immune regulation in plasma cell myeloma - opus at uts: home · woodland n, nassif n, hart d,...

142
IMMUNE REGULATION IN PLASMA CELL MYELOMA Esther Aklilu Submitted for the degree of Master of Science at the University of Technology, Sydney in December 2013

Upload: ngoquynh

Post on 20-Jan-2019

213 views

Category:

Documents


0 download

TRANSCRIPT

IMMUNE REGULATION IN

PLASMA CELL

MYELOMA

Esther Aklilu

Submitted for the degree of Master of Science at the University of Technology, Sydney in December 2013

i

CERTIFICATE OF ORIGINAL AUTHORSHIP

I certify that the work in this thesis has not previously been submitted for a degree nor has it been submitted as part of requirements for a degree except as fully acknowledged within the text.

I also certify that the thesis has been written by me. Any help that I have received in my research work and the preparation of the thesis itself has been acknowledged. In addition, I certify that all information sources and literature used are indicated in the thesis.

Signature of Student:

Date:

ii

Acknowledgements

I wish to express my sincere gratitude to my supervisors Dr Ross Brown, Mrs Narelle

Woodland and Dr Najah Nassif for their magnanimous support, encouragement and

patience throughout the duration of my degree.

To Ross, thank you for the opportunity to work for you and for allowing me to take on such

an interesting project for my degree. I truly appreciate your exceptional advice and guidance

throughout this project.

To Narelle and Najah, thank you for accepting me as your Masters Degree student and for

all the excellent advice, support and encouragement. I truly appreciate all your immense

help.

To my fellow research team and colleagues, your assistance and support is greatly

appreciated. Thank you to Shihong Yang for imparting to me your knowledge of flow

cytometry, cell sorting and cell culture work. It has been an absolute joy working with you.

Thank you to James Favaloro for your editorial advice, support and friendship. Although we

had only worked together for a short time, it has been a real pleasure working with you.

Also, I would like to appreciate your assistance in the analysis of the 10 year survivor

samples.

Thank you to the haematology staff of RPAH, for making my time in RPA thoroughly

enjoyable and to the volunteers, for the kind donation of your blood.

Thank you to Professor Doug Joshua and Dr P Joy Ho, for your suggestions and advice during

our weekly meetings.

I also wish to acknowledge the financial support provided by the Sydney Foundation for

Medical Research and the Cancer Institute, NSW.

Finally, I wish to thank my family and friends for their support and encouragement.

iii

Manuscripts and presentations arising from the work completed as part of this thesis

Articles published in peer-reviewed journals

Favaloro J, Brown R, Aklilu E, Yang S, Suen H, Hart D, Fromm P, Gibson J, Khoo L, Ho PJ, Joshua D (2013) Myeloma skews regulatory T and pro-inflammatory T helper 17 cell balance in favour of a suppressive state. Leukaemia & lymphoma. 55(5):1090-8

Bryant C, Suen H, Brown R, Yang S, Favaloro J, Aklilu E, Gibson J, Ho PJ, Iland H, Fromm P, Woodland N, Nassif N, Hart D, Joshua D (2013) Long-term survival in multiple myeloma is associated with a distinct immunological profile, which includes proliferative cytotoxic T-cell clones and a favourable Treg/Th17 cell balance. Blood Cancer journal 3:e148.

Conference abstracts

Favaloro J, Brown R, Aklilu E, Yang S, Suen H, Gibson J, Ho PJ, Joshua D. (2013) The ratio of Treg/Th17 cells is of prognostic significance in multiple myeloma. Australian Flow Cytometry group (AFCG) meeting.

Bryant CE, Brown RD, Yang S, Suen H, Aklilu E, Favaloro J, Hart DNJ, Fromm P, Woodland N, Nassif N, Iland H, Gibson J, Ho PJ, Joshua DE. (2011) Immunological Biomarkers in 10 year Survivors of Multiple Myeloma. Proceedings of the Haematology Society of Australia and New Zealand. O/021.

Bryant CE, Brown RD, Yang S, Suen H, Aklilu E, Favaloro J, Hart DNJ, Fromm P, Woodland N, Nassif N, Iland H, Gibson J, Ho PJ, Joshua DE. (2011) Ten year survivors of multiple myeloma demonstrate a differential expression of immunological biomarkers including a high incidence of cytotoxic T-cell clones which have not acquired myeloma-associated anergy. Blood. 118:S38865.

Aklilu E, Brown R, Yang S, Kabani K, Woodland N, Nassif N, Ho P, Gibson J, Joshua D. (2010) Treg number, Treg function and Th17 cells in plasma cell dyscrasias. Proceedings of the Haematology Society of Australia and New Zealand. A94.

Aklilu E, Brown R, Yang S, Kabani K, Woodland N, Nassif N, Ho PJ, Gibson J, Joshua D (2010) Treg number, Treg function and Th17 cells in plasma cell dyscrasias. Australian Flow Cytometry group (AFCG) meeting. A87.

Aklilu E, Brown R, Yang S, Kabani K, Woodland N, Nassif N, Ho PJ, Gibson J, Joshua D (2010) Treg number, Treg function and Th17 cells in plasma cell dyscrasias, The XXVIIth Annual scientific research meeting. P42.

iv

Abbreviations

Ab/ Abs Antibody (-ies)

AF-647 Alexa Fluor 647

AML Acute myeloid leukaemia

Anti- Antibody against

APC Allophycocyanin

APCs Antigen presenting cells

ATLL Adult T-cell leukaemia/lymphoma

B cell B lymphocyte

2M Beta-2 microglobulin

BM Bone marrow

BMMC Bone marrow mononuclear cells

Bort- Bortezomib (Velcade)

C Degrees Celsius

CD Cluster of differentiation

CFSE Carboxyfluorescein succinimidyl ester

CLL

CLMF

Chronic lymphocytic leukaemia

Cytotoxic lymphocyte maturation factor

CRAB Hypercalcaemia, renal failure, anaemia and bone lesion

CRP C-reactive protein

CTLA-4 Cytotoxic T lymphocyte-associated antigen-4

CTLA-8 Cytotoxic T lymphocyte-associated antigen-8

DC Dendritic cells

Dex Dexamethasone

DMSO Dimethyl sulfoxide

DVT Deep vein thrombosis

EDTA Ethylenediamine tetra-acetic acid

FACS Fluorescence activated cell sorting

FISH Fluorescent in situ hybridisation

FITC Fluorescein isothiocyanate

v

FoxP3 Forkhead box P3

FSC Forward scatter

g Gravitational force

G-CSF Granulocyte-colony stimulating factor

GITR Glucocorticoid-induced tumour necrosis factor -receptor-related

GvHD Graft versus host disease

HCl Hydrochloric acid

HEPES 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid

hi High

HLA Human leukocyte antigen

IFN Interferon

Ig Immunoglobulin

IL Interleukin

IMiD/ IMiDs Immunomodulatory drug (s)

iTreg Induced regulatory T cell

ISS International staging system

kDa- Kilo Dalton

LAP Latency associated peptide

Len Lenalidomide

Len + Dex Lenalidomide in combination with dexamethasone

mAB Monoclonal Antibody

MFI Mean fluorescence intensity

MGUS Monoclonal gammopathy of undetermined significance

MHC Major histocompatibility complex

min Minutes

MM

MRI

Plasma cell myeloma

Magnetic resonance imaging

NK

NKSF

Natural killer

Natural killer cell stimulatory factor

NS Not significant (P>0.05)

nTreg Natural regulatory T cell

vi

PB Peripheral blood

PBMC Peripheral blood mononuclear cells

PBS Phosphate buffered saline

PCLI Plasma cell labelling index

PE Phycoerythrin

PerCP Peridinin chlorophyll protein

PMA Phorbol 12-myristate 13-acetate

Pom Pomalidomide

Rh Recombinant human

ROR Retinoid-related orphan receptor

RPMI Roswell Park Memorial Institute

RT Room temperature

SAM Significance analysis of microarray

SSC Side scatter

SCT Stem cell transplantation

SD Standard deviation

SEM Standard error of mean

SM Smouldering myeloma

SPE Serum protein electrophoresis

STAT3 Signal transducer and activator of transcription 3

T cell T lymphocyte

TCR T cell receptor

TGF- Transforming growth factor

Th T helper cell

Thal- Thalidomide

TILs Tumour infiltrating lymphocytes

TNF- Tumour necrosis factor -

Treg Regulatory T lymphocyte

WM Waldenström macroglobulinaemia

vii

Abstract

The pathogenesis of monoclonal gammopathies, particularly plasma cell myeloma (MM), is

multifaceted and complex. In recent years, Treg and Th17 cells have emerged as key factors

in the development and progression of malignancies including MM. However, there are still

conflicting reports on whether regulatory T (Treg) cells and Th17 cells are increased or

decreased in the peripheral blood (PB) of patients with MM. This is partly due to technical

difficulties associated with the use of the transcriptional repressor, forkhead box P3 (FoxP3)

to identify Treg cells. Studies have shown FoxP3 results to be dependent on the clones,

fluorochromes attached and fixation/permeabilisation methods used. More recent studies

have defined Treg cells as CD4+CD25hi cells which do not express CD127, an IL-7 receptor.

This methodology was used to determine Treg cell number and develop assays for the

assessment of Treg cell function. The study also extends to exploring Th17 cell number in

plasma cell dyscrasias and the overall effect of the Treg and Th17 cell equilibrium on the

survival of patients with MM.

CD4+CD25hiCD127- expression was used to quantitate Treg cell numbers and an intracellular

IL-17 assay on CD3+CD4+ cells was used for Th17 cell enumeration. Treg cell function was

determined using carboxyfluorescein succinimidyl ester (CFSE) tracking of Treg depleted

lymphocyte preparations stimulated by anti-CD3,CD2,CD28 beads at a 1:1 ratio for 4 days

1:1 fluorescence-activated cell sorted Treg cells. This functional assay was also used to

investigate the effect of recombinant human (rh) TGF- and rhIL-12 on Treg cells.

The mean proportion of Treg cells in the CD4+ compartment of PB of patients with MM

(n=32) was 8.9±0.6% and this was increased compared to the mean of the normal cohort

(n=36) at 6.5±0.4% (p=0.009). However, no significant difference was observed between the

frequency of PB Treg cells in the control group compared to patients with monoclonal

gammopathy of undetermined significance (MGUS) (n=20) (mean=7.5±0.8%; P=0.24) and

patients with Waldenström macroglobulinaemia (WM) (n=13) (mean=6.0±0.5%; P=0.48).

Interestingly, a comparison of the absolute numbers exhibited different results. A

significantly lower number of Treg cells was observed in patients with MM [(3.2±0.4) x107/L;

P<0.01] and WM [(3.0±0.6) x107/L; P<0.01] compared to the control group [(6.4±0.7)

x107/L]. However, no significant difference was observed when comparing patients with

MGUS [(4.3±0.8) x107/L; P= 0.06] to the normal cohort. It was observed that a significantly

viii

higher proportion of PB Treg cells in patients with MM (85.9±1.8%; P<0.01) and WM

(86.4±2.1%; P=0.02) to be of the CD45RO+ memory phenotype compared to the normal

cohort (76.7±2.5%). However this was not observed in patients with MGUS (73.4±4.0%;

P=0.47). In addition, the study revealed that PB Treg cell proportions were not influenced by

MM stage. Thalidomide treated patients with MM appeared to have an increased PB Treg

cell proportion, however only a small number of thalidomide treated patients were tested

due to the use of thalidomide therapy being phased out and its replacement with

lenalidomide. Treg cells from bone marrow (BM) were compared to matched PB samples

from patients with MM, demonstrating a significantly greater proportion (p=0.02) of Treg

cells in the CD4+ compartment of the BM (9.7±1.2%) compared to PB (6.7±1.4%).

Regarding Th17 cells, a significant decrease (p=0.03) in the mean proportion and absolute

number of Th17 cells was observed in the PB of patients with MM (n=22) compared with the

controls (n=20) (0.7±0.1% and 2.0±0.6% respectively). However, the mean number of Th17

cells in patients with MGUS (2.2±0.6%; n=15) and WM (1.1±0.2%; n=12) was not significantly

different from normal. No correlation was observed between Th17 cell number and MM

staging or therapy.

Additionally, the study explored the Treg/Th17 cell ratio in PB of patients with monoclonal

gammopathies with comparison made to normal subjects. The mean Treg/Th17 cell ratio of

patients with MM (16.1±2.4) was significantly higher (p=0.0002) than the healthy control

group (6.6±1.0). The Treg/Th17 cell ratio of WM and MGUS patients was 7.0±1.0 and

4.9±0.5 respectively, neither of which were statistically different to the ratio of the normal

controls. Most interestingly, patients who have survived with MM for 10 or more years

possessed a Treg/Th17 cell ratio similar to the normal controls (7.04±2.47; p=0.84) and this

was shown to affect overall survival. The data demonstrated that patients with MM

observed to have a high Treg/Th17 cell ratio had an overall shorter survival compared to

those whose Treg/Th17 cell ratio was lower (p<0.025).

The suppressive capability of Treg cells from MM patients (n=15) was variable. The Treg cell

function of patients treated with lenalidomide (n=5) was increased (mean=68%) compared

to patients treated with thalidomide (n=5; mean=23%), Velcade (n=3; mean=12%),

untreated patients (n=5; mean=36%) and normal controls (n=11; mean=31%). The

suppression exerted upon the CD4+ T cell subset in patients treated with bortezomib was

ix

significantly lower when compared to the normal cohort. However, no significant difference

in CD8+ T cell suppression was found between patients with MM and the normal controls.

rhTGF- increased the suppressive capabilities and rhIL-12 reduced the function of Treg cells

from both MM and normal PB samples.

In conclusion, immune regulation is dysfunctional in patients with MM as the proportion of

PB Treg cells is increased and Th17 cells are reduced. Also, the cytokine microenvironment

and treatment have a major impact on the function of Treg cells. The data clearly delineate

the importance of the PB Treg/Th17 cell equilibrium, revealing a strong association between

the Treg/Th17 cell homeostatic balance and disease progression and survival in MM,

indicating an imbalance may cause either or both the innate and adaptive immune system

to be dormant and incapacitate the anti-tumour response.

x

Table of Contents

ACKNOWLEDGEMENTS .................................................................................................................. II

MANUSCRIPTS AND PRESENTATIONS ARISING FROM THE WORK COMPLETED AS PART OF THIS THESIS ........... III

ABBREVIATIONS ......................................................................................................................... IV

ABSTRACT .............................................................................................................................. .. VII

TABLE OF CONTENTS .................................................................................................................... X

LIST OF FIGURES ........................................................................................................................ XV

LIST OF TABLES ......................................................................................................................... XVII

INTRODUCTION................................................................................................... 1

PLASMA CELL MYELOMA................................................................................................................ 2

1.1 INTRODUCTION TO PLASMA CELL MYELOMA: EPIDEMIOLOGY, INCIDENCE AND MORTALITY ............... 2

1.2 CLINICAL PRESENTATION, DIAGNOSIS AND PROGNOSTIC INDICATORS IN MM ................................ 2

1.2.1 Clinical presentation ............................................................................................ 2

1.2.2 Diagnosis and classification of monoclonal gammopathies ................................ 3

1.2.3 Staging and prognosis .......................................................................................... 5

1.3 THERAPY FOR MM ........................................................................................................... 8

1.3.1 Thalidomide ......................................................................................................... 8

1.3.2 Lenalidomide and Pomalidomide ........................................................................ 9

1.3.3 Bortezomib ......................................................................................................... 10

1.3.4 Haematopoietic stem cell transplantation ........................................................ 11

1.3.4.1 Autologous haematopoietic stem cell transplantation ................................. 11

1.3.4.2 Allogeneic haematopoietic stem cell transplantation ................................... 12

IMMUNE REGULATION ................................................................................................................ 12

1.4 THE IMMUNE SYSTEM ...................................................................................................... 12

1.4.1 Lymphocytes ...................................................................................................... 12

1.5 REGULATORY T (TREG) CELLS ............................................................................................ 15

1.5.1 Treg cells in PB ................................................................................................... 15

1.5.2 Classes of Treg cells............................................................................................ 15

1.5.3 Treg cell identification by flow cytometry ......................................................... 16

xi

1.5.4 Treg cells and cancer .......................................................................................... 17

1.5.5 Treg cells and haematological malignancies ..................................................... 18

1.5.6 Treg cells and GvHD ........................................................................................... 18

1.5.7 Controversy ........................................................................................................ 19

1.6 TH17 CELLS AND THEIR CYTOKINES ..................................................................................... 21

1.6.1 Th17 cell development and differentiation ....................................................... 21

1.6.2 The IL-17 cytokine .............................................................................................. 21

1.6.3 Th17 cells in cancer ............................................................................................ 22

1.7 THE ROLE OF THE CYTOKINES TGF- AND IL-12 IN CANCER ..................................................... 23

1.7.1 TGF- .................................................................................................................. 23

1.7.2 IL-12 .................................................................................................................... 23

1.8 BACKGROUND TO THE PROJECT .......................................................................................... 24

1.9 HYPOTHESIS .................................................................................................................. 24

1.10 AIMS OF THE PROJECT ...................................................................................................... 25

MATERIALS AND METHODS .................................................................................. 26

2.1 PARTICIPANTS ................................................................................................................ 27

2.2 SAMPLE COLLECTIONS ...................................................................................................... 27

2.2.1 Blood sample collection ..................................................................................... 27

2.2.2 BM collection ..................................................................................................... 27

2.3 METHODOLOGY FOR PREPARATION OF COMMON REAGENTS ................................................... 28

2.3.1 Preparation of ammonium chloride lysing reagent ........................................... 28

2.3.2 Preparation of phosphate buffered saline ........................................................ 28

2.3.3 Preparation of Roswell Park Memorial Institute -10 medium........................... 28

2.3.4 Preparation of RPMI cell culture medium ......................................................... 29

2.3.5 Preparation of MACSBeads ................................................................................ 29

2.3.6 Preparation of carboxyfluorescein succinimidyl ester stain .............................. 29

2.4 GENERAL TECHNIQUES ..................................................................................................... 31

2.4.1 Cell wash ............................................................................................................ 31

2.4.2 Isolation of mononuclear cells using Ficoll-Paque density gradient centrifugation ................................................................................................................ ... 31

xii

2.5 MONOCLONAL ANTIBODIES USED DURING EXPERIMENTATION .................................................. 33

2.6 ASSAY FOR IDENTIFICATION OF TREG CELLS .......................................................................... 34

2.6.1 Cell staining for flow cytometric identification of CD127lo/- Treg cells .............. 34

2.6.2 Preparation of PBMC Treg cell identification using FoxP3 ................................ 34

2.6.3 Preparation of BM Treg cells for flow cytometric analysis ................................ 35

2.7 METHODOLOGY FOR THE IDENTIFICATION OF TH17 CELLS IN PB .............................................. 36

2.7.1 Preparation of Th17 cells for flow cytometric analysis ..................................... 36

2.8 TREG CELL FUNCTIONAL ASSAY USING CFSE ......................................................................... 37

2.8.1 Fluorescence-activated cell sorting of PBMC .................................................... 37

2.8.2 FACS purity test .................................................................................................. 38

2.8.3 CFSE staining of target cells ............................................................................... 38

2.8.4 Cell count ........................................................................................................... 39

2.8.5 Preparation of the MACSbead suspension ........................................................ 39

2.8.6 Cell culture of Treg and CFSE stained target cell ............................................... 39

2.8.7 Analysis of the CFSE assay after culture ............................................................ 41

2.9 EVALUATION OF THE IMPACT OF CYTOKINES (IL-12 AND TGF- ) AND ANTI-TGF- ON TREG CELLS 41

2.9.1 IL-12 preparation ............................................................................................... 41

2.9.2 rhTGF- 1 preparation ........................................................................................ 42

2.9.3 Anti TGF- preparation ...................................................................................... 42

2.9.4 Plating of the cell culture wells with addition of cytokines ............................... 42

2.10 STATISTICS AND ANALYSIS ................................................................................................. 44

RESULTS ......................................................................................................... 45

3.1 OVERVIEW .................................................................................................................... 46

3.2 TREG CELLS IN MONOCLONAL GAMMOPATHIES ..................................................................... 46

3.2.1 Identification of Treg cells using CD127 ............................................................ 46

3.2.2 Identification of Treg cells using FoxP3 ............................................................. 49

3.2.3 Enumeration of Treg cells in PB ......................................................................... 52

3.2.4 Phenotypic analysis of Treg cells ....................................................................... 55

3.2.5 Effect of IMiD treatment on Treg cell proportions ............................................ 58

xiii

3.2.6 MM staging and Treg cell quantitation ............................................................. 60

3.2.7 Enumeration of Treg cells in BM versus PB ....................................................... 60

3.2.8 Treg cells in patients with MM who have survived more than 10 years ........... 63

3.3 TH17 CELLS IN MM AND OTHER MONOCLONAL GAMMOPATHIES ............................................. 66

3.3.1 Identification of Th17 cells in PB........................................................................ 66

3.3.2 Enumeration of Th17 cells in PB ........................................................................ 68

3.3.3 Th17 cells and stage of MM ............................................................................... 70

3.3.4 Th17 cells in MM patients who have survived more than 10 years .................. 71

3.4 TREG AND TH17 CELL EQUILIBRIUM ................................................................................... 73

3.4.1 Treg/Th17 cell ratio in monoclonal gammopathies........................................... 73

3.4.2 Effect of a high Treg/Th17 cell ratio on overall survival in MM ........................ 73

3.5 TREG CELL FUNCTIONAL ASSAY ........................................................................................... 77

3.5.1 Development and optimisation of Treg cell functional assay ........................... 77

3.5.2 Treg cell functional assay results ....................................................................... 79

3.5.3 Suppression of T cell proliferation ..................................................................... 79

3.6 EFFECT OF CYTOKINES ON TREG CELL FUNCTION .................................................................... 84

DISCUSSION ..................................................................................................... 86

4.1 OVERVIEW .................................................................................................................... 87

4.2 THE ROLE OF TREG CELLS IN MONOCLONAL GAMMOPATHIES ................................................... 88

4.2.1 The phenotype and identification of Treg cells ................................................. 88

4.2.2 Enumeration and phenotype of Treg cells in monoclonal gammopathies ....... 89

4.2.3 Naïve and memory Treg cells in monoclonal gammopathies ........................... 91

4.2.4 Effect of MM treatment on Treg cell proportion .............................................. 91

4.2.5 Effect of MM stage on Treg cell proportion ...................................................... 92

4.2.6 Quantification of Treg cells in the PB and BM of MM patients ......................... 93

4.2.7 Treg cell function in MM .................................................................................... 93

4.2.8 Treg cell suppression on the CD4 and CD8 T cell subsets.................................. 95

4.2.9 Modification of Treg cell function ..................................................................... 95

4.3 THE ROLE OF TH17 CELLS IN THE MONOCLONAL GAMMOPATHIES ............................................. 97

xiv

4.3.1 Th17 cell enumeration in MM and other monoclonal gammopathies ............. 97

4.3.2 Association of MM stage and Th17 cell proportion .......................................... 99

4.4 TREG AND TH17 EQUILIBRIUM ........................................................................................ 100

4.4.1 Treg/Th17 cell ratio in monoclonal gammopathies......................................... 100

4.4.2 Treg/Th17 cell ratio and survival ..................................................................... 101

4.5 CONCLUSIONS AND FUTURE DIRECTIONS ............................................................................ 102

4.5.1 Future directions .............................................................................................. 102

4.5.2 Conclusion ........................................................................................................ 103

REFERENCES .................................................................................................. 105

xv

List of Figures

Figure 1.1: Diagnostic characteristics of MM ............................................................................ 4

Figure 1.2: Gene expression patterns and overall survival in the different molecular

subgroups of MM ............................................................................................................... ........ 7

Figure 1.3: The chemical structure of the IMiDs thalidomide, lenalidomide and

pomalidomide .................................................................................................................. ........ 10

Figure 1.4: Hierarchy of lymphocytes and T cell lineages involved in the innate and adaptive

immune response ............................................................................................................... ..... 14

Figure 2.1: Flow cytometric analysis of a CFSE stained cell population .................................. 30

Figure 2.2: Whole blood before and after centrifugation with Ficoll-Paque .......................... 31

Figure 2.3: Cell culture plate set up for investigation of Treg cell function ............................ 40

Figure 2.4: Cell culture plate set up to evaluate the effect of IL-12, TGF- , and anti-TGF- on

the function of Treg cells .................................................................................................... ..... 43

Figure 3.1: Isotype control for each fluorochrome used in the Treg cell assay ...................... 47

Figure 3.2: Flow cytometry results from a representative normal subject showing the gating

strategy for identification of Treg cells in PB ........................................................................... 48

Figure 3.3: Flow cytometry results from a representative normal subject showing the gating

strategy for identification of PB Treg cells using FoxP3 ........................................................... 50

Figure 3.4: Comparison of FoxP3 expression on sorted CD25hiCD127lo/- Treg cells compared

to the Treg depleted T cells .................................................................................................. ... 51

Figure 3.5: Enumeration of PB Treg cells in patients with monoclonal gammopathies and

normal control individuals .................................................................................................... ... 54

Figure 3.6: Flow cytometry results showing CD45RA and CD45RO expression on Treg cells in

PB of a representative normal subject .................................................................................... 56

Figure 3.7: Expression levels of CD45RA and CD45RO on Treg cells in PB of patients with

monoclonal gammopathies and normal individuals ............................................................... 57

Figure 3.8: Effect of IMiD treatment on PB Treg cell proportions in patients with MM ......... 59

Figure 3.9: Comparison of the percentage of PB Treg cells and the different stages of MM . 61

Figure 3.10: Comparison of Treg cell numbers in the BM and PB of patients with MM......... 62

Figure 3.11: Relative Treg cell numbers in PB of normal subjects, patients with MM for less

than 10 years and MM patients who have survived for 10 or more years since diagnosis .... 64

xvi

Figure 3.12: Absolute number of PB Treg cells in normal subjects, patients with MM for less

than 10 years and patients who have survived 10 or more years with MM ........................... 65

Figure 3.13: Flow cytometric results from a PB sample of a representative normal subject

showing the gating strategy for the identification of Th17 cells ............................................. 67

Figure 3.14: Relative Th17 cells number in the PB of patients with monoclonal

gammopathies and normal controls ........................................................................................ 69

Figure 3.15: Comparison of the percentage of Th17 cells of the CD4+ compartment in PB and

different stages of MM ........................................................................................................ .... 70

Figure 3.16: Enumeration of Th17 cells in PB of patients with MM and normal subjects ...... 72

Figure 3.17: Treg/Th17 cell ratio in patients with monoclonal gammopathies and normal

individuals ................................................................................................................... ............. 74

Figure 3.18: Treg/Th17 cell ratio in 10 year MM survivors ..................................................... 75

Figure 3.19: The effect of Treg/Th17 cell ratio on overall survival of patients with MM ....... 76

Figure 3.20: Optimisation of MACSbead stimulation for the Treg functional assay ............... 78

Figure 3.21: Flow cytometry gating strategy for FACS of Treg cells in PB ............................... 81

Figure 3.22: A representative flow cytometric analysis of CFSE stained CD3+ cells after 4 days

culture ....................................................................................................................... ............... 82

Figure 3.23: Effect of treatment on Treg cells suppressive activity on lymphocytes target

cells ......................................................................................................................... ................. 83

Figure 3.24: Effect of IL-12, TGF- and anti-TGF- cytokines on the suppressive function of

Treg cells .................................................................................................................... .............. 85

xvii

List of Tables

Table 1.1: International Staging System for MM ....................................................................... 5

Table 1.2: Comparison of studies of Treg identification, enumeration and function in MM. 20

Table 2.1: List of monoclonal antibodies (mAb) used during experimentation ...................... 33

Table 2.2: Fluorescent antibodies used for the flow cytometric identification of Treg cells

using CD127 ................................................................................................................... .......... 34

Table 2.3: Fluorescent antibodies used for the flow cytometric identification of Treg cells

using FoxP3 ................................................................................................................... ........... 35

Table 2.4: Fluorescent antibodies used for the flow cytometric identification of BM Treg cells

............................................................................................................................... ................... 36

Table 2.5: Fluorescent antibodies used to identify Th17 cells for analysis by flow cytometry

............................................................................................................................... ................... 37

Table 2.6: Fluorescent antibodies used to identify Treg cells for flow cytometric sorting ..... 38

Table 2.7: Fluorescent antibodies used for analysis of the CFSE assay by flow cytometry .... 41

1

CHAPTER ONE

INTRODUCTION

2

PLASMA CELL MYELOMA

1.1 Introduction to plasma cell myeloma: Epidemiology, incidence and mortality

Plasma cell myeloma (MM), more commonly known as multiple myeloma, is a malignancy of

plasma cells that is characterised morphologically by an excessive number of abnormal

plasma cells in the bone marrow (BM). The Australian Institute of Health and Welfare data

from 2007 indicates that in Australia, MM accounts for approximately 1.2% of all cancers

with approximately 1200 people, most of whom are above the age of 40, diagnosed each

year. The median age of onset is in the early 60s, and it is more common in males than

females, occurring at a ratio of 1.6:1. Overall, MM accounts for approximately 10% of all

haematological malignancies (Welfare, 2010).

In patients with MM, malignant plasma cells (MM cells) produce excessive amounts of a

monoclonal immunoglobulin, called M protein or paraprotein. The type of paraprotein

produced by the plasma cell determines the MM classification. The paraprotein produced by

the malignant plasma cells is Immunoglobulin G (IgG) in 60% of cases, Immunoglobulin A

(IgA) in 20% and Immunoglobulin D (IgD) in 2% of cases. MM characterised by excessive

immunoglobulin M (IgM) is rare. Some patients produce a paraprotein that does not have a

heavy chain and this is referred to as light chain disease (monoclonal kappa or lambda).

There are also a few patients who are non-secretors and have no paraprotein despite the

presence of a large number of malignant plasma cells. MM can be a seriously debilitating

disease with multi-organ involvement. Currently it is incurable, although there are a number

of therapies that improve quality of life and prolong survival.

1.2 Clinical presentation, diagnosis and prognostic indicators in MM

1.2.1 Clinical presentation

The clinical presentation of patients with MM can be quite variable. Twenty percent of

patients are diagnosed by chance without any characteristic symptoms (Sagar, 2010). The

most common symptom of MM is bone pain. Patients usually develop osteolytic lesions in

multiple sites around the body (Figure 1.1A), hence the term “multiple” myeloma. Bone

3

pain may be caused by fractures, spinal compression and/or the formation of lesions by

osteoclasts or osteoblasts proliferating to replace missing bone.

According to the review of Kyle and Rajkumar (2008), the most common clinical findings in

patients with MM at diagnosis are the presence of paraprotein (>90%) and bone lesions

(80%), followed by anaemia (72%), renal failure (19%) and hypercalcaemia (13%).

1.2.2 Diagnosis and classification of monoclonal gammopathies

Physical examination, blood tests, urine tests, BM biopsy and X-ray are all required for the

diagnosis of MM (Marc et al., 2009). At presentation, all patients with a monoclonal

gammopathy are categorised into one of three groups according to the published criteria of

the International Myeloma Working Group (Table 1.1). The original classification of 2005

(Greipp et al., 2005) was revised in 2008 (Kyle and Rajkumar, 2008). The three groups are:

(1) MM, (2) smouldering myeloma (SM) and (3) monoclonal gammopathy of undetermined

significance (MGUS). In addition to the three major groups of monoclonal gammopathies,

there are a number of other, rare, associated conditions that include solitary plasmacytoma,

amyloidosis and osteosclerotic myeloma (POEMS syndrome) (Kyle and Rajkumar, 2008) and

Waldenström macroglobulinaemia (WM), which is now classified as a lymphoplasmacytic

lymphoma (Swerdlow et al., 2008).

While a X-ray (Figure 1.1A), BM biopsy (Figure 1.1B), serum protein electrophoresis (SPE)

(Figure 1.1C) and immunofixation (Figure 1.1D) are the basic tests used for the diagnosis of

MM, a range of laboratory tests are performed to provide prognostic information and to

detect, or monitor, multi-organ failure. A full blood count, biochemistry screen (including

calcium and creatinine), serum 2microglobulin ( 2M), cytogenetics and fluorescent in situ

hybridisation (FISH) are also studied.

4

A B

C D

Figure 1.1: Diagnostic characteristics of MM A. X-rays showing lytic lesions in the skull and bones. B. BM biopsy stained with May-

Grünwald Giemsa showing an increased number of plasma cells. These are large

mononuclear cells with an eccentrically placed nucleus, basophilic cytoplasm and a nuclear

hoff. C. Serum protein electrophoresis showing an abnormal peak in the gamma-globulin

region. D. Immunofixation demonstrating the presence of an IgG paraprotein in a serum

sample from a patient with MM. (Images courtesy of R. Brown, Haematology, RPAH).

5

1.2.3 Staging and prognosis

There are a number of staging systems to classify patients with MM based on pathological

findings. These staging systems are designed to predict the prognosis of each patient. The

Durie-Salmon staging system was originally used as the main staging system for MM (Durie

and Salmon, 1975), however this was replaced in 2005 by the International Staging System

(ISS) (Greipp et al., 2005). The ISS only requires the use of two simple markers, serum

albumin and 2M, yet provides discrimination between each stage, and is superior to other

staging systems.

Table 1.1: International Staging System for MM Serum 2-

microglobulin

Serum albumin Median survival

Stage I <3.5 mg/L 35 g/L 62 months

Stage II Neither stage I or III 44 months

Stage III 5.5 mg/L 29 months

(Greipp et al., 2005)

A range of prognostic factors has been proposed for patients with MM, which may include:

1. Parameters reflecting the inherent proliferative capacity of the malignant clone

2. Parameters reflecting tumour bulk

3. Parameters reflecting renal failure

4. Parameters reflecting host-tumour interactions (Joshua et al., 1994, Joshua, 1996).

Other prognostic tests for MM include plasma cell morphology (Greipp et al., 1998), C-

reactive protein (CRP) levels (Bataille et al., 1992) and plasma cell labelling index (PCLI) to

determine the percentage of plasma cells that are in S-phase of the cell cycle (Pope et al.,

1999, Greipp et al., 2005). A patient with a high percentage of plasma cells in S-phase is

interpreted as having a poor prognosis.

In addition, molecular genetic studies of MM cells, including cytogenetic and FISH analysis,

also provide prognostic indicators. MM is divided into two fairly equally distributed groups

based on karyotypic abnormalities (Dewald et al., 1985, Gould et al., 1988, Lai et al., 1995,

6

Tricot et al., 1995). These comprise the hyperdiploid group, with mostly numerical gains in

chromosomes and few structural changes, and the non-hyperdiploid group, with many

chromosomal rearrangements and occasional numerical loss of chromosomes (Wuilleme et

al., 2004). This cytogenetic classification is of clinical importance as patients with

hyperdiploidy appear to have a better prognosis (Smadja et al., 2001, Debes-Marun et al.,

2003). Genetic aberrations such as chromosome 13 deletion, non-hyperdiploidy, t(4;14) and

t(14;16) translocations confer a poor prognosis while the 11q13 and 6p21 cytogenetic

abnormalities are associated with a better prognosis (Kyle and Rajkumar, 2008, Sagar,

2010).

Although not used in routine clinical practice, a molecular classification of MM, based on the

presence of translocations and hyperdiploidy, has been proposed by Zhan et al. (2006). This

classification consists of seven groups (Figure 1.2A) (Zhan et al., 2006) which have been

defined by two letter names and are characterised as follows:

a) PR – over-expression of genes responsible for cell cycle and cell proliferation

b) MS – over-expression of fibroblast growth factor receptor 3 (FGFR3) and multiple

myeloma SET domain (MMSET) genes

c) HY – hyperdiploid signature

d) CD-1 – over-expression of cyclin D1 and cyclin D3

e) CD-2 – over-expression of cyclin D2

f) MF – over-expression of MAF and MAFB genes

g) LB – low expression of genes involved in bone disease, such as Wnt signalling

antagonists Dickkopf (DKK1) and Frizzled B, and a low number of magnetic resonance

imaging (MRI) defined focal lesions.

With a 36-month median follow-up, the HY, CD-1, CD-2, and LB groups had a higher event-

free survival and overall survival compared with the PR, MS, and MF groups. In fact the high

proliferation group (PR) had the most significantly poor prognosis (Figure 1.2B) (Zhan et al.,

2006).

7

A B

Figure 1.2: Gene expression patterns and overall survival in the different molecular subgroups of MM A. Supervised clustergram of the expression of genes (50 significance analysis of microarray

(SAM)-defined over-expressed and under-expressed genes from each of the 7 defined

molecular subgroups). The vertical axis represents different genes, and the horizontal axis

represents samples. High expression is represented in red and low expression is represented

in blue. B. Kaplan-Meier estimates of overall survival for the 7 molecular subgroups. (Zhan

et al., 2006).

Months from start of therapy

Proportion of patients

8

1.3 Therapy for MM

There is a wide range of treatment options available for patients with MM. Current therapy

for patients under 65 years of age generally involves high dose induction chemotherapy

followed by autologous haematopoietic stem cell transplantation (SCT) (Quach and Prince,

2012). The main therapeutic agents are thalidomide or its analogue, lenalidomide, usually in

combination with dexamethasone. Other agents used are bortezomib and

cyclophosphamide, also in combination with dexamethasone. Patients often have low-dose

chemotherapy for maintenance after transplantation (Marc et al., 2009, Spencer et al.,

2009). Elderly patients may receive a less intensive therapy such as melphalan and

prednisone. Patients with MGUS or smouldering disease are likely not to receive therapy

(Kyle et al., 2007) as it has been shown in two randomised control trials that there is no

clinical benefit (Hjorth et al., 1993, Riccardi et al., 2000). Conversely, a recent study by

Mateos et al. (2013) has shown that early treatment of patients with high-risk smouldering

MM, with lenalidomide and dexamethasone, followed by maintenance therapy with

lenalidomide, would significantly delay the time to progression to symptomatic disease and

improve overall survival.

1.3.1 Thalidomide

Thalidomide ( -N-phthalimido-glutarimide) is a synthetic derivative of glutamic acid that has

been used successfully in the treatment of MM. Thalidomide has a number of different

modes of action that include immunomodulatory, anti-angiogenic, inhibition of the

microenvironment and a direct inhibition of the proliferation of the malignant cells

(D'Amato et al., 1994, Singhal et al., 1999, Teo, 2005). In the early 1960s, thalidomide was

withdrawn from the market as an antiemetic in pregnancy as it was associated with causing

foetal malformations (Lenz et al., 1962, Speirs, 1962). Reports of thalidomide’s efficacy in

graft versus host disease (GvHD) in animal models (Field et al., 1966), and early clinical

studies (Vogelsang et al., 1992), suggested that the drug may modify a T cell response. It is

currently thought that thalidomide initiates immunomodulation through its role as a potent

co-stimulator of human T cells, acting through the T cell receptor complex to increase

interleukin (IL) -2 mediated T cell proliferation and interferon (IFN) - production (Haslett et

al., 1998, Hayashi et al., 2005, Quach et al., 2009). The drug is associated with a range of

9

side effects, including neuropathy, deep vein thrombosis (DVT), fatigue and constipation. In

addition, patients may develop resistance to the drug and eventually relapse. A range of

thalidomide analogues, classified as immunomodulatory drugs (IMiDs), including

lenalidomide and pomalidomide have been developed. These analogues have greater anti-

cancer potency with fewer side effects, however the high cost of these drugs has been a

major factor in their delayed introduction into Australia.

1.3.2 Lenalidomide and Pomalidomide

The thalidomide derivatives lenalidomide (CC-5013; Revlimid) and pomalidomide (CC-4047;

Actimid) (Figure 1.3) are both effective IMiDs in the treatment of MM (Quach et al., 2009).

Compared to thalidomide, these IMiDs have improved potency and reduced toxicity (Quach

et al., 2009).

10

Thalidomide: 2-(2, 6-dioxopiperidin-3-yl)-1H-isoindole-1, 3(2H)-dione

Lenalidomide: 3-(4-amino-1-oxo 1, 3-dihydro-2H-isoindol-2-yl) piperidine-2, 6-dione

Pomalidomide: 4-Amino-2-(2, 6-dioxopiperidin-3-yl) isoindole-1, 3-dione

Figure 1.3: The chemical structure of the IMiDs thalidomide, lenalidomide and pomalidomide An amino group is added to the fourth carbon of the phthaloyl ring of thalidomide to

produce lenalidomide. The chemical structure of pomalidomide is an amalgamation of

thalidomide and lenalidomide (Kotla et al., 2009).

1.3.3 Bortezomib

Another new drug used to treat MM is the proteasome inhibitor bortezomib (Velcade),

which works by inhibiting intracellular protein degradation (Marc et al., 2009). In addition,

bortezomib induces apoptosis and inhibits growth of MM cells (Hideshima et al., 2001,

Mitsiades et al., 2002, Mitsiades et al., 2003). The drug may be beneficial for the treatment

of bone disease, as it increases bone formation by stimulation of osteoblasts and inhibition

of osteoclasts (Giuliani et al., 2007). Due to its different mode of action, bortezomib is a

11

useful agent in patients who are resistant to other therapies, however in Australia it is only

available to patients who meet set criteria. Toxicities associated with bortezomib include

peripheral neuropathy, transient thrombocytopaenia, fatigue, and gastrointestinal disorders

(Marc et al., 2009). A range of next generation protease inhibitors are currently being

trialled, including carfilzomib (Kuhn et al., 2007).

1.3.4 Haematopoietic stem cell transplantation

Haematopoietic SCT is used to facilitate the numerical restoration of cellular elements and

functional recovery of BM cellular interactions (Guillaume et al., 1998). Although autologous

SCT is not curative, it is used to induce long term disease-free survival in MM. In 1996, a

randomised trial by Intergoupe Francophone du Myélome demonstrated a better overall

survival after SCT compared to conventional chemotherapy (Attal et al., 1996).

Haematopoietic SCT can be either autologous, utilising stem cells from the patient’s own

BM or peripheral blood (PB), or allogeneic, utilising stem cells sourced from human

leukocyte antigen (HLA) matched normal donors.

1.3.4.1 Autologous haematopoietic stem cell transplantation

Autologous haematopoietic SCT is preferred in most cases as it is less complex and there is

minimal risk of GvHD (Lennard and Jackson, 2000). It involves high dose induction therapy,

which could involve agents such as high dose dexamethasone and thalidomide, followed by

mobilisation of stem cells to the blood using granulocyte-colony stimulating factor (G-CSF)

and leukapheresis to harvest the patient’s stem cells. The patient then receives high dose

chemotherapy to destroy remaining tumour bulk and the harvested stem cells are reinfused

into the patient. Some patients also undergo tandem autologous SCT that involves infusion

of a second transplant once a patient has recovered from the first. Studies have shown that

this improves event free and overall survival compared to a single autologous transplant

(Attal et al., 2003, Rajkumar and Kyle, 2005).

12

1.3.4.2 Allogeneic haematopoietic stem cell transplantation

Allografts are currently rarely performed. Unfortunately, due to old age, poor organ

function and HLA matched donor availability, only 5-10% of patients with MM are eligible

for allogeneic SCT. While autologous SCT is preferred, allogeneic SCT can be advantageous

as the transplant lacks tumour cell contamination, provides additional immunologic graft-

versus-myeloma effect and is potentially curative (Tricot et al., 1996, Bensinger, 2009,

Koehne and Giralt, 2012). The delay in disease relapse, however, does not offset the effects

of GvHD, which may lead to non-relapse mortality and an overall shortened survival

(Pasquini, 2008). Unless the HLA match is identical, the patient will almost certainly develop

GvHD.

IMMUNE REGULATION

1.4 The immune system

The immune system is composed of many cell types and mediators that interact with one

another. It is divided into two components, the innate and the adaptive immune system

based on antigen specificity and timing of activation (de Visser et al., 2006). The innate

immune system is the first line of defence against pathogens, and comprises macrophages,

natural killer (NK) cells, neutrophils, basophils and eosinophils. The adaptive immune

response has the ability to utilise immunological memory to recognise and retain antigen

specific epitopes. Dendritic cells (DC) are specialised antigen presenting cells (APCs) that

cross the divide between innate and adaptive immunity. Upon subsequent exposure to a

previously encountered antigen, the adaptive immune system is able to mount a more

specific and rapid response. In recent times, it has become apparent that immune cells of

patients with malignancies have impaired function and this may play a critical role in cancer

pathobiology.

1.4.1 Lymphocytes

Lymphocytes are classified into three groups: cells associated with the adaptive immune

response, T (thymus) cells (CD3+) and B (bursa of Fabricius) cells (CD19+) and a key

13

component of the innate immune system, natural killer (NK) cells. T cells are responsible for

cellular immunity, whereas B cells are primarily involved with humoral immunity.

T cells are further subdivided into T helper (Th) cells (CD4+) and cytotoxic T cells (CD8+)

(Figure 1.4). Cytotoxic T cells monitor other cells of the body and are capable of destroying

infected, damaged or dysfunctional cells and micro-organisms (Andersen et al., 2006). T

helper CD4+ cells are an important component of the immune system, that coordinates the

immune response of other cell types (Ji and Zhang, 2010). Upon activation, CD4+ cells are

able to differentiate into different Th subsets. In cases of autoimmunity and immunity

against intracellular pathogens, CD4+ cells are induced to differentiate into the T helper type

1 (Th1) cell type. This subset is distinguished by the expression of the transcription factor T-

bet and production of IFN- and IL-2 (Peck and Mellins, 2010). T helper type 2 (Th2) cell

types are involved with humoral immunity against parasites and allergic reactions and are

characterised by the expression of transcription factor GATA-3 and production of IL-4, IL-5

and IL-13. In recent years, a further two lineages of T helper cells have been identified and

these have been termed T regulatory (Treg) cells (Sakaguchi et al., 1995) and T helper 17

(Th17) cells (Harrington et al., 2005). These cell subsets will be described in greater detail in

the following sections.

In MM, the absolute numbers of T cells, in particular the CD4+ cells, are reduced, especially

in cases of progressive disease (Mills and Cawley, 1983, Pilarski et al., 1989, Kay et al., 2001,

Raitakari et al., 2003). In addition, abnormal T cell function has been observed in MM with

reports of enhanced susceptibility to apoptosis (Massaia et al., 1995) and an impaired CD8+

cell response (Maecker et al., 2003).

14

Figure 1.4: Hierarchy of lymphocytes and T cell lineages involved in the innate and adaptive immune response Depending on cytokine stimulation, T helper cells can differentiate into Th1, Th2, Treg or

Th17 cells.

NK cell CD16/56+

B cell CD19+

T cell CD3+

Th17 cell Inflammation

Treg cell Suppression

Th2 cell Humoral immunity

Th1 cell Cellular immunity

T cytotoxic cell CD8+

T helper cell CD4+

Lymphocyte

15

1.5 Regulatory T (Treg) cells

The concept of Treg cells as a novel CD4+ cell subset was first described in the early 1970s

(Gershon and Kondo, 1971, Gershon et al., 1972), however, there was great scepticism

about the existence of such cells. In 1995, Sakaguchi et al. identified a population of CD4+

cells which constitutively expressed CD25, the IL-2 receptor- chain (Sakaguchi et al., 1995,

Sakaguchi, 2004). It was reported that these cells were involved in maintaining self-

tolerance and eradication of these cells would lead to autoimmunity (Sakaguchi et al., 1995,

Chen and Oppenheim, 2009, Ha, 2009). These cells were termed Treg cells.

1.5.1 Treg cells in PB

Treg cells occupy 5-10% of the CD4+ compartment in PB (Sakaguchi, 2004, Prabhala et al.,

2006, Bronte, 2008, Ha, 2009). They are involved in the maintenance of self-tolerance,

control of autoimmunity, and modulation of the immune response against infections and

tumour cells (Sakaguchi et al., 2001). These cells are crucial in maintaining immunological

homeostasis (Fontenot and Rudensky, 2005, Chen and Oppenheim, 2009) by the

suppression of immune cells. Studies have shown that Treg cells are able to influence the

activation, proliferation and function of a range of immune cells such as T helper cells,

cytotoxic T cells, NK cells, DC, macrophages, osteoblasts, mast cells and B cells (Ha, 2009,

Chen and Oppenheim, 2009). Antigenic stimulation through the T cell receptor (TCR) is

required to induce the suppressive activity of Treg cells, however suppression exerted by

Treg cells appears to be non-specific (Liu et al., 2006, Chen and Oppenheim, 2009). It has

been demonstrated that Treg cells are able to exert their suppressive capabilities using two

mechanisms. Treg cells can inhibit the proliferation of CD4+CD25- cells by cell to cell contact

(Prabhala et al., 2006) or, alternatively, they can secrete immunosuppressive cytokines,

including IL-10 and transforming growth factor (TGF) - , to suppress an immune response

(Chen et al., 2003, Prabhala et al., 2006, Ji and Zhang, 2010).

1.5.2 Classes of Treg cells

There are two classes of Treg cells; the natural Treg (nTreg) cells and the adaptive or

induced Treg (iTreg) cells. nTreg cells are derived from the thymus, then exported into the

16

peripheral circulation to reside in the lymph nodes and spleen (Joshua et al., 2008, Ha,

2009). Naïve T cells can differentiate into iTreg cells in the periphery when induced by TGF-

and IL-10 (Bettelli et al., 2006, Ha, 2009). Moreover, Treg cells can also be further

characterised into at least three subsets: (1) CD4+CD25+FoxP3+, (2) Type 1 Treg cells (T 1)

which secrete IL-10 and (3) Th3 cells which secrete TGF- (Liu et al., 2006, Ha, 2009).

1.5.3 Treg cell identification by flow cytometry

Study in the field of Treg cell immunology has been difficult due to the lack of specific cell

surface biomarkers to identify Treg cells from regulator and effector T cell subsets (Liu et al.,

2006). The earliest studies used CD4 and CD25 as markers to identify the Treg cell

population. In animal models held under pathogen free conditions, CD25 is an adequate

marker for Treg cells. However, the use of CD4 and CD25 in the human setting is insufficient,

as humans are continuously exposed to foreign antigens and therefore constantly possess a

population of activated CD25+ effector T cells (Beyer and Schultze, 2006, Liu et al., 2006).

Currently, the most common phenotype identified and used to define a Treg cell is

CD4+CD25hiFoxP3+. FoxP3 is an X chromosome linked forkhead/winged helix family

transcriptional repressor (Fontenot and Rudensky, 2005, Joshua et al., 2008) that is a crucial

component in the development and function of Treg cells (Liu et al., 2006, Ha, 2009). The

over-expression of FoxP3 is associated with the suppressive function of Treg cells (Prabhala

et al., 2006). However, there are problems associated with the use of FoxP3 as a marker for

Treg cells. Law et al. (2009) demonstrated that the use of different clones of FoxP3, different

fluorochromes attached to the FoxP3 antibody and different fixation/permeabilisation

methods can give different results.

Glucocorticoid-induced tumour necrosis factor (TNF) -receptor-related (GITR) protein

(McHugh et al., 2002) and cytotoxic lymphocyte-associated antigen-4 (CTLA-4 [CD152])

(Takahashi et al., 2000) are reported to be expressed on Treg cells and important for their

development and function (Beyer and Schultze, 2006, Ha, 2009). Other surface antigens that

are purported to be expressed on Treg cells are CD103, CD62L, CD69, CD134, CD71, CD54

and CD45RA (Belkaid and Rouse, 2005, Prabhala et al., 2006).

17

It has been shown by Liu et al. (2006) that the expression of CD127, the IL-7 receptor, is

down-regulated in a subset of CD4+ cells that are also FoxP3+ and that these cells are highly

suppressive. Hence, Treg cells can now be defined as CD4+CD25hiCD127lo/-. This is a

significant advancement in the identification of Treg cells. As FoxP3 is an intracellular

protein, until recently, its use was limited to enumeration of Treg cells and functional

studies, as well as in vitro expansions, had not been possible (Liu et al., 2006). However,

with the use of the surface biomarker CD127 to identify and isolate the Treg cell population,

further studies on Treg cells can be performed.

1.5.4 Treg cells and cancer

Tumours may develop ways to tactically exploit the immune system by evading or

suppressing the immune response. Two principal mechanisms for immune evasion by

tumours have been described. In the first mechanism, the tumour may directly suppress an

immune response by releasing factors like TGF- or by down-regulating major

histocompatibility complex (MHC) class I molecules (Pardoll, 2002). The second mechanism

may be by secreting inhibitory cytokines causing T cell anergy, depletion, or inhibition of the

T cell response (Bourgeois et al., 2002, Joshua et al., 2008).

The role of Treg cells in malignancies is currently an area of intense research interest and

the subject of many reviews. Increasing evidence suggests that high Treg cell levels are

found in the tumour microenvironment, leading to the hypothesis that Treg cells play a

crucial role in tumour immunity (Beyer and Schultze, 2006, Kryczek et al., 2007). A study by

Woo et al. (2001) reported an elevated number of CD4+CD25hi Treg cells in tumour

infiltrating lymphocytes (TILs) in non-small cell lung cancer and ovarian cancer. An increase

in the prevalence of CD4+CD25hi Treg cells in the tumour microenvironment and PB of

patients with breast and pancreatic cancer has also been observed (Liyanage et al., 2002). A

higher proportion of CD4+CD25hi Treg cells has been demonstrated in the PB of patients with

gastrointestinal malignancies (Sasada et al., 2003, Kono et al., 2006), as well as in melanoma

(Javia and Rosenberg, 2003, Viguier et al., 2004). It has also been suggested that poor

prognosis is related to a higher prevalence of Treg cells in patients with gastric carcinoma

(Sasada et al., 2003, Kono et al., 2006) and TILs of patients with advanced disease have a

18

significantly higher percentage of Treg cells compared to early stage disease (Ichihara et al.,

2003, Kono et al., 2006). Treg cells are also purported to be expanded in ovarian cancer

(Curiel et al., 2004), squamous cell carcinoma of the head and neck (Schaefer et al., 2005)

and hepatocellular carcinoma (Ormandy et al., 2005). Overall, it appears that Treg cell

numbers are elevated in the majority of solid tumours investigated and that the increased

percentage of Treg cells infers a poor prognosis.

1.5.5 Treg cells and haematological malignancies

In haematological malignancies, because the malignant cells are often also immune cells,

the interaction between Treg cells and malignant cells may differ compared to solid

tumours. In chronic lymphocytic leukaemia (CLL), an increase in the number of Treg cells in

PB has been reported and this appears to correlate with a poor prognosis (Beyer et al.,

2005, Motta et al., 2005) and treatment with lenalidomide and fludarabine appears to

reduce Treg cell numbers in patients with CLL (Beyer et al., 2005, Idler et al., 2010).

Increased frequency of Treg cells has also been observed in acute myeloid leukaemia (AML)

(Wang et al., 2005, Szczepanski et al., 2009), Hodgkin disease (Marshall et al., 2004) and

non-Hodgkin lymphoma (Yang et al., 2006). In patients with AML, a lower percentage of

circulating Treg cells at diagnosis correlated with better response to induction

chemotherapy (Szczepanski et al., 2009). Conversely, in follicular lymphoma, it has been

demonstrated that higher frequencies of Treg cells correlate with a better prognosis (Álvaro

et al., 2006, Carreras et al., 2006, Lee et al., 2006), possible due to either the direct or

indirect suppression of Treg cells upon B cell proliferation. Karube et al. (2004) report that

Treg cells are down-regulated in adult T-cell leukaemia/lymphoma (ATLL), however, in ATLL,

the tumour cells have been reported to be FoxP3 positive and may function as Treg cells

(Yano et al., 2007).

1.5.6 Treg cells and GvHD

It has been reported that Treg cells suppress GvHD in animal models, however, there are

conflicting reports on the impact of Treg cells on GvHD following SCT. Rezvani et al. (2006)

have reported that there is a greater risk of developing GvHD in patients who receive SCT if

19

they have lower absolute numbers of CD4+FoxP3+ cells. Also, Brunstein et al. (2011) have

suggested that Treg cells inhibit the development of GvHD. Recently, it has been

demonstrated that the infusion of Treg cells at the time of transplantation can preserve the

graft versus tumour effect and at the same time suppress the GvHD incidence and lethality

(Taylor et al., 2002, Edinger, 2009, Colonna et al., 2011). Cohen et al. (2002) have

demonstrated that Treg cells regulate GvHD and removal of these cells will accelerate the

disease. Furthermore, the addition of ex vivo expanded Treg cells at the time of grafting has

been shown to significantly reduce the incidence of GvHD (Cohen et al., 2002). This concept

has resulted in a series of clinical trials, the first of which are now published (Brunstein et al.,

2011, Colonna et al., 2011, Di Ianni et al., 2011), demonstrating the infusion of Treg cells

prevents GvHD in the absence of post transplantation immunosuppression.

Based on current knowledge, it is possible that Treg cells could potentially be used as a

therapeutic agent in preventing GvHD in patients with MM undergoing SCT. The potential

for Treg cells to attenuate GvHD is currently being investigated in both animal models and in

humans (Taylor et al., 2002, Taylor et al., 2004). Treg cells may have the ability to facilitate

engraftment (Shevach, 2002) and reduce the risk of GvHD after allogeneic transplantation

(Sakaguchi et al., 1995, Baecher-Allan et al., 2001, Brunstein et al., 2011).

1.5.7 Controversy

The role of Treg cells in MM remains highly controversial with the existence of conflicting

literature in regards to Treg cell number and function in MM, as shown in Table 1.2. This is

presumably due to the aforementioned problems with Treg cell identification as well as

patient heterogeneity, particularly with treatment.

20

Table 1.2: Comparison of studies of Treg identification, enumeration and function in MM.

*PB: Peripheral blood. BM**: Bone Marrow. Len#: Lenalidomide. Pom^: Pomalidomide.

Reference Treg cell identification

Treg cell frequency in PB*

Suppressive function of Treg cell

Other

Banerjee et al. (2006)

CD4+CD25+FoxP3+ Not studied

Not studied DC injection stimulates Treg cell expansion

Beyer and Schultze (2006)

CD4+CD25+FoxP3+

Foglietta et al. (2006)

CD4+CD25hi Normal Normal

Normal Treg cell number in BM**

Noonan et al. (2006)

CD4+CD25+ Not studied

Not studied Reduced Treg cell in BM** compared to *PB

Prabhala et al. (2006)

CD4+CD25+

Th17 cells increased

Minnema et al. (2007)

CD4+FoxP3+ Not studied

Not studied Len# increased Treg cell

Quach et al. (2008)

CD4+CD25hiCD127lo

FoxP3+ Not studied Len# increases

Treg cell number

Chiarenza et al. (2009)

CD4+CD25+FoxP3+ Not studied Len# increased Treg cell

Feyler et al. (2009)

CD4+CD25+FoxP3+ Normal

Galustian et al. (2009)

CD4+CD25+CTLA4+

FoxP3+ Not

studied Not studied Len# and Pom^

inhibit Treg cell function

Raja et al. (2009)

CD4+CD25hiCD127lo

FoxP3+ Not studied Frequency of

BM** Treg cell proportional to increase severity of disease

Gupta et al. (2011)

CD4+CD25++CD127lo

FoxP3+ Normal Treg cells

increasing after treatment with IMiD

Carter, et al. (2012)

CD4+CD25++FoxP3+ Not studied

Clave et al. (2013)

CD4+CD25++CD127lo Not studied

21

1.6 Th17 cells and their cytokines

The range of effector CD4+ T cell lineages has been expanded with the description of a novel

CD4 sub-population called Th17 cells (Harrington et al., 2005, Kryczek et al., 2007). Th17

cells are characterised by their production of IL-17A, IL-17F, IL-21, IL-22 and IL-26. Th17 cells

also express chemokine receptor CCR6 and its ligand, CCL20 (Fouser et al., 2008, Ji and

Zhang, 2010). They have been observed in both humans and mice and are responsible for

the clearance of extracellular pathogens, including bacteria and fungi, and are pathogenic in

inflammation and autoimmunity (Fouser et al., 2008, Ji and Zhang, 2010, Prabhala et al.,

2010).

1.6.1 Th17 cell development and differentiation

The predominance of Th17 cells in PB and the conditions for Th17 cell development are not

completely understood (Ji and Zhang, 2010). Some groups have reported that IL-6 and TGF-

are responsible for the differentiation of Th17 cells (Bettelli et al., 2006), with contributions

by IL-1, IL-23, TNF- , and other cytokines (Fouser et al., 2008, Noonan et al., 2010).

However, subsequent studies have reported that IL-6, IL-1 and IL-23 were sufficient to

promote Th17 differentiation and that TGF- was not needed (Ji and Zhang, 2010).

Maintenance and expansion of Th17 cells is driven by IL-21 and IL-23. The key

transcriptional regulator for Th17 cytokines is retinoid-related orphan receptor (ROR) - t

(Ivanov et al., 2006, Fouser et al., 2008) and it is the over-expression of ROR- t that

promotes Th17 cell differentiation and up-regulation of IL-17 (Ji and Zhang, 2010). However,

other transcription factors critical in the regulation of Th17 cell development are ROR- and

signal transducer and activator of transcription 3 (STAT3) (Fouser et al., 2008, Ji and Zhang,

2010). Studies have shown that mutation in the STAT3 gene is detrimental to Th17 cell

differentiation and IL-17 production (Fouser et al., 2008, Prabhala et al., 2010) and that IL-2

reduces Th17 cell differentiation (Kryczek et al., 2007).

1.6.2 The IL-17 cytokine

IL-17, initially termed cytotoxic T lymphocyte-associated antigen-8 (CTLA-8), is a T-cell

derived pro-inflammatory cytokine (Rouvier et al., 1993, Nakae et al., 2002). It is a 32-kDa

22

structural homologue of the cystine knot family of proteins with intra-chain disulphide

bonds (Hymowitz et al., 2001, Prabhala et al., 2010). The structural motif of IL-17 is closely

related to TGF- , nerve growth factor, platelet derived growth factor and bone

morphogenetic proteins (Prabhala et al., 2010). The majority of IL-17 is produced by

activated Th17 cells, however, variable amounts of IL-17 are also produced by CD8 cells

(Shin et al., 1999, Ferretti et al., 2003), T cells (Lockhart et al., 2006), NK cells and

neutrophils (Ferretti et al., 2003, Prabhala et al., 2010).

1.6.3 Th17 cells in cancer

The prevalence and function of Th17 cells in cancer is unknown. The role of Th17 cells in

tumour growth and development, particularly in haematological malignancies, is

undoubtedly complex and remains controversial.

Wróbel et al. (2003) report that the concentration of IL-17 in serum is not significantly

elevated in patients with AML. However, a subsequent study by Wu et al. (2009)

demonstrated an increased frequency of Th17 cells in the PB, and a corresponding elevation

in IL-17 levels, in AML. The study also reported a reduction in Th17 levels once a patient had

achieved complete remission after chemotherapy (Wu et al., 2009). In non-Hodgkin

lymphoma, a low frequency of Th17 cells was observed in the tumour microenvironment,

and it was proposed that lymphoma B cells may favour Treg cells and down-regulate the

Th17 cell generation (Yang et al., 2009). In ovarian cancer, a high percentage of Th17 cells

was observed at the tumour site, but a low percentage of Th17 cells was present in PB

(Miyahara et al., 2008). Further, Su et al. (2010) demonstrated elevated Th17 cell frequency

in TILs from melanoma, breast and colon cancer. An increase in Th17 cells, and Th17-related

cytokines, has been reported in PB and tumour draining lymph nodes and this correlated

with clinical stage (Zhang et al., 2008). It has been suggested that Th17 cells may promote

tumour progression and these cells have been proposed as a potential prognostic marker in

hepatocellular carcinoma (Zhang et al., 2009). Studies have also shown that in prostate

cancer, Th17 cell numbers are inversely proportional to results of the Gleason test, a

prostate cancer staging and prognostic test (Sfanos et al., 2008).

23

1.7 The role of the cytokines TGF- and IL-12 in cancer

Cellular homeostasis requires an intricate balance between cells and the cytokine milieu.

The role of cytokines in the tumour microenvironment is an area that is under investigation

and the possibility of utilising cytokines to improve cancer immunotherapy has been

proposed. The effects of TGF- and IL-12 on immunomodulation (Brunda et al., 1993,

Cerwenka et al., 1999, Chen et al., 2003, King et al., 2005, Tahara et al., 1995, Xu et al.,

2010) suggested that these may be two of the most important immunomodulatory

cytokines and thus have been investigated in this study.

1.7.1 TGF-

TGF- is a widely distributed cytokine which has been the subject of many studies involving

cancer. In brief TGF- , a prototypical member of the TGF- superfamily, is a multifunctional

polypeptide that regulates multiple cellular processes including cellular proliferation,

differentiation, survival and adhesion. There are three known isoforms of TGF- called TGF-

1, TGF- 2, and TGF- 3. It has been reported that TGF- , which may be present in the

tumour microenvironment to prevent premalignant progression, may, however, be

eventually exploited by malignant cells (Massagué, 2008). It has been purported that cancer

cells increase the production of TGF- to allow the cells to be more invasive (Blobe et al.,

2000). Matsumoto and Abe (2011) have suggested that TGF- may, in part, suppress BM

formation in MM and inhibition of TGF- signalling may enhance bone formation and

suppress MM cell growth. Growing evidence suggests that TGF- acts as a tumour-derived

immunosuppressor, prompting interest in TGF- as a therapeutic target.

1.7.2 IL-12

The IL-12 family of cytokines, which includes IL-12, IL-23, IL-27 and IL-35, is produced by

APCs, such as DC and macrophages. IL-12 was formerly termed cytotoxic lymphocyte

maturation factor (CLMF) (Stern et al., 1990) or natural killer cell stimulatory factor (NKSF)

(Kobayashi et al., 1989). The cytokine is required for the differentiation to Th1 cells and is a

crucial mediator of inflammatory disease. The critical role of IL-12 in immune regulation has

drawn attention to its potential use for cancer immunotherapy. Studies have investigated

24

the anti-tumour and anti-metastatic effect of IL-12 in a range of murine tumour models such

as melanomas, sarcomas, mammary, colon and renal carcinomas (Brunda et al., 1993,

Colombo and Trinchieri, 2002). IL-12 induces IFN- production by NK cells and T cells, which

is thought to be essential for the anti-tumour effects of IL-12 (Car et al., 1995, Ryffel, 1997,

Wigginton et al., 2001). Administration of IL-12 has been shown to reduce or eradicate

tumour development in tumour bearing mice (Tahara and Lotze, 1995, Tahara et al., 1995).

However, clinical trials have shown limited efficacy of IL-12 therapy, due to systemic toxicity

including splenomegaly, leucopenia and gastrointestinal toxicity (Ryffel, 1997, Car et al.,

1999). Xu et al. (2010) have suggested administering IL-12 with Treg-depleting antibodies to

minimise toxicity and improve therapeutic efficacy.

1.8 Background to the project

The role of Treg and Th17 cells in MM remains highly controversial. As indicated in Table

1.2, the role of Treg cells in MM is not well defined and conflicting literature exists with

respect to Treg cell number and function. In addition, the proportion of Th17 cells in PB of

patients with MM has not been determined although these patients are reported to have a

higher proportion of Th17 cells in their BM compared to BM of patients with the pre-

neoplastic condition MGUS (Dhodapkar et al., 2008). Alexandrakis et al. (2006) have

suggested that elevated serum IL-17 levels may correlate with higher disease stage in MM.

The reciprocal relationship between Treg and Th17 cells has been reported in the literature,

however the Treg/Th17 cell balance has not been described in cancer or in MM in particular.

An imbalance of Treg and Th17 cells may play a crucial role in the dysfunction observed in

MM. Further work in this area of research will aid in the understanding of the aetiology and

pathogenesis of MM and to enhance progress towards the eventual development of a cure

for this disease.

1.9 Hypothesis

The hypothesis of this study is that dysfunction of normal immune control occurs in patients

with malignancies such as MM and that restoration of immune regulation may be beneficial

to overall survival.

25

1.10 Aims of the project

There are conflicting reports about the quantity and function of Treg and Th17 cells in MM.

Thus the overall aim of this study is to clarify any discrepancies in the literature regarding

Treg and Th17 cell number and function in MM. Furthermore, the immunomodulatory

mechanisms utilised by MM cells to cause immune dysfunction were studied.

The overall aim of the project will be achieved through completion of the following specific

aims:

1. Enumeration of Treg and Th17 cell number in the PB of patients with MM, in normal

individuals and in patients with other B cell malignancies.

2. Determination of the importance of the Treg/Th17 cell balance on survival of

patients with MM.

3. Determination the functional status of Treg cells from patients with MM and from

healthy control individuals.

4. Determination of the effect of specific factors derived from tumour cells (e.g. TGF- ,

IL-12) on Treg cell function.

26

CHAPTER TWO

MATERIALS AND METHODS

27

2.1 Participants

Whole blood was collected from patients with MM, MGUS and WM who attend the

haematology department of Royal Prince Alfred Hospital (RPAH) as part of a routine check.

In this study the mean age was 67 ± 10 yrs, 70 ± 12 yrs, and 79 ± 6 yrs for patients with MM,

MGUS and WM respectively. The patients with MM were dichotomised based on 10 year

survival since diagnosis. Patients surviving for greater than 10 years since diagnosis had a

mean age of 73 ± 9 yrs. An age-matched control group was also recruited for the study with

a mean age of 53 ± 21 yrs. Ethics approval was granted by the Sydney South West Area

Health Service Human Research Ethics Committee (RPAH Zone, Protocol X07-0201).

2.2 Sample collections

2.2.1 Blood sample collection

Venepuncture was performed by trained haematology nursing staff in RPAH. PB was

collected in a 4.0mL BD Vacutainer® blood collection tube containing either 5.4mg K2

Ethylenediamine tetra-acetic acid (EDTA) or lithium heparin (BD Biosciences, New Jersey,

USA) as an anticoagulant. The PB was inverted 3 times immediately upon collection. Using

an automated haematology analyser (Cell-Dyn Sapphire; Abbott Diagnostics, Illinois, USA), a

full blood count was performed on each sample on the day of collection. Samples not tested

on the day of collection were stored overnight at 4°C and tested within 48hrs of collection.

For patients on therapy, blood samples were collected immediately prior to the next course

of therapy to minimise the effects of therapy on the blood cells being investigated.

2.2.2 BM collection

BM was aspirated from consenting patients with MM by trained haematology registrars.

The BM was aspirated into a 20mL tube (Sarstedt, Nümbrecht, Germany) containing 1mL of

Hanks Balanced Salt Solution (HBSS) (Thermo Scientific, Massachusetts, USA), supplemented

with foetal calf serum (FCS) (SAFC® Biosciences, Sigma-Aldrich Missouri, USA) and heparin

(Pfizer, New South Wales, Australia). 20mL of sterile RPMI-10 (described in section 2.3.3)

28

was added to the BM aspirate of samples not tested on the day of collection. BM aspirates

were stored at 4°C and tested within 24hrs of collection.

2.3 Methodology for preparation of common reagents

2.3.1 Preparation of ammonium chloride lysing reagent

Ammonium chloride lysing reagent was prepared by dissolving 4.15g ammonium chloride

(Scharlau Chemie, Spain), 0.45g Sodium hydrogen carbonate (VWR, New Jersey, USA) and

0.018g Na2 EDTA.2H2O (Searle, Nebraska, USA) in 500mL Milli-Q™ water. The Milli-Q™ water

was obtained from the Synergy UV filtering system (Millipore, Madison, USA). The pH of the

lysing reagent was determined immediately before use, using a Beckman 44 pH meter

(Beckman, Fullerton, USA). The desired pH range was between 7.25-7.30.

2.3.2 Preparation of phosphate buffered saline

10 phosphate buffered saline (PBS) (Dulbecco A) tablets (Oxoid Ltd., Hampshire, England)

were dissolved in 1L of Milli-Q™ water (Millipore). The manufacturer’s recommended pH

range was 7.30 ± 0.02. pH was optimised by the addition of either 1N HCl or 1N NaOH where

required.

This PBS method was used for all tests unless stated otherwise.

2.3.3 Preparation of Roswell Park Memorial Institute -10 medium

The Roswell Park Memorial Institute (RPMI) -10 medium was prepared under sterile

conditions in an Airpure biological safety cabinet class II (Westinghouse, Australia).

RPMI-1640 cell culture medium with 20mM HEPES, without L-glutamine, sodium

bicarbonate (MP Biomedicals, LLC, Ohio, USA) was supplemented with 10% (v/v) foetal

bovine serum (SAFC Biosciences, Australia).

The RPMI-10 medium was stored at 4°C until required.

29

2.3.4 Preparation of RPMI cell culture medium

The RPMI cell culture medium was prepared under sterile conditions in an Airpure biological

safety cabinet class II (Westinghouse). The following were added to 500mL of RPMI-1640

cell culture medium containing 20mM HEPES, without L-glutamine or sodium bicarbonate

(MP Biomedicals):

50mL heat inactivated, irradiated foetal bovine serum (SAFC Biosciences)

5mL of 1.0mM sodium pyruvate (Invitrogen™, Beijing, China)

5mL of 2mM L-glutamine, 10 l/mL Penicillin/ Streptomycin antibiotics (Invitrogen™,

California, USA)

2mL of 1.5g/L sodium bicarbonate (Invitrogen™, Mulgrave, Australia )

The RPMI cell culture medium was stored at 4°C until required.

2.3.5 Preparation of MACSBeads

Anti-CD2, CD3, CD28 beads were used to stimulate the proliferation of T cells.

For preparation of the MACSbead suspension, a T cell activation/expansion kit containing

anti-biotin MACSiBead (Miltenyi Biotec, Bergisch Gladbach, Germany) was used, in

accordance with the manufacturer’s instructions. 0.4mL CD2-Biotin, 0.4mL CD3-Biotin, and

0.4mL CD28-Biotin, were added to 2mL MACS® anti-biotin MACSiBead™ particles and spun

using a Clements mixer (H.I. Clements Pty Ltd, North Ryde, NSW) for 2hrs at 4°C, yielding a

final concentration of 1x105 beads/ L.

2.3.6 Preparation of carboxyfluorescein succinimidyl ester stain

Carboxyfluorescein succinimidyl ester (CFSE) is a fluorescent cell permeable dye that can be

used to monitor the proliferation of cells. CFSE passively diffuses into cells and binds to

intracellular molecules where it is cleaved by intracellular esterases to yield a fluorescent

dye. As CFSE stained cells divide the daughter cells retain half the original CFSE fluorescence.

30

CFSE

Coun

t

When analysed by flow cytometry (Figure 2.1), each decreasing peak in fluorescence

corresponds to each cell division.

Figure 2.1: Flow cytometric analysis of a CFSE stained cell population The histogram shows the proliferation of a CFSE stained cell population. As each cell divides

the daughter cells retain half the amount of CFSE, seen by the sequential reduction in

fluorescence. Each peak is proportional to the number of cell division. 6 divisions are

displayed in the above flow plot.

The CellTrace™ CFSE Cell Proliferation Kit (Invitrogen™, Mulgrave, Australia) was used to

prepare CFSE. 1 vial of CFSE containing 50 g of lyophilised powder was dissolved in 50 L of

dimethyl sulfoxide (DMSO) (Sigma Aldrich, St Louis, MO, USA). The suspension of CFSE at a

concentration of 5mM was then divided into 2 L aliquots and stored in a Sanyo MDF-U50V

freezer (Sanyo, Japan) at -80°C until required.

Immediately prior to use, the 2 L aliquot was resuspended into 48 L of 37oC RPMI cell

culture medium.

31

2.4 General techniques

2.4.1 Cell wash

Cell washes were generally with 2mL PBS for 2min at 300g, unless otherwise stated.

2.4.2 Isolation of mononuclear cells using Ficoll-Paque density gradient centrifugation

Ficoll-Paque Plus (GE Healthcare, Uppsala, Sweden) was used for the isolation of peripheral

blood mononuclear cells (PBMC) for analysis of Th17 cells and bone marrow mononuclear

cells (BMMC) for identification of BM Treg cells. Ficoll-Paque is a density gradient

centrifugation medium for separation of PB or BM into its constituent components. After

centrifugation of PB or BM with Ficoll-Paque, shown in Figure 2.2B, the bottom layer

contains aggregated erythrocytes and granulocytes; above is the Ficoll-Paque layer; the

lymphocytes, monocytes and platelets sit above the Ficoll-Paque; and the top layer contains

plasma.

Figure 2.2: Whole blood before and after centrifugation with Ficoll-Paque A. Image of diluted PB over a layer of Ficoll-Paque prior to centrifugation. B. After

centrifugation using Ficoll-Paque the PB is separated into its constituent components.

A B

Diluted whole blood

Ficoll-Paque

Peripheral blood mononuclear cells

Plasma

Ficoll-Paque, granulocytes and Red blood cells

32

PB was diluted with PBS (Oxoid, England) at a 1:1 ratio and, using a plastic transfer pipette

(Samco Scientific, California, USA), was gently layered over the Ficoll-Paque in an 8mL

polycarbonate round bottom tube (Sarstedt, South Australia, Australia) at a ratio of 3mL

Ficoll-Paque to 4mL diluted PB. The Ficoll-Paque and diluted PB were then centrifuged at

360g for 15min at 20oC (room temperature; RT) using a Spintron GT10S-V3 centrifuge

(Spintron, Australia). The PBMC layer was collected using a plastic transfer pipette and

transferred into another 8mL polycarbonate tube. The PBMC were then washed in

approximately 7mL PBS and centrifuged at 360g for 10min at RT. The remaining cell pellet

was then resuspended in 1mL RPMI-10.

33

2.5 Monoclonal antibodies used during experimentation

Table 2.1: List of monoclonal antibodies (mAb) used during experimentation mAb name/ specificity

Fluorophore Isotype Clone Company details

CD3 FITC Ms IgG1, SK7 BD, San Jose, CA, USA

CD3 PerCP-Cy5.5 Ms IgG1, SK7 BD

CD4 APC Ms IgG1, SK3 BD

CD4 PE-Cy7 Ms IgG1, SK3 BD

CD4 PerCP-Cy5.5 Ms IgG1, SK3 BD

CD25 APC Ms IgG1, 2A3 BD

CD45RA FITC Ms IgG2b, HI100 BD

CD45RO FITC Ms IgG2a, UCHL1 BD

CD127 PE Ms IgG1, hIL-7R-M21 BD

IL-17 PE Ms IgG1 41802 R&D Systems, Minneapolis, MN, USA

FoxP3 Alexa Fluor® 647 (AF-647)

Ms IgG1, 206D Biolegend, San Diego, CA, USA

FoxP3 PE Ms IgG1 259D/C7 BD

Isotype control APC Ms IgG2a, G155-178 BD

Isotype control FITC Ms IgG1, 1 BD

Isotype control PE Ms IgG1, X40 BD

Isotype control PerCP-Cy™ 5.5 Ms IgG2a, G155-178 BD

FITC: Fluorescein isothiocyanate, PE: Phycoerythrin, PerCP-Cy™ 5.5: Peridinin chlorophyll

protein conjugate with a cyanine-5.5 protein, APC: Allophycocyanin, PE-Cy7: Phycoerythrin

conjugated with cyanine-7 protein

34

2.6 Assay for identification of Treg cells

2.6.1 Cell staining for flow cytometric identification of CD127lo/- Treg cells

100 L of PB from participants was lysed with 2mL lysing reagent (described in section 2.3.1)

for 10min at RT. The lysed blood was then centrifuged for 2min at 300g in an EBA 21

centrifuge (Hettich Zentrifugen, Tuttlingen, Germany) and the supernatant, containing lysed

red cells, was discarded. The cell wash was repeated with PBS to remove any residual lyse

buffer or red cells. The remaining leukocytes were then incubated at RT for 15min with

fluorescent labelled mAb (Table 2.2) in the dark. Following the incubation period, the cells

were washed and the supernatant was discarded. The resulting cell pellet was resuspended

in 0.5mL of PBS. Analysis of the sample was performed on the BD FACSAria II using DIVA

software for acquisition (BD Biosciences, San Jose, CA).

Table 2.2: Fluorescent antibodies used for the flow cytometric identification of Treg cells using CD127

mAb Fluorophore Volume per test

Tube ID

CD45RA CD45RO

CD4 PerCP-Cy5.5 20 L

CD25 APC 5 L

CD127 PE 10 L

CD45RA FITC 20 L

CD45RO FITC 20 L

: indicates mAb in use

2.6.2 Preparation of PBMC Treg cell identification using FoxP3

PBMCs, were fluorescently labelled with the surface antibodies against CD3, CD4, and CD25

(see Table 2.3) for 15min at RT in the dark. The cells were then washed and the supernatant

discarded. 250 L CytoFix/CytoPerm BD™ Fixation/Permeabilisation solution (Becton

Dickinson, SanDiego, CA, USA) was added to the PBMC and incubated for 20min at RT in the

dark. The labelled PBMC were washed once with the Cytofix/Cytoperm washing solution

35

and again with PBS. The suspension of permeabilised cells was incubated with fluorescent-

labelled intracellular antibody against FoxP3 for 30min in the dark at 4°C. The cells were

washed and resuspended in 0.5mL of PBS for acquisition on the BD FACSAria II.

Table 2.3: Fluorescent antibodies used for the flow cytometric identification of Treg cells using FoxP3

mAb Fluorophore Volume per test

Tube ID

FoxP3 AF-647

FoxP3 PE

CD3 PerCP-Cy5.5 20 L

CD4 PE-Cy7 5 L

CD25 FITC 5 L

FoxP3 AlexaFluor647 5 L

FoxP3 PE 10 L

: indicates mAb in use

2.6.3 Preparation of BM Treg cells for flow cytometric analysis

100 L of BMMC suspension was incubated for 15min at RT with mAb in the dark, as per

Table 2.4. The cells were then washed in PBS and the supernatant was discarded. The

resulting cell pellet was resuspended in 0.5mL of PBS. Analysis of the sample was performed

on the BD FACSAria II using DIVA software (BD Bioscience) for acquisition.

36

Table 2.4: Fluorescent antibodies used for the flow cytometric identification of BM Treg cells

mAb Fluorophore Volume per test

CD3 PerCP-Cy5.5 20 L

CD4 PE-Cy7 5 L

CD25 APC 5 L

CD127 PE 10 L

2.7 Methodology for the identification of Th17 cells in PB

2.7.1 Preparation of Th17 cells for flow cytometric analysis

1x106 PBMCs, obtained using the Ficoll-Paque method described in section 2.4.2, was

diluted up to 1mL with RPMI-10. The cell suspension was then incubated with 50ng/mL

phorbol 12-myristate 13-acetate (PMA) (Sigma Aldrich, Missouri, USA) and 750ng/mL

Ionomycin calcium salt, from Streptomyces conglobatus (Sigma Aldrich, Israel) in a Ratek

WB14 water bath (Ratek Instruments, Australia) at 37°C for 2.5hrs. The use of PMA and

Ionomycin stimulates the production of cytokines within cells. The release of cytokines from

the cell was blocked by the addition of 10 L of a 1/10 solution of Brefeldin A diluted with

PBS (Becton Dickinson, USA) followed by a further 2.5hrs incubation in a 37°C water bath.

The cell suspension was then washed in 3mL PBS (Oxoid, England) for 5min at 300g using an

EBA 21 centrifuge (Hettich Zentrifugen). The supernatant was discarded and the resulting

cells were incubated with surface antibodies, as per Table 2.5 for 15min at RT in the dark.

The cells were then washed in 2mL PBS for 3min at 300g. The supernatant was discarded

and the cells were incubated with 250 L CytoFix/CytoPerm BD™ Fixation/Permeabilisation

solution (Becton Dickinson) for 20min at RT in the dark. After washing twice in PBS, the

permeabilised cells were divided into two separate 5mL, 12mm x 75mm, polystyrene round-

bottom tubes (BD Bioscience) and incubated with intracellular antibodies to either human

IL-17 (R&D Systems) or mouse IgG1 (BD Bioscience) for 30min at RT in the dark. The cells

37

were washed and resuspended in 0.5mL PBS. The cell suspension was analysed on the BD

FACSAria II.

Table 2.5: Fluorescent antibodies used to identify Th17 cells for analysis by flow cytometry

mAb Fluorophore Volume Isotype control

Th17 test

Surface mAb

CD3 PerCP-Cy5.5 20 L

CD4 APC 5 L

Intracellular mAb

Mouse IgG1 PE 10 L

IL-17 PE 10 L

: indicates mAb in use

2.8 Treg cell functional assay using CFSE

2.8.1 Fluorescence-activated cell sorting of PBMC

Fluorescence-activated cell sorting (FACS) is a process that allows cells of interest to be

physically separated from a heterogeneous population. All blood samples were prepared

under sterile conditions in an Airpure biological safety cabinet class II (Westinghouse).

PB was diluted at a 1:1 ratio with sterile PBS pH7.4 without calcium chloride and magnesium

chloride (GIBCO Invitrogen, Grand Island, New York, USA). PBMC were obtained by Ficoll-

Paque separation (as previously described in section 2.4.2). Briefly, 30mL of diluted whole

blood was layered over 20mL of Ficoll-Paque in a sterile Blue Max 50mL polypropylene

conical tube (BD Bioscience). The tube was then centrifuged for 25min at 360g, without

brakes, using a Spintron GT10S-V3 centrifuge (Spintron, Australia). The PBMC were

harvested into a separate tube and washed in 45mL sterile PBS (GIBCO Invitrogen) by

centrifugation for 15min at 360g. The supernatant was discarded and the cells were stained

with antibodies, as per Table 2.6, followed by a 30min incubation period, on ice, in the dark.

The PBMC were then washed in sterile PBS (GIBCO Invitrogen) for 5min at 300g, the

supernatant was discarded and the cells were resuspended in 1mL sterile PBS (GIBCO

38

Invitrogen). The suspension was filtered into a 5mL polystyrene round bottom tube with a

cell strainer cap (BD Bioscience) and then sorted using the BD FACSAria II. Both Treg cells

(referred to hereafter as ‘purified Treg’); defined as CD3+CD4+CD25hiCD127lo/-) and

remaining CD3+ cells (referred to hereafter as ‘target’) were sorted into a 5mL polystyrene

round bottom tube (BD Bioscience). Once the sorting process was complete, the cells were

washed for 5min at 300g. The supernatant was discarded and the cells were resuspended in

1mL RPMI cell culture medium.

Table 2.6: Fluorescent antibodies used to identify Treg cells for flow cytometric sorting

Ab Specificity Fluorophore Volume per test

CD3 FITC 40 L

CD4 PE-Cy7 10 L

CD25 APC 10 L

CD127 PE 20 L

2.8.2 FACS purity test

Once the FACS was complete, 10 L of the sorted target cells and purified Treg cell

suspension were each diluted in 200 L of PBS and analysed on the BD FACSAria II. In this

study the criteria for a successful FACS process was to yield a purity of greater than 90%. If

this was not achieved the experiment would be abandoned.

2.8.3 CFSE staining of target cells

2mM CFSE was added to the sorted target cells suspension and incubated at 37°C for 10min.

To quench the reaction, 3mL of cold RPMI cell culture medium was added to the cell

suspension and placed on ice for 5min. The cell suspension was then spun at 300g for 5min

and the supernatant was discarded. The CFSE labelled cells were resuspended in 1mL RPMI

cell culture medium.

39

2.8.4 Cell count

A cell count was performed on both the target and purified Treg cell suspensions. The

sorted cells were counted on a Neubauer Improved Hemocytometer counting chamber

(American Optical Corporation, New York, USA) using a Carl Zeiss light microscope (Carl

Zeiss, New York, USA).

The following formula was used to determine the number of cells in the suspension:

N= number of cells counted in 25 squares of the centre grid

Volume= Volume of cell suspension (mL)

2.8.5 Preparation of the MACSbead suspension

5 L of MACSbeads from stock (equivalent to 5x105 beads) (described in section 2.3.5) was

suspended in 1mL RPMI cell culture medium in a 2mL microtube (Sarstedt, Numbrecht,

Germany) followed by a 5min centrifugation at 300g using an Eppendorf centrifuge 5415D

(Eppendorf AG, Hamburg, Germany) to remove any preservatives. The supernatant was

discarded and the MACSbeads were resuspended in 1mL RPMI cell culture medium.

2.8.6 Cell culture of Treg and CFSE stained target cell

All samples were plated under sterile conditions in an Airpure biological safety cabinet class

II (Westinghouse). The cells were placed in a 96 well U bottom tissue culture plate with low

evaporation lid (Becton Dickinson Labware, Franklin Lakes, NJ, USA) and cultured for four

days at 37oC with 5% CO2 in a NuAire NU-5100E DH Automatic CO2 Direct incubator (Nuaire,

Plymouth, MN, USA).

As displayed in Figure 2.3, to monitor normal proliferation of target cells, a cell culture well

containing CFSE stained target cells was cultured with MACSbeads at a 1:1 ratio. To

determine the suppressive capabilities of Treg cells, a 1:1 ratio of purified Treg cells to

target cells was added to a well containing MACSbeads. As a control, target cells were

Total number of cells/mL = N X 104 X Volume (mL)

40

cultured without MACSbeads and purified Treg cells, as this would demonstrate where the

non-proliferating target cells were positioned on the flow cytometric histogram. Another

control well was set up with MACSbeads and target cells at a 1:2 ratio.

RPMI cell culture medium was added to each well for a final concentration of 25x104

cells/mL.

Figure 2.3: Cell culture plate set up for investigation of Treg cell function

This figure displays the set-up of the 96 well cell culture plates, described in section 2.8.6, to

investigate the suppressive activity of Treg cells. The cell culture wells A and D were used as

a control to monitor proliferation without beads (well A) and with twice the cells (well D).

Cell culture wells B and C were used to evaluate the suppressive activity of Treg cells.

*Please note a 96 well U bottom tissue culture plate was used.

Target cells No beads

Target cells with beads &

Treg cells 1:1:1

Target cells & beads

2:1

Target cells & beads

1:1

A B C D

41

2.8.7 Analysis of the CFSE assay after culture

After 4 days, the cultured cells were transferred from the cell culture plate to a 5mL

polystyrene round-bottom tube (BD Bioscience). The cells were then washed in 2mL PBS at

300g for 3min in an EBA 21 centrifuge (Hettich Zentrifugen). The supernatant was discarded

and the cells were incubated for 15min with the antibodies detailed in Table 2.7. FITC

conjugated antibodies could not be used simultaneously with CFSE due to the similar

excitation and emission properties. The cells were washed and the supernatant discarded.

The remaining cells were resuspended in 0.5mL PBS. Flow cytometric acquisition of the

sample was performed on a BD FACSAria II (BD Bioscience).

Table 2.7: Fluorescent antibodies used for analysis of the CFSE assay by flow cytometry

Ab Specificity Fluorophore Volume per test

CD3 PerCP-Cy5.5 10 L

CD4 PE-Cy7 5 L

CD25 APC 5 L

2.9 Evaluation of the impact of cytokines (IL-12 and TGF- ) and anti-TGF- on Treg cells

To determine the effect of IL-12, TGF- , and anti-TGF- on Treg cells, the same methodology

described in section 2.8.1 to 2.8.5 was followed for FACS, CFSE staining of the target cell

suspension and preparation of MACSbead suspension.

2.9.1 IL-12 preparation

Recombinant human (rh)- IL-12 (R&D Systems, Minneapolis, MN, USA) was stored at -20oC

at a concentration of 10 g/mL. Stock solution was diluted with RPMI cell culture medium to

a final working concentration of 100ng/mL per well.

42

2.9.2 rhTGF- 1 preparation

rhTGF- 1 (R&D Systems, Minneapolis, MN, USA) was stored at -20oC at a concentration of

1 g/mL. Stock solution was diluted with RPMI cell culture medium to a final working

concentration of 100pg/mL per well.

2.9.3 Anti TGF- preparation

Monoclonal anti-human latency associated peptide (LAP) (TGF- 1) antibody (R&D Systems,

Minneapolis, MN, USA) was stored at -20oC at a concentration of 500 g/mL. Stock solution

was diluted with RPMI cell culture medium to a final working concentration of 10ng/mL per

well.

2.9.4 Plating of the cell culture wells with addition of cytokines

All samples were plated under sterile conditions in an Airpure biological safety cabinet class

II (Westinghouse). The cells were placed in a 96 well U bottom tissue culture plate with low

evaporation lid (Becton Dickinson Labware, USA) and cultured for 4 days at 37oC with 5%

CO2 in a NuAire NU-5100E DH Automatic CO2 Direct incubator (Nuaire, USA).

As demonstrated in Figure 2.4, the CFSE stained target cells were cultured with MACSbeads

at a 1:1 ratio. To determine the normal suppressive capabilities of Treg cells, equal numbers

of purified Treg cells were co-cultured with MACSbead stimulated target cells. To determine

the effect of cytokines on Treg cell suppression, either 100ng/mL of IL-12, 100pg/mL TGF-

or 10ng/mL anti-TGF- was added to the cell culture wells with and without purified Treg

cells.

As a proliferation control, target cells were cultured without either MACSbeads or purified

Treg cells. Another control well was set up with beads and target cells at a 1:2 ratio.

43

Figure 2.4: Cell culture plate set up to evaluate the effect of IL-12, TGF- , and anti-TGF-

on the function of Treg cells

This figure displays the set-up of the cell culture plates, described in section 2.9.4, to

determine the effect of IL-12, TGF- , and anti-TGF- on the suppressive activity of Treg cells.

Cell culture wells A and D were used as a control to monitor proliferation without

MACSbeads (well A) and with twice the cells (well D). The cell culture wells in column B and

C were used to evaluate the suppressive activity of Treg cells and the effect of IL-12, TGF- ,

and anti-TGF- on the suppressive activity of Treg cells. ppppppppppppppppppppppppppp

*Please note a 96 well U bottom tissue culture plate was used.

A B C D Target cells No beads

CTarget cells with beads & Treg cells

1:1:1

Target cells & beads

2:1

Target cells & beads

1:1

Target cells & beads

1:1 with rhIL-12

Target cells with beads &

Treg cells 1:1:1

with rhIL-12

Target cells & beads

1:1 with rhTGF-

Target cells with beads &

Treg cells 1:1:1 with rhTGF-

Target cells & beads

1:1 with anti-TGF-

Target cells with beads &

Treg cells 1:1:1 with anti-TGF-

44

2.10 Statistics and analysis

FlowJo software (TreeStar Inc, USA) Version 7.6.4 (PC compatible) was used for analysis of

flow cytometric data.

Statistical analysis of data was performed on GraphPad Prism software versions 5 (GraphPad

Inc, San Diego, California, USA) and calculated using a two-tailed unpaired Students T-test

when of equal variance. For statistical analysis of the Treg cell proportion in BM and PB

comparison a paired Student T-test was performed. The Kaplan-Meier survival curve was

used for statistical comparison between high and normal Treg/Th17 ratio, this was assessed

using a chi-square analysis. P values less than 0.05 were deemed statistically significant. All

experimental data are shown as a mean values ± standard error of mean (SEM) in the text

and the mean is also indicated in the figures.

45

CHAPTER THREE

RESULTS

46

3.1 Overview

The enumeration of Treg and Th17 cells in the PB of patients with MM has been

controversial. Treg and Th17 cells are thought to provide immunological homeostasis and

any imbalance in the number and function of these cells could impact immune control and

potential clinical recovery of patients with MM. The discrepancies that exist in the literature

regarding Treg and Th17 cell number and function in MM, summarised in Table 1.2,

substantiate the need for more research in this field. Therefore, the quantity of Treg and

Th17 cells in the PB of patients with MM, WM, MGUS and the normal control group was

examined using a CD4, CD25 and CD127 staining method for identifying Treg cells (Figure

3.2). Further, a comparison of Treg cell function between normal individuals and patients

with MM was also investigated. In addition, the effect of cytokine treatment on the function

of Treg cells is also detailed. These studies support the proposal that it is the homeostatic

balance between Treg and Th17 cells which is more important than the actual number of

either cell type alone.

3.2 Treg cells in monoclonal gammopathies

3.2.1 Identification of Treg cells using CD127

In order to enumerate Treg cells in the PB of normal subjects and patients with monoclonal

gammopathies, Treg cells needed to be clearly identified amongst other leucocytes. For this

portion of the study, Treg cells were defined as CD4+CD25hiCD127lo/-. The flow cytometric

gating strategy used to obtain Treg cell proportions of lymphocytes is displayed in Figure 3.2

using a representative normal PB sample. Using the size (forward scatter; FSC) and

granularity (side scatter; SSC) parameters, the lymphocyte population was able to be

identified amongst the leucocytes, as depicted in Figure 3.2A. Anti-CD4-PerCP-Cy5.5 was

then used to isolate the T helper cells (Figure 3.2B), of which Treg cells are a subset. To

distinguish the Treg cell population amongst the CD4+ cells, anti-CD25-APC and anti-CD127-

PE were used. The Treg cell population was then identified according to high surface

expression of CD25 and low or negative expression of CD127 (Figure 3.2C). Relevant isotype

controls were performed during the optimisation process, as shown in Figure 3.1, but this

was not required once the assay had been established.

47

SSC

-APC

G1 isotype control for PE G1 isotype control for FITC

G1 isotype control for APC G1 isotype control for PerCP-Cy5.5

Figure 3.1: Isotype control for each fluorochrome used in the Treg cell assay

An anti-mouse IgG1 antibody conjugated to relevant fluorochromes was used as an isotype

control to determine the level of non-specific staining and auto-fluorescence. This was

performed during the optimisation process, but was not required once the assay had been

established.

FSC

Coun

t -FITC -PE

-PerCP-Cy5.5

48

Figure 3.2: Flow cytometry results from a representative normal subject showing the gating strategy for identification of Treg cells in PB

The figure presents the flow cytometric analysis used to identify Treg cells. CD4+CD25hiCD127lo/- expression was used to enumerate Treg cell

numbers in PB. In the Treg assay, cells were analysed after a gating strategy that included a lymphocyte gate on the FSC/SSC scattergram and

the identification of the CD4+ population (Figure 3.2B). Treg cells were then identified from a scattergram which identified a population of CD4+

cells which were CD25hi and CD127lo/- (Figure 3.2C). The results were expressed as a percentage of the CD4+ cells and also as an absolute

number, calculated using the lymphocyte count. CD

127

A B C

CD4 CD25 SSC

FSC

49

3.2.2 Identification of Treg cells using FoxP3

Early research into Treg cells used FoxP3, a transcriptional repressor, to identify Treg cell

populations, defined as CD4+CD25hiFoxP3+. FoxP3 is required in Treg cells as it is associated

with its suppressive capabilities. However, there are problems associated with the use of

FoxP3 as a marker for Treg cells. The use of FoxP3 limited research to only the identification

of Treg cells and functional assays could not be performed, as this required permeabilisation

of the cells, therefore destroying the cells of interest. In addition, Law et al. (2009)

demonstrated that FoxP3 results were dependent on the clones, fluorochromes attached

and fixation/permeabilisation methods used. These difficulties were also encountered

during early optimisation experiments in this project whereby in some cases it was difficult

to achieve a clear separation between FoxP3 positive and FoxP3 negative populations using

flow cytometry (Figure 3.3). Additionally, identifying the CD25hi population was found to be

subjective. Therefore, due to the complications and limitations associated with the use of

FoxP3, the lack of CD127 expression was preferred as the primary marker for identification

of Treg cells, and FoxP3 was omitted from the Treg cell identification panel.

To determine whether Treg cells, defined as CD4+CD25hiCD127lo/- expressed FoxP3, the Treg

cells were sorted from PBMC, using a BD FACSAria II flow cytometric sorter. FoxP3

expression was determined on the purified Treg cells and remaining Treg depleted CD3+ T

cells. As expected, the CD127lo/- Treg cells were found to express intracellular FoxP3 at a

greater level of fluorescence than other CD4+ cells (Figure 3.4).

50

FoxP3 AlexaFluor647

CD25 FITC

CD4

PEcy

7

CD4 PECy7

CD3

Perc

Pcy5

.5

SSC

FSC

FoxP3 PE

Figure 3.3: Flow cytometry results from a representative normal subject showing the gating strategy for identification of PB Treg cells using FoxP3 The figure presents the flow cytometric gating strategy to identify Treg cells defined as

CD3+CD4+CD25hiFoxP3+. Treg cells were analysed after a gating strategy that included a

lymphocyte gate on the FSC/SSC scattergram and the identification of the CD3+CD4+

population. The CD4+CD25hi (Treg) and CD4+CD25- T cells were analysed for their FoxP3

expression. The overlay histogram (bottom row) highlights the difference in FoxP3

expression in Treg cells and other CD4+CD25- T cells, as well as revealing the difference in

expression when using different fluorochrome labelled FoxP3 antibodies.

CD4+CD25- cells

CD4+CD25+ cells

51

Coun

t

Figure 3.4: Comparison of FoxP3 expression on sorted CD25hiCD127lo/- Treg cells compared to the Treg depleted T cells

The figure demonstrates the flow cytometric gating strategy used to determine whether the CD4+CD25hiCD127lo/- Treg cells also expressed

FoxP3. After sorting PBMC for Treg cells, both the isolated Treg cells and the Treg depleted cells were examined for FoxP3 expression. Row A

displays the FoxP3 expression of Treg depleted CD4+ T cells. Row B displays the FoxP3 expression results of the purified Treg cells suspension.

The lymphocyte population was gated based on FSC/SSC characteristics. CD4+ cells were gated based on the constitutive expression of CD3 and

CD4. These CD3+CD4+ cells were then gated and analysed for their FoxP3 expression. The overlay histogram (far right) highlights the difference

in FoxP3 expression in Treg cells and other T cells.

Treg depleted

Purified Treg cells CD

4

FSC

CD3 FoxP3

A

B

SSC

52

3.2.3 Enumeration of Treg cells in PB

There are conflicting results in the literature with regards to PB Treg cell proportions in

patients with MM and in normal subjects. Early studies used CD25hiFoxP3+ cells to identify

Treg cells, and the discrepancies in the literature may be due to the difficulties associated

with FoxP3 staining, discussed in section 3.2.2. Through the use of CD127, the limitations

and difficulties were diminished due to the simplified methodology and reduced processing

required for the identification of Treg cells. Therefore, the use of CD127 provided clearer

indication of Treg cell quantitation in PB.

Flow cytometry was used to enumerate Treg cells in PB of patients with MM (n=32), MGUS

(n=20), WM (n=13) and the normal controls (n=36). After analysis of the flow cytometry

results, Treg cell numbers, as a proportion of CD4+ lymphocytes, were attained. The

lymphocyte count obtained from the Cell-Dyn Sapphire haematology analyser of RPAH, was

used in conjunction with the CD4+ cell percentage obtained by flow cytometry to calculate

the absolute CD4 count. In addition the Treg cell percentage was obtained by flow

cytometry to determine the absolute Treg cell count.

The percentage of Treg cells in the CD4+ compartment (Figure 3.5A) and absolute number

(Figure 3.5B) in PB were statistically compared between the different patient groups and the

normal controls. The mean percentage of Treg cells in the PB of patients with MM was

8.9±0.6% and was significantly elevated (P<0.01) compared to the mean percentage of Treg

cells of the normal controls at 6.5±0.4%. On average, patients with MGUS appear to have a

higher percentage of PB Treg cells compared to normal controls (7.5±0.8%), however this

was not found to be significant (P=0.24). In addition, no significant difference was observed

between the percentage of Treg cells in the control group and patients with WM (6.0±0.5%;

P=0.48). Interestingly, however, the absolute number of Treg cells in PB revealed a different

outcome between the patients with WM and the control group; suggesting that there is a

significant difference in absolute number of Treg cells in patients with WM [(3.0±0.6) x107/L]

compared to the normal control cohort [(6.4±0.7) x107/L; P<0.01]. A significantly lower

absolute number of Treg cells was also observed in patients with MM [(3.2±0.4) x107/L;

Absolute Treg cells = Absolute CD4 count X Per cent Treg cells of CD4+ cells

53

P<0.01] compared to healthy individuals. However, absolute Treg cell numbers in patients

with MGUS [(4.3±0.8) x107/L; P=0.06] did not differ significantly from those of the normal

control cohort.

54

A

B

Normal MM MGUS WM0

5

10

15

20 * *NS

Abso

lute

Tre

g (x

107 /L

)

Figure 3.5: Enumeration of PB Treg cells in patients with monoclonal gammopathies and normal control individuals A. The data are presented as a percentage of Treg cells, as part of the CD4+ cell

compartment, in PB of normal individuals (n=36) and patients with MM (n=32), MGUS

(n=20) and WM (n=13) respectively. B. The data demonstrate the absolute number of Treg

cells in PB of normal individuals (n=39) and patients with MM (n=32), MGUS (n=20) and WM

(n=13) respectively. The mean of each cohort is represented by the horizontal black line. For

analysis, an unpaired two-tailed t-test was performed with a comparison between the

outcomes of patients with monoclonal gammopathies compared to the normal control.

*P<0.05, NS- Not significant.

Normal MM MGUS WM0

5

10

15

20* NS NS

% T

reg

cells

of C

D4+ T

cel

ls

55

3.2.4 Phenotypic analysis of Treg cells

To determine whether the Treg cells were of a naïve or memory cell phenotype, cell surface

expression of CD45RA and CD45RO were determined using anti-CD45RA-FITC and anti-

CD45RO-FITC. Naïve T cells have surface expression of CD45RA whereas CD45RO is

expressed on memory T cells. Displayed in Figure 3.6 is a representative flow cytometric

analysis sequence used to determine the naïve or memory phenotype of PB Treg cells. It

appears that the majority of PB Treg cells were of a memory cell phenotype (Figure 3.7) and

this was consistent amongst both normal individuals and those with monoclonal

gammopathies.

The nature of Treg cells was determined in the PB of the normal cohort (n=26) and

compared to the PB of patients with MM (n=31), MGUS (n=20) and WM (n=13). In

comparison to the normal cohort (16.3±2.1%), the proportion of naïve Treg cells in PB of

patients with MM (12.0±1.9%; P=0.13), MGUS (17±2.4%; P=0.83), and WM (9.8±2.2%;

P=0.06) was not significantly different. Although no significant difference was observed in

CD45RA+ Treg cells proportions in patients with MM they appeared to have a significantly

higher proportion of CD45RO+, memory T cell phenotype, compared to normal subjects

(85.9±1.8; 76.7±2.5 respectively P<0.01). Similarly, Treg cells in patients with WM had a

significantly higher proportion of memory Treg cells compared to the normal cohort

(86.4±2.1%; P=0.02). No significant difference was observed in the CD45RO+ Treg cell

proportion between patients with MGUS and the control group (73.4±4.0%; P=0.47).

56

A

B

Figure 3.6: Flow cytometry results showing CD45RA and CD45RO expression on Treg cells in PB of a representative normal subject PB Treg cells from patients with monoclonal gammopathies and normal individuals was

analysed for their expression of CD45RA+ (naïve phenotype) or CD45RO+ (memory

phenotype). The gating strategy displayed in Figure 3.2 was followed for the identification of

Treg cells. A. Displays a representative result of CD45RA expression on PB Treg cells. B

Displays a representative example of CD45RO expression on PB Treg cells. The majority of

Treg cells appear to be of the memory cell phenotype.

57

CD45RA CD45RO CD45RA CD45RO CD45RA CD45RO CD45RA CD45RO0

20

40

60

80

100

Normal MM MGUS WM

* *%

of t

otal

Tre

g ce

lls

Figure 3.7: Expression levels of CD45RA and CD45RO on Treg cells in PB of patients with monoclonal gammopathies and normal individuals The data are presented as a percentage of CD45RA or CD45RO expression on Treg cells in PB of patients with MM (n=31), MGUS (n=20) and

WM (n=13) compared to the normal cohort. The results of the experiment are presented as a mean ± SEM, shown by the error bars. For

analysis, an unpaired two-tailed t-test was performed with a comparison between the results of patients with monoclonal gammopathies

compared to the normal control. The CD45RO expression on PB Treg cells was found to be significantly higher in patients with MM and WM

compared to normal individuals. *P<0.05

58

3.2.5 Effect of IMiD treatment on Treg cell proportions

In patients with MM, the effect of IMiD therapy on Treg cell proportion was investigated.

Patients undergoing lenalidomide treatment (7.9±0.9%; P=0.89) did not appear to have a

significantly different percentage of Treg cells compared with patients who had not

undergone IMiD therapy. However, a trend for an increase in the proportion of Treg cells

was observed in patients with MM treated with thalidomide (12.7±2.1%) compared to

patients with MM not treated with IMiD therapy (8.1±0.7%; P=0.02), this was found to be

significant as shown in Figure 3.8. For a more conclusive analysis however, additional

patients would need to be tested. Difficulty in testing more patients treated with

thalidomide was encountered, as the use of thalidomide therapy was being phased out and

replaced with lenalidomide. The effect of IMiD therapy on the absolute number of Treg cells

was not analysed due to the limited sample number.

In accordance with previous publications (Chiarenza et al., 2009, Quach et al.,

2008,Minnema et al., 2007)

59

no-IMiD Thalidomide Lenalidomide0

5

10

15

20

NS*

% T

reg

cells

of C

D4+ T

cel

ls

Figure 3.8: Effect of IMiD treatment on PB Treg cell proportions in patients with MM The data are presented as a percentage of Treg cells in the CD4+ compartment of PB in

patients treated with thalidomide (n=4), lenalidomide (n=7) and patients not treated with

IMiDs (n=21). The black horizontal line represents the mean of each cohort. For analysis, an

unpaired two-tailed t-test was performed with a comparison between the percentage of PB

Treg cell in patients with IMiD therapy and those that had not undergone treatment with

IMiDs. The results indicate a significant increase in PB Treg cells of MM patients treated with

thalidomide compared to patients not treated with IMiDs. However, no significant change

was observed in PB Treg cells of patients treated with lenalidomide. *P<0.05, NS- Not

significant.

60

3.2.6 MM staging and Treg cell quantitation

Currently, MM staging is based on the ISS criteria, which uses two biomarkers, serum

albumin and 2microglobulin levels. Patient data were categorised based on the ISS stage to

determine whether there was any correlation between different stages of MM and the

percentage of Treg cells in PB. As evident in Figure 3.9, no correlation between percentage

of Treg cells in PB and MM ISS stage was observed. The percentage of Treg cells in PB of

patients with stage I (8.9±1.0%) MM did not differ significantly from patients with stage II

(9.2±1.2%) or stage III (8.5±1.4%) MM. Further, the absolute number of Treg cells did not

differ between the stages.

3.2.7 Enumeration of Treg cells in BM versus PB

The BM is the site of tumour development in MM, therefore the study aimed to establish

the Treg cell proportion in matched samples of BM compared to PB in patients with MM.

The PB and BM Treg cell proportions were enumerated and statistically compared as shown

in Figure 3.10. BM Treg cells were identified as CD3+CD4+CD25hiCD127lo/-, using the same

gating strategy as shown in Figure 3.21. The CD3+CD4+ cells were distinguished from the

lymphocytes, of which the CD25hiCD127lo/- cells were identified as Treg cells. The BM of

patients with MM comprise 9.7±1.2% of CD4+ T cells and was significantly elevated (P=0.02)

compared to the relative proportion of Treg cells in PB which was observed to be 6.7±1.4%

of CD4+ T cells.

61

Stage I Stage II Stage III0

5

10

15

20NS NS

% T

reg

cells

of C

D4+ T

cel

ls

Figure 3.9: Comparison of the percentage of PB Treg cells and the different stages of MM The data are presented as a percentage of Treg cells as part of the CD4+ cell compartment in

PB of patients with stage I (n=14), stage II (n=9) and stage III (n=9) MM. Staging of patients

with MM was based on the ISS criteria. The results of the experiment are presented as a

mean represented by the black horizontal line. For analysis, an unpaired two-tailed t-test

was performed with a comparison between the outcomes of patients with stage II and stage

III MM compared to patients with stage I MM. *P<0.05, NS- Not significant.

62

Figure 3.10: Comparison of Treg cell numbers in the BM and PB of patients with MM The data are presented as a percentage of Treg cells as part of the CD4+ T cell compartment

in matched BM and PB of patients with MM (n=9). The horizontal black line represents the

mean of each cohort. For analysis, a paired two-tailed t-test was performed with a

comparison between proportions of BM Treg cells to PB Treg cells of the same patients.

*P<0.05, NS- Not significant.

Blood Bone Marrow0

5

10

15

*

% T

reg

of C

D3+ C

D4+ s

ubse

t

63

3.2.8 Treg cells in patients with MM who have survived more than 10 years

To expand our current knowledge of the PB composition of patients with MM and establish

the significance of Treg cell quantity in MM, this study investigated the difference in the

proportion and numbers of PB Treg cells in patients surviving with MM for greater than 10

years since diagnosis. The MM cohort was divided into less than 10 years and greater than

10 years according to the number of years since diagnosis. The data demonstrate, in Figure

3.11, that patients with MM appear to have a significantly higher percentage of Treg cells in

PB compared to normal individuals (8.9±0.6%; 6.5±0.4% respectively; P=0.003). However,

patients who have survived for 10 or more years with MM (5.5±0.8%) were found to have a

significantly lower proportion of Treg cells in PB compared to their MM counterparts who

have survived less than 10 years (P=0.005). The data imply that the proportion of Treg cells

in PB of 10 year survivors appears to normalise to the same proportion as the normal

control group (P=0.21). This same phenomenon was also observed with the absolute PB

Treg cell count of 10 year survivors. Interestingly, substantially lower absolute numbers of

Treg cells were observed in patients with MM for less than 10 years [(3.2±0.4) x107/L]

compared to normal individuals [(6.4±0.7) x107/L; P<0.01]. However, the Treg cell count of

patients who have survived with MM for more than 10 years [(5.1±1.1) x107/L] did not

significantly differ from the normal cohort (P=0.37). Intriguingly, the PB Treg cell count of

MM survivors of greater than 10 years was observed to be significantly higher than those

with MM for less than 10 years (P=0.04).

64

Normal <10Y MM >10Y MM0

5

10

15

20 * *NS

% T

reg

cells

of C

D4+ c

ells

Figure 3.11: Relative Treg cell numbers in PB of normal subjects, patients with MM for less than 10 years and MM patients who have survived for 10 or more years since diagnosis The data are presented as a percentage of Treg cells as part of the CD4+ cell compartment in

PB with a distinction between MM patients who have survived with MM greater than (n=13)

or less than (n=32) 10 years since diagnosis. This was compared to the normal cohort

(n=36). The results of the experiment are presented as a mean, represented by the

horizontal black lines. For analysis, an unpaired two-tailed t-test was performed with a

comparison between the Treg cell proportion of patients with different length of survival

with MM and normal controls. *P<0.05, NS- Not significant.

65

Normal <10Y MM >10Y MM0

5

10

15

20* *

NS

Abso

lute

Tre

g (x

107 /L

)

Figure 3.12: Absolute number of PB Treg cells in normal subjects, patients with MM for less than 10 years and patients who have survived 10 or more years with MM The data are presented as absolute numbers of Treg cells with a distinction between

patients with MM who have survived greater than (n=13) or less than (n=32) 10 years

compared to the normal control cohort (n=36). The horizontal black line represents the

mean of each cohort. For analysis, an unpaired two-tailed t-test was performed with a

comparison between the absolute Treg cells count of patients with different length of

survival with MM compared to the normal cohort. *P<0.05, NS- Not significant.

66

3.3 Th17 cells in MM and other monoclonal gammopathies

Th17 cells are responsible for the clearance of extracellular pathogens and are pathogenic in

inflammation and autoimmunity (Fouser et al., 2008, Ji and Zhang, 2010, Prabhala et al.,

2010). This study examined the quantity of Th17 cells in PB of patients with MM, WM,

MGUS and in normal individuals using a CD3, CD4, and IL-17 phenotype after PMA

stimulation to identify Th17 cells (Figure 3.13). The proportions of PB Th17 cells of the CD4+

compartment were statistically compared to the normal cohort (Figure 3.14). Patients were

categorised based on the type of monoclonal gammopathy, the stage of MM and length of

survival with MM.

3.3.1 Identification of Th17 cells in PB

Th17 cells were identified according to surface expression of CD3, using anti-CD3-FITC, CD4,

using anti-CD4-PE-Cy7 and intracellular expression of IL-17, using anti-IL-17- PE. Figure 3.13

depicts the flow cytometric results from a representative normal PB sample. For each

patient a mouse anti-IgG1-PE isotype control was used. The results for the isotype control

assisted in determining where the negative –PE expressing population was located, and

therefore set the region for the positive IL-17-PE expressing population. In addition, the

isotype control provided an indication of the level of non-specific binding and these results

were considered when determining the IL-17 expression for each patient.

67

CD4

CD3 SSC

FSC

Figure 3.13: Flow cytometric results from a PB sample of a representative normal subject showing the gating strategy for the identification of Th17 cells The figure portrays the flow cytometric gating strategy to identify Th17 cells. CD3+CD4+IL-17+ expression was used to enumerate PB Th17 cell

numbers. In the Th17 assay, PBMC were analysed after a gating strategy that included a lymphocyte gate on the FSC/SSC scattergram and the

identification of the IL-17 expression on the CD3+CD4+ population. An anti-mouse IgG1 isotype control was used to assist with setting the

gating for the PE fluorochrome.

Th17

G1

68

3.3.2 Enumeration of Th17 cells in PB

As illustrated in Figure 3.14, a significant decrease was observed in the mean percentage of

Th17 cells in PB of patients with MM (0.7±0.1%), which includes both treated and untreated

patients, compared to the normal cohort (2.0±0.6%; p=0.03). However, the mean

percentage of Th17 cells in MGUS (2.2±0.6%) and WM (1.1±0.2%) patients was not

significantly different from the control group. Similarly, the absolute number of Th17 cells in

PB was significantly lower in patients with MM [(0.6±0.1) x107/L] compared to the control

group [(2.9±0.7) x107/L] (p=0.004). However, no significant variation was observed in

absolute Th17 cell numbers in patients with WM [(1.1±0.3) x107/L] and MGUS [(2.0±0.5)

x107/L] when compared to the normal cohort.

69

A

B

Normal MM MGUS WM0

5

10

15

20 * NS NS

Abso

lute

Th1

7 (x

107 /L

)

Figure 3.14: Relative Th17 cells number in the PB of patients with monoclonal gammopathies and normal controls A. Data are presented as a percentage of Th17 cells in the CD4+ compartment of PB of

normal individuals (n=20) and patients with MM (n=22), WM (n=12) and MGUS (n=15)

respectively. B. The data demonstrate the absolute number of Th17 cells in PB of normal

individuals (n=23) and patients with MM (n=21), WM (n=12), and MGUS (n=14) respectively.

The results of the experiment are presented as a mean, represented by the horizontal black

lines. For analysis an unpaired two-tailed t-test was performed with a comparison between

the proportions of patients with monoclonal gammopathies compared to the normal

cohort. *P<0.05, NS- Not significant.

Normal MM MGUS WM0

5

10

15 * NS NS

%Th

17 c

ells

of C

D4+ c

ells

70

3.3.3 Th17 cells and stage of MM

To determine whether there was a correlation between the proportion of Th17 cells in PB

and different stages of MM, the tested patients with MM were stratified into different

stages using the ISS criteria. As shown in Figure 3.15, no correlation between the

percentages of Th17 cells in PB and MM stage was observed. The percentage of Th17 cells in

patients with stage I (0.6±0.1%) MM did not differ significantly from patients with stage II

(1.1±0.3%) or stage III (0.6 ±0.2%) MM.

Stage I Stage II Stage III0.0

0.5

1.0

1.5

2.0

%Th

17 c

ells

of C

D4+ c

ells

NS NS

Figure 3.15: Comparison of the percentage of Th17 cells of the CD4+ compartment in PB and different stages of MM The data are presented as a percentage of Th17 cells as part of the CD4+ cell compartment

of PB in patients with stage I (n=10), stage II (n=4) and stage III (n=8) MM. Staging of

patients was based on the international staging system criteria. The horizontal black line

represents the mean of each cohort. For analysis an unpaired two-tailed t-test was

performed with a comparison between patients with stage II and stage III MM compared to

patients with stage I MM. *P<0.05, NS- Not significant.

71

3.3.4 Th17 cells in MM patients who have survived more than 10 years

In an attempt to elucidate the significance of Th17 cells in MM, the relative proportion of

Th17 cells and absolute numbers was investigated in patients with MM. The data depicted

in Figure 3.16 includes patients surviving with MM with minimal disease progression for 10

or more years since diagnosis. The results indicate that patients who have survived greater

than 10 years with MM have a higher proportion of PB Th17 cells compared to their MM

counterparts who have experienced the malignancy for less than 10 years (3.3±0.9% and

0.72±0.1% respectively; P<0.01). Although a significant decrease is observed in Th17 cell

proportions in patients with MM for less than 10 years compared to the normal cohort

(2.0±0.6; P=0.03), this was not the same for the 10-year MM survivors. The data indicates

that 10-year survivors of MM possess similar proportions of PB Th17 cells to that of the

normal subjects (P=0.21). The same trend was also found for the absolute Th17 cell counts

in 10-year survivors of MM. The Th17 cell count in patients surviving greater than 10 years

did not differ significantly (P=0.29) from those of the normal cohort [(1.9±0.5 x107/L) and

(2.9±0.7 x107/L) respectively], although patients with MM who have not yet achieved long

term survival possessed a significantly lower (P<0.01) number of PB Th17 cells (0.58±0.1

x107/L) compared to the normal group. The PB Th17 cell number in 10-year survivors of MM

was also significantly higher than their MM counterparts of less than 10-years survival

(P<0.01).

72

A

B

Figure 3.16: Enumeration of Th17 cells in PB of patients with MM and normal subjects A. The data are presented as a percentage of Th17 cells in the CD4+ compartment of PB of

normal individuals (n=20), and patients with MM who have survived less than (n=22) or

greater than (n=16) 10 years. B. The data demonstrate the absolute number of Th17 cells in

PB of normal individuals (n=23), and patients with MM who have survived less than (n=21)

or greater than (n=16) 10 years. The horizontal black line represents the mean of each

cohort. For analysis an unpaired two-tailed t-test was performed comparing the outcomes

of MM patients with different lengths of survival and to the normal cohort. *P<0.05, NS- Not

significant.

Normal <10Y MM >10Y MM0

5

10

15

20**

NS

Abso

lute

Th1

7 (x

107 /L

)

Normal <10Y MM > 10Y MM0

5

10

15**

NS

%Th

17 c

ells

of C

D4+ c

ells

73

3.4 Treg and Th17 cell equilibrium

As Treg cells and Th17 cells have opposing functions, it is imperative that a balance of the

two cell subsets is maintained in PB. A possible cause for degeneration in haematological

malignancies may be that cell subset proportions may deviate from the homeostatic

equilibrium and cause an imbalance in the cellular environment of PB. To test this, the

Treg/Th17 ratio was determined by dividing the percentage of Treg cells by the percentage

of Th17 cells found in PB for each patient.

3.4.1 Treg/Th17 cell ratio in monoclonal gammopathies

The data, shown in Figure 3.17, suggests that patients with MM have a significantly higher

Treg/Th17 ratio (16.1±2.4) compared to the normal cohort (6.6±1.0; p<0.005). Conversely,

the PB Treg/Th17 cell ratio of patients with WM (7.0±1.0; p=0.79) or MGUS (4.9±0.5;

p=0.23) did not differ significantly from that of the control group. A high Treg/Th17 cell ratio

in patients with MM may contribute to a suppression of the immune control of the

malignancy.

3.4.2 Effect of a high Treg/Th17 cell ratio on overall survival in MM

Interestingly, the data presented in Figure 3.18 indicates that Treg/Th17 ratio of patients

who have survived greater than 10 years with MM (7.04±2.47) was similar to that of the

normal cohort (6.57±1.01; P=0.84), whereas patients who had not reached 10 year survival

with MM had a significant increase in their Treg/Th17 cell ratio (16.1±2.38; p=0.01).

However, a bimodal distribution is observed in the greater than 10 year survivor cohort

(Figure 3.18) particularly, 4 patients exhibiting a higher Treg/Th17 ratio. In addition, the

patients with MM, dichotomised into high and normal Treg/Th17 cell ratio, was further

analysed to assess whether the Treg/Th17 cell ratio influenced the survival time of patients.

As demonstrated in Figure 3.19, the patients with a high Treg/Th17 cell ratio appear to have

Treg/ Th17 cell ratio = % Treg cells % Th17 cells

74

a significantly lower overall survival compared to patients with a normal Treg/Th17 cell ratio

(p=0.025). Thus, taken together, this shows the clinical significance of the Treg/Th17 cell

ratio in MM.

Normal MM MGUS WM0

20

40

60 * NS NS

Treg

to T

h17

cell

ratio

Figure 3.17: Treg/Th17 cell ratio in patients with monoclonal gammopathies and normal individuals The Treg/Th17 cell ratio of patients with MM (n=20), WM (n=12) and MGUS (n=15) was

determined using the percentage values of these cells in PB and compared to the ratio of

the control group (n=28). For analysis, an unpaired two-tailed t-test was performed with a

comparison between the Treg/Th17 cell ratio of patients with monoclonal gammopathies

compared to the normal cohort. *P<0.05, NS- Not significant.

75

Normal <10Y MM >10Y MM0

10

20

30

40 **NS

Treg

to T

h17

cell

ratio

Figure 3.18: Treg/Th17 cell ratio in 10 year MM survivors The Treg/Th17 cell ratio of patients with MM for less than 10 years (<10Y MM; n=20) and

patients with MM for more than 10 years (>10Y MM; n=15) was determined using the

percentage values of these cells in PB and compared to the ratio of the normal control

group (n=28). For analysis, an unpaired two-tailed t-test was performed with a comparison

between the Treg/Th17 cell ratio of patients with MM compared to the normal control

group as well as comparing the different MM cohorts. *P<0.05, NS- Not significant.

76

Figure 3.19: The effect of Treg/Th17 cell ratio on overall survival of patients with MM The Kaplan-Meier curve shows the survival of patients with MM stratified by high Treg/Th17

cell ratio in PB (i.e. +2SD above the mean for patients with MM) and patients with a normal

Treg/Th17 cell ratio in PB. The data demonstrate MM patients with a high Treg/Th17 cell

ratio (dotted line) had a significantly shortened overall survival compared to patients with a

normal Treg/Th17 cell ratio (solid line). For analysis, a chi-square analysis was performed

with comparison between the high Treg/Th17 cell ratio compared to the low Treg/Th17 cell

ratio ( 2=5.1; p<0.025).

0 20 40 600

20

40

60

80

100

Months

Ove

rall

surv

ival

Normal Treg/Th17 cell ratio High Treg/Th17 cell ratio

(%)

77

3.5 Treg cell functional assay

As shown in section 3.2.3, there was a higher proportion of Treg cells in PB of patients with

MM compared to the normal group. However, there is little in the literature about the

functional status of Treg cells in MM. This information could indicate whether Treg cells are

recruited by the tumour to exploit the immune system and inhibit an immune response.

Thus either aberrant or increased numbers of Treg cells may suppress an anti-tumour

immune response. The functional status of PB Treg cells in patients with MM compared to

the normal cohort was investigated using a CFSE suppression assay. Treg cells were

identified, using the gating strategy shown in Figure 3.21, and isolated from PBMC by FACS

using a BD FACSAria II. The Treg cells were then cultured with CFSE stained target cells (Treg

depleted CD3+ T cells) and proliferation of the target T cells was monitored whilst in culture

with and without Treg cells. The percentage of proliferation was obtained by flow

cytometric analysis, and used to calculate the percentage of suppression exerted by the

Treg cells.

The suppression exerted by PB Treg cells on CD3+ target cells was statistically compared

between the Treg cells from patients with MM (n=19) and the normal control group (n=11).

The MM cohort was categorised based on the current treatment the patient was

undergoing, as treatment may have an effect on the activity of Treg cells. The patients

tested were either untreated patients (n=6) or treated with thalidomide (n=5), lenalidomide

(n=5) and bortezomib (n=3).

3.5.1 Development and optimisation of Treg cell functional assay

For optimisation of the Treg functional assay, CD3+ cells were cultured for four days, with

proliferation stimulated by anti-CD3, CD2, CD28 MACSbeads at different concentrations. A

proliferation of approximately 80% was preferred, as this was thought to provide enough

stimulation to the cells without overpowering any potential inhibition by Treg cells. Equally,

if the proliferation of cells was too low the effect of the Treg cell suppression would be

% suppression = % proliferation without Treg - % proliferation with Treg X 100 % proliferation without Treg

78

negligible. As evident in Figure 3.20, a ratio of 1:1 MACSbead to target cell provided enough

stimulation for the target cells.

Figure 3.20: Optimisation of MACSbead stimulation for the Treg functional assay As part of optimisation, different concentrations of beads were used. The figure shows the

proliferation of CD3+ ‘target’ cells using different concentrations of beads. Row A shows

0.06% proliferation with no beads. Row B displays 12.6% proliferation of target cells with

MACSbeads at a ratio of 1 MACSbead to 10 target cells. Row C presents 83.5% proliferation

of target cells with a ratio of 1 MACSbead per target cell.

FSC

CFSE SSC

A

B

C

CD4

CD3

79

3.5.2 Treg cell functional assay results

CD3+CD4+CD25hiCD127lo/- Treg cells were identified by the gating strategy illustrated in

Figure 3.21. The remaining CD3+ cells (‘target cells’) were also collected (Gate A (CD3+CD4-)

and Gate B (CD3+CD4+) of Figure 3.21). The cells were co-cultured with Treg cells for four

days and the suppression exerted upon CD3+ cells was calculated, as described in section

3.5.

Figure 3.22 shows the analysis strategy in which the percentage proliferation of T cells and

their subsets were obtained. As activated lymphocytes are increased in size, they have a

higher FSC value hence, as shown in Figure 3.22; a large gate was created to incorporate

these cells in analysis. Cells without any stimulation (without beads) were used to

determine where the non-proliferating (highest fluorescing) cells were positioned on the

flow cytometric CFSE histogram (Figure 3.22). As the cells divide the daughter cells retain

half the CFSE fluorescence, detected by flow cytometry, and each peak on the CFSE

histogram corresponds to a cell division.

3.5.3 Suppression of T cell proliferation

Untreated patients with MM had a mean suppression of 30.2±10.9% which was similar to

the normal cohort, whose mean Treg cell suppression was 31.4±4.8% (p=0.79), inferring that

Treg cells of patients with MM exhibit normal suppressive activity (Figure 3.23). However,

Treg cells of treated patients with MM exhibited varying degrees of suppression on

lymphocytes which appear to be influenced by treatment. The mean percentage of

suppression of lymphocytes exerted by PB Treg cells in thalidomide treated patients was

17.7±8.0%, which was reduced, however not significantly different from the normal cohort

(p=0.15). Similarly, patients treated with bortezomib had reduced suppressive activity of

12.0±7.7%, however this too was not observed to differ significantly from the normal cohort

(p=0.08). In contrast, patients undergoing combination treatment with lenalidomide and

dexamethasone had a mean suppression exerted by Treg cells of 68.9±6.4%, which is

significantly increased compared to the normal control (P<0.0001) and approximately 3

times more suppressive than observed with thalidomide treatment.

80

The suppression of CD4+ and CD8+ cell proliferation was similar to the suppression exerted

on T cells as a whole. Interestingly, however, Treg cells were significantly more suppressive

on CD4+ cells in the patients treated with bortezomib (p=0.04) compared to the normal

cohort, which was not observed on the CD8+ subset or on T cells as a whole. The

suppression upon CD4+ and CD8+ cells were also statistically compared in each cohort, but

were not found to be significantly different. .

81

Figure 3.21: Flow cytometry gating strategy for FACS of Treg cells in PB The figure demonstrates the flow cytometric analysis used to identify Treg cells for sorting. For the Treg cell sorting assay, cells were analysed

after a gating strategy that included a lymphocyte gate on the FSC/SSC scattergram and the identification of the CD3+CD4+ population. Gate C,

shown in red, were identified as the Treg cells, and cells in gates A and B, shown in black, were combined and identified as target cells.

82

Normal

MM

N

M

Figure 3.22: A representative flow cytometric analysis of CFSE stained CD3+ cells after 4 days culture The figure represents the flow cytometric analysis to determine the suppressive activity of Treg cells on CD3+ target cells. The FSC/SSC

scattergram, shown in the far left, was used to gate on the lymphocytes population. Activated lymphocytes are increased in size and thus have

a high FSC value; therefore a large gate was created to incorporate these cells. The second graph displays CD3 and CD4 for analysis on

individual T cell subsets and T cells as a whole. The histograms demonstrate the proliferation of the cells, whereby each peak is equivalent to

one cell division. The CFSE histograms represent proliferative responses for a patient with MM (top row) and a normal control (bottom row). In

this example the patients with MM has a percent suppression of 35.7%, whereas the normal control has a percent suppression of 22.5%.

83

Figure 3.23: Effect of treatment on Treg cells suppressive activity on lymphocytes target cells The box and whiskers graph presents the percentage of in vitro suppression by PB Treg cells, obtained by FACS, on target T cells in patients

with MM treated with Len + Dex (Lenalidomide in combination with dexamethasone; n=5), Thalidomide (n=5), Bortezomib (n=3) and untreated

patients with MM (n=6) as well as percentage of normal suppression (n=11). The results of the experiment are presented as a mean

represented by the cross, the horizontal line within the box represents the median and the whiskers identify the minimum and maximum

values for each group. For analysis, unpaired two-tailed t-test was performed with a comparison between the suppression of CD3, CD4 and

CD8 cells in patients with MM compared to the normal control. *P<0.05 when compared to the respective normal cohort.

CD3 suppression

CD4 suppression

CD8 suppression

-20

0

20

40

60

80

100

Normal Len + Dex Thalidomide Bortezomib Untreated

* * * *%

sup

pres

sion

84

3.6 Effect of cytokines on Treg cell function

Treg cells play an important role in managing an immune response. The aim of this section

of the study was to investigate whether modification of Treg cell function can be

manipulated to help restore immunological homeostasis. Altering the intensity of Treg cell

suppression may assist the immune system in its anti-tumour response.

Similar methodology to the functional assay, described in Section 3.5, was performed with

the addition of cytokines to target cells with and without Treg cells. As a control, the

cytokine or antibody was added with the target cells without the presence of Treg cells. This

was to determine the direct effect of the cytokine on the target cells. The difference

between the direct effect of the cytokine or antibody on the target and the effect via the

Treg cell was calculated using the formula given below.

This study examined the effect of IL-12, TGF- and an antibody against TGF- on the

suppressive activity of Treg cells from both patients with MM and healthy individuals. rhIL-

12 was observed to decrease the functionality of Treg cells (Figure 3.24). The overall

difference in suppression was -45.6±10.5%. In contrast, rhTGF- increased the suppression

of T cells by 40.8±25%. Anti-TGF- however, was not observed to greatly influence the

suppression of Treg cells (4.2±27%). It should be noted that these suppression assays were

technically difficult to perform, due to the long processing time and low cell numbers, and

this may account for some of the variability observed in the results. Nevertheless, the

results indicate that IL-12 can significantly inhibit (p<0.01) and TGF- can significantly

enhance (p<0.01) the suppression caused by Treg cells.

% change in % proliferation without cytokines - % proliferation with cytokines X 100 suppression % proliferation without cytokines

85

IL12 rhTGF- anti-TGF--200-100

-50

0

50

100200

% c

hang

e in

sup

pres

sion

Figure 3.24: Effect of IL-12, TGF- and anti-TGF- cytokines on the suppressive function of Treg cells The figure demonstrates the change in in vitro suppression by PB Treg cells on target cells

due to the addition of IL-12 (n=10), rhTGF- (n=5) and anti-TGF- (n=8). The mean is

represented by the black horizontal line. The results infer that the addition of IL-12 opposes

the suppression exerted by Treg cell. In contrast, the results indicate rhTGF- to enhance

the suppressive capacity of Treg cells. Anti-TGF- was not shown to greatly influence the

Treg cell suppression. The assay was technically difficult to perform and this may account for

some variability on the results.

86

CHAPTER FOUR

DISCUSSION

87

4.1 Overview

MM is an incurable malignancy characterised by the clonal expansion of abnormal plasma

cells in the BM. Recent developments in treatment, such as the use of IMiDs, proteasome

inhibitors and high dose chemotherapy with autologous SCT, have improved the quality of

life and the length of progression free and overall survival of patients suffering with the

disease. However, despite improvements in both the treatment and management of MM,

most patients eventually relapse. Immune dysfunction is an important characteristic of MM

leading to infections, progression of tumour growth and resistance to different therapies. In

order to further understand the complex malignancy that is MM and develop potential

cures, it is essential to understand the nature of any defects in the immune response in

patients with MM.

Since their first description in the early 1970s, Treg cells have engendered much attention

and in recent years and have become one of the focal points of cancer research. These cells

play a crucial role in immunological homeostasis, autoimmune diseases and possibly in

malignancies. Their role in maintaining self-tolerance leads to question whether these cells

are also responsible for the immune system tolerating tumour cells. Despite the range of

malignancies in which Treg cells are implicated, studies in this field have been highly

controversial. There are discordant findings in the literature which mainly stem from the

difficulty in the identification of these cells, the use of different criteria to identify the cells

and the selection of patients at different stages of disease and treated with different

therapies. There is also still ambiguity regarding enumeration and function of Treg cells in

patients with malignancies, particularly MM.

Another lymphocyte subset of interest in this study was Th17 cells, which are functionally

mutually exclusive to Treg cells. Their predominance and function in PB is still not

completely understood, however studies have proposed Th17 cells as a potential prognostic

indicator in patients with hepatocellular carcinoma (Zhang et al., 2009) and prostate cancer

(Sfanos et al., 2008). Their role in MM and other monoclonal gammopathies, however, still

needs to be investigated and this was addressed in this study.

To facilitate advances in treatment and potential cures for MM, the aims and hypotheses of

the study addressed the role of Treg and Th17 cells in monoclonal gammopathies.

88

Specifically, the study aimed to clarify some discrepancies in the literature with regard to

Treg and Th17 cell proportions in the PB of patients with MM, in normal individuals and in

patients with other monoclonal gammopathies. It also aimed to determine the functional

status of Treg cells in patients with MM and the impact of factors derived from tumour cells

(e.g. TGF- , IL-12) on Treg cell populations and function as well as clinical and prognostic

significance. This chapter discusses the approach, outcome and significant findings for each

aim of the study.

4.2 The role of Treg cells in monoclonal gammopathies

There are inconsistent reports on the role of Treg cells in malignancies. These discordant

findings, which have been outlined in chapter one, are due, in part, to the lack of

standardisation in Treg cell identification across different studies (Law et al., 2009). Previous

attempts to enumerate and understand Treg cell populations in PB have also been

complicated by the lack of surface markers that clearly identify Treg cells. The role of Treg

cells could be of biological and clinical relevance in patients with MM and other monoclonal

gammopathies and therefore their quantification and functional status requires clarification.

The present study was undertaken to elucidate the relative proportion and number of Treg

cells in the PB of patients with MM, WM and MGUS, and to compare with the normal

cohort. The study also aimed to establish whether Treg cell number alterations correlate

with disease severity and/or treatment regime. A further investigation to determine the

proportions of naïve and memory Treg cells was also undertaken, along with a comparison

between the percentage of Treg cells in the PB and the BM. In addition, the current study

explored the suppressive capacity of Treg cells and the effect of exogenous cytokines on

their function.

4.2.1 The phenotype and identification of Treg cells

Due to the complications associated with the use of FoxP3 expression to identify Treg cells,

FoxP3 was not used as the primary marker in the assay. The current study aimed to develop

a simplified cell staining method, defining Treg cells as CD3+CD4+CD25hiCD127lo/-, since this

89

would provide a flexible alternative to FoxP3 (Seddiki et al., 2006) by reducing variables in

identification and eliminating limitations that plagued earlier literature and prevented a

thorough phenotypic and functional assessment of Treg cells. Firstly, it was imperative to

demonstrate that the CD25hiCD127lo/- Treg cell subset was the same subset defined as

CD25hiFoxP3+ in earlier publications. To confirm these populations were the same cell type,

the expression of the FoxP3 transcriptional repressor was determined in the CD25hiCD127lo/-

population. Previous publications have demonstrated an inverse correlation between FoxP3

and CD127 (Liu et al., 2006, Seddiki et al., 2006). The CD25hiCD127lo/- Treg cells identified

herein were found to have a higher expression, and therefore a greater proportion, of FoxP3

compared to the rest of the CD4+ T cell subsets. These results are consistent with the

observation by Liu et al. (2006) that the expression of CD127 is down-regulated in a highly

suppressive subset of CD4+ cells that are also FoxP3+. Thus, it can be concluded that the

CD25hiCD127lo/- cell population possesses the phenotypic characteristics of Treg cells.

Therefore, by virtue of its cell surface expression, the preferred marker for the isolation and

identification of Treg cells in the current study was the lack of CD127.

4.2.2 Enumeration and phenotype of Treg cells in monoclonal gammopathies

Due to the inconsistent findings in previous studies (Table 1.2) regarding Treg cell

proportion and number, it is important to clarify these discrepancies in patients with MM as

well as patients with other monoclonal gammopathies.

The literature indicates that Treg cells constitute 5-10% of the CD4+ lymphocyte population

of healthy individuals (Wolf et al., 2003, Prabhala et al., 2006). This study found the mean

percentage of CD25hiCD127lo/- Treg cells in PB to be 6.5±0.4% for the normal cohort, which is

within the reported mean range. The results obtained from this study demonstrate an

expanded pool of PB Treg cells in patients with MM which corroborate previous studies

(Beyer et al., 2006, Feyler et al., 2009, Brimnes et al., 2010, Muthu Raja et al., 2012). In

terms of absolute number of Treg cells however, there was a reduced number of PB Treg

cells in patients with MM compared to the normal cohort. Albeit it is evident that patients

with MM have lower absolute Treg cell numbers than normal, this could be attributed to the

overall reduction in CD4+ lymphocytes in patients with MM (Mills and Cawley, 1983, Pilarski

90

et al., 1989, Kay et al., 2001, Raitakari et al., 2003). This decrease could also be, at least

partially, a result of treatment, such as chemotherapy, which is known to render patients

lymphopenic (Mackall et al., 1994). Moreover, this study observed an increased frequency

of PB Treg cell in patients with MGUS, however this was not found to be significant and

there was no significant difference in absolute number of Treg cells in patients with MGUS,

compared to the normal cohort. Additionally, no significant difference in Treg cell

proportion was observed in patients with WM when compared to the normal cohort,

however, the reduced absolute number of Treg cells was observed to be significant in these

patients. This too may be a result of treatment such as chemotherapy-induced lymphopenia

(Mackall et al., 1994).

Most interestingly, this study observed that patients who have survived with MM for

greater than 10 years since diagnosis have a normal proportion of PB Treg cells, whereas,

other patients with MM for less than 10 years since diagnosis have a significantly elevated

proportion of PB Treg cells, and, as stated previously, patients with MGUS have an increased

percentage of Treg cells. As most patients with MM have preceding MGUS (Landgren et al.,

2009, Weiss et al., 2009, Korde et al., 2011), the results from this study suggest that an

increase in PB Treg cells in patients with MM may be a result of disease progression, as

opposed to patients with MGUS who have yet to develop significant disease burden. It is

therefore possible that the increased frequencies of Treg cells may abrogate anti-tumour

immunity and that tumour cells may exploit Treg cells to elude immune surveillance and

facilitate tumour progression. This theory is strengthened by the evidence that patients with

MM who have survived greater than 10 years have a normal Treg cell numbers, suggesting

they may have developed an ability to avoid the detrimental increase in PB Treg cells. Thus

the results suggest that the number of PB Treg cells may be of clinical and possible

prognostic value. Similarly, a recent study has shown the relationship between increased

Treg cells in patients with MM and adverse clinical features such as hypercalcaemia,

reduced normal plasma cell count and IgA MM (Muthu Raja et al., 2012). These findings

strengthen the notion of Treg cells as a prognostic indicator in predicting survival and

progression risk of patients with MM.

91

4.2.3 Naïve and memory Treg cells in monoclonal gammopathies

The current study characterised the Treg cells further by categorising them into naïve

(CD45RA+) and memory (CD45RO+) Treg phenotypic groups and comparing the proportions

in the PB of patients with monoclonal gammopathies. Muthu Raja et al. (2012) documented

an increase in naïve Treg cells in patients with MM, however, in sharp contrast, the current

study did not demonstrate any significant difference in the naïve, CD45RA+, Treg cell

proportions amongst the cohorts tested. The study by Muthu Raja et al. (2012) assessed the

naïve Treg cell proportions in 5 patients with MM and this may not be a sufficient number to

accurately represent the disease. The current study has assessed more than 31 patients and

the results may be more of a reflection of the cell proportions in MM. Studies of solid

malignancies, such as lung and breast carcinoma, as well as healthy volunteers, have shown

the majority of Treg cells to be of the memory phenotype (Dieckmann et al., 2001, Jonuleit

et al., 2001, Ng et al., 2001, Liyanage et al., 2002). In accordance with previous publications,

the majority of Treg cells observed in this study were found to constitutively express

CD45RO+, thus resembling a memory T cell phenotype. Interestingly, it was found that of the

total proportion of Treg cells, there was a significant increase in memory Treg cells in the PB

of patients with MM and WM compared to the normal cohort. Memory Treg cell quantity

did not differ between patients with MGUS and individuals within the normal cohort. Lin et

al. (2009) proposed that memory Treg cells may be responsible for the loss of concomitant

tumour immunity, assisting tumour progression. The phenomenon of concomitant tumour

immunity may be utilised in MM, where the memory Treg cells may be repressing the

immune system, possibly cytotoxic T cells and inflammatory Th17 cells from inhibiting

tumour progression.

4.2.4 Effect of MM treatment on Treg cell proportion

The current study also focused on the effect of MM treatments, particularly IMiDs, on PB

Treg cell proportion. Although the exact mechanisms of action are not clear, IMiDs have

been shown to exhibit anti-tumour activities and, in recent years, have been widely adopted

for use in the treatment of MM, with, the previously banned drug, thalidomide, as the first

IMiD utilised in this capacity. Lenalidomide, a second generation analogue of thalidomide,

92

has been shown to be more potent and less toxic than thalidomide in the treatment of MM

(Anderson, 2005, Quach et al., 2009). With increasing availability in Australia, more patients

are now treated with lenalidomide. Quach et al. (2009) detailed some of the purported

methods in which IMiDs are proposed to work including both directly against the tumour as

well as through immune related mechanisms. IMiDs have been shown to activate and

induce expansion of both T and NK cells in a number of models, including both mouse

models (Hernandez-Ilizaliturri et al., 2005), normal subjects and patients with MM (Chang et

al., 2006). It has been postulated that IMiDs such as lenalidomide are also active against

MM by their anti Treg cell properties (Kuhn et al., 2007, Galustian et al., 2009). A study by

Galustian et al (2009) showed that lenalidomide inhibits the in vitro expansion and function

of Treg cells. However, thalidomide was not observed to exhibit the same mechanism and

this is almost certainly due to the lack of potency of thalidomide in vitro (Quach et al., 2009).

In the present study, the effect of IMiD treatment on PB Treg cell proportions was examined

in patients with MM. No significant change was observed in the proportion of PB Treg cell in

MM patients receiving lenalidomide treatment compared to untreated patients. Due to the

decreasing number of patients undergoing thalidomide treatment, it was difficult to test a

sufficient number of such patients for a conclusive comparison. The results, however, imply

that thalidomide treated patients have increased PB Treg cell quantity, suggesting that the

mode of action of thalidomide may not be dependent on Treg cell control. Rather, it has

been shown that thalidomide works directly on the tumour by inhibiting vascular

angiogenesis (Quach et al., 2009).

4.2.5 Effect of MM stage on Treg cell proportion

Patients with MM can be categorised into clinical stages based on disease severity using the

ISS. There is currently no consensus as to the association between Treg cell frequency and

MM stage, although a recent study by Muthu Raja et al. (2012) has suggested an association

between the development of adverse clinical features and Treg cell proportion in patients

with MM. The study by Muthu Raja et al. (2012) found a stage dependent increase in Treg

cell frequency, however the observation did not reach statistical significance. Treg cells

have, however, been reported to be associated with disease severity in MM (Feyler et al.,

93

2009), B-CLL (Giannopoulos et al., 2008a), cutaneous T cell lymphoma (Gjerdrum et al.,

2007) and papillary thyroid cancer (French et al., 2010). The present study aimed to

elucidate the relationship between the proportion of Treg cells in the PB and clinical stage of

MM. An increase in Treg cell frequency was observed in patients with MM and premalignant

stage MGUS, although the increase was only found to be significant in MM. The Treg cell

frequency, however, was not observed to differ with disease stage, indicating the increase

in Treg cell number may be a tumour induced mechanism aimed to resist the initial immune

response, and the tumour may then progress through other mechanisms. Further work

needs to be performed to clarify these mechanisms.

4.2.6 Quantification of Treg cells in the PB and BM of MM patients

This study aimed to examine Treg cell quantities in the BM of patients with MM and

compare this to Treg cell quantities in their corresponding PB. It has been previously shown

that Treg cells can accumulate at the site of various cancers such as ovarian cancer (Curiel et

al., 2004), follicular lymphoma (Carreras et al., 2006), gastric and oesophageal cancers

(Ichihara et al., 2003) and that this accumulation may hinder the host’s response to the

tumours. It has also been postulated that the BM may be a reservoir for Treg cells in healthy

subjects (Zhan et al., 2006). Given that MM is a cancer of the BM, it is hypothesised that

Treg cells may accumulate in the BM. This notion has been supported by the observation of

increased Treg cell numbers in the BM of patients with MM compared to the corresponding

PB, demonstrated as part of this study. Thus Treg cells may be localised in the tumour

microenvironment in MM.

4.2.7 Treg cell function in MM

There is little in the literature about the function of Treg cells in malignancies. This may be,

at least in part, because the Treg functional assay is technically difficult, requiring sorting (by

FACS) of Treg cells and sufficient purification to perform the assay. Adding to these

complications, it is especially difficult to obtain a sufficient number of cells to perform the

experiment from patients who are often leucopenic. This study has been the first to

94

compare the effect of different MM therapies on the function of PB Treg cells using a purely

autologous model. Treg cells were isolated from PB of patients with MM already undergoing

different treatments as well as the normal control subjects. The Treg cells were then co-

cultured with purified CD3+ T cells depleted of Treg cells and the proliferation of the T cells

was observed. Further, to ascertain whether Treg cells preferentially suppress a particular T

cell subset, the effect of Treg cell suppression on the CD4+ and CD8+ T cell subsets was also

investigated.

Lenalidomide has been reported to down-regulate FoxP3 and therefore inhibit the function

and proliferation of Treg cells in vitro (Giannopoulos et al., 2008b, Galustian et al., 2009).

Further, it has been documented that treatment with lenalidomide restores the function of

T cell subsets in CLL (Lee et al., 2011). Both thalidomide and lenalidomide are usually used in

combination with dexamethasone as they have been shown to increase rate of clinical

response (Dimopoulos et al., 2007, Richardson et al., 2007, Hideshima et al., 2008, Galustian

and Dalgleish, 2009, Kastritis et al., 2009, Richardson et al., 2009, Stadtmauer et al., 2009,

Hsu et al., 2011). Patients included in this study had been treated with both an IMiD and

dexamethasone.

The current study observed no significant difference in suppressive activity when comparing

Treg cells from healthy individuals and untreated MM patients, inferring that patients with

MM exhibit normal Treg cell function. The Treg cell function of treated patients with MM,

however, was found to be treatment dependent. A trend was observed for lower

suppressive capabilities of Treg cells from patients treated with thalidomide and bortezomib

compared to those from the normal cohort and untreated patients. Interestingly, however,

it was found that Treg cells of patients treated with lenalidomide in conjunction with

dexamethasone were significantly more suppressive when tested in vitro. In addition, these

suppressive capabilities were effective on both T helper and cytotoxic T cells.

Dexamethasone has been shown to be antagonistic to the immunostimulatory capacity of

lenalidomide (Hsu et al, 2011). Prado et al. (2011) demonstrated dexamethasone’s ability to

increase FoxP3 expression, paradoxically in cells that do not have suppressive activity. On

the contrary, a study by Hu et al. (2012) has shown an improved function of Treg cells in

Graves’ disease patients treated with dexamethasone. Favaloro et al. (2013) have shown

that patients receiving high dose dexamethasone had more suppressive Treg cells compared

95

to patients treated with low dose dexamethasone, suggesting that dexamethasone may be

responsible for the increased Treg cell suppression. Low dose dexamethasone has been

shown to be associated with better outcomes than high dose dexamethasone (Rajkumar et

al., 2010). Aside from increased toxicity, it is possible that high dose dexamethasone may

increase the suppressive capacity of Treg cells and therefore low dose dexamethasone, in

combination with lenalidomide, may be the most effective way to restore Treg cell function.

4.2.8 Treg cell suppression on the CD4 and CD8 T cell subsets

This section investigates the in vitro suppression of the two major subsets of T cells, CD4+

and CD8+ T cells, by Treg cells from treated MM patients. The literature has proposed that

Treg cells substantially suppress memory CD8+ cell responses (Murakami et al., 2002)

abrogating the induction of effector CD8+ T cell-mediated tumour rejection. Also, Dai et al.

(2004) have revealed that the presence of antigen induced Treg cells increases the apoptosis

of memory CD8+ cells.

The current study did not observe Treg cells to preferentially suppress either CD4+ or CD8+ T

cells, despite Treg cells appearing slightly more suppressive on CD4+ cells compared to CD8+

in normal individuals and in thalidomide treated MM patients. Interestingly, untreated

patients were not observed to exhibit the same trend as the normal cohort. Treg cells of

untreated MM patients appear equally as suppressive upon T helper and cytotoxic T cells

and this may be a mechanism utilised by MM cells to facilitate their expansion. Intriguingly,

the Treg cells of patients treated with bortezomib were found to have a significantly lower

suppression upon CD4+ cells compared to the normal cohort, presumably revealing a

mechanism by which bortezomib may exert its anti-MM effect.

4.2.9 Modification of Treg cell function

The aim of this section was to investigate whether the addition of exogenous cytokines,

particularly rhIL-12 and rhTGF- would be able to modify the suppressive activity of Treg

cells in vitro. Further, the effect of anti-TGF- on the suppressive function of Treg cells was

examined. This is of particular interest as the observed increase in Treg cell proportions may

96

be the cause of immune tolerance in MM, with malignant cells exploiting the suppressive

nature of the Treg cells. Utilising cytokines to neutralise Treg cell function could expand

therapeutic approaches for MM and other malignancies (Tang et al., 2004).

Although the exact mechanisms used by Treg cells to exert suppression is unknown, Treg

cells are thought to utilise at least two mechanisms, that of cell to cell contact and cytokine

release, with their function thought to be at least partially mediated by the release of TGF-

and IL-10. TGF- is a widely distributed immunomodulatory cytokine with pleiotropic

functions in T cell development, homeostasis and survival (Cerwenka and Swain, 1999,

Gorelik and Flavell, 2002, Li et al., 2006). Studies have shown that TGF- plays a pivotal role

in the generation of Treg cells from CD4+CD25- T cells (Chen et al., 2003, Fantini et al., 2004,

Tang et al., 2004). Further, Marie et al. (2005) have reported that TGF- 1 is imperative in

the maintenance of FoxP3, regulatory function and Treg cell homeostasis. The results

obtained from this study show that rhTGF- appears to increase the suppressive activity of

Treg cells. Interestingly, anti-TGF- had little effect on the in vitro suppression exerted by

the Treg cells.

On the other hand, IL-12 is purported to be involved in the instigation of the Th1 immune

response (King and Segal, 2005). The cytokine has been shown to induce CD4+ T cell

activation during co-culture with Treg cells (King and Segal, 2005) and is thought to reduce

the frequency of Treg cells by induction of IFN- expression by both conventional T cells and

Treg cells (Zhao et al., 2012). This current study has shown that the addition of exogenous

IL-12 is able to reduce the suppressive capability of Treg cells, and allow effector cells to

proliferate. This may offer a unique therapeutic approach, as IL-12 is found to oppose the

function of Treg cells while simultaneously inducing effector T cell responses.

97

4.3 The role of Th17 cells in the monoclonal gammopathies

In recent years, subsets of the T helper cell lineage have been expanded with the

identification of Th17 cells. Increased numbers of these cells are associated with

autoimmunity and inflammation (Dong et al., 2003, Park et al., 2005, Cooke, 2006, Fouser et

al., 2008, Oukka, 2008, Tesmer et al., 2008, Traves and Donnelly, 2008, Yamada, 2009).

Furthermore, Th17 cells have been implicated in the pathogenesis of solid tumours, and

increased numbers have been observed in the PB of patients with malignancies such as

advanced ovarian cancer, gastric cancer and AML and also in tumour tissues of patients with

pancreatic and renal cell carcinoma (Kryczek et al., 2007, Zhang et al., 2008, Wu et al.,

2009). IL-17 has been demonstrated to promote tumour neovascularisation (Inozume et al.,

2009) and a study conducted by Alexandrakis et al. (2006) suggested that IL-17, as a

mediator of angiogenesis, has been associated with disease progression in MM. Further, IL-

17 production has been shown to correlate with osteoclast mediated lytic bone disease in

MM (Noonan et al., 2010). In sharp contrast, it has also been suggested that Th17 cells have

the ability to eradicate melanoma (Muranski et al., 2008) and may enhance the anti-tumour

effect of a lymphoma vaccine (Alvarez et al., 2010). Other studies have suggested that Th17

cells may possess anti-tumour activities and therefore be beneficial to cancer patients,

particularly those with advanced disease (Kryczek et al., 2007, Kryczek et al., 2009a, Kryczek

et al., 2009b). The role of Th17 cells in cancer progression and development remains

controversial and poorly understood due to the scarce and contradictory reports on Th17

cells. The current study aimed to enumerate Th17 cells in the PB of patients with

monoclonal gammopathies and in a normal cohort to allow a comparison of cell numbers to

be made. Additionally, the study aimed to assess whether factors such as disease stage and

length of survival time had an impact on the predominance of these cells.

4.3.1 Th17 cell enumeration in MM and other monoclonal gammopathies

This study found that patients with MM had a significantly lower proportion of Th17 cells in

their PB compared to healthy volunteers. In addition, the absolute Th17 cell count was also

reduced in patients with MM compared to the normal cohort, albeit this may be a reflection

98

of chemotherapy induced lymphopoenia (Mackall et al., 1994). There was no significant

change in Th17 cell proportions in the PB of patients with WM and MGUS compared to the

normal cohort, inferring that amongst the monoclonal gammopathies studied, the decrease

in Th17 cells is unique to patients with MM. Interestingly, MM patients who had survived 10

or more years since diagnosis possessed PB Th17 cell proportions that were similar to those

of the normal cohort.

Some studies have suggested that Th17 cells are associated with a favourable prognosis and

may be involved in anti-tumour activity (Kryczek et al., 2009b). It has been purported that

this may be mediated through the activation of the innate immune system by IL-17. Kryczek

et al. (2009b) have reported that mice deficient in IL-17 had enhanced tumour growth in

subcutaneous and lung metastasis. This supports the notion that the decrease in Th17 cells

may be evident in malignancies such as MM. In non-Hodgkin lymphoma, a low frequency of

Th17 cells was observed in the tumour microenvironment, and it was proposed that

lymphoma B cells may favour the expansion of Treg cells and down-regulate the generation

of Th17 cells (Yang et al., 2009). This may be similar for patients with MM where malignant

cells may allow for the expansion of Treg cells, simultaneously inhibiting Th17 cell

development. A study of patients with ovarian cancer demonstrated a higher percentage of

Th17 cells at the tumour site, however a low presence of Th17 cells was observed in PB

(Miyahara et al., 2008). While the pathogenesis of solid tumours may be different from that

of haematological malignancies, this still provides a sound basis for the investigation of the

BM of MM patients.

It has been suggested that Th17 cells could generate an immune response against an

established tumour (Chen and Oppenheim, 2009), as studies have proposed exploiting the

autoimmune response of Th17 cells as an immunotherapy (Kottke et al., 2007). In addition,

the adoptive transfer of Th17 cells was potent in inhibiting tumour progression in a study

conducted by Muranski et al. (2008). The study illustrated that, in a B16 melanoma murine

model, animals receiving Th0 or Th1 cells relapsed, whereas animals treated with Th17 cells

remained tumour free. The suggestion that Th17 cells may be potent in inducing an anti-

tumour response and potentially eradicating the tumour provides evidence of possible value

in exploiting the Th17 immune response as an immunotherapy. The results of this study

suggest that Th17 cells may be of clinical and biological importance in MM due to their pro-

99

inflammatory capacity and the decrease in Th17 cells observed may be a result of a tumour

induced mechanism aimed to incapacitate the immune response.

4.3.2 Association of MM stage and Th17 cell proportion

The effect of MM stage and treatment on Th17 cell quantity was also considered in this

study. Although an increase in Th17 cell percentage was observed in patients with stage II

MM, no significant difference was observed amongst the different stages. This shows that

patients with MM have an overall decrease in Th17 cell percentage regardless of their stage

classification, suggesting that the reduction in Th17 cell frequency may be a result of tumour

pathogenesis as opposed to tumour progression. Th17 cells are thought to be potent

activators of the innate immune system and an overall decrease in Th17 cell number may

hinder the process of eradicating the malignant plasma cells. This may possibly be a

mechanism by which the tumour evades an immune response and may be the effect of the

increased Treg cells in the PB of patients with MM. Contrary to these results, an association

between Th17 cells and disease progression in MM has been suggested, proposing the use

of Th17 cells as a possible prognostic predictor in MM (Shen et al., 2012) however, due to

the limited number of patients studied, that particular study also recommended an

additional study with a greater number of MM patients and normal individuals to support

the results. The current study, however, evaluated 22 patients with MM, a much greater

number than that used in the study by Shen et al. and therefore may be more of a reflection

on the effect of MM stage on Th17 cell proportion. However, other factors (eg. therapy,

transfusion etc) being administered to each patient may influence the Th17 cell proportion

in MM and should be considered in any prospective study.

100

4.4 Treg and Th17 equilibrium

The data taken together so far is in accordance with the notion of reciprocal generation of

Treg and Th17 cells. It is thought that there is reciprocal generation of Th17 and Treg cells in

the PB, as these cells have mutually exclusive functions (Bettelli et al., 2006, Joshua et al.,

2008). Presumably, the expanded PB Treg cell pool may be a cause, or effect, of the reduced

PB Th17 cell proportion. Treg cells are known to inhibit the expansion of Th17 cells in the

tumour microenvironment (Chen and Oppenheim, 2009). This decrease in Th17 cell

proportions may enable the expansion of the malignant cells in patients with MM.

4.4.1 Treg/Th17 cell ratio in monoclonal gammopathies

Accumulating evidence suggests that Treg and Th17 cells are mutual antagonists (Afzali et

al., 2007, Joshua et al., 2008). The inverse relationship of Treg and Th17 cells has been

illustrated in a number of studies (Bettelli et al., 2006, Mangan et al., 2006, Veldhoen et al.,

2006). An imbalance in the Treg/Th17 cell ratio can be detrimental to the health of patients

and has been suggested as a factor in the development or progression of various cancers

(Sfanos et al., 2008, Yang et al., 2009, Chi et al., 2010). In the current study, an imbalance in

the ratio of Treg/Th17 cells was observed in patients with MM and such an imbalance was

non-existent in MGUS and WM. This is the first time such an imbalance has been reported

and hence is an original observation in this study. It suggests that measuring Treg cell

numbers alone does not provide sufficient details of the swing of the Treg/Th17 axis. Thus, it

is likely to be more important to evaluate the Treg and Th17 cell homeostatic balance,

rather than PB Treg cell numbers alone, as it appears to be a greater predictor of survival in

patients with MM.

The current study found a clear favouring of PB Treg cells over Th17 cells in patients with

MM compared to the normal cohort. Giannopoulos et al. (2012) reported an association

between increased Treg cell percentage and poor prognosis in patients with MM. However,

they did not determine the number or frequency of Th17 cells. The current study

demonstrates a significant difference in patients with MM, who have a mean Treg/Th17 cell

ratio of 16.1:1, and the normal control cohort, observed to have a Treg/Th17 cell ratio of

6.6:1, indicating a tumour-induced defect in the immune regulation of patients with MM to

101

maintain cellular homeostatic balance in PB. In contrast, the difference in Treg/Th17 cell

ratio was not observed in patients with MGUS or WM, thus suggesting, the imbalance in the

Treg/Th17 cell ratio is dependent on tumour load and disease type. Further, Yang et al.

(2009) reported a similar observation in non-Hodgkin lymphoma. It was reported that

lymphoma B cells may be responsible for the deleterious skewing of the Treg and Th17 cell

balance. Similarly, MM cells may be responsible for the abnormal imbalance between Treg

and Th17 cells by up-regulating Treg cells and down-regulating Th17 cells. Consequently, the

skewed Treg/Th17 cell balance may result in suppression of the immune response, thus

inducing immune tolerance towards the malignant cells.

4.4.2 Treg/Th17 cell ratio and survival

The current study was also able to compare the outcome of patients surviving 10 years or

greater with MM to those with MM for less than 10 years since diagnosis. Most

interestingly, the patients who have survived for 10 or more years with MM possessed a

similar Treg/Th17 cell ratio to the normal cohort, suggesting that survival with MM is

associated with a normal Treg/Th17 cell ratio. This implicates the critical nature of the Treg

and Th17 cell balance on survival with MM. The malignant cells may attempt to alter the

cellular environment for optimisation of their expansion and long term surviving patients

may have the ability to resist the MM cell induced Treg/Th17 cell ratio deviation from

homeostatic equilibrium. Consistent with this idea, the current study is the first to

demonstrate that MM patients with a high Treg/Th17 cell ratio had a significantly lower

survival compared to patients who were able to retain a normal Treg/Th17 cell ratio,

indicating an association between a high Treg/Th17 cell ratio and survival. A bimodal

distribution was observed in patients surviving with MM for more than 10 years (Figure

3.18). 4 patients in particular exhibited a higher Treg/Th17 cell ratio, which may possibly be

associated with relapse or even death. Overall however, the data suggest that the 10 year

survivor cohort had the ability to maintain their Treg and Th17 cell equilibrium and

therefore resistance to the progression of the malignancy. Further, the data emphasise the

importance of preservation of the Treg and Th17 cell homeostatic balance as this may

impact tumour immunity.

102

The delicate balance of both TGF- and IL-6 is documented to be important in the

development of either Treg or Th17 cell lineages. TGF- alone skews precursors towards the

development of the Treg cell lineage, whereas a combination of TGF- and IL-6 induces

differentiation of naïve T cells into Th17 cells (Bettelli et al., 2006, Mangan et al., 2006,

Veldhoen et al., 2006). Evidence indicates that IL-6, produced by BM stromal cells and MM

cells, is a major growth and survival factor for malignant plasma cells (Zhang et al., 1992,

Hallek et al., 1998, Zdzisi ska et al., 2008). The expanding malignant clones may possibly

consume the majority of the available IL-6 in the BM, rendering the environment depleted

of IL-6. Furthermore, MM cells secrete large amounts of TGF- (Cook et al., 1999), which

may induce the expansion of Treg cells, hence the increased Treg cell proportion observed

in both the BM and PB of patients with MM. This relative skewing of IL-6 and TGF- may

partly explain the reciprocal Treg/Th17 cell relationship observed in patients with MM, of

which the cytokine milieu in MM may favour the expansion of the immunosuppressive Treg

cell lineage and impede the expansion of the pro-inflammatory Th17 cell lineage.

4.5 Conclusions and future directions

4.5.1 Future directions

There is a potential for either Treg or Th17 cells to be the target of therapies for

malignancies and autoimmunity and this should be investigated in future studies.

Identification of surface markers for the detection of Th17 cells would enable easier

enumeration of this cell type in the BM as well as the development of functional studies for

comparison of their role in MM and other monoclonal gammopathies to healthy individuals.

The current method is timely and far more complicated than a simple surface stain. A

number of novel monoclonal antibodies are currently under investigation but were not

available for these studies (Prof Mark Hogarth, personal communication).

This study reinforces the need to study the Treg/Th17 cell balance in the BM

microenvironment of MM patients with a comparison being made with healthy individuals

to enable a better understanding of the pathogenesis of the malignancies and development

of curative immunotherapies. A prospective study to clarify the prognostic significance of

Treg cells in MM as well as clarification in terms of the Treg/Th17 dynamic, is required to

103

determine whether the high Treg/Th17 ratio is causative of a poor prognosis or a biomarker

of immune dysfunction. Further investigation is warranted to elucidate the factors involved

in the Treg/Th17 dynamic in MM and to study the overall quantitation and interaction

between cytokines and lymphocytes in the PB and BM of patients with MM and other

monoclonal gammopathies. Changing the Treg/Th17 cell ratio may be possible by treatment

with cytokines or by cell therapeutic physical removal of Treg cells. In patients with

autoimmune disease or GvHD, ex vivo expansion and infusion of Treg cells has already been

considered (Brunstein et al., 2011, Colonna et al., 2011, Di Ianni et al., 2011). It is important

that laboratories can provide monitoring support for such clinical activities. The clear bi-

modal distribution of Treg/Th17 cell ratio, evident in patients with MM for greater than 10

years (Figure 3.18), warrants further investigation, and may serve to strengthen the

argument that the ratio, and not the parameters by themselves, is the important factor in

determining the prognostic value of this test.

Considering the complexity of the cytokine network, targeted immunotherapy requires a

delicate balance between increasing the immune response and severe autoimmunity. The

finding that Treg cell function is altered by therapy and by cytokines, elucidates the need to

study the cytokine milieu in the BM and PB of patients with MM and in healthy individuals.

So far studies have suggested the inhibition of TGF- as a new therapeutic approach as it

simultaneously enhances bone formation and assists in the suppression of MM cell growth

(Matsumoto and Abe, 2011). Also, the use of CTLA-4 and PD-1 blockers have emerged as

successful treatment approaches in melanoma, after being explored in phase 1 clinical trials

(Lotem et al., 2012). Thus the results of these studies may facilitate the development of new

immunotherapies, which may assist in alleviating the cancer and may ultimately lead to the

development of a cure through reinstatement of the immunological balance.

4.5.2 Conclusion

The pathogenesis of monoclonal gammopathies is multifaceted and complex. This study

aimed to clarify some aspects of the dysfunctional immunogenic control in these patients.

The results demonstrate a skewed Treg/Th17 cell ratio in the PB of patients with MM

compared to healthy individuals. The study observed an increase in Treg cell percentage,

104

which largely consisted of memory Treg cells, in patients with MM compared to healthy

individuals, as well as a reduced Th17 cell frequency in the PB of patients with MM and this

reciprocal combination was not observed in the other monoclonal gammopathies studied.

Additionally, the BM of patients with MM was found to consist of a higher frequency of Treg

cells compared to the PB of the same patients. The PB Treg and Th17 cell proportions in

patients with MM were not observed to fluctuate based on treatment or disease stage. The

function of Treg cells, however, was observed to differ based on the treatment being

received by the patient and, if required, could be altered using exogenously administered

cytokines. Most interestingly, 10 year survivors of MM possess similar proportions of Treg

and Th17 in their PB compared to healthy individuals, and therefore a similar PB Treg/Th17

cell ratio. Further studies of Treg/Th17 cell ratio in MM demonstrated patients with a high

Treg/Th17 cell ratio had a significantly lower survival than those with a normal Treg/Th17

cell ratio. The results of this study clearly delineate for the first time the importance of the

Treg and Th17 cell equilibrium in the PB of patients with MM. The results of the 10 year

survivors reveal a strong association between the Treg and Th17 cell homeostatic balance

and survival, particularly in MM. Therefore, this indicates that the imbalance favouring

suppressive Treg cells may cause the innate and/or the adaptive immune system to be

quiescent and limited in any anti-tumour response.

105

CHAPTER FIVE

REFERENCES

106

AFZALI, B., LOMBARDI, G., LECHLER, R. I. & LORD, G. M. 2007. The role of T helper 17 (Th17) and regulatory T cells (Treg) in human organ transplantation and autoimmune disease. Clinical& Experimental Immunology, 148, 32-46.

ALEXANDRAKIS, M. G., PAPPA, C. A., MIYAKIS, S., SFIRIDAKI, A., KAFOUSI, M., ALEGAKIS, A. & STATHOPOULOS, E. N. 2006. Serum interleukin-17 and its relationship to angiogenic factors in multiple myeloma. European Journal of Internal Medicine, 17, 412-416.

ALVAREZ, E., MOGA, E., BARQUINERO, J., SIERRA, J. & BRIONES, J. 2010. Dendritic and tumor cell fusions transduced with adenovirus encoding CD40L eradicate B-cell lymphoma and induce a Th17-type response. Gene Therapy, 17, 469-77.

ÁLVARO, T., LEJEUNE, M., SALVADÓ, M.-T., LOPEZ, C., JAÉN, J., BOSCH, R. & PONS, L. E. 2006. Immunohistochemical Patterns of Reactive Microenvironment Are Associated With Clinicobiologic Behavior in Follicular Lymphoma Patients. Journal of Clinical Oncology, 24, 5350-5357.

ANDERSEN, M. H., SCHRAMA, D., STRATEN, P. T. & BECKER, J. C. 2006. Cytotoxic T Cells. Journal of Investigative Dermatology, 126, 32-41.

ANDERSON, K. C. 2005. Lenalidomide and Thalidomide: Mechanisms of Action—Similarities and Differences. Seminars in Hematology, 42, S3-S8.

ATTAL, M., HAROUSSEAU, J.-L., FACON, T., GUILHOT, F., DOYEN, C., FUZIBET, J.-G., MONCONDUIT, M., HULIN, C., CAILLOT, D., BOUABDALLAH, R., VOILLAT, L., SOTTO, J.-J., GROSBOIS, B. & BATAILLE, R. 2003. Single versus Double Autologous Stem-Cell Transplantation for Multiple Myeloma. New England Journal of Medicine, 349, 2495-2502.

ATTAL, M., HAROUSSEAU, J.-L., STOPPA, A.-M., SOTTO, J.-J., FUZIBET, J.-G., ROSSI, J.-F., CASASSUS, P., MAISONNEUVE, H., FACON, T., IFRAH, N., PAYEN, C. & BATAILLE, R. 1996. A Prospective, Randomized Trial of Autologous Bone Marrow Transplantation and Chemotherapy in Multiple Myeloma. New England Journal of Medicine, 335, 91-97.

BAECHER-ALLAN, C., BROWN, J. A., FREEMAN, G. J. & HAFLER, D. A. 2001. CD4+CD25high Regulatory Cells in Human Peripheral Blood. The Journal of Immunology, 167, 1245-1253.

BANERJEE, D. K., DHODAPKAR, M. V., MATAYEVA, E., STEINMAN, R. M. & DHODAPKAR, K. M. 2006. Expansion of FOXP3high regulatory T cells by human dendritic cells (DCs) in vitro and after injection of cytokine-matured DCs in myeloma patients. Blood, 108, 2655-2661.

BATAILLE, R., BOCCADORO, M., KLEIN, B., DURIE, B. & PILERI, A. 1992. C-reactive protein and beta-2 microglobulin produce a simple and powerful myeloma staging system. Blood, 80, 733-737.

BELKAID, Y. & ROUSE, B. T. 2005. Natural regulatory T cells in infectious disease. Nature Immunology, 6, 353-360.

BENSINGER, W. I. 2009. Role of autologous and allogeneic stem cell transplantation in myeloma. Leukemia, 23, 442-448.

107

BETTELLI, E., CARRIER, Y., GAO, W., KORN, T., STROM, T. B., OUKKA, M., WEINER, H. L. & KUCHROO, V. K. 2006. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature, 441, 235-238.

BEYER, M., KOCHANEK, M., DARABI, K., POPOV, A., JENSEN, M., ENDL, E., KNOLLE, P. A., THOMAS, R. K., VON BERGWELT-BAILDON, M., DEBEY, S., HALLEK, M. & SCHULTZE, J. L. 2005. Reduced frequencies and suppressive function of CD4+CD25hi regulatory T cells in patients with chronic lymphocytic leukemia after therapy with fludarabine. Blood, 106, 2018-2025.

BEYER, M., KOCHANEK, M., GIESE, T., ENDL, E., WEIHRAUCH, M. R., KNOLLE, P. A., CLASSEN, S. & SCHULTZE, J. L. 2006. In vivo peripheral expansion of naive CD4+CD25high FoxP3+ regulatory T cells in patients with multiple myeloma. Blood, 107, 3940-3949.

BEYER, M. & SCHULTZE, J. L. 2006. Regulatory T cells in cancer. Blood, 108, 804-811.

BLOBE, G. C., SCHIEMANN, W. P. & LODISH, H. F. 2000. Role of transforming growth factor beta in human disease. New England Journal of Medicine, 342, 1350-8.

BOURGEOIS, C., VEIGA-FERNANDES, H., JORET, A.-M., ROCHA, B. & TANCHOT, C. 2002. CD8 lethargy in the absence of CD4 help. European Journal of Immunology, 32, 2199-2207.

BRIMNES, M. K., VANGSTED, A. J., KNUDSEN, L. M., GIMSING, P., GANG, A. O., JOHNSEN, H. E. & SVANE, I. M. 2010. Increased Level of both CD4+FOXP3+ Regulatory T Cells and CD14+HLA-DR /low Myeloid-Derived Suppressor Cells and Decreased Level of Dendritic Cells in Patients with Multiple Myeloma. Scandinavian Journal of Immunology, 72, 540-547.

BRONTE, V. 2008. Th17 and cancer: friends or foes? Blood, 112, 214.

BRUNDA, M. J., LUISTRO, L., WARRIER, R. R., WRIGHT, R. B., HUBBARD, B. R., MURPHY, M., WOLF, S. F. & GATELY, M. K. 1993. Antitumor and antimetastatic activity of interleukin 12 against murine tumors. Journal of Experimental Medicine, 178, 1223-30.

BRUNSTEIN, C. G., MILLER, J. S., CAO, Q., MCKENNA, D. H., HIPPEN, K. L., CURTSINGER, J., DEFOR, T., LEVINE, B. L., JUNE, C. H., RUBINSTEIN, P., MCGLAVE, P. B., BLAZAR, B. R. & WAGNER, J. E. 2011. Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics. Blood, 117, 1061-1070.

CAR, B. D., ENG, V. M., LIPMAN, J. M. & ANDERSON, T. D. 1999. The Toxicology of Interleukin-12: A Review. Toxicologic Pathology, 27, 58-63.

CAR, B. D., ENG, V. M., SCHNYDER, B., LEHIR, M., SHAKHOV, A. N., WOERLY, G., HUANG, S., AGUET, M., ANDERSON, T. D. & RYFFEL, B. 1995. Role of interferon- in interleukin 12-induced pathology in mice. American Journal of Pathology, 147, 1693-1707.

CARRERAS, J., LOPEZ-GUILLERMO, A., FOX, B. C., COLOMO, L., MARTINEZ, A., RONCADOR, G., MONTSERRAT, E., CAMPO, E. & BANHAM, A. H. 2006. High numbers of tumor-infiltrating FOXP3-positive regulatory T cells are associated with improved overall survival in follicular lymphoma. Blood, 108, 2957-2964.

108

CERWENKA, A. & SWAIN, S. L. 1999. TGF-beta1: immunosuppressant and viability factor for T lymphocytes. Microbes Infection, 1, 1291-6.

CHANG, D. H., LIU, N., KLIMEK, V., HASSOUN, H., MAZUMDER, A., NIMER, S. D., JAGANNATH, S. & DHODAPKAR, M. V. 2006. Enhancement of ligand-dependent activation of human natural killer T cells by lenalidomide: therapeutic implications. Blood, 108, 618-621.

CHEN, W., JIN, W., HARDEGEN, N., LEI, K.-J., LI, L., MARINOS, N., MCGRADY, G. & WAHL, S. M. 2003. Conversion of Peripheral CD4+CD25 Naive T Cells to CD4+CD25+ Regulatory T Cells by TGF- Induction of Transcription Factor FoxP3. The Journal of Experimental Medicine, 198, 1875-1886.

CHEN, X. & OPPENHEIM, J. J. 2009. Regulatory T Cells, Th17 Cells, and TLRs: Crucial Roles in Inflammation, Autoimmunity, and Cancer. Pathways.e-pub

CHI, L. J., LU, H. T., LI, G. L., WANG, X. M., SU, Y., XU, W. H. & SHEN, B. Z. 2010. Involvement of T helper type 17 and regulatory T cell activity in tumour immunology of bladder carcinoma. Clinical & Experimental Immunology, 161, 480-489.

CHIARENZA, A., PARRINELLO, N., LA CAVA, P., SPINA, E., TIBULLO, D., GIALLONGO, C., CAVALLI, M., ROMANO, A., FIUMARA, P., PALUMBO, G. A. & DI RAIMONDO, F. 2009. Lenalidomide IS Able to Restore Immune SYSTEM IN MULTIPLE MYELOMA PATIENTS. ASH Annual Meeting Abstracts, 114, 4909.

CLAVE, E., DOUAY, C., COMAN, T., BUSSON, M., BOMPOINT, C., MOINS-TEISSERENC, H., GLAUZY, S., CARMAGNAT, M., GORIN, N. C., TOUBERT, A. & GARDERET, L. 2013. Lenalidomide consolidation and maintenance therapy after autologous stem cell transplantation for multiple myeloma induces persistent changes in T-cell homeostasis. Leukemia/Lymphoma.

COHEN, J. L., TRENADO, A., VASEY, D., KLATZMANN, D. & SALOMON, B. L. 2002. CD4+CD25+ Immunoregulatory T Cells. The Journal of Experimental Medicine, 196, 401-406.

COLOMBO, M. P. & TRINCHIERI, G. 2002. Interleukin-12 in anti-tumor immunity and immunotherapy. Cytokine Growth Factor Review, 13, 155-68.

COLONNA, L., SEGA, E. I. & NEGRIN, R. S. 2011. Natural and Expanded CD4+CD25+ Regulatory T Cells in Bone Marrow Transplantation. Biology of Blood and Marrow Transplantation : Journal of the American Society for Blood and Marrow Transplantation, 17, S58-S62.

COOK, G., CAMPBELL, J. D., CARR, C. E., BOYD, K. S. & FRANKLIN, I. M. 1999. Transforming growth factor beta from multiple myeloma cells inhibits proliferation and IL-2 responsiveness in T lymphocytes. Journal of Leukocyte Biology, 66, 981-8.

COOKE, A. 2006. Th17 Cells in Inflammatory Conditions. The review of diabetic studies, 3, 72-75.

109

CURIEL, T. J., COUKOS, G., ZOU, L., ALVAREZ, X., CHENG, P., MOTTRAM, P., EVDEMON-HOGAN, M., CONEJO-GARCIA, J. R., ZHANG, L., BUROW, M., ZHU, Y., WEI, S., KRYCZEK, I., DANIEL, B., GORDON, A., MYERS, L., LACKNER, A., DISIS, M. L., KNUTSON, K. L., CHEN, L. & ZOU, W. 2004. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nature Medicine, 10, 942-949.

D'AMATO, R. J., LOUGHNAN, M. S., FLYNN, E. & FOLKMAN, J. 1994. Thalidomide is an inhibitor of angiogenesis. Proceedings of the National Academy of Sciences, 91, 4082-4085.

DAI, Z., LI, Q., WANG, Y., GAO, G., DIGGS, L. S., TELLIDES, G. & LAKKIS, F. G. 2004. CD4+CD25+ regulatory T cells suppress allograft rejection mediated by memory CD8+ T cells via a CD30-dependent mechanism. The Journal of Clinical Investigation, 113, 310-317.

DE VISSER, K. E., EICHTEN, A. & COUSSENS, L. M. 2006. Paradoxical roles of the immune system during cancer development. Nature Reviews Cancer, 6, 24-37.

DEBES-MARUN, C. S., DEWALD, G. W., BRYANT, S., PICKEN, E., SANTANA-DAVILA, R., GONZALEZ-PAZ, N., WINKLER, J. M., KYLE, R. A., GERTZ, M. A., WITZIG, T. E., DISPENZIERI, A., LACY, M. Q., RAJKUMAR, S. V., LUST, J. A., GREIPP, P. R. & FONSECA, R. 2003. Chromosome abnormalities clustering and its implications for pathogenesis and prognosis in myeloma. Leukemia, 17, 427-436.

DEWALD, G., KYLE, R., HICKS, G. & GREIPP, P. 1985. The clinical significance of cytogenetic studies in 100 patients with multiple myeloma, plasma cell leukemia, or amyloidosis. Blood, 66, 380-390.

DHODAPKAR, K. M., BARBUTO, S., MATTHEWS, P., KUKREJA, A., MAZUMDER, A., VESOLE, D., JAGANNATH, S. & DHODAPKAR, M. V. 2008. Dendritic cells mediate the induction of polyfunctional human IL17-producing cells (Th17-1 cells) enriched in the bone marrow of patients with myeloma. Blood, 112, 2878-2885.

DI IANNI, M., FALZETTI, F., CAROTTI, A., TERENZI, A., CASTELLINO, F., BONIFACIO, E., DEL PAPA, B., ZEI, T., OSTINI, R. I., CECCHINI, D., ALOISI, T., PERRUCCIO, K., RUGGERI, L., BALUCANI, C., PIERINI, A., SPORTOLETTI, P., ARISTEI, C., FALINI, B., REISNER, Y., VELARDI, A., AVERSA, F. & MARTELLI, M. F. 2011. Tregs prevent GVHD and promote immune reconstitution in HLA-haploidentical transplantation. Blood, 117, 3921-3928.

DIECKMANN, D., PLOTTNER, H., BERCHTOLD, S., BERGER, T. & SCHULER, G. 2001. Ex Vivo Isolation and Characterization of Cd4+Cd25+ T Cells with Regulatory Properties from Human Blood. The Journal of Experimental Medicine, 193, 1303-1310.

DIMOPOULOS, M., SPENCER, A., ATTAL, M., PRINCE, H. M., HAROUSSEAU, J.-L., DMOSZYNSKA, A., MIGUEL, J. S., HELLMANN, A., FACON, T., FOÀ, R., CORSO, A., MASLIAK, Z., OLESNYCKYJ, M., YU, Z., PATIN, J., ZELDIS, J. B. & KNIGHT, R. D. 2007. Lenalidomide plus Dexamethasone for Relapsed or Refractory Multiple Myeloma. New England Journal of Medicine, 357, 2123-2132.

110

DONG, G., YE, R., SHI, W., LIU, S., WANG, T., YANG, X., YANG, N. & YU, X. 2003. IL-17 induces autoantibody overproduction and peripheral blood mononuclear cell overexpression of IL-6 in lupus nephritis patients. Chinese Medical Journal (Engl), 116, 543-8.

DURIE, B. G. M. & SALMON, S. E. 1975. A clinical staging system for multiple myeloma correlation of measured myeloma cell mass with presenting clinical features, response to treatment, and survival. Cancer, 36, 842-854.

EDINGER, M. 2009. Regulatory T cells for the prevention of graft-versus-host disease: Professionals defeat amateurs. European Journal of Immunology, 39, 2966-2968.

FANTINI, M. C., BECKER, C., MONTELEONE, G., PALLONE, F., GALLE, P. R. & NEURATH, M. F. 2004. Cutting Edge: TGF- Induces a Regulatory Phenotype in CD4+CD25 T Cells through Foxp3 Induction and Down-Regulation of Smad7. The Journal of Immunology, 172, 5149-5153.

FAVALORO, J., BROWN, R., AKLILU, E., YANG, S., SUEN, H., HART, D., FROMM, P., GIBSON, J., KHOO, L., HO, P. J. & JOSHUA, D. 2013. Myeloma skews regulatory T and pro-inflammatory T helper 17 cell balance in favor of a suppressive state. Leukemia Lymphoma. 55, 1090-1098

FERRETTI, S., BONNEAU, O., DUBOIS, G. R., JONES, C. E. & TRIFILIEFF, A. 2003. IL-17, produced by lymphocytes and neutrophils, is necessary for lipopolysaccharide-induced airway neutrophilia: IL-15 as a possible trigger. The Journal of Immunology, 170, 2106-12.

FEYLER, S., VON LILIENFELD-TOAL, M., JARMIN, S., MARLES, L., RAWSTRON, A., ASHCROFT, A. J., OWEN, R. G., SELBY, P. J. & COOK, G. 2009. CD4+CD25+FoxP3+ regulatory T cells are increased whilst CD3+CD4 CD8 TCR+ Double Negative T cells are decreased in the peripheral blood of patients with multiple myeloma which correlates with disease burden. British Journal of Haematology, 144, 686-695.

FIELD, E. O., GIBBS, J. E., TUCKER, D. F. & HELLMANN, K. 1966. Effect of Thalidomide on the Graft versus Host Reaction. Nature, 211, 1308-1310.

FOGLIETTA, M., MARIANI, S., CASTELLA, B., COSCIA, M., GODIO, L., ZAPPIA, S., BOCCADORO, M. & MASSAIA, M. 2006. Regulatory T Cells (Tregs) Are Highly Preserved in Multiple Myeloma Patients and Can Dominate Effector Functions. ASH Annual Meeting Abstracts, 108, 658.

FONTENOT, J. D. & RUDENSKY, A. Y. 2005. A well adapted regulatory contrivance: regulatory T cell development and the forkhead family transcription factor FoxP3. Nature Immunology, 6, 331-337.

FOUSER, L., WRIGHT, J., DUNUSSI-JOANNOPOULOS, K. & COLLINS, M. 2008. Th17 cytokines and their emerging roles in inflammation and autoimmunity. Immunological reviews, 226, 87-102.

FRENCH, J. D., WEBER, Z. J., FRETWELL, D. L., SAID, S., KLOPPER, J. P. & HAUGEN, B. R. 2010. Tumor-Associated Lymphocytes and Increased FoxP3+ Regulatory T Cell Frequency Correlate with More Aggressive Papillary Thyroid Cancer. Journal of Clinical Endocrinology & Metabolism, 95, 2325-2333.

GALUSTIAN, C. & DALGLEISH, A. 2009. Lenalidomide: a novel anticancer drug with multiple modalities. Expert Opinion on Pharmacotherapy, 10, 125-33.

111

GALUSTIAN, C., MEYER, B., LABARTHE, M.-C., DREDGE, K., KLASCHKA, D., HENRY, J., TODRYK, S., CHEN, R., MULLER, G., STIRLING, D., SCHAFER, P., BARTLETT, J. & DALGLEISH, A. 2009. The anti-cancer agents lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory cells. Cancer Immunology Immunotherapy, 58, 1033-1045.

GERSHON, R. K., COHEN, P., HENCIN, R. & LIEBHABER, S. A. 1972. Suppressor T Cells. The Journal of Immunology, 108, 586-590.

GERSHON, R. K. & KONDO, K. 1971. Infectious Immunological Tolerance. Immunology, 21, 903.

GIANNOPOULOS, K., KAMINSKA, W., HUS, I. & DMOSZYNSKA, A. 2012. The frequency of T regulatory cells modulates the survival of multiple myeloma patients: detailed characterisation of immune status in multiple myeloma. British Journal of Cancer, 106, 546-552.

GIANNOPOULOS, K., SCHMITT, M., KOWAL, M., WLASIUK, P., BOJARSKA-JUNAK, A., CHEN, J., ROLINSKI, J. & DMOSZYNSKA, A. 2008a. Characterization of regulatory T cells in patients with B-cell chronic lymphocytic leukemia. Oncology reports, 20, 677-682.

GIANNOPOULOS, K., SCHMITT, M., WLASIUK, P., CHEN, J., BOJARSKA-JUNAK, A., KOWAL, M., ROLINSKI, J. & DMOSZYNSKA, A. 2008b. The high frequency of T regulatory cells in patients with B-cell chronic lymphocytic leukemia is diminished through treatment with thalidomide. Leukemia, 22, 222-4.

GIULIANI, N., MORANDI, F., TAGLIAFERRI, S., LAZZARETTI, M., BONOMINI, S., CRUGNOLA, M., MANCINI, C., MARTELLA, E., FERRARI, L., TABILIO, A. & RIZZOLI, V. 2007. The proteasome inhibitor bortezomib affects osteoblast differentiation in vitro and in vivo in multiple myeloma patients. Blood, 110, 334-338.

GJERDRUM, L. M., WOETMANN, A., ODUM, N., BURTON, C. M., ROSSEN, K., SKOVGAARD, G. L., RYDER, L. P. & RALFKIAER, E. 2007. FOXP3+ regulatory T cells in cutaneous T-cell lymphomas: association with disease stage and survival. Leukemia, 21, 2512-2518.

GORELIK, L. & FLAVELL, R. A. 2002. Transforming growth factor-beta in T-cell biology. Nature reviews Immunology, 2, 46-53.

GOULD, J., ALEXANIAN, R., GOODACRE, A., PATHAK, S., HECHT, B. & BARLOGIE, B. 1988. Plasma cell karyotype in multiple myeloma. Blood, 71, 453-456.

GREIPP, P. R., LEONG, T., BENNETT, J. M., GAILLARD, J. P., KLEIN, B., STEWART, J. A., OKEN, M. M., KAY, N. E., VAN NESS, B. & KYLE, R. A. 1998. Plasmablastic Morphology---An Independent Prognostic Factor With Clinical and Laboratory Correlates: Eastern Cooperative Oncology Group (ECOG) Myeloma Trial E9486 Report by the ECOG Myeloma Laboratory Group. Blood, 91, 2501-2507.

GREIPP, P. R., MIGUEL, J. S., DURIE, B. G. M., CROWLEY, J. J., BARLOGIE, B., BLADÉ, J., BOCCADORO, M., CHILD, J. A., AVET-LOISEAU, H., KYLE, R. A., LAHUERTA, J. J., LUDWIG, H., MORGAN, G., POWLES, R., SHIMIZU, K., SHUSTIK, C., SONNEVELD, P., TOSI, P., TURESSON, I. & WESTIN, J. 2005. International Staging System for Multiple Myeloma Journal of Clinical Oncology, 23, 3412-3420.

112

GUILLAUME, T., RUBINSTEIN, D. B. & SYMANN, M. 1998. Immune Reconstitution and Immunotherapy After Autologous Hematopoietic Stem Cell Transplantation. Blood, 92, 1471-1490.

GUPTA, R., GANESHAN, P., HAKIM, M., VERMA, R., SHARMA, A. & KUMAR, L. 2011. Significantly reduced regulatory T cell population in patients with untreated multiple myeloma. Leuk Res, 35, 874-8.

HA, T.-Y. 2009. The Role of Regulatory T Cells in Cancer. Immune Network, 9, 209-235.

HALLEK, M., LEIF BERGSAGEL, P. & ANDERSON, K. C. 1998. Multiple Myeloma: Increasing Evidence for a Multistep Transformation Process. Blood, 91, 3-21.

HARRINGTON, L. E., HATTON, R. D., MANGAN, P. R., TURNER, H., MURPHY, T. L., MURPHY, K. M. & WEAVER, C. T. 2005. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nature Immunology, 6, 1123-1132.

HASLETT, P. A. J., CORRAL, L. G., ALBERT, M. & KAPLAN, G. 1998. Thalidomide Costimulates Primary Human T Lymphocytes, Preferentially Inducing Proliferation, Cytokine Production, and Cytotoxic Responses in the CD8+ Subset. The Journal of Experimental Medicine, 187, 1885-1892.

HAYASHI, T., HIDESHIMA, T., AKIYAMA, M., PODAR, K., YASUI, H., RAJE, N., KUMAR, S., CHAUHAN, D., TREON, S. P., RICHARDSON, P. & ANDERSON, K. C. 2005. Molecular mechanisms whereby immunomodulatory drugs activate natural killer cells: clinical application. British Journal of Haematology, 128, 192-203.

HERNANDEZ-ILIZALITURRI, F. J., REDDY, N., HOLKOVA, B., OTTMAN, E. & CZUCZMAN, M. S. 2005. Immunomodulatory Drug CC-5013 or CC-4047 and Rituximab Enhance Antitumor Activity in a Severe Combined Immunodeficient Mouse Lymphoma Model. Clinical Cancer Research, 11, 5984-5992.

HIDESHIMA, T., RAJE, N., RICHARDSON, P. G. & ANDERSON, K. C. 2008. A review of lenalidomide in combination with dexamethasone for the treatment of multiple myeloma. Therapy and Clinical Risk Management, 4, 129-36.

HIDESHIMA, T., RICHARDSON, P., CHAUHAN, D., PALOMBELLA, V. J., ELLIOTT, P. J., ADAMS, J. & ANDERSON, K. C. 2001. The Proteasome Inhibitor PS-341 Inhibits Growth, Induces Apoptosis, and Overcomes Drug Resistance in Human Multiple Myeloma Cells. Cancer Research, 61, 3071-3076.

HJORTH, M., HELLQUIST, L., HOLMBERG, E., MAGNUSSON, B., RODJER, S. & WESTIN, J. 1993. Initial versus deferred melphalan-prednisone therapy for asymptomatic multiple myeloma stage I--a randomized study. Myeloma Group of Western Sweden. European Journal of Haematology, 50, 95-102.

HSU, A. K., QUACH, H., TAI, T., PRINCE, H. M., HARRISON, S. J., TRAPANI, J. A., SMYTH, M. J., NEESON, P. & RITCHIE, D. S. 2011. The immunostimulatory effect of lenalidomide on NK-cell function is profoundly inhibited by concurrent dexamethasone therapy. Blood, 117, 1605-13.

113

HU, Y., TIAN, W., ZHANG, L.-L., LIU, H., YIN, G.-P., HE, B.-S. & MAO, X.-M. 2012. Function of regulatory T-cells improved by dexamethasone in Graves' disease. European Journal of Endocrinology, 166, 641-646.

HYMOWITZ, S. G., FILVAROFF, E. H., YIN, J., LEE, J., CAI, L., RISSER, P., MARUOKA, M., MAO, W., FOSTER, J., KELLEY, R. F., PAN, G., GURNEY, A. L., DE VOS, A. M. & STAROVASNIK, M. A. 2001. IL-17s adopt a cystine knot fold: structure and activity of a novel cytokine, IL-17F, and implications for receptor binding. European Molecular Biology Organization Journal, 20, 5332-5341.

ICHIHARA, F., KONO, K., TAKAHASHI, A., KAWAIDA, H., SUGAI, H. & FUJII, H. 2003. Increased Populations of Regulatory T Cells in Peripheral Blood and Tumor-Infiltrating Lymphocytes in Patients with Gastric and Esophageal Cancers. Clinical Cancer Research, 9, 4404-4408.

IDLER, I., GIANNOPOULOS, K., ZENZ, T., BHATTACHARYA, N., NOTHING, M., DÖHNER, H., STILGENBAUER, S. & MERTENS, D. 2010. Lenalidomide treatment of chronic lymphocytic leukaemia patients reduces regulatory T cells and induces Th17 T helper cells. British Journal of Haematology, 148, 948-950.

INOZUME, T., HANADA, K., WANG, Q. J. & YANG, J. C. 2009. IL-17 secreted by tumor reactive T cells induces IL-8 release by human renal cancer cells. Journal of Immunotherapy, 32, 109-17.

IVANOV, I., MCKENZIE, B., ZHOU, L., TADOKORO, C., LEPELLEY, A., LAFAILLE, J., CUA, D. & LITTMAN, D. 2006. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell, 126, 1121-33.

JAVIA, L. R. & ROSENBERG, S. A. 2003. CD4+CD25+ Suppressor Lymphocytes in the Circulation of Patients Immunized Against Melanoma Antigens. Journal of Immunotherapy, 26, 85-93.

JI, Y. & ZHANG, W. 2010. Th17 cells: positive or negative role in tumor? Cancer Immunology Immunotherapy, 59, 979-987.

JONULEIT, H., SCHMITT, E., STASSEN, M., TUETTENBERG, A., KNOP, J. & ENK, A. H. 2001. Identification and Functional Characterization of Human Cd4+Cd25+ T Cells with Regulatory Properties Isolated from Peripheral Blood. The Journal of Experimental Medicine, 193, 1285-1294.

JOSHUA, D. E. 1996. Myeloma: New aspects of biology, prognosis and treatment. Pathology, 28, 2-7.

JOSHUA, D. E., BROWN, R. D. & GIBSON, J. 1994. Prognostic Factors in Myeloma: What They Tell Us About the Pathophysiology of the Disease. Leukemia & Lymphoma, 15, 375-381.

JOSHUA, D. E., BROWN, R. D., HO, P. J. & GIBSON, J. 2008. Regulatory T Cells and Multiple Myeloma Clinical Lymphoma, Myeloma & Leukemia, 8, 283-286.

KARUBE, K., OHSHIMA, K., TSUCHIYA, T., YAMAGUCHI, T., KAWANO, R., SUZUMIYA, J., UTSUNOMIYA, A., HARADA, M. & KIKUCHI, M. 2004. Expression of FoxP3, a key molecule in CD4+CD25+ regulatory T cells, in adult T-cell leukaemia/lymphoma cells. British Journal of Haematology, 126, 81-84.

114

KASTRITIS, E., PALUMBO, A. & DIMOPOULOS, M. A. 2009. Treatment of Relapsed/Refractory Multiple Myeloma. Seminars in Hematology, 46, 143-157.

KAY, N. E., LEONG, T. L., BONE, N., VESOLE, D. H., GREIPP, P. R., VAN NESS, B., OKEN, M. M. & KYLE, R. A. 2001. Blood levels of immune cells predict survival in myeloma patients: results of an Eastern Cooperative Oncology Group phase 3 trial for newly diagnosed multiple myeloma patients. Blood, 98, 23-28.

KING, I. L. & SEGAL, B. M. 2005. Cutting Edge: IL-12 Induces CD4+CD25 T Cell Activation in the Presence of T Regulatory Cells. The Journal of Immunology, 175, 641-645.

KOBAYASHI, M., FITZ, L., RYAN, M., HEWICK, R. M., CLARK, S. C., CHAN, S., LOUDON, R., SHERMAN, F., PERUSSIA, B. & TRINCHIERI, G. 1989. Identification and purification of natural killer cell stimulatory factor (NKSF), a cytokine with multiple biologic effects on human lymphocytes. Journal of Experimental Medicine, 170, 827-845.

KOEHNE, G. & GIRALT, S. 2012. Allogeneic hematopoietic stem cell transplantation for multiple myeloma: curative but not the standard of care. Current Opinion in Oncology, 24, 720-6.

KONO, K., KAWAIDA, H., TAKAHASHI, A., SUGAI, H., MIMURA, K., MIYAGAWA, N., OMATA, H. & FUJII, H. 2006. CD4(+)CD25high regulatory T cells increase with tumor stage in patients with gastric and esophageal cancers. Cancer Immunology Immunotherapy, 55, 1064-1071.

KORDE, N., KRISTINSSON, S. Y. & LANDGREN, O. 2011. Monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM): novel biological insights and development of early treatment strategies. Blood, 117, 5573-5581.

KOTLA, V., GOEL, S., NISCHAL, S., HEUCK, C., VIVEK, K., DAS, B. & VERMA, A. 2009. Mechanism of action of lenalidomide in hematological malignancies. Journal of Hematology and Oncology, 2, 36.

KOTTKE, T., SANCHEZ-PEREZ, L., DIAZ, R. M., THOMPSON, J., CHONG, H., HARRINGTON, K., CALDERWOOD, S. K., PULIDO, J., GEORGOPOULOS, N., SELBY, P., MELCHER, A. & VILE, R. 2007. Induction of hsp70-Mediated Th17 Autoimmunity Can Be Exploited as Immunotherapy for Metastatic Prostate Cancer. Cancer Research, 67, 11970-11979.

KRYCZEK, I., BANERJEE, M., CHENG, P., VATAN, L., SZELIGA, W., WEI, S., HUANG, E., FINLAYSON, E., SIMEONE, D., WELLING, T. H., CHANG, A., COUKOS, G., LIU, R. & ZOU, W. 2009a. Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments. Blood, 114, 1141-1149.

KRYCZEK, I., WEI, S., SZELIGA, W., VATAN, L. & ZOU, W. 2009b. Endogenous IL-17 contributes to reduced tumor growth and metastasis. Blood, 114, 357-359.

KRYCZEK, I., WEI, S., ZOU, L., ALTUWAIJRI, S., SZELIGA, W., KOLLS, J., CHANG, A. & ZOU, W. 2007. Cutting Edge: Th17 and Regulatory T Cell Dynamics and the Regulation by IL-2 in the Tumor Microenvironment. The Journal of Immunology, 178, 6730-6733.

115

KUHN, D. J., CHEN, Q., VOORHEES, P. M., STRADER, J. S., SHENK, K. D., SUN, C. M., DEMO, S. D., BENNETT, M. K., VAN LEEUWEN, F. W. B., CHANAN-KHAN, A. A. & ORLOWSKI, R. Z. 2007. Potent activity of carfilzomib, a novel, irreversible inhibitor of the ubiquitin-proteasome pathway, against preclinical models of multiple myeloma. Blood, 110, 3281-3290.

KYLE, R. A. & RAJKUMAR, S. V. 2008. Criteria for diagnosis, staging, risk stratification and response assessment of multiple myeloma. Leukemia, 23, 3-9.

KYLE, R. A., REMSTEIN, E. D., THERNEAU, T. M., DISPENZIERI, A., KURTIN, P. J., HODNEFIELD, J. M., LARSON, D. R., PLEVAK, M. F., JELINEK, D. F., FONSECA, R., MELTON, L. J. & RAJKUMAR, S. V. 2007. Clinical Course and Prognosis of Smoldering (Asymptomatic) Multiple Myeloma. New England Journal of Medicine, 356, 2582-2590.

LAI, J., ZANDECKI, M., MARY, J., BERNARDI, F., IZYDORCZYK, V., FLACTIF, M., MOREL, P., JOUET, J., BAUTERS, F. & FACON, T. 1995. Improved cytogenetics in multiple myeloma: a study of 151 patients including 117 patients at diagnosis. Blood, 85, 2490-2497.

LANDGREN, O., KYLE, R. A., PFEIFFER, R. M., KATZMANN, J. A., CAPORASO, N. E., HAYES, R. B., DISPENZIERI, A., KUMAR, S., CLARK, R. J., BARIS, D., HOOVER, R. & RAJKUMAR, S. V. 2009. Monoclonal gammopathy of undetermined significance (MGUS) consistently precedes multiple myeloma: a prospective study. Blood, 113, 5412-5417.

LAW, J. P., HIRSCHKORN, D. F., OWEN, R. E., BISWAS, H. H., NORRIS, P. J. & LANTERI, M. C. 2009. The importance of Foxp3 antibody and fixation/permeabilization buffer combinations in identifying CD4+CD25+Foxp3+ regulatory T cells. Cytometry Part A, 75A, 1040-1050.

LEE, A. M., CLEAR, A. J., CALAMINICI, M., DAVIES, A. J., JORDAN, S., MACDOUGALL, F., MATTHEWS, J., NORTON, A. J., GRIBBEN, J. G., LISTER, T. A. & GOFF, L. K. 2006. Number of CD4+ cells and location of forkhead box protein P3-positive cells in diagnostic follicular lymphoma tissue microarrays correlates with outcome. Journal of Clinical Oncology, 24, 5052-5059.

LEE, B.-N., GAO, H., COHEN, E. N., BADOUX, X., WIERDA, W. G., ESTROV, Z., FADERL, S. H., KEATING, M. J., FERRAJOLI, A. & REUBEN, J. M. 2011. Treatment with lenalidomide modulates T-cell immunophenotype and cytokine production in patients with chronic lymphocytic leukemia. Cancer, 117, 3999-4008.

LENNARD, A. L. & JACKSON, G. H. 2000. Stem cell transplantation. British Medical Journal, 321, 433-437.

LENZ, W., PFEIFFER, R. A., KOSENOW, W. & HAYMAN, D. J. 1962. Thalidomide and congenital abnormalities. The Lancet, 279, 45-46.

LI, M. O., WAN, Y. Y., SANJABI, S., ROBERTSON, A.-K. L. & FLAVELL, R. A. 2006. Transforming growth factor- regulation of immune responses. Annual Review of Immunology, 24, 99-146.

LIN, Y.-C., CHANG, L.-Y., HUANG, C.-T., PENG, H.-M., DUTTA, A., CHEN, T.-C., YEH, C.-T. & LIN, C.-Y. 2009. Effector/Memory but Not Naive Regulatory T Cells Are Responsible for the Loss of Concomitant Tumor Immunity. The Journal of Immunology, 182, 6095-6104.

116

LIU, W., PUTNAM, A. L., XU-YU, Z., SZOT, G. L., LEE, M. R., ZHU, S., GOTTLIEB, P. A., KAPRANOV, P., GINGERAS, T. R., DE ST. GROTH, B. F., CLAYBERGER, C., SOPER, D. M., ZIEGLER, S. F. & BLUESTONE, J. A. 2006. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. The Journal of Experimental Medicine, 203, 1701-1711.

LIYANAGE, U., MOORE, T., JOO, H.-G., TANAKA, Y., HERRMANN, V., GERARD, D., DREBIN, J. A., STRASBERG, S. M., EBERLEIN, T. J., GOEDEGEBUURE, P. S. & LINEHAN, D. C. 2002. Prevalence of Regulatory T Cells Is Increased in Peripheral Blood and Tumor Microenvironment of Patients with Pancreas or Breast Adenocarcinoma. The Journal of Immunology, 169, 2756-2761.

LOCKHART, E., GREEN, A. M. & FLYNN, J. L. 2006. IL-17 Production Is Dominated by T Cells rather than CD4 T Cells during Mycobacterium tuberculosis Infection. The Journal of Immunology, 177, 4662-4669.

LOTEM, M., MERIMS, S., FRANK, S., OSPOVAT, I. & PERETZ, T. 2012. Ctla-4 blockade: a new hope for the immunotherapy of malignant melanoma. Harefuah, 151, 585-8.

MACKALL, C. L., FLEISHER, T. A., BROWN, M. R., MAGRATH, I. T., SHAD, A. T., HOROWITZ, M. E., WEXLER, L. H., ADDE, M. A., MCCLURE, L. L. & GRESS, R. E. 1994. Lymphocyte depletion during treatment with intensive chemotherapy for cancer. Blood, 84, 2221-8.

MAECKER, B., ANDERSON, K. S., VON BERGWELT-BAILDON, M. S., WELLER, E., VONDERHEIDE, R. H., RICHARDSON, P. G., SCHLOSSMAN, R. L., MENEZES, I. A., XIA, Z., MUNSHI, N. C., ANDERSON, K. C., NADLER, L. M. & SCHULTZE, J. L. 2003. Viral antigen-specific CD8+ T-cell responses are impaired in multiple myeloma. British Journal of Haematology, 121, 842-848.

MANGAN, P. R., HARRINGTON, L. E., O'QUINN, D. B., HELMS, W. S., BULLARD, D. C., ELSON, C. O., HATTON, R. D., WAHL, S. M., SCHOEB, T. R. & WEAVER, C. T. 2006. Transforming growth factor-[beta] induces development of the TH17 lineage. Nature, 441, 231-234.

MARC, S. R., KLAUS, P., IRIS, B., PAUL, G. R. & KENNETH, C. A. 2009. Multiple myeloma. The Lancet, 374, 324-339.

MARIE, J. C., LETTERIO, J. J., GAVIN, M. & RUDENSKY, A. Y. 2005. TGF- 1 maintains suppressor function and Foxp3 expression in CD4+CD25+ regulatory T cells. The Journal of Experimental Medicine, 201, 1061-1067.

MARSHALL, N. A., CHRISTIE, L. E., MUNRO, L. R., CULLIGAN, D. J., JOHNSTON, P. W., BARKER, R. N. & VICKERS, M. A. 2004. Immunosuppressive regulatory T cells are abundant in the reactive lymphocytes of Hodgkin lymphoma. Blood, 103, 1755-1762.

MASSAGUÉ, J. 2008. TGF in Cancer. Cell, 134, 215-230.

MASSAIA, M., BORRIONE, P., ATTISANO, C., BARRAL, P., BEGGIATO, E., MONTACCHINI, L., BIANCHI, A., BOCCADORO, M. & PILERI, A. 1995. Dysregulated Fas and Bcl-2 expression leading to enhanced apoptosis in T cells of multiple myeloma patients. Blood, 85, 3679-3687.

117

MATEOS, M.-V., HERNÁNDEZ, M.-T., GIRALDO, P., DE LA RUBIA, J., DE ARRIBA, F., CORRAL, L. L., ROSIÑOL, L., PAIVA, B., PALOMERA, L., BARGAY, J., ORIOL, A., PROSPER, F., LÓPEZ, J., OLAVARRÍA, E., QUINTANA, N., GARCÍA, J.-L., BLADÉ, J., LAHUERTA, J.-J. & SAN MIGUEL, J.-F. 2013. Lenalidomide plus Dexamethasone for High-Risk Smoldering Multiple Myeloma. New England Journal of Medicine, 369, 438-447

MATSUMOTO, T. & ABE, M. 2011. TGF-[beta]-related mechanisms of bone destruction in multiple myeloma. Bone,48, 129-34.

MCHUGH, R. S., WHITTERS, M. J., PICCIRILLO, C. A., YOUNG, D. A., SHEVACH, E. M., COLLINS, M. & BYRNE, M. C. 2002. CD4+CD25+ Immunoregulatory T Cells: Gene Expression Analysis Reveals a Functional Role for the Glucocorticoid-Induced TNF Receptor. Immunity, 16, 311-323.

MILLS, K. H. G. & CAWLEY, J. C. 1983. Abnormal monoclonal antibody-defined helper/suppressor T-cell subpopulations in multiple myeloma: relationship to treatment and clinical stage. British Journal of Haematology, 53, 271-275.

MINNEMA, M. C., VAN DER VEER, M., AARTS, T., EMMELOT, M., MUTIS, T. & LOKHORST, H. M. 2007. Lenalidomide Is Highly Effective in Patients with Relapsed Multiple Myeloma Following Allogeneic Stem Cell Transplantation and Increases the Frequency of CD4+FOXP3+ T Cells. ASH Annual Meeting Abstracts, 110, 2722.

MITSIADES, N., MITSIADES, C. S., POULAKI, V., CHAUHAN, D., RICHARDSON, P. G., HIDESHIMA, T., MUNSHI, N., TREON, S. P. & ANDERSON, K. C. 2002. Biologic sequelae of nuclear factor-kappa B blockade in multiple myeloma: therapeutic applications. Blood, 99, 4079-4086.

MITSIADES, N., MITSIADES, C. S., RICHARDSON, P. G., POULAKI, V., TAI, Y.-T., CHAUHAN, D., FANOURAKIS, G., GU, X., BAILEY, C., JOSEPH, M., LIBERMANN, T. A., SCHLOSSMAN, R., MUNSHI, N. C., HIDESHIMA, T. & ANDERSON, K. C. 2003. The proteasome inhibitor PS-341 potentiates sensitivity of multiple myeloma cells to conventional chemotherapeutic agents: therapeutic applications. Blood, 101, 2377-2380.

MIYAHARA, Y., ODUNSI, K., CHEN, W., PENG, G., MATSUZAKI, J. & WANG, R.-F. 2008. Generation and regulation of human CD4+ IL-17-producing T cells in ovarian cancer. Proceedings of the National Academy of Sciences, 105, 15505-15510.

MOTTA, M., RASSENTI, L., SHELVIN, B. J., LERNER, S., KIPPS, T. J., KEATING, M. J. & WIERDA, W. G. 2005. Increased expression of CD152 (CTLA-4) by normal T lymphocytes in untreated patients with B-cell chronic lymphocytic leukemia. Leukemia, 19, 1788-1793.

MURAKAMI, M., SAKAMOTO, A., BENDER, J., KAPPLER, J. & MARRACK, P. 2002. CD25+CD4+ T cells contribute to the control of memory CD8+ T cells. Proceedings of the National Academy of Sciences, 99, 8832-8837.

MURANSKI, P., BONI, A., ANTONY, P. A., CASSARD, L., IRVINE, K. R., KAISER, A., PAULOS, C. M., PALMER, D. C., TOULOUKIAN, C. E., PTAK, K., GATTINONI, L., WRZESINSKI, C., HINRICHS, C. S., KERSTANN, K. W., FEIGENBAUM, L., CHAN, C.-C. & RESTIFO, N. P. 2008. Tumor-specific Th17-polarized cells eradicate large established melanoma. Blood, 112, 362-373.

118

MUTHU RAJA, K. R., RIHOVA, L., ZAHRADOVA, L., KLINCOVA, M., PENKA, M. & HAJEK, R. 2012. Increased T Regulatory Cells Are Associated with Adverse Clinical Features and Predict Progression in Multiple Myeloma. PLoS ONE, 7, e47077.

NAKAE, S., KOMIYAMA, Y., NAMBU, A., SUDO, K., IWASE, M., HOMMA, I., SEKIKAWA, K., ASANO, M. & IWAKURA, Y. 2002. Antigen-Specific T Cell Sensitization Is Impaired in IL-17-Deficient Mice, Causing Suppression of Allergic Cellular and Humoral Responses. Immunity, 17, 375-387.

NG, W. F., DUGGAN, P. J., PONCHEL, F., MATARESE, G., LOMBARDI, G., EDWARDS, A. D., ISAACS, J. D. & LECHLER, R. I. 2001. Human CD4+CD25+ cells: a naturally occurring population of regulatory T cells. Blood, 98, 2736-2744.

NOONAN, K., MARCHIONNI, L., ANDERSON, J., PARDOLL, D., ROODMAN, G. D. & BORRELLO, I. 2010. A novel role of IL-17-producing lymphocytes in mediating lytic bone disease in multiple myeloma. Blood, 116, 3554-3563.

NOONAN, K. A., MGEBROFF, S., SERAFINI, P., MECKEL, K., BONYHADI, M. & BORRELLO, I. 2006. CD4+CD25+ Marrow Infiltrating Lymphocytes in Myeloma Patients Display an Activated Phenotype and Lack Suppressive Function. ASH Annual Meeting Abstracts, 108, 1741.

ORMANDY, L. A., HILLEMANN, T., WEDEMEYER, H., MANNS, M. P., GRETEN, T. F. & KORANGY, F. 2005. Increased Populations of Regulatory T Cells in Peripheral Blood of Patients with Hepatocellular Carcinoma. Cancer Research, 65, 2457-2464.

OUKKA, M. 2008. Th17 cells in immunity and autoimmunity. Annals of the Rheumatic Diseases, 67 Suppl 3, iii26-9.

PARDOLL, D. M. 2002. Spinning molecular immunology into successful immunotherapy. Nature Reviews Immunology, 2, 227-238.

PARK, H., LI, Z., YANG, X., CHANG, S., NURIEVA, R., WANG, Y., WANG, Y., HOOD, L., ZHU, Z., TIAN, Q. & DONG, C. 2005. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nature Immunology, 6, 1133-41.

PASQUINI, M. C. 2008. Impact of graft-versus-host disease on survival. Best practice & research. Clinical haematology, 21, 193-204.

PECK, A. & MELLINS, E. D. 2010. Plasticity of T-cell phenotype and function: the T helper type 17 example. Immunology, 129, 147-153.

PILARSKI, L. M., ANDREWS, E. J., SERRA, H. M., RUETHER, B. A. & MANT, M. J. 1989. Comparative Analysis of Immunodeficiency in Patients with Monoclonal Gammopathy of Undetermined Significance and Patients with Untreated Multiple Myeloma. Scandinavian Journal of Immunology, 29, 217-228.

POPE, B., BROWN, R., GIBSON, J. & JOSHUA, D. 1999. The bone marrow plasma cell labeling index by flow cytometry. Cytometry, 38, 286-292.

PRABHALA, R. H., NERI, P., BAE, J. E., TASSONE, P., SHAMMAS, M. A., ALLAM, C. K., DALEY, J. F., CHAUHAN, D., BLANCHARD, E., THATTE, H. S., ANDERSON, K. C. & MUNSHI, N. C. 2006. Dysfunctional T regulatory cells in multiple myeloma. Blood, 107, 301-304.

119

PRABHALA, R. H., PELLURU, D., FULCINITI, M., PRABHALA, H. K., NANJAPPA, P., SONG, W., PAI, C., AMIN, S., TAI, Y.-T., RICHARDSON, P. G., GHOBRIAL, I. M., TREON, S. P., DALEY, J. F., ANDERSON, K. C., KUTOK, J. L. & MUNSHI, N. C. 2010. Elevated IL-17 produced by TH17 cells promotes myeloma cell growth and inhibits immune function in multiple myeloma. Blood, 115, 5385-5392.

PRADO, C., GÓMEZ, J., LÓPEZ, P., DE PAZ, B., GUTIÉRREZ, C. & SUÁREZ, A. 2011. Dexamethasone upregulates FOXP3 expression without increasing regulatory activity. Immunobiology, 216, 386-392.

QUACH, H. & PRINCE, M. 2012. Clinical Practice Guildeline: Multiple Myeloma, Sydney.

QUACH, H., RITCHIE, D., NEESON, P., HARRISON, S., TAI, T., TAINTON, K., LYNCH, K. & PRINCE, M. 2008. Regulatory T Cells (Treg) Are Depressed in Patients with Relapsed/Refractory Multiple Myeloma (MM) and Increases towards Normal Range in Responding Patients Treated with Lenalidomide (LEN). ASH Annual Meeting Abstracts, 112, 1696.

QUACH, H., RITCHIE, D., STEWART, A. K., NEESON, P., HARRISON, S., SMYTH, M. J. & PRINCE, H. M. 2009. Mechanism of action of immunomodulatory drugs (IMiDS) in multiple myeloma. Leukemia, 24, 22-32.

RAITAKARI, M., BROWN, R. D., GIBSON, J. & JOSHUA, D. E. 2003. T cells in myeloma. Hematological Oncology, 21, 33-42.

RAJA, K. R. M., ZAHRADOVA, L., KOVAROVA, L., MIKULKOVA, Z., BURESOVA, I., MICHALEK, J. & HAJEK, R. 2009. Flow Cytometric Phenotyping and Analysis of T Regulatory Cells in Monoclonal Gammopathy Patients. ASH Annual Meeting Abstracts, 114, 2819-.

RAJKUMAR, S. V., JACOBUS, S., CALLANDER, N. S., FONSECA, R., VESOLE, D. H., WILLIAMS, M. E., ABONOUR, R., SIEGEL, D. S., KATZ, M. & GREIPP, P. R. 2010. Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma: an open-label randomised controlled trial. Lancet Oncology, 11, 29-37.

RAJKUMAR, S. V. & KYLE, R. A. 2005. Multiple Myeloma: Diagnosis and Treatment. Mayo Clinic Proceedings, 80, 1371-1382.

REZVANI, K., MIELKE, S., AHMADZADEH, M., KILICAL, Y., SAVANI, B. N., ZEILAH, J., KEYVANFAR, K., MONTERO, A., HENSEL, N., KURLANDER, R. & BARRETT, A. J. 2006. High donor FOXP3-positive regulatory T-cell (Treg) content is associated with a low risk of GVHD following HLA-matched allogeneic SCT. Blood, 108, 1291-1297.

RICCARDI, A., MORA, O., TINELLI, C., VALENTINI, D., BRUGNATELLI, S., SPANEDDA, R., DE PAOLI, A., BARBARANO, L., DI STASI, M., GIORDANO, M., DELFINI, C., NICOLETTI, G., BERGONZI, C., RINALDI, E., PICCININI, L. & ASCARI, E. 2000. Long-term survival of stage I multiple myeloma given chemotherapy just after diagnosis or at progression of the disease: a multicentre randomized study. Cooperative Group of Study and Treatment of Multiple Myeloma. British Journal of Cancer, 82, 1254-60.

120

RICHARDSON, P., JAGANNATH, S., HUSSEIN, M., BERENSON, J., SINGHAL, S., IRWIN, D., WILLIAMS, S. F., BENSINGER, W., BADROS, A. Z., VESCIO, R., KENVIN, L., YU, Z., OLESNYCKYJ, M., ZELDIS, J., KNIGHT, R. & ANDERSON, K. C. 2009. Safety and efficacy of single-agent lenalidomide in patients with relapsed and refractory multiple myeloma. Blood, 114, 772-8.

RICHARDSON, P., MITSIADES, C., SCHLOSSMAN, R., GHOBRIAL, I., HIDESHIMA, T., CHAUHAN, D., MUNSHI, N. & ANDERSON, K. 2007. The Treatment of Relapsed and Refractory Multiple Myeloma. ASH Education Program Book, 2007, 317-323.

ROUVIER, E., LUCIANI, M. F., MATTÉI, M. G., DENIZOT, F. & GOLSTEIN, P. 1993. CTLA-8, cloned from an activated T cell, bearing AU-rich messenger RNA instability sequences, and homologous to a herpesvirus saimiri gene. The Journal of Immunology, 150, 5445-56.

RYFFEL, B. 1997. Interleukin-12: Role of interferon- in IL-12 adverse effects. Clinical Immunology and Immunopathology, 83, 18-20.

SAGAR, L. 2010. Presentation and risk stratification – improving prognosis for patients with multiple myeloma. Cancer treatment reviews, 36, S12-S17.

SAKAGUCHI, S. 2004. Naturally Arising CD4+ Regulatory T Cells for Immunologic Self-Tolerance and Negative Control of Immune Responses. Annual Review of Immunology, 22, 531-562.

SAKAGUCHI, S., SAKAGUCHI, N., ASANO, M., ITOH, M. & TODA, M. 1995. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. The Journal of Immunology, 155, 1151-1164.

SAKAGUCHI, S., SAKAGUCHI, N., SHIMIZU, J., YAMAZAKI, S., SAKIHAMA, T., ITOH, M., KUNIYASU, Y., NOMURA, T., TODA, M. & TAKAHASHI, T. 2001. Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunological Reviews, 182, 18-32.

SASADA, T., KIMURA, M., YOSHIDA, Y., KANAI, M. & TAKABAYASHI, A. 2003. CD4+CD25+ regulatory T cells in patients with gastrointestinal malignancies: possible involvement of regulatory T cells in disease progression. Cancer, 98, 1089-1099.

SCHAEFER, C., KIM, G. G., ALBERS, A., HOERMANN, K., MYERS, E. N. & WHITESIDE, T. L. 2005. Characteristics of CD4+CD25+ regulatory T cells in the peripheral circulation of patients with head and neck cancer. British Journal of Cancer, 92, 913-920.

SEDDIKI, N., SANTNER-NANAN, B., MARTINSON, J., ZAUNDERS, J., SASSON, S., LANDAY, A., SOLOMON, M., SELBY, W., ALEXANDER, S. I., NANAN, R., KELLEHER, A. & DE ST. GROTH, B. F. 2006. Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. The Journal of Experimental Medicine, 203, 1693-1700.

SFANOS, K. S., BRUNO, T. C., MARIS, C. H., XU, L., THOBURN, C. J., DEMARZO, A. M., MEEKER, A. K., ISAACS, W. B. & DRAKE, C. G. 2008. Phenotypic Analysis of Prostate-Infiltrating Lymphocytes Reveals TH17 and Treg Skewing. Clinical Cancer Research, 14, 3254-3261.

121

SHEN, C. J., YUAN, Z. H., LIU, Y. X. & HU, G. Y. 2012. Increased numbers of T helper 17 cells and the correlation with clinicopathological characteristics in multiple myeloma. The Journal of International Medicine Research, 40, 556-64.

SHEVACH, E. M. 2002. CD4+CD25+ suppressor T cells: more questions than answers. Nature Reviews Immunology, 2, 389-400.

SHIN, H. C., BENBERNOU, N., ESNAULT, S. & GUENOUNOU, M. 1999. Expression of IL-17 in human memory CD45RO+ T lymphocytes and its regulation by protein kinase A pathway. Cytokine, 11, 257-66.

SINGHAL, S., MEHTA, J., DESIKAN, R., AYERS, D., ROBERSON, P., EDDLEMON, P., MUNSHI, N., ANAISSIE, E., WILSON, C., DHODAPKAR, M., ZELDIS, J., SIEGEL, D., CROWLEY, J. & BARLOGIE, B. 1999. Antitumor Activity of Thalidomide in Refractory Multiple Myeloma. New England Journal of Medicine, 341, 1565-1571.

SMADJA, N. V., BASTARD, C., BRIGAUDEAU, C., LEROUX, D. & FRUCHART, C. 2001. Hypodiploidy is a major prognostic factor in multiple myeloma. Blood, 98, 2229-2238.

SPEIRS, A. L. 1962. Thalidomide and congenital abnormalities. The Lancet, 279, 303-305.

SPENCER, A., PRINCE, H. M., ROBERTS, A. W., PROSSER, I. W., BRADSTOCK, K. F., COYLE, L., GILL, D. S., HORVATH, N., REYNOLDS, J. & KENNEDY, N. 2009. Consolidation therapy with low-dose thalidomide and prednisolone prolongs the survival of multiple myeloma patients undergoing a single autologous stem-cell transplantation procedure. Journal of Clinical Oncology, 27, 1788-93.

STADTMAUER, E. A., WEBER, D. M., NIESVIZKY, R., BELCH, A., PRINCE, M. H., SAN MIGUEL, J. F., FACON, T., OLESNYCKYJ, M., YU, Z., ZELDIS, J. B., KNIGHT, R. D. & DIMOPOULOS, M. A. 2009. Lenalidomide in combination with dexamethasone at first relapse in comparison with its use as later salvage therapy in relapsed or refractory multiple myeloma. European Journal of Haematology, 82, 426-432.

STERN, A. S., PODLASKI, F. J., HULMES, J. D., PAN, Y. C. E., QUINN, P. M., WOLITZKY, A. G., FAMILLETTI, P. C., STREMLO, D. L., TRUITT, T., CHIZZONITE, R. & GATELY, M. K. 1990. Purification to homogeneity and partial characterization of cytotoxic lymphocyte maturation factor from human B-lymphoblastoid cells. Proceedings of the National Academy of Sciences of the United States of America, 87, 6808-6812.

SU, X., YE, J., HSUEH, E. C., ZHANG, Y., HOFT, D. F. & PENG, G. 2010. Tumor Microenvironments Direct the Recruitment and Expansion of Human Th17 Cells. The Journal of Immunology, 184, 1630-1641.

SWERDLOW, S., CAMPO, E., HARRIS, N. L., JAFFE, E. S., PILERI, S. A., STEIN, H., THIELE, J. & VARDIMAN, J. W. (eds.) 2008. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues, Lyon.

SZCZEPANSKI, M. J., SZAJNIK, M., CZYSTOWSKA, M., MANDAPATHIL, M., STRAUSS, L., WELSH, A., FOON, K. A., WHITESIDE, T. L. & BOYIADZIS, M. 2009. Increased Frequency and Suppression by Regulatory T Cells in Patients with Acute Myelogenous Leukemia. Clinical Cancer Research, 15, 3325-3332.

122

TAHARA, H. & LOTZE, M. T. 1995. Antitumor effects of interleukin-12 (IL-12): applications for the immunotherapy and gene therapy of cancer. Gene Therapy, 2, 96-106.

TAHARA, H., ZITVOGEL, L., STORKUS, W. J., ZEH, H. J., 3RD, MCKINNEY, T. G., SCHREIBER, R. D., GUBLER, U., ROBBINS, P. D. & LOTZE, M. T. 1995. Effective eradication of established murine tumors with IL-12 gene therapy using a polycistronic retroviral vector. Journal of Immunology, 154, 6466-74.

TAKAHASHI, T., TAGAMI, T., YAMAZAKI, S., UEDE, T., SHIMIZU, J., SAKAGUCHI, N., MAK, T. W. & SAKAGUCHI, S. 2000. Immunologic Self-Tolerance Maintained by Cd25+Cd4+Regulatory T Cells Constitutively Expressing Cytotoxic T Lymphocyte–Associated Antigen 4. The Journal of Experimental Medicine, 192, 303-310.

TANG, Q., BODEN, E. K., HENRIKSEN, K. J., BOUR-JORDAN, H., BI, M. & BLUESTONE, J. A. 2004. Distinct roles of CTLA-4 and TGF- in CD4+CD25+ regulatory T cell function. European Journal of Immunology, 34, 2996-3005.

TAYLOR, P. A., LEES, C. J. & BLAZAR, B. R. 2002. The infusion of ex vivo activated and expanded CD4+CD25+ immune regulatory cells inhibits graft-versus-host disease lethality. Blood, 99, 3493-3499.

TAYLOR, P. A., PANOSKALTSIS-MORTARI, A., SWEDIN, J. M., LUCAS, P. J., GRESS, R. E., LEVINE, B. L., JUNE, C. H., SERODY, J. S. & BLAZAR, B. R. 2004. L-Selectinhi but not the L-selectinlo CD4+25+ T-regulatory cells are potent inhibitors of GVHD and BM graft rejection. Blood, 104, 3804-3812.

TEO, S. 2005. Properties of thalidomide and its analogues: Implications for anticancer therapy. The AAPS Journal, 7, E14-E19.

TESMER, L. A., LUNDY, S. K., SARKAR, S. & FOX, D. A. 2008. Th17 cells in human disease. Immunological reviews, 223, 87-113.

TRAVES, S. L. & DONNELLY, L. E. 2008. Th17 cells in airway diseases. Current Molecular Medicine, 8, 416-26.

TRICOT, G., BARLOGIE, B., JAGANNATH, S., BRACY, D., MATTOX, S., VESOLE, D., NAUCKE, S. & SAWYER, J. 1995. Poor prognosis in multiple myeloma is associated only with partial or complete deletions of chromosome 13 or abnormalities involving 11q and not with other karyotype abnormalities. Blood, 86, 4250-4256.

TRICOT, G., VESOLE, D., JAGANNATH, S., HILTON, J., MUNSHI, N. & BARLOGIE, B. 1996. Graft-versus-myeloma effect: proof of principle. Blood, 87, 1196-1198.

VELDHOEN, M., HOCKING, R. J., ATKINS, C. J., LOCKSLEY, R. M. & STOCKINGER, B. 2006. TGF² in the Context of an Inflammatory Cytokine Milieu Supports De Novo Differentiation of IL-17-Producing T Cells. Immunity, 24, 179-189.

VIGUIER, M., LEMAÎTRE, F., VEROLA, O., CHO, M.-S., GOROCHOV, G., DUBERTRET, L., BACHELEZ, H., KOURILSKY, P. & FERRADINI, L. 2004. Foxp3 Expressing CD4+CD25high Regulatory T Cells Are Overrepresented in Human Metastatic Melanoma Lymph Nodes and Inhibit the Function of Infiltrating T Cells. The Journal of Immunology, 173, 1444-1453.

123

VOGELSANG, G. B., FARMER, E. R., HESS, A. D., ALTAMONTE, V., BESCHORNER, W. E., JABS, D. A., CORIO, R. L., LEVIN, L. S., COLVIN, O. M., WINGARD, J. R. & SANTOS, G. W. 1992. Thalidomide for the Treatment of Chronic Graft-versus-Host Disease. New England Journal of Medicine, 326, 1055-1058.

WANG, X., ZHENG, J., LIU, J., YAO, J., HE, Y., LI, X., YU, J., YANG, J., LIU, Z. & HUANG, S. 2005. Increased population of CD4+CD25high regulatory T cells with their higher apoptotic and proliferating status in peripheral blood of acute myeloid leukemia patients. European Journal of Haematology, 75, 468-476.

WEISS, B. M., ABADIE, J., VERMA, P., HOWARD, R. S. & KUEHL, W. M. 2009. A monoclonal gammopathy precedes multiple myeloma in most patients. Blood, 113, 5418-5422.

WELFARE, A. I. O. H. A. 2010. Australian Cancer Incidence and Mortality Books: Myeloma for Australia [Online]. Canberra. Available: http://www.aihw.gov.au/acim-books/.

WIGGINTON, J. M., GRUYS, E., GEISELHART, L., SUBLESKI, J., KOMSCHLIES, K. L., PARK, J.-W., WILTROUT, T. A., NAGASHIMA, K., BACK, T. C. & WILTROUT, R. H. 2001. IFN- and Fas/FasL are required for the antitumor and antiangiogenic effects of IL-12/pulse IL-2 therapy. The Journal of Clinical Investigation, 108, 51-62.

WOLF, A. M., WOLF, D., STEURER, M., GASTL, G., GUNSILIUS, E. & GRUBECK-LOEBENSTEIN, B. 2003. Increase of Regulatory T Cells in the Peripheral Blood of Cancer Patients. Clinical Cancer Research, 9, 606-612.

WOO, E. Y., CHU, C. S., GOLETZ, T. J., SCHLIENGER, K., YEH, H., COUKOS, G., RUBIN, S. C., KAISER, L. R. & JUNE, C. H. 2001. Regulatory CD4+CD25+ T Cells in Tumors from Patients with Early-Stage Non-Small Cell Lung Cancer and Late-Stage Ovarian Cancer. Cancer Research, 61, 4766-4772.

WRÓBEL, T., MAZUR, G., JAZWIEC, B. & KULICZKOWSKI, K. 2003. Interleukin-17 in acute myeloid leukemia. Journal of Cellular and Molecular Medicine, 7, 472-474.

WU, C., WANG, S., WANG, F., CHEN, Q., PENG, S., ZHANG, Y., QIAN, J., JIN, J. & XU, H. 2009. Increased frequencies of T helper type 17 cells in the peripheral blood of patients with acute myeloid leukaemia. Clinical & Experimental Immunology, 158, 199-204.

WUILLEME, S., ROBILLARD, N., LODE, L., MAGRANGEAS, F., BERIS, H., HAROUSSEAU, J. L., PROFFITT, J., MINVIELLE, S. & AVET-LOISEAU, H. 2004. Ploidy, as detected by fluorescence in situ hybridization, defines different subgroups in multiple myeloma. Leukemia, 19, 275-278.

XU, M., MIZOGUCHI, I., MORISHIMA, N., CHIBA, Y., MIZUGUCHI, J. & YOSHIMOTO, T. 2010. Regulation of Antitumor Immune Responses by the IL-12 Family Cytokines, IL-12, IL-23, and IL-27. Clinical and Developmental Immunology, 2010.

YAMADA, H. 2009. Th17 cells in human rheumatoid arthritis. Nihon Rinsho Meneki Gakkai Kaishi, 32, 249-55.

YANG, Z.-Z., NOVAK, A. J., STENSON, M. J., WITZIG, T. E. & ANSELL, S. M. 2006. Intratumoral CD4+CD25+ regulatory T-cell-mediated suppression of infiltrating CD4+ T cells in B-cell non-Hodgkin lymphoma. Blood, 107, 3639-3646.

124

YANG, Z.-Z., NOVAK, A. J., ZIESMER, S. C., WITZIG, T. E. & ANSELL, S. M. 2009. Malignant B Cells Skew the Balance of Regulatory T Cells and TH17 Cells in B-Cell Non-Hodgkin's Lymphoma. Cancer Research, 69, 5522-5530.

YANO, H., ISHIDA, T., INAGAKI, A., ISHII, T., KUSUMOTO, S., KOMATSU, H., IIDA, S., UTSUNOMIYA, A. & UEDA, R. 2007. Regulatory T-cell function of adult T-cell leukemia/lymphoma cells. International Journal of Cancer, 120, 2052-2057.

ZDZISI SKA, B., BOJARSKA-JUNAK, A., DMOSZY SKA, A. & KANDEFER-SZERSZE , M. 2008. Abnormal cytokine production by bone marrow stromal cells of multiple myeloma patients in response to RPMI8226 myeloma cells. Archivum Immunologiae et Therapiae Experimentalis, 56, 207-221.

ZHAN, F., HUANG, Y., COLLA, S., STEWART, J. P., HANAMURA, I., GUPTA, S., EPSTEIN, J., YACCOBY, S., SAWYER, J., BURINGTON, B., ANAISSIE, E., HOLLMIG, K., PINEDA-ROMAN, M., TRICOT, G., VAN RHEE, F., WALKER, R., ZANGARI, M., CROWLEY, J., BARLOGIE, B. & SHAUGHNESSY, J. D., JR 2006. The molecular classification of multiple myeloma. Blood, 108, 2020-2028.

ZHANG, B., RONG, G., WEI, H., ZHANG, M., BI, J., MA, L., XUE, X., WEI, G., LIU, X. & FANG, G. 2008. The prevalence of Th17 cells in patients with gastric cancer. Biochemical and Biophysical Research Communications, 374, 533-537.

ZHANG, J.-P., YAN, J., XU, J., PANG, X.-H., CHEN, M.-S., LI, L., WU, C., LI, S.-P. & ZHENG, L. 2009. Increased intratumoral IL-17-producing cells correlate with poor survival in hepatocellular carcinoma patients. Journal of Hepatology, 50, 980-989.

ZHANG, X. G., BATAILLE, R., WIDJENES, J. & KLEIN, B. 1992. Interleukin-6 dependence of advanced malignant plasma cell dyscrasias. Cancer, 69, 1373-6.

ZHAO, J., ZHAO, J. & PERLMAN, S. 2012. Differential Effects of IL-12 on Tregs and Non-Treg T Cells: Roles of IFN- , IL-2 and IL-2R. PLoS ONE, 7, e46241.