaminolysis to proliferating CLL cells’ metabolism

and sensitivity to GC treatment can be assessed using the in vitro PCs model.

A key glutaminolysis enzyme, glutaminase catalyzes the conversion of glutamine to

glutamate. There are two isozymes of glutaminase: glutaminase (GLS), the liver isozyme,

and glutaminase 2 (GLS2), the mitochondrial isozyme. GCs are known to up-regulate

GLS expression in human lymphocytes and intestinal epithelial cells (Dudrick et al.,

1993; Sarantos et al., 1994). It has been shown inhibiting mitochondrial GLS activity

prevents oncogenic transformation (Wang et al., 2010). GC-regulation of the expression

of these two GLS isozymes can be determined by Q-PCR and Western blotting. There are

GLS inhibitors available such as 6-diazo-5-oxo-L-norleucine (DON) (Willis and

Page 67: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

59

59

Seegmiller, 1977), which can be tested to see if they can overcome GC resistance in

proliferating CLL cells.

4.3 Rationale for combinatorial GC treatment of CLL disease GCs are commonly combined with other therapeutic modalities like chemotherapy and

immunotherapy for the treatment of CLL disease. The efficacy of monoclonal antibodies

such as Rituximab, Ofatumumab and Alemtuzumab, is significantly increased when

combined with HDGCs (Bowen et al., 2007; Spaner et al., 2011; Pettitt et al., 2012). The

work in this thesis shows that circulating CLL cells are sensitive to GC treatment in the

presence of membrane-damaging agents (Chapter 2).

GC treatment causes bioenergetic restriction, which prevents cells from repairing

the membrane damage caused by processes such as complement-mediated cytotoxicity.

This observation provides an explanation for the enhanced efficacy of both chemotherapy

and immunotherapy treatment modalities that have been successfully combined with GCs

for the treatment of leukemias. Also, considering that the GC mechanism of action is

metabolic in nature and there does not appear to be any correlation between cytogenetic

abnormalities and GC efficacy, this suggests that GC treatment is a valid option for all

CLL patients regardless of cytogenetic status.

4.4 Mechanism of GC-mediated down-regulation of PKM2 Following my findings that GC treatment causes bioenergetic restriction in circulating

CLL cells, I also determined that GC treatment resulted in a rapid down-regulation of the

enzyme, pyruvate kinase muscle isozyme 2 (PKM2) (Chapter 2). PKM2 is a kinase that is

responsible for transferring a phosphate group from phosphoenolpyruvate (PEP) to

adenosine diphosphate (ADP) to generate pyruvate and adenosine triphosphate (ATP)

(Mazurek, 2011). Pyruvate feeds into the tricarboxylic acid (TCA) cycle in the

mitochondria and goes on to generate acetyl coenzyme A. The TCA cycle is responsible

for the generation of ATP, which is the major source of cellular energy (DeBerandinis et

al., 2008). Given that the mRNA and protein expression level of an enzyme may not

necessarily reflect its enzymatic activity, it would be important to directly measure

Page 68: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

60

60

PKM2 activity in response to GC treatment. Measuring PKM2 enzymatic activity can be

achieved using an enzymatic cycling microplate-based system developed for the

quantitative determination of low levels of mammalian metabolic enzymes (Janke et al.,

2010).

Another important aspect for consideration is the mechanism of GC-mediated

down-regulation of PKM2. Down-regulation of PKM2 mRNA transcript was detected

early following GC administration, thereby implicating a direct effect. GCs can

differentially regulate the expression of metabolic genes, like PKM2 by directly

interacting with the promoters of target genes. GCs are also known to regulate the

expression of microRNAs (miRs), which can inhibit the expression of a variety of genes

(Harada et al., 2012; Smith et al., 2010). miR-mediated repression of target gene

expression is a quick process, which corresponds with the early event of GC-mediated

PKM2 down-regulation. Future studies will investigate the involvement of miRs with the

GC-mediated down-regulation of PKM2. Additionally, a potential mechanism of action

for proliferating CLL cells’ GC resistance (Section 4.1) could be the prevention of PKM2

down-regulation, which will also be investigated in future studies.

4.5 Involvement of miR-17 in GC-induced cell death miRs are a class of small, non-coding RNAs around 22 nucleotides in length that

negatively regulate gene expression by either inducing degradation or translational

inhibition of target mRNAs (Babashah and Soleimani, 2011). miRs are initially

transcribed and processed in the nucleus before being exported into the cytoplasm where

they undergo further processing to generate mature miRs (Figure 13). Mature miRs

recognize and target the 3’-untranslated region (3’-UTR) of specific mRNAs that contain

a complementary target site. Depending on the degree of complementarity between the

mRNA target sites and the nucleotide sequence from position 2-8 at the 5’ end of the

miRs (the seed region), there are two potential mechanisms of regulating target gene

expression: Ago-catalyzed cleavage of target mRNA when the miR has perfect or near

perfect complementarity or repression of translation when the miR has imperfect

complementarity to the target mRNA (Hutvagner and Zamore, 2002).

Page 69: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

61

61

The aberrant expression of miRs has been found in various types of solid tumours

and leukemias (Calin and Croce, 2006; Babashah et al., 2012). The first evidence of the

involvement of miRs in hematological malignancies was actually described in CLL. The

miR-15a/miR-16-1 cluster resides on chromosome 13q14.3, a genomic region frequently

lost or down-regulated in CLL, which was found to be associated with the indolent form

of disease (Calin et al., 2002). Interestingly, some of the direct targets of the miR-

15a/miR-16-1 cluster are mRNAs encoding gene products that are involved in regulating

cell proliferation and apoptosis such as cyclins (CCND1 and CCND3), cyclin-dependent

kinases (CDK6) and B-cell lymphoma-2 (Bcl-2) (Cimmino et al., 2005; Klein and Dalla-

Favera, 2010). Other miRs that are known to be involved in CLL disease include miR-

155, miR-34a, miR29b, and miR-181b (Dijkstra et al., 2009; Fulci et al., 2007; Pekarsky

et al., 2006).

miRs are involved in controlling several cellular processes altered in cancer, such

as proliferation, differentiation and apoptosis (Babashah et al., 2012). Considering the

ability of GCs to induce apoptosis in lymphocytes (Dougherty et al., 1943; Pearson et al.,

1949), recent studies have started investigating the relationship between GCs and miRs

particularly with regards to leukemia. It has been shown that several miRs were regulated

by GCs such as the myeloid-specific miR-223 and the apoptosis and cell cycle arrest-

inducing miR-15a/miR-16-1 cluster in ALL cell lines (Rainer et al., 2009).

Other groups have also shown that miRs are severely repressed and the expression

of miR processing enzymes are significantly reduced during GC-induced apoptosis

(Harada et al., 2012; Smith et al., 2010). Specifically, the conserved miR-17-92 cluster

was found to be a prime target of GC-induced repression, which mediated subsequent cell

death in ALL cell lines (Harada et al., 2012; Smith et al., 2010). Inhibition of the level of

miR-17 expression was shown to increase GC sensitivity and overexpression of miR-17

was found to induce GC resistance (Harada et al., 2012). The target of miR-17 was

determined to be the pro-apoptotic protein, Bim. These findings suggest a model of GC-

induced cell death that is mediated by the sequential down-regulation of miR-17 and up-

regulation of Bim.

No specific miR response has been determined in GC-treated CLL cells.

However, based upon the quick down-regulation of PKM2 in response to GC treatment

Page 70: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

62

62

(Chapter 2), it is possible that GCs could up-regulate the expression of miR-17 in CLL

cells. Future studies would focus on determining the effects of GCs on miR-17

expression in CLL cells. The potential miR-17 binding site on the PKM2 mRNA

transcript needs to be identified and verified using a luciferase reporter assay. The CD5+

Daudi cell line would also need to be engineered to overexpress miR-17 in order to assess

the effects of miR-17 on PKM2 expression and GC-induced cell death. In addition, there

are commercially available miR inhibitors (Qiagen miScript miRNA inhibitors) that can

be used to inhibit miR-17 expression in primary CLL cells to verify its effects PKM2 and

GC-induced cell death. To assess the in vivo effects of miR-17 on CLL cells, there are

miR mimics available from Qiagen that can be injected into the CLL mouse model used

in these studies.

4.6 Concluding remarks Based on the findings presented in this thesis, emphasis needs to be placed on exploring

metabolic inhibitors as a valid option for enhancing the effects of GC therapy in CLL.

These metabolic inhibitors would allow for more efficient bioenergetic restriction by

preventing tumour cells from harnessing energy from alternative sources, which would

prevent GC resistance and enhance GC efficacy. More specifically, fatty acid oxidation is

emerging as a common metabolic strategy of resistance in tumour cells. In the case of

CLL, PPARα has emerged as a mediator of resistance against metabolic and cytotoxic

stresses (Spaner et al., 2012) and therefore a promising therapeutic target especially with

regards to GC treatment (Chapter 2).

Page 71: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

63

63

LIST OF REFERENCES Adcock IM. Glucocorticoid-regulated transcription factors. Pulm Pharmacol Ther. 2001;14:211-219. Adcock IM, Nasuhara Y, Stevens DA, et al. Ligand-induced differentiation of glucocorticoid receptor (GR) transrepression and transactivation: preferential targeting NF-κB and lack of IκB involvement. Br J Pharmacol. 1999;127:1003:1011. Agarwai MK. The antiglucocorticoid action of Mifepristone. Pharmacol.Ther. 1996;70:183-213.

Ahn YS, Harrington WJ, Simon SR, et al. Danazol for the treatment of idiopathic thyombocytopenic purpura. N Engl J Med. 1983;308:1396-1399. Ahn YS, Harrington WJ, Mylvaganam R, et al. Danazol therapy for autoimmune hemolytic anemia. Ann Intern Med. 1985;102:298-301. Alvarez G, Gómez-Galicia D, Rodríguez-Fragoso L, et al. Danazol improves thrombocytopenia in HCV patients treated with peginterferon and ribavirin. Ann Hepatol 2011; 10: 458-468. Auphan N, DiDonato JA, Rosette C, et al. Immunosuppression by glucocorticoids:inhibition of NF-κB activity through induction of IκB synthesis. Science. 1995;270:286:290. Babashah S and Soleimani M. The oncogenic and tumour suppressive roles of microRNAs in cancer and apoptosis. Eur J Cancer. 2011;47:1127-1137.

Babashah S, Sadeghizadeh M, Rezaei M, et al. Aberrant microRNA expression and its implications in the pathogenesis of leukemias. Cell Oncol. 2012;35:317-334.

Bamberger CM, Bamberger AM, de Castro M, et al. Glucocorticoid receptor beta, a potential endogenous inhibitor of glucocorticoid action in humans. J Clin Invest. 1995;95:2435:2441. Banerji V and Gibson SB. Targeting metabolism and autophagy in the context of haematologic malignancies. Int J Cell Biol. 2012;2012:595976. Barbieri RL. Danazol inhibition of steroidgenesis in the human corpus luteum. Obstet Gynecol. 1981;57:722-724. Binet JL, Auquier A, Dighiero G, et al. A new prognostic classification of chronic lymphocytic leukemia derived from a multivariate survival analysis. Cancer. 1981;48:198-206.

Page 72: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

64

64

Beesley AH, Firth MJ, Ford J, et al. Glucocorticoid resistance in T-lineage acute lymphoblastic leukaemia is associated with a proliferative metabolism. Br J Cancer. 2009;100:1926-1936. Belfroy DE, Petersen KF, Dulfour S, et al. Increase substrate oxidation and mitochondrial uncoupling in skeletal muscle of endurance-trained individuals. Proc Natl Acad Sci U S A. 2008;105:16701-16706. Beum PV, Lindorfer MA, Beurskens F, et al. Complement activation of B lymphocytes opsonized with rituximab or ofatumumab produces substantial changes in membrane structure preceding cell lysis. J Immunol. 2008;181:822-832. Boag JM, Beesley AH, Firth MJ, et al. Altered glucose metabolism in childhood pre-B acute lymphoblastic leukaemia. Leukemia. 2006;20:1731-1737. Boruchov DM, Gururangan S, Driscoll MC, Bussel JB. Multiagent induction and maintenance therapy for patients with refractory ITP. Blood 2007; 110: 3526- 3531. Bosanquet AG, McCann SR, Crotty GM, et al. Methylprednisolone in advanced chronic lymphocytic leukaemia: rationale for, and effectiveness of treatment suggested by DiSC assay. Acta Haematol. 1995;93:73-79. Bowen DA, Call TG, Jenkins GD, et al. Methylprednisolone-rituximab is an effective salvage therapy for patients with relapsed chronic lymphocytic leukemia including those with unfavorable cytogenetic features. Leuk Lymphoma. 2007;48:2412-2417. Bremer J. Carnitine—metabolism and functions. Phyiol Rev. 1983;63:1420-1480. Brunengraber H, Roe CR. Anaplerotic molecules: current and future. J.Inherit.Metab Dis. 2006;29:327-331.

Buentke E, Nordstrom A, Lin H, et al. Glucocorticoid-induced cell death is mediated through reduced glucose metabolism in lymphoid leukemia cells. Blood Cancer J. 2011;1:e31. Buttergeit F, Burmester GR and Brand MD. Therapeutically targeting lymphocyte energy metabolism by high-dose glucocorticoids. Biochem Pharmacol. 2000;59:597-603. Calin GA and Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006;6:857-866.

Page 73: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

65

65

Calin GA, Dumitru CD, Shimizu M, et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A. 2002;99:15524-15529.

Carracedo A, Weiss D, Leliaert AK et al. A metabolic prosurvival role for PML in breast cancer. J.Clin.Invest. 2012;122:3088-3100. Castro JE, Barajas-Gamboa JS, Melo-Cardenas J, et al. Ofatumumab and high-dose methylprednisolone is an effective salvage treatment for heavily pretreated, unfit or refratory patients with chronic lymphocytic leukemia: single institution experience. ASH Annual Meeting Abstracts. 2010;116:4638. Castro JE, James DF, Sandoval-Sus JD, Jain S, Bole J, Rassenti L, Kipps TJ. Rituximab in combination with high-dose methylprednisolone for the treatment of CLL. Leukemia 2009; 23:1779-1789. Castro JE, Sandoval-Sus JD, Bole J, et al. Rituximab I combination with high-dose methylprednisolone for the treatment of fludarabine refractory high-risk chronic lymphocytic leukemia. Leukemia. 2008;22:2048-2053. Catovsky D, Fooks J and Richards S. The UK Medical Research Council CLL trials 1 and 2. Nouv Rev Fr Hematol. 1988;30:423-427. Cheung J and Smith DF. Molecular chaperone interactions with steroid receptors : An update. Mol Endocrinol. 2000;14 :936:946. Chiorazzi N, Rai KR and Ferrarini M. Chronic lymphocytic leukemia. N Engl J Med. 2005;352:804-815.

Cimmino A, Calin GA, Fabbri M, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad U S A. 2005;102:13944-13949.

Cicardi M, Zingale LC, Pappalardo E, Folcioni A, Agostoni A. Autoantibodies and lymphoproliferative diseases in acquired C1-inhibitor deficiencies. Medicine (Baltimore) 2003; 82: 274-281. Cols M, Barra CM, He B, et al. Stromal endothelial cells establish a bidirectional crosstalk with CLL cells through the TNF-related factors BAFF, APRIL, and CD40L. J Immunol. 2012;188:6071-6083.

Daye D and Wellen KE. Metabolic reprogramming in cancer: unraveling the role of glutamine in tumorigenesis. Semin Cell dev Biol. 2012;23:362-369. de Viron E, Michaux L, Put N, et al. Present status and perspectives in functional analysis of p53 in chronic lymphocytic leukemia. Leuk Lymphoma. 2012;53:1445-1451.

Page 74: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

66

66

DeBerandinis RJ, Lum JL, Hatzivassilou G, et al. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 2008;7:11-20.

Dijkstra MK, van Lom K, Tielemans D, et al. 17p13/TP53 deletion in B-CLL patients is associated with microRNA-34a downregulation. Leukemia. 2009;23:625-627.

Ding W, Knox TR, Tschumper RC, et al. Platelet-derived growth factor (PDGF)-PDGF receptor interaction activates bone marrow-derived mesenchymal stromal cells derived from CLL: implications for an angiogenic switch. Blood. 2010;116:2984- 2993.

Dmowski WP, Scholer HF, Mahesh VB, et al. Danazol—a synthetic steroid derivative with interesting physiologic properties. Fertil Steril. 1971;22:9-18. Döhner H, Stilgenbauer S, Benner A, et al. Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med. 2000;343:1910-1916. Dougherty TF and White A. Influence of adrenal cortisol secretion on blood elements. Science. 1943;98:367-369. Du J, Yang H, Guo Y, et al. Structure of the Fab fragment of therapeutic antibody Ofatumumab provides insights into the recognition mechanism with CD20. Mol Immunol. 2009;46:2419-2423. Dudrick PS, Sarantos P, Ockert K, et al. Dexamethasone stimulation of glutaminase expression in mesenteric lymph nodes. Am J Surg. 1993;165:34-39.

Dungarwalla M, Evans SO, Riley U, et al. High dose methylprednisolone and rituximab is an effective therapy in advanced refractory chronic lymphocytic leukemia resistant to fludarabine therapy. Haematologica. 2008;93:475-476. Eaton S, Bartlett K and Pourfarzam M. Mammalian mitochondrial β-oxidation. Biochem J. 1996;320:345-357. Eberhart K, Rainer J, Bindreither D, et al. Glucocorticoid-induced alterations in mitochondrial membrane properties and respiration in childhood acute lymphoblastic leukemia. Biochim Biophys Acta. 2011;1807:719-725. el Rouby S, Thomas A, Costin D, et al. p53 Gene mutation in B-cell chronic lymphocytic leukemia is associated with drug resistance and is independent of MDR1/MDR3 gene expression. Blood. 1993;82:3452-3259. Elbi C, Walker DA, Romero G et al. Molecular chaperones function as steroid receptor nuclear mobility factors. Proc Natl Acad of Sci U.S.A. 2004;94:6803-6808.

Page 75: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

67

67

Elzein E, Ibrahim P, Koltun DO et al. CVT-4325: a potent fatty acid oxidation inhibitor with favorable oral bioavailability. Bioorg.Med.Chem.Lett. 2004;14:6017-6021.

Faderi S, Ferrajoli A, Frankfurt O and Pettitt A. Treatment of B-cell chronic lymphocytic leukemia with nonchemotherapeutic agents: experience with single-agent and combination therapy. Leukemia. 2009;23:457-466. Flammer JR, Rogatsky I. Minireview: Glucocorticoids in autoimmunity: unexpected targets and mechanisms. Mol.Endocrinol. 2011;25:1075-1086.

Fontana V, Dudkiewicz P, Ahn ER, Horstman L, Ahn YS. Danazol therapy combined with intermittent application of chemotherapy induces lasting remission in myeloproliferative disorder (MPD). Hematology 2011;16: 90-94. Frankfurt O and Rosen ST. Mechanisms of glucocorticoid-induced apoptosis in hematologic malignancies: updates. Curr Opin Oncol. 2004;16:553-563. Freedman LP and Luisi BF. On the mechanism of DNA binding by nuclear hormone receptor: A structural and functional perspective. J Cell Biochem. 1993;51:140- 150. Freedman ND and Yamamoto KR. Importin 7 and importin alpha/importin beta are nuclear import receptor for the glucocorticoid receptor. Mol Biol Cell. 2004;15:2276- 2286. Friday E, Ledet J and Turturro F. Response to dexamethasone is glucose-sensitive in multiple myeloma cell lines. J Exp Clin Cancer Res. 2011;30:81. Friedlander RL. The treatment of endometriosis with Danazol. J Reprod Med. 1973;10:197-199.

Fulci V, Chiaretti S, Goldoni M, et al. Quantitative technologies establish a novel microRNA profile of chronic lymphocytic leukemia. Blood. 2007;109:4944-4951.

Gametchu B, Watson CS, Wu S. Use of receptor antibodies to demonstrate membrane glucocorticoid receptor in cells from human leukemic patients. FASEB J. 1993;7:1283-1292. Garcia-Gonzalo FR, Izpisua Belmonte JC. Albumin-associated lipids regulate human embryonic stem cell self-renewal. PLoS.One. 2008;3:e1384.

Garfinkel AG, Nilsson-ehle P and Schotz MC. Regulation of lipoprotein lipase. Induction by insulin. Biochim Biophys Acta. 1976;424:264-273.

Page 76: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

68

68

Giguere V, Hollenberg SM, Rosenfeld GM, et al. Functional domains of the human glucocorticoid receptor. Cell. 1986;46:645-652. Ginaldi L, De Martinis M, Matutes E, et al. Levels of expression of CD19 and CD20 in chronic B cell leukemias. J Clin Pathol. 1998;51:364-369. Glass CK, Saijo K. Nuclear receptor transrepression pathways that regulate inflammation in macrophages and T cells. Nat Rev Immunol. 2010;10:365-376. Glatz JF, Luiken JJ, Bonen A. Membrane fatty acid transporters as regulators of lipid metabolism: implications for metabolic disease. Physiol Rev. 2010;90:367-417.

Grander D, Kharaziha P, Laane E, Pokrovskaja K, Panaretakis T. Autophagy as the main means of cytotoxicity by glucocorticoids in hematological malignancies. Autophagy. 2009;5:1198-1200.

Gribben JG and O’Brien S. Update on therapy of chronic lymphocytic leukemia. J Clin Oncol. 2011;29:544-550. Haarman EG, Kaspers GJ and Veerman AJ. Glucocorticoid resistance in childhood leukaemia: mechanisms and modulation. Br J Haematol. 2003;120:919:929. Hallek M, Cheson BD, Catovsky D, et al. Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines. Blood. 2008;111:5446-5456. Hallek M, Fischer K, Fingerle-Rowson G, et al. Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukemia: A randomized, open-label, phase 3 trial. Lancet. 2010;376:1164-1174. Hamblin TJ. Autoimmune complications of chronic lymphocytic leukemia. Semin Oncol. 2006;33:230-239. Harada M, Pokrovskaja-Tamm K, Soderhall S, et al. Involvement of miR17 pathway in glucocorticoid-induced cell death in pediatric acute lymphoblastic leukemia. Leuk Lymphoma. 2012; [Epub ahead of print]. Harmon GS, Lam MT, Glass CK. PPARs and lipid ligands in inflammation and metabolism. Chem.Rev. 2011;111:6321-6340.

Harper ME, et al. Characterization of novel metabolic strategy used by drug-resistant tumor cells. FASEB J. 2002;16:1550-1557.

Hench P, Kendall E, Slocumb C, Polley H. Adrenocortical Hormone in Arthritis : Preliminary Report. Ann.Rheum.Dis. 1949;8:97-104.

Page 77: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

69

69

Herishanu Y, Perez-Galan P, Liu D, et al. The lymph node microenvironment promotes B-cell receptor signaling, NF-kappaB activation, and tumor proliferation in CLL. Blood. 2011;117:563-574.

Hillmen P. Using the biology of chronic lymphocytic leukemia to choose treatment. Hematology Am Soc Hematol Educ Program. 2011;2011:104-109. Hirsch HA, Iliopoulos D, Joshi A, et al. A transcription signature and common gene networks link cancer with lipid metabolism and diverse human disease. Cancer Cell. 2010;17:348-361. Holleman A, Cheok MH, den Boer ML, et al. Gene expression patterns in drug-resistant acute lymphoblastic leukemia cells and response to treatment. N Engl J Med. 2004;351:533-542. Hollenberg SM, Evans RM. Multiple and cooperative trans-activation domains of the human glucocorticoid receptor. Cell. 1988.55:899-906. Hollenberg SM, Weinberger C, Ong ES, et al. Primary structure and expression of a functional human glucocorticoid receptor cDNA. Nature. 1985;318:635-641. Holzer RG, Park EJ, Li N et al. Saturated fatty acids induce c-Src clustering within membrane subdomains, leading to JNK activation. Cell. 2011;147:173-184.

Horstman L, Jy W, Arce M, Ahn YS. Danazol distribution in plasma and cell membranes as related to altered cell properties: implications for mechanism. Am J Hematol 1995; 50: 179–187. Hulleman E, Kazemier KM, Holleman A, et al. Inhibition of glucolysis modulates prednisolone resistance in acute lymphoblastic leukemia cells. Blood. 2009;113:2014- 2021. Hutvagner G and Zamore PD. A microRNA in multiple-turnover RNA I enzyme complex. Science. 2002;297:2056-2060.

Janke R, Genzel Y, Wahl A, et al. Measurement of key metabolic enzyme activities in mammalian cells using rapid and sensitive microplate-based assays. Biotechnol Bioeng. 2010;107:566-581. Johanssen S, Allolio B. Mifepristone (RU 486) in Cushing's syndrome. Eur J Endocrinol 2007; 157: 561-569. Johnston JB, Daeninck P, Verburg L, et al. P53, MDM-2, BAX and BCL-2 and drug resistance in chronic lymphocytic leukemia. Leuk Lymphoma. 1997;26:435-449.

Page 78: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

70

70

Kalant K and Cianflone K. Regulation of fatty acid transport. Curr Opin Lipidol. 2004;15:309-314. Keating MJ, O’Brien S, Albitar M, et al. Early results of a chemoimmunotherapy regimen of fludarabine, cyclophosphamide, and rituximab as initial therapy for chronic lymphocytic leukemia. J Clin Oncol. 2005;23:4079:4088. Keating MJ, O’Brien S, Lerner S, et al. Long-term follow up of patients with chronic lymphocytic leukemia (CLL) receiving fludarabine regimens as initial therapy. Blood. 1998;92:1165-1171 Kfir-Erenfeld S, Sionov RV, Spokoini R, Cohen O, Yefenof E. Protein kinase networks regulating glucocorticoid-induced apoptosis of hematopoietic cancer cells: fundamental aspects and practical considerations. Leuk.Lymphoma 2010;51:1968-2005. Klein U and Dalla-Favera R. New insights into the pathogenesis of chronic lymphocytic leukemia. Semin Cancer Bio. 2010;20:377-383.

Klein U, Lia M, Crespo M, et al. The DLEU/miR-15a/16-1 cluster controls B cell proliferation and its deletion leads to CLL. Cancer Cell. 2010;17:28-40 Krauss S, Brand MD, Buttgereit F. Signaling takes a breath--new quantitative perspectives on bioenergetics and signal transduction. Immunity. 2001;15:497- 502.

Landgren O, Rapkin JS, Caporaso NE, et al. Respiratory tract infections and subsequent risk of CLL. Blood. 2007;109:2198-2201 Liden J, Delaunay F, Rafter I, et al. A new function for the C-terminal zinc finfer of the glucocorticoid receptor: repression of RelA transactivation J Biol Chem. 1997;272:21467-21472. Lin X, Adams SH, Odle J. Acetate represents a major product of heptanoate and octanoate beta-oxidation in hepatocytes isolated from neonatal piglets. Biochem.J. 1996;318:235-240.

Linder C, Chernick SS, Fleck TR, et al. Lipoprotein lipase and uptake of chylomicron triglyceride by skeletal muscle of rats. Am J Physiol. 1976;231:860-864. Lynen F Die Rolle der Phosphosaeure bei Dehydrierungsvorgaengen und ihre biologische Bedeutung. Naturwissenschaften. 1951;30:398-406. Macfarlane DP, Forbes S and Walker BR. Glucocorticoids and fatty acid metabolism in humans: fueling fat redistribution in the metabolic syndrome. J Endocrinol. 2008;197:189-204.

Page 79: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

71

71

Matalliotakis I, Neonaki M, Koumantaki Y, Goumenou A, Kyriakou D, Koumantakis E. A randomized comparison of danazol and leuprolide acetate suppression of serum-soluble CD23 levels in endometriosis. Obstet Gynecol 2000; 95: 810-813. Mazurek S. Pyruvate kinase type M2: a key regulator of the metabolic budget system in tumor cells. Int J Biochem Cell Biol. 2011;43:969-980.

Mazurek S and Eigenbrodt E. The tumor metabolome. Anticancer Res. 2003;23:1149- 1154.

McGarry JD and Brown NF. The mitochondrial carnitine palmitoyl transferase system. From concept to molecular analysis. Eur J Biochem. 1997;244:1-14. McKay LI and Cidlowski JA. CBP (CREB binding protein) integrates NF-kappaB (nuclear factor-kappaB) and glucocorticoid receptor physical interactions and antagonism. Mol Endocrinol. 2000;14:1222-1234. Melarangi T, Zhuang J, Lin K, et al. Glucocorticoid resistance in chronic lymphocytic leukaemia is associated with a failure of upregulated Bim/Bcl-2 complexes to activate Bax and Bak. Cell Death Dis. 2012;3:e372. Moalli PA, Pillay S, Krett NL, et al. Alternatively spliced glucocorticoid receptor messenger RNAs in glucocorticoid-resistant human multiple myeloma cells. Cancer Res. 1993;95:3877-3879. Morcavallo PS, Leonida A, Rossi G, Mingardi M, Martini M, et al. Hereditary angioedema in oral surgery. J Oral Maxillofac Surg 2010; 68: 2307-2311. Newsholme EA, Carbtree B and Ardawi MS. Glutamine metabolism in lymphocytes: its biochemical, physiological and clinical importance. Q J Exp Physiol. 1985;70:473-489.

Nissen RM and Yamamoto KR. The glucocorticoid receptor inhibits NF-κB by interfering with serine-2-phosphorylation of the RNA polymerase II carboxy- terminal domain. Genes Dev. 2000;14:2314-2329. Nomura DK, Long JZ, Niessen S, et al. Monoacylglycerol lipase regulates a fatty acid network that promotes cancer pathogenesis. Cell. 2010;140:49-61. O’Brien S, Kantarjian H, Beran M, et al. Results of fludarabine and prednisone therapy in 264 patients with chronic lymphocytic leukemia with multivariate analysis- derived prognostic model for response to treatment. Blood. 1993;82:1695-1700.

Page 80: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

72

72

Odukoya OA, Bansal A, Wilson AP, et al. Serum-soluble CD23 in patients with endometriosis and the effect of treatment with Danazol and leuprolide acetate depot injection. Human Reprod. 1995;10:942-946. Pearson OH, Eliel LP, et al. Adrenocorticotropic hormone- and incortisone-induced regression of lymphoid tumors in man; a preliminary report. Cancer. 1949;2:943- 945. Pecquer C, Bui T, Gelly C, et al. Uncoupling protein-2 controls proliferation by promoting fatty acid oxidation and limiting glycolysis-derived pyruvate utilization. FASEB J. 2008;22:9-18. Pekarsky Y, Santanam U, Cimmino A, et al. Tc11 expression in chronic lymphocytic leukemia is regulated by miR-29 and miR-181. Cancer Res. 2006;66:11590- 11593.

Pettitt AR, Jackson R, Carruthers S, et al. Alemtuzumab in combination with methylprednisolone is a highly effective induction regimen for patients with chronic lymphocytic leukemia and deletion of TP53: final results of the national cancer research institute CLL206 trial. J Clin Oncol. 2012;30:1647-1655.

Pettitt AR, Matutes E and Oscier D. Alemtuzumab in combination with high-dose methylprednisolone is a logical, feasible and highly active therapeutic regimen in chronic lymphocytic leukaemia patients with p53 defects. Leukemia. 2006;201:441-445.

Picard D, Yamamoto KR. Two signals mediate hormone-dependent nuclear localization of the glucocorticoid receptor. EMBO J. 1987;6:3333-3340. Pond CM. Adipose tissue and the immune system. Prostaglandins Leukot.Essent.Fatty Acids. 2005;73:17-30.

Ponzoni M, Doglioni C and Caligaris-Cappio F. Chronic lympohcytic leukemia: the pathologists’s view of lymph node microenvironment. Semin Diagn Pathol. 2011;28:161-166.

Pratt WB and Toft, DO. Steroid receptor interactions with heat shock protein and immunophilin chaperones. Endocr Rev. 1997;18:306-360. Rai KR, Peterson BL, Appelbaum FR, et al. Fludarabine compared with chlorambucil as primary therapy for chronic lymphocytic leukemia. N Engl J Med. 2000;343:1750-1757. Rai KR, Sawitsky A, Cronkite EP, et al. Clinical staging of chronic lymphocytic leukemia. Blood. 1975;46:219-234.

Page 81: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

73

73

Rainer J, Ploner C, Jesacher S, et al. Glucocorticoid-regulated microRNAs and mitrons in acute lymphoblastic leukemia. Leukemia. 2009;23:746-752. Ralph SJ, Rodriguez-Enriquez S, Neuzil J, et al. Bioenergetic pathways in tumor mitochondria as targets for cancer therapy and the importance of the ROS-induced apoptotic trigger. Ray A and Prefontaine KE. Physical association and functional antagonism between the p65 subunit of transcription factor NF-kappa B and the glucocorticoid receptor. Proc Natl Acad Sci U.S.A. 1994;91:752:756. Rivers C, Levy A, Hancock J, et al. Insertion of an amno acid in the DNA-binding domain of the glucocorticoid receptor as a results of alternative splicing. J Clin Endocrinol and Metab. 1999;84:4283-4286. Rose AL, Smith BE, Maloney DG. Glucocorticoids and rituximab in vitro: synergistic direct antiproliferative and apoptotic effects. Blood. 2002;100:1765-1773. Rossmeisl M, Syrovy I, Baumruk F, et al. Decreased fatty acid synthesis due to mitochondrial uncoupling in adipose tissue. FASEB J. 2000;14:1793-17800. Sadek I, Zayed E, Hayne O, et al. Prolonged complete remission of myelodysplastic syndrome treated with Danazol, retinoic acid and low dose prednisone. Am J Hematol. 2000;64:306-310. Sakai DD, Helms S, Carlstedt-Duke J, et al. Hormone mediated repression: A negative glucocorticoid response element from the bovine prolactin gene. Genes Dev. 1988;2:1144-1154. Samudio I, Fiegl M, McQueen T, et al. The Warburg effect in leukemia-stroma cocultures is mediated by mitochondrial uncoupling associated with uncoupling protein 2 activation. Cancer Res. 2008;68:5198-5205. Samudio I, Harmancey R, Fiegl M et al. Pharmacologic inhibition of fatty acid oxidation sensitizes human leukemia cells to apoptosis induction. J.Clin.Invest. 2010;120:142-156. Sankari L, Masthan k, Babu A, et al. Apoptosis in cancer – an update. Asian Pac J Cancer Prev. 2012;13:4873-4878. Sapolsky RM, Romero LM, Munck AU. How do glucocorticoids influence stress responses? Integrating permissive, suppressive, stimulatory, and preparative actions. Endocr Rev. 2000;21:55-89.

Sarantos P, Abouhamze Z, Copeland EM, et al. Glucocorticoids regulate glutaminase gene expression in human intestinal epithelial cells. J Surg Res. 1994;57:227-231.

Page 82: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

74

74

Schafer ZT, Grassian AR, Song L et al. Antioxidant and oncogene rescue of metabolic defects caused by loss of matrix attachment. Nature. 2009;461:109-113.

Schnaffer C, Stilgenbauer S, Gudrun A, et al. Somatic ATM mutations indicate a pathogenic role of ATM in B-cell chronic lymphocytic leukemia. Blood. 1999;94:748-753. Scheinman RI, Gualberto A, Jewell CM, et al. Characterization of mechanisms involved in transrepression of NF-κB by activated glucocorticoid receptors. Mol Cell Biol. 1995;15:943:953. Schulze A, Downward J. Flicking the Warburg switch-tyrosine phosphorylation of pyruvate dehydrogenase kinase regulates mitochondrial activity in cancer cells. Mol.Cell 2011;44:846-848.

Schultze SM, Hemmings BA, Niessen M, Tschopp O. PI3K/AKT, MAPK and AMPK signaling: protein kinases in glucose homeostasis. Expert Rev Mol Med. 2010;14e1.

Shearer BG, Billin AN. The next generation of PPAR drugs: do we have the tools to find them? Biochim.Biophys.Acta. 2007;1771:1082-1093.

Shepherd J. Danazol and plasma lipoprotein metabolism. Int J Gynaecol Obstet 1995; 50 Suppl 1: S23-S26. Shipp LE, Lee JV, Yu CY, et al. Transcriptional regulation of human dual specificity protein phosphatase 1 (DUSP1) gene by glucocorticoids. PLoS One. 2010;5(10):e13753 Smith LK and Cidlowski JA. Glucocorticoid-induced apoptosis of healthy and malignant lymphocytes. Prog Brain Res. 2010;182:1-30. Smith LK, Shah, RR and Cidlowski, JA. Glucocorticoids modulate microRNA expression and processing during lymphocyte apoptosis. J Biol Chem. 2010;285:36698-36708. Spaner DE. Oral high-dose glucocorticoids and Ofatumumab in fludarabine-resistant Chronic Lymphocytic Leukemia. Leukemia. 2011;26:1144-1145.

Spaner DE, Lee E, Shi Y, et al. PPAR-alpha is a therapeutic target for chronic lymphocytic leukemia. Leukemia. 2012; [Epub ahead of print] Stocklin E, Wissler M, Gouilleux F, et al. Functional interactions between Stat5 and the glucocorticoid receptor. Nature. 1996;383:726-728. Spector AA. Fatty acid binding to plasma albumin. J Lipid Research. 1975;16:165-179.

Page 83: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

75

75

Tam CS, O’Brien S, Wierda W, et al. Long-term results of the fludarabine, cyclophosphamide, and rituximab regimen as initial therapy of chronic lymphocytic leukemia. Blood. 2008;112:975-980. Tamada M, Suematsu M, Saya H. Pyruvate kinase m2: multiple faces for conferring benefits on cancer cells. Clin.Cancer Res. 2012;18:5554-5561.

Tanaka T. Danazol regulates the functions of normal human endometrial stromal cell subpopulations by modifying endometrial cytokine networks. Int J Mol Med 2009; 23: 421-428. Tao Y, Williams-Skipp C, Scheinman RI. Mapping of glucocorticoid receptor DNA binding domain surfaces contributing to transrepression of NF-kappa B and induction of apoptosis. J Biol Chem. 2001:276:2329:2332. Teichman ML, Ho VQ, Balducci L, et al. Efficacy of ofatumumab and high-dose methylprednisolone for the treatment of relapsed or refractory chronic lymphocytic leukemia (CLL). ASH Annual Meeting Abstracts. 2011;118:4619. Theriault A, Boyd E, Harrap SB, et al. Regional chromosomal assignment of the human glucocorticoid receptor gene to 5q31. Hum Genet. 1989;83:289-291. Thornton PD, Hamblin M, Treleaven JG, et al. High dose methylprednisolone in refractory chronic lymphocytic leukaemia. Leuk Lymphoma. 1999;34:167-170. Thornton PD, Matutes E, Bonsanquet AG, et al. High dose methylprednisolone can induce remissions in CLL patients with p53 abnormalities. Ann Hematol. 2003;82:759-765. Tiefenthaler M, Amberger A, Bacher N, et al. Increased lactate production follows loss of mitochondrial membrane potential during apoptosis of human leukaemia cells. Br J Haematol. 2001;114:574-580. Tissing WJ, Meijerink JP. Den Boer ML, et al. Genetic variations in the glucocorticoid receptor gene are not related to glucocorticoid resistance in childhood actue lymphoblastic leukemia. Clin Cancer Res. 2005;11:6050-6056. Tomic J, Lichty B, Spaner DE. Aberrant interferon-signaling is associated with aggressive CLL. Blood 2011; 117: 2668-2680. Tomic J, White D, Shi Y, et al. Sensitization of IL-2 signaling through TLR-7 enhances B lymphoma cell immunogenicity. J.Immunol. 2006;176:3830-3839.

Toze CL, Dalal CB, Nevill TJ, et al. Allogeneic haematopoietic stem cell transplantation for chronic lymphocytic leukaemia: outcome in a 20-year cohort. Br J Haematol. 2012;158:174-185.

Page 84: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

76

76

Vegiopoulos A and Herzig S. Glucocorticoids, metabolism and metabolic diseases. Mol Cell Endocrinol. 2007;275:43-61. Wallberg AE, Neely KE, Hassan AH, et al. Recruitment of the SWI/SNF chromatin remodeling complex as a mechanism of gene activation by the glucocorticoid receptor tau1 activation domain. Mol Cell Biol. 2000;20:2004-2013. Wang JB, Erickson JW, Fuji R, et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell. 2010;18:207-219.

Warburg O. On respiratory impairment in cancer cells. Science. 1956;124:269-270. Watanabe T, Hotta T, Ichikawa A, et al. The MDM2 oncogene overexpression in chronic lymphocytic leukemia and low-grade lymphoma of B-cell origin. Blood. 1994;84:3158-3165. Wei G, Twomey D, Lamb J, et al. Gene expression-based chemical genomics identifies rapamycin as a modulator of MCL1 and glucocorticoid resistance. Cancer Cell. 2006;10:331-342. Weirda WG, Kipps TJ, Mayer J, et al. Ofatumumab as single-agent CD20 immunotherapy in fludarabine –refractory chronic lymphocytic leukemia. J Clin Oncol. 2010;28:1749-1755. Wiestner A. Targeting B-Cell Receptor Signaling for Anticancer Therapy: The Bruton's Tyrosine Kinase Inhibitor Ibrutinib Induces Impressive Responses in B-Cell Malignancies. J Clin Oncol. 2012 Oct 22. [Epub ahead of print].

Willis RC and Seegmiller JE. The inhibition by 6-diazo-5-oxo-l-norleucine of glutamine catabolism of the cultured human lymphoblast. J Cell Physiol. 1977;93:375-382.

Wolins NE, Quaynor BK, Skinner JR et al. OP-9 mouse stromal cells rapidly differentiate into adipocytes: characterization of a useful new model of adipogenesis. J.Lipid Res. 2006;47:450-460.

Wong KK, Brenneman F, Chesney A, et al. Soluble CD200 Is Critical to Engraft CLL Cells in Immunocompromised Mice. Cancer Res. 2012;72:4931-4943.

Xia Y, Lu RN and Li J. Angiogenic factors in CLL. Leuk.Res. 2012;36:1211-1217.

Xu W, Miao KR, Hong M, et al. High-dose methylprednisolone can induce remissions in patients with fludarabine-refractory chronic lymphocytic leukaemia. Eur J Cancer. 2010;46:2145-2149.

Page 85: IMPROVING GLUCOCORTICOID THERAPY IN CHRONIC … · iv List of Figures Figure 1 Effects of Dexamethasone (DEX) on circulating CLL cells 25 Figure 2 Effects of DEX on mitochondrial

77

77

Xu HE, Stanley TB, Montana VG et al. Structural basis for antagonist-mediated recruitment of nuclear co-repressors by PPARalpha. Nature. 2002;415:813-817.

Yang-Yen H, Chambard JC. Sun YL, et al. Transcriptional interference between c-Jun an the glucocorticoid receptor: Mutual inhibition of DNA binding due to direct protein-protein interaction. Cell. 1990;62:1205-1215. Zaugg K, Yao Y, Reilly PT et al. Carnitine palmitoyltransferase 1C promotes cell survival and tumor growth under conditions of metabolic stress. Genes Dev. 2011;25:1041-1051.

Zent CS and Kay NE. Autoimmune complications in chronic lymphocytic leukaemia (CLL). Best Pract Res Clin Haematol. 2010;23:47-59. Zenz T, Gribben JG, Hallek M et al. Risk categories and refractory CLL in the era of chemoimmunotherapy. Blood 2012;119:4101-4107.

Zhang Z, Jones S, Hagood JS, et al. STAT3 acts as a co-activator of glucocorticoid receptor signaling. J Biol Chem. 1997;272:30607-30610. Zhang C, Ryu YK, Chen TZ, et al. Synergistic activity of rapamycin and dexamethasone in vitro and in vivo in acute lymphoblastic leukemia via cell-cycle arrest and apoptosis. Leuk Res. 2012;36:342-349. Zhou J, Cidlowski JA. The human glucocorticoid receptor: one gene, multiple proteins and diverse responses. Steroids. 2005;70:407-417.


Recommended