266
Eradication of established murine mesothelioma tumours by combined immunotherapy Shruti Krishnan, B.Sc (Micro), Grad Dip (ForenSc), M.Sc (Micro), M (ForSci) This thesis is presented for the degree of Doctor of Philosophy The University of Western Australia School of Pathology and Laboratory Medicine Faculty of Medicine, Dentistry and Health Sciences 2015

Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

Eradication of established murine mesothelioma tumours by combined immunotherapy

Shruti Krishnan, B.Sc (Micro), Grad Dip (ForenSc), M.Sc (Micro), M (ForSci)

This thesis is presented for the degree of Doctor of Philosophy The University of Western Australia

School of Pathology and Laboratory Medicine Faculty of Medicine, Dentistry and Health Sciences

2015

Page 2: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti
Page 3: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

iii

ABSTRACT

Despite the recent progress made in applying immunotherapy for the treatment

of various cancers, such a therapy has not been developed for treating mesothelioma.

Mesothelioma has a latency period of 30-40 years with a median survival being 9-10

months from diagnosis. Due to the lack of early diagnostic capabilities, patients in

general, present with well-advanced disease that renders many therapeutic options

ineffective. Standard treatments for the management of mesothelioma are limited due to

late diagnoses, significant toxicities and the occurrences of relapses. Targeted therapies

such as immunotherapy are selective and are aimed at stimulating the host’s immune

system to produce complete tumour destruction. The work described in this thesis, and

the work of others in both mouse tumour models and clinical settings has shown that

simultaneous targeting of multiple immune mechanisms results in better outcomes.

A major deterrent to boosting anti-tumour immune activity was the

immunosuppressive environment within the growing tumours themselves. Work carried

out by our laboratory, and others, defined the role of Tregs and TGF-β in suppressing

immune responses to growing tumours. This led to the hypothesis that overcoming

immune suppression alone was not sufficient, and perhaps an additional boost to

effector T cell activity would improve the outcome.

Chapter 3 investigated the improved efficacy of the timed combination of three

immune modulators (anti-CD25mAb, anti-TGF-βmAb and anti-CTLA-4mAb) in the

AE17 model (timed triple immunotherapy, TTI). Treatment with the TTI completely

eradicated tumours in 50% of the C57BL/6J mice, with maintenance of low Treg

numbers nine days after treatment initiation. The cured mice also showed complete

resistance to re-challenge with the same tumour cells and had an increased percentage

Page 4: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

iv

of CD4+CD44+ T cells, indicative of immune memory creation. Increased effector T cell

function was also observed during the rechallenge (Kissick et al., 2012).

Chapter 4 shows that the 50% cure rate of AE17 tumours could be raised to

100% by doubling the dosage of anti-TGF-βmAb in the TTI protocol. It was also found

that the standard TTI treatment produced complete clearance in the non-TGF-β AB1

murine mesothelioma tumours in all BALB/c mice. Additionally, combining all three

agonist antibodies into a single intra-tumoural triple immunotherapy cocktail (TIC) for

injection into the established tumours was found to be as effective as the TTI in the

AB1 model.

Chapter 5 investigated the role of B cells in TIC and TTI mediated tumour

eradication. Mice cured by the treatment showed elevated levels of tumour specific IgG

antibodies. These antibodies were higher against whole live tumour cells than cell

lysates. Time-course studies of tumour clearance showed; (a) that IgG levels were not

elevated during tumour clearance and (b) that B cell numbers were increased in the

tumour draining lymph nodes and spleens during tumour clearance. Finally,

employment of B-cell knock-out mice indicated a significant role for B cells in the

successful eradication of the established tumours by the TIC.

Chapter 6 detailed preliminary studies examining the efficacy of the triple

immunotherapy on eradicating non-mesothelioma tumours. TIC treatment tested on the

murine tumours (B16 melanoma, EO771 and 4T1 breast carcinoma tumours) showed

that it was ineffective in eradicating tumours. However, a transient delay in tumour

growth and improvement in survival times were observed in the EO771 and 4T1 breast

carcinoma models. Further optimisations of the concentration of the triple

immunotherapy components, and/or the addition of a fourth component are discussed.

Additional work in this chapter showed that the 100% cure rate observed against AB1

Page 5: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

v

tumours at 9mm2 dropped to 20% when the tumours were bigger (25mm2). When two

9mm2 tumours were grown on each mouse and only one tumour was treated, the results

showed reduced efficacy in eradication of the treated tumour and delayed growth of the

untreated tumour.

Overall, the findings of this thesis indicate that: a) targeting multiple immune

mechanisms simultaneously can completely eradicate established tumours, b) immunity

to the eradicated tumour is achieved and c) that clinical translation to mesothelioma

patients (phase I) is warranted.

Page 6: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

vi

TABLE OF CONTENTS

ABSTRACT .................................................................................................................... iii

TABLE OF CONTENTS............................................................................................... vi

ACKNOWLEDGEMENTS.......................................................................................... xii

STATEMENT OF CANDIDATE CONTRIBUTION .............................................. xvi

ABBREVIATIONS ..................................................................................................... xvii

LIST OF FIGURES ..................................................................................................... xxi

LIST OF TABLES ..................................................................................................... xxiv

PUBLICATIONS AND PROCEEDINGS ................................................................ xxv

Chapter 1 Literature Review ......................................................................................... 1

1.1 Introduction ....................................................................................................... 3

1.2 Etiology of mesothelioma ................................................................................. 4

1.3 Importance of early diagnosis of mesothelioma ............................................... 6

1.4 Standard treatments for mesothelioma .............................................................. 6

1.4.1 Surgery .......................................................................................................... 6

1.4.2 Chemotherapy ............................................................................................... 7

1.4.3 Radiation therapy .......................................................................................... 8

1.5 Murine models of mesothelioma ....................................................................... 9

1.6 Immunosurveillance and cancer control ......................................................... 10

1.7 Immunotherapy ............................................................................................... 12

1.7.1 Enhancing anti-tumour effector activity by immunotherapy ...................... 13

1.7.1.1 Importance of immune checkpoint blockade ...................................... 15

1.7.2 Overcoming immune suppressive mechanisms employed by growing

tumours.................................................................................................................... 17

1.7.2.1 Regulatory T cells ............................................................................... 17

1.7.2.2 TGF-β, a key immunosuppressive cytokine ....................................... 18

1.7.2.3 Strategies targeting tumour-intrinsic evasion mechanisms ................. 19

1.7.2.4 Role of Tregs in mesothelioma ............................................................. 22

1.7.2.4.1 Impact of Treg cell depletion on survival ....................................... 22

1.8 Combination therapies are necessary for overcoming immune suppressive

mechanisms ................................................................................................................. 25

Page 7: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

vii

1.8.1 Simultaneous targeting of Tregs and TGF-β is more beneficial ................... 27

1.8.2 Boosting effector cell function .................................................................... 27

1.9 Role of B cells in immunotherapy for mesothelioma ..................................... 28

1.10 Extending combination therapy to other tumours ........................................... 29

1.11 Aims of the thesis ............................................................................................ 29

Chapter 2 Materials and methods ............................................................................... 31

2.1 Murine tumour cell lines ................................................................................. 33

2.2 Tissue culture .................................................................................................. 33

2.2.1 Storage of tumour cell lines ........................................................................ 33

2.2.2 Resuscitation of frozen stocks ..................................................................... 33

2.2.3 Passage of tumour cell lines ........................................................................ 33

2.2.4 Harvesting cells for in vivo use ................................................................... 34

2.2.5 Cell counting ............................................................................................... 34

2.3 In vivo use of tumour cell lines ....................................................................... 35

2.3.1 Inoculation of tumour cells ......................................................................... 35

2.3.2 Measurement of subcutaneous tumours ...................................................... 35

2.3.3 Euthanasia of tumour bearing mice............................................................. 36

2.3.4 Treatment regimens ..................................................................................... 36

2.3.1 Preparation of anti-CD25mAb treatments .................................................. 38

2.3.2 Preparation of anti-CTLA-4mAb treatments .............................................. 38

2.3.3 Preparation of anti-TGF-βmAb treatments ................................................. 38

2.3.4 Preparation of triple immunotherapy cocktail (TIC) .................................. 38

2.4 Treatment administration ................................................................................ 38

2.4.1 Timed triple immunotherapy (TTI) treatment administration at 9mm2 ...... 38

2.4.2 Intra-tumoural (i.t) administration of a single dose of triple immunotherapy

cocktail (TIC) .......................................................................................................... 39

2.5 Sample preparation and analysis by flow cytometry ...................................... 42

2.5.1 Preparation of lymph nodes and tumours ................................................... 42

2.5.2 Preparation of spleen cells .......................................................................... 42

2.5.3 Live/dead viability dye (eFlour 780) staining ............................................. 43

2.5.4 Cell surface staining for flow cytometry ..................................................... 43

2.5.5 Intracellular Fox P3 and Ki-67 staining ...................................................... 45

2.5.6 Preparation of compensation controls for flow cytometry .......................... 45

2.5.7 Flow cytometric analysis of samples .......................................................... 46

Page 8: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

viii

2.5.7.1 Analysis of T cells............................................................................... 46

2.5.7.2 Analysis of Regulatory T cells ............................................................ 46

2.5.7.3 Analysis of DC cells ........................................................................... 46

2.5.7.4 Determination of relative expression of CD80 ................................... 46

2.5.7.5 Determination of cell numbers ............................................................ 51

2.5.8 Isolation of immune cells by Fluorescence-activated cell sorting (FACS) 51

2.5.9 Adoptive transfer of immune cells into mice .............................................. 51

2.6 Tumour specific Immunoglobulin detection analysis ..................................... 52

2.6.1 Preparation of serum ................................................................................... 52

2.6.2 Preparation of cell lysate coated ELISA immunosorbent plates................. 52

2.6.3 Detection of tumour cell lysate specific IgG in serum ................................ 52

2.6.4 Detection of IgG in serum specific to live tumour cells ............................. 53

2.6.5 Measurement of optical density of serum IgG specificity .......................... 53

2.7 Statistical analysis ........................................................................................... 54

2.8 Materials and Reagents ................................................................................... 54

2.8.1 Mice ............................................................................................................ 54

2.8.2 Reagents ...................................................................................................... 55

2.8.3 ELISA reagents ........................................................................................... 56

2.9 Equipment ....................................................................................................... 56

2.10 Buffers, media and solutions ........................................................................... 58

CHAPTER 3 Development and characterisation of an effective triple

immunotherapy in a TGF-β secreting murine mesothelioma model (AE17 model)

......................................................................................................................................... 61

3.1 Introduction ..................................................................................................... 63

3.2 Results ............................................................................................................. 67

3.2.1 Intra-tumoural administration of anti-CD25mAb together with intra-

peritoneal anti-CTLA-4mAb and sequential anti-TGF-βmAb, completely

eradicates established AE17 murine mesothelioma tumours .................................. 67

3.2.2 Administration of timed triple immunotherapy into established AE17

tumours inhibits re-accumulation of Tregs in tumour draining lymph nodes ........... 69

3.2.2.1 TTI treatment results in greater maturation of DCs and subsequent

activation of effector T cells in the TDLNs ........................................................ 72

3.2.3 A specific anti-tumour memory response results from TTI treatment ........ 76

Page 9: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

ix

3.2.3.1 Sustained depletion of Tregs in the TDLNs of re-challenged AE17

cured mice ........................................................................................................... 78

3.2.3.2 Induction of memory T cells by the TTI treatment ............................. 80

3.3 Discussion ....................................................................................................... 82

CHAPTER 4 Improved efficacy of the triple immunotherapy in the non-TGF-β

secreting murine mesothelioma model (AB1 model) ................................................. 89

4.1 Introduction ..................................................................................................... 91

4.2 Results ............................................................................................................. 92

4.2.1 Timed administration of antibodies targeting CD25, TGF-β and CTLA-4

completely cleared established AB1 sub-cutaneous tumours in 100% of BALB/c

mice…. .................................................................................................................... 92

4.2.2 A combined, single administration of the triple treatment as a cocktail

(TIC) is sufficient to induce complete clearance of AB1 tumours in close to 90% of

animals .................................................................................................................... 94

4.2.3 Induction of systemic immune response in cured mice .............................. 96

4.2.3.1 Susceptibility of cured mice to re-challenge with syngeneic alternate

tumour type-4T1 breast carcinoma ..................................................................... 98

4.2.4 Incomplete neutralisation of TGF-β within the AE17 tumour

microenvironment is integral to the suboptimal response generated in AE17

tumour bearing mice treated with TTI. ................................................................. 100

4.2.4.1 Sub-optimal response observed in partial responders despite attempts

at recovering them with additional top-up of immunotherapy treatment ......... 106

4.2.4.2 Increased anti-TGF-βmAb dosage in the original TTI results in

complete tumour eradication in AE17 tumour bearing mice ............................ 110

4.2.4.2.1 Induction of systemic immune response in the mice cured using

mTTI…….. ................................................................................................... 112

4.3 Discussion ..................................................................................................... 114

Chapter 5 Evidence of B cell involvement in triple immunotherapy ..................... 121

5.1 Introduction ................................................................................................... 123

5.2 Result ............................................................................................................ 125

5.2.1 Detection of tumour specific IgG antibodies in the serum of AE17 tumour-

bearing mice by ELISA ......................................................................................... 125

5.2.2 Elevated levels of tumour specific IgG antibodies in the sera of AE17

tumour-bearing mice cured by TTI ....................................................................... 127

Page 10: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

x

5.2.2.1 Partial cross-reactivity of IgG antibodies in TTI cured mice to B16

melanoma cell lysates and spleen cell lysates ................................................... 129

5.2.3 Development of a living whole cell ELISA for detecting serum IgG levels

against AE17 tumour cells .................................................................................... 132

5.2.3.1 Increased reactivity of serum IgG to whole AE17 live cells in TTI

long-term cured mice ........................................................................................ 134

5.2.4 Increased levels of AB1 tumour specific IgG in sera of mice 95 days post

treatment with the TTI or TIC ............................................................................... 136

5.2.4.1 Partial cross-reactivity of IgG antibodies in TTI and TIC cured mice to

syngeneic 4T1breast carcinoma cells ................................................................ 139

5.2.4.2 IgG antibodies present in the TTI and TIC cured BALB/c mice are

tumour specific and not auto-reactive ............................................................... 141

5.2.5 Tumour specific antibodies are not elevated during tumour eradication .. 143

5.2.5.1 Elevated B cell numbers during TIC induced tumour eradication .... 145

5.2.5.2 B cells are vital to the efficacy of the combined triple

immunotherapy…….. ....................................................................................... 147

5.2.5.2.1 B cells are required for successful treatment of AB1 tumours by

TIC……… .................................................................................................... 150

5.2.5.2.2 Overcoming the immunosuppressive tumour microenvironment is

critical to tumour clearance ........................................................................... 152

5.3 Discussion ..................................................................................................... 154

Chapter 6 Preliminary investigations of the intra-tumoural triple

immunotherapy in other tumour models and its effects on distal tumours ......... 163

6.1 Introduction ................................................................................................... 165

6.2 Results ........................................................................................................... 167

6.2.1 Detection of high levels of Tregs within the tumour microenvironment of

non-mesothelioma tumours by flow cytometry .................................................... 167

6.2.2 TIC treatment is not effective in eradicating B16 melanoma tumours in

C57BL/6J mice. .................................................................................................... 170

6.2.2.1 Increased dosage of all three components of TIC has no effect on

tumour retardation. ............................................................................................ 172

6.2.3 Transient period of EO771 breast carcinoma tumour growth retardation in

C57BL/6J mice treated with TIC. ......................................................................... 174

Page 11: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xi

6.2.4 Increased survival time in 4T1 tumour-bearing BALB/c mice treated with

either TIC or dTIC. ............................................................................................... 176

6.2.5 Efficacy of the triple immunotherapy is diminished with an increase in

tumour burden. ...................................................................................................... 179

6.2.5.1 TIC treatment was effective in generating partial concomitant

immunity– a preliminary study on the effect of TIC on distal tumours............ 181

6.3 Discussion ..................................................................................................... 184

6.4 Conclusion and future perspectives .............................................................. 192

6.4.1 Future of the triple immunotherapy .......................................................... 193

6.4.1.1 Addition of more agonist antibodies to the TIC treatment to improve

tumour regression .............................................................................................. 194

Chapter 7 Bibliography .............................................................................................. 197

Page 12: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xii

ACKNOWLEDGEMENTS

I would like to express my gratitude to my supervisors Assoc. Prof. Manfred

Beilharz, Asst. Prof. Demelza Ireland and Dr. Haydn Kissick.

Manfred, I would like to thank you for accepting me into your lab and for teaching me

everything I know now. I would especially like to thank you for your patience and

constant diligence when I needed help with my research. I am very grateful that I could

approach you to ask questions irrespective of whether they were related to lab work,

designing experiments, candidature or scholarship. Your encouragement and advice in

regard to research and a future scientific career have been invaluable. I have learnt a

great deal on how to further my career as a research scientist, and it is all thanks to you.

Thank you for having faith in me.

Dem, I am very grateful for your patient guidance and for always being there for me.

You have always been very helpful with everything regarding the experimental designs,

the data interpretation, critical reading of manuscripts and of course, my thesis chapters.

Your insights have been invaluable, and I will always be very grateful.

Haydn, you were there in the beginning when I was barely equipped with the task at

hand. Thank you for patiently helping me with so many things initially when I had just

started my Ph.D. All the animal work, flow cytometry, tissue culturing and data

interpretation, I learnt from you. Thank you.

Parts of the experimental work described in this thesis were performed by other

members of the Beilharz laboratory. Specifically, Dr. Haydn Kissick assisted me with

the performance and interpretation of experiments described in chapter 3 at the

beginning of my Ph.D. work in March 2011. The following experimental work was

conducted with the 2012 Honours student Emily Bakker: The initial TTI (see section

4.2.1) and TIC (see section 4.2.2) treatment experiments in the AB1 murine

Page 13: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xiii

mesothelioma model; initial experiments on IgG detection following the cure of AB1

tumours (see section 5.2.4) and the single tumour burden experiment described in

section 6.2.5. The following year, a new Honours student, Cassandra Lee assisted in the

following experiments: the IgG detection experiments in sections 5.2.4.1 and 5.2.4.2

and the time-course experiments described in 5.2.5 and 5.2.5.1.

I would also like to thank Dr. Sara Greay, Dr. Cornelia Hooper and Dr. Erika

Bosio for their support and assistance throughout the difficult times faced during my

candidature. I would not be the person or a scientist that I am today if it had not been for

you. Not only have you guys been there for support during this whole experience but

you have also shared your knowledge as young successful scientists and have given me

the confidence that I could make a good researcher as well. Chandelle, you have been a

great office buddy- thanks for cheering me up in the laboratory when it seemed

impossible to smile. Thank you for sharing my enthusiasm for the goodies available at

conferences and for distracting me from my nervousness for public speaking. Cassandra

and Milly, thank you for being such exceptionally good students! Your input into the

project has been invaluable. I will miss all of you, and I wish, wherever life takes us, we

will always find a way to stay in touch (Thank god for social networking sites!!).

I would like to thanks the Dust Disease Board of NSW for funding part of the

research presented in my thesis. I am very grateful to the University of Western

Australia for awarding me with a Scholarship for International Research Fees (SIRF),

University Postgraduate Award for International Students (UPAIS) and the UWA

Travel Award. Without the financial support provided, none of this would have been

possible. I would also like to acknowledge the School of Pathology and Laboratory

Medicine (UWA) for allowing me to undertake my research within the school.

Page 14: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xiv

Furthermore, I would like to acknowledge some of our collaborators. I would

like to thank Tracy Lee-Pullen, Irma Larma and Matthew Linden from CMCA for their

excellent assistance with operating the flow cytometer, designing my antibody panels

and data analyses on FlowJo. A special thanks to Kathy Heel for patiently teaching me

the importance of maintaining gating strategies for all samples being analysed. I

appreciate the assistance provided by Sandy Goodin, Kelly Hunt, Neill Wilson and the

rest of the staff at M block, Animal Care Unit of the University of Western Australia for

assisting with animal monitoring. Thank you!

I feel incredibly blessed to have so many amazing people in my life. I would like

to take this opportunity to thank my friends and family for their love and support; when

I needed them the most. Dibakar, Paritosh and Prashant: you guys are the best! You had

been there to lend an ear when I needed someone to talk to and your humour, it

astonishes me but yet, I do not think I have ever laughed so much. Thanks, also for

having me over at your houses and for taking time to teach me how to cook. I would

also like to thank all the new friends I made since I first arrived in Australia and for all

the good times we have had together. Thanks for being such great friends.

Chloe and Jerome, you guys have been the best housemates, and I am extremely

blessed to have you guys in my life. You welcomed me into your home and made me

feel like I was part of your family. Thank you for all the fun moments we have had

together: the yummy salads, pasta, interesting conversations, “Zombie marathon”, a

shared passion for “star wars”, dancing, the dinner parties, the list is endless…..

Page 15: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xv

To my most treasured family: Amma, Appa, Vasanth, Santosh and Chitra. All of

you have been integral to making me a strong and independent person. If it were not for

your love and support, none of this would have been possible.

To my dear husband Vasanth, you have made me a better person from the moment you

came into my life. You are definitely, the person who has motivated and inspired me the

most; and made me try harder to achieve my goals and to never give up hope. You are

unquestionably my better half, and I love you for that. Thanks for being there for me.

Amma and Appa, what can I say: you have inspired and encouraged me every step of

the way! I am forever grateful to you for always believing in me and for the

unconditional love you have been giving me. I honestly believe that I could not have

finished my candidature without my parents and my husband. Love you guys.

My thoughts also go to my beloved great-uncle Seetharaman, who believed in me long

before I believed in myself.

Page 16: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xvi

STATEMENT OF CANDIDATE CONTRIBUTION

I hereby declare that all of the work described within this thesis was performed

by myself. Exceptions are the few experimental contributions from other members of

the Beilharz laboratory detailed in the acknowledgements.

_____________________ _______________________

Shruti Krishnan A/Prof Manfred Beilharz

Candidate Co-ordinating supervisor

April 2015

Page 17: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xvii

ABBREVIATIONS

+ Immunopositive

- Immunonegative

# Number

oC Degrees (Celsius)

µg Microgram

µl Microlitre

2-ME 2-Mercaptoethanol

AON Antisense Oligonucleotides

APC Allophycocyanin

APCs Antigen Presenting Cells

ARC Animal Resource Centre

ATP Adenosine Tri-phosphate

BCG Bacillus of Calmette and Guerin

BD Becton Dickinson

BKO B-cell Knockout

CD Cluster of differentiation

Cm2 Centimetres squared

CTL Cytotoxic T Lymphocyte

CTLA-4 Cytotoxic T lymphocyte Antigen-4

DAPI 4’,6-diamidino-2-phenylindole

DC Dendritic Cell

DD Denileukin Diftitox

ddH2O Distilled deionised water

DMSO Dimethyl Sulphoxide

DNA Deoxyribonucleic acid

EDTA Ethylenediaminetetraacetic acid

Page 18: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xviii

ELISA Enzyme-linked immunosorbent assay

ELISPOT Enzyme-linked Immunospot

EPP Extrapleural Pneumonectomy

FACS Fluorescence-activated cell sorting

FCS Foetal Calf Serum

FDA Food and Drug Administration

FITC Fluorescein isothiocyanate

FoxP3 Forkhead box P3

FSC Forward Scatter

GCV Ganciclovir

GITR Glucocorticoid Induced TNF Factor

GM-CSF Granulocyte/Macrophage Colony-Stimulating Factor

Gy Gray (unit)

hr Hour

HRP Horseradish Peroxidase

HSV Herpes Simplex Virus

i.p Intra-peritoneal

i.t Intra-tumoural

IDO Indoleamine -2,-3 -dioxygenase

IFN Interferon

IGF Insulin-like Growth Factor

IgG Immunoglobulin G

IL Interleukin

IMRT Intensity-Modulated Radiotherapy

LCMV Lymphocytic Chorio-Meningitis Virus

mAb Monoclonal antibody

MDSC Myeloid-Derived Suppressor Cells

MFI Mean Fluorescence Intensity

Page 19: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xix

MHC Major histocompatibility complex

min Minute

ml Millilitre

MM Murine Mesothelioma

NK cells Null Killer cells

NKT Natural Killer T cells

OD Optical Density

PBS Phosphate Buffered Saline

PD-1 Programmed Death-1

PDGF Platelet-Derived Growth Factor

PD-L1 Programmed Death Ligand-1

PE R-Phycoerythrin

PE-Cy 7 Phycoerythrin-Cyanine 7

PerCP-Cy5.5 Peridinin-chlorophyll proteins-Cyanine5.5

pfu Plaque-forming units

RCC Renal Cell Carcinoma

RPMI Roswell Park Memorial Institute

RT Radiation Therapy

s.c Subcutaneous

SCC Squamous Cell Carcinoma

SD Standard Deviation

SMART Surgery for mesothelioma after radiation therapy

SMRP Soluble Mesothelin-Related Protein

SSC Side Scatter

SV40 Simian Virus 40

TAA Tumour Associated Antigen

TCR T Cell Receptor

TDLN Tumour Draining Lymph Node

Page 20: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xx

Teff Effector T cells

TGF-β Transforming Growth Factor-β

TIC Triple Immunotherapy Cocktail

tk Thymidine kinase

TNF Tumour Necrosis Factor

TPA Tissue Polypeptide Antigen

Tregs Regulatory T cells

TTI Timed Triple Immunotherapy

UT Untreated

UWA University of Western Australia

VEGF Vascular Endothelial Growth Factor

WT Wild-type

Page 21: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xxi

LIST OF FIGURES

Figure 1. 1 Major mechanisms by which potent anti-tumour immune responses can be

generated ......................................................................................................................... 21

Figure 2. 1 Timed Triple Immunotherapy (TTI) experiment protocol. .......................... 41

Figure 2. 2 Gating strategy for analysing CD4+ and CD8+ T cells. ................................ 48

Figure 2. 3 Gating strategy for Treg estimation in TDLNs and tumours. ........................ 49

Figure 2. 4 Gating strategy for analysing dendritic cells. ............................................... 50

Figure 3. 1. Improved survival with complete tumour eradication in 46% of mice treated

with timed triple immunotherapy (TTI) a significant improvement over double

immunotherapy (DI)........................................................................................................ 68

Figure 3. 2. Sustained depletion of Treg cells in TDLNs by TTI treatment even 3 days

post treatment completion. .............................................................................................. 71

Figure 3. 3 Enhanced CD80 expression by dendritic cells and increased effector T cell

levels in mice treated with TTI. ...................................................................................... 75

Figure 3. 4 Sustained depletion of Tregs in the TDLNs of re-challenged cured mice. ..... 79

Figure 3. 5 Induction of memory T cells in mice cured of AE17 murine mesothelioma

by TTI treatment. ............................................................................................................ 81

Figure 4.1 Complete tumour eradication in 100% of AB1 tumour bearing BALB/c mice

treated with TTI............................................................................................................... 93

Figure 4. 2 Tumour growth kinetics and survival of AB1 tumour bearing BALB/c mice

following treatment with the timed triple immunotherapy (TTI) or the triple

immunotherapy cocktail (TIC). ....................................................................................... 95

Page 22: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xxii

Figure 4. 3 Tumour growth of partial responders to the TTI treatment is not significantly

different from the DI treated mice. ............................................................................... 103

Figure 4. 4 Failed recovery of partial responders despite attempts with secondary round

of immunotherapy. ........................................................................................................ 109

Figure 4. 5 Complete tumour eradication in 100% of AE17 tumour bearing C57BL/6J

mice treated with mTTI. ................................................................................................ 111

Figure 5. 1 Elevated AE17 cell lysate specific IgG antibodies detected in the sera of

AE17 end-point tumour bearing untreated mice. .......................................................... 126

Figure 5. 2 Elevated levels of tumour specific IgG observed in sera of TTI cured mice

compared to untreated controls. .................................................................................... 128

Figure 5. 3 Partial cross-reactivity of IgG antibodies in the serum of TTI long-term

cured mice to syngeneic B16 melanoma tumour cell lysates. ...................................... 131

Figure 5. 4 Detection of increased reactivity of serum IgG in TTI cured mice to whole

live AE17 cells. ............................................................................................................. 133

Figure 5. 5 Increased reactivity of serum IgG to whole AE17 live cells in TTI cured

mice compared to AE17 cell lysates. ............................................................................ 135

Figure 5. 6 Elevated levels of tumour specific IgG antibodies detected in combined

immunotherapy (TTI and TIC) treated mice. ................................................................ 138

Figure 5. 7 Partial cross-reactivity high against live syngeneic 4T1 tumour cells in TTI

and TIC cured mice. ...................................................................................................... 140

Figure 5. 8 Low levels of auto-reactive antibodies in TTI and TIC cured mice. .......... 142

Figure 5. 9 No significant change in serum IgG levels in TIC treated mice up to 20 days

post treatment. ............................................................................................................... 144

Figure 5. 10 Changes in B cell percentage post TIC treatment. ................................... 146

Figure 5. 11 Successful tumour eradication with TIC requires B cells. ...................... 149

Page 23: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xxiii

Figure 5. 12 Anti-tumour efficacy of TIC treatment is augmented by B cells. ............ 151

Figure 5. 13 Manipulation of tumour microenvironment is important for successful

clearance of established tumours. ................................................................................. 153

Figure 6. 1 Detection of Tregs within non-mesothelioma tumours grown in both

C57BL/6J and BALB/c mice. ....................................................................................... 169

Figure 6. 2 Growth of melanoma tumours in C57BL/6J mice is unhindered by TIC

treatment. ....................................................................................................................... 171

Figure 6. 3 Double the dosage of all three components in the TIC has no effect on

melanoma growth in C57BL/6J mice. .......................................................................... 173

Figure 6. 4 Treatment with TIC or dTIC did not significantly improve survival of mice

bearing EO771 tumours. ............................................................................................... 175

Figure 6. 5 Improved survival with TIC or dTIC in BALB/c mice bearing 4T1 tumours,

compared to mice left untreated. ................................................................................... 177

Figure 6. 6 Central zone of tumour clearance observed in mice post treatment with

dTIC. ............................................................................................................................. 178

Figure 6. 7 Tumour eradication efficacy of TTI treatment lowered with increase in

tumour burden in AB1 tumour bearing BALB/c mice. ................................................. 180

Figure 6. 8 TIC treatment of single primary tumour in mice co-challenged

simultaneously with AB1 tumours was effective in generating partial concomitant

immunity to secondary tumours. ................................................................................... 183

Page 24: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xxiv

LIST OF TABLES

Table 1. 1 Monotherapies used for the depletion of Tregs in murine mesothelioma models

......................................................................................................................................... 24

Table 2. 1 Mono-clonal antibodies ................................................................................. 37

Table 2. 2 Antibodies used for flow cytometry .............................................................. 44

Table 3. 1 TTI cured mice are resistant to re-challenge with original inoculum (AE17)

and partially resistant to syngeneic B16 melanoma re-challenge. .................................. 77

Table 4. 1 BALB/c mice cured of established AB1 tumours with TTI or TIC are

resistant to re-challenge. ................................................................................................. 97

Table 4. 2 Immunological memory generated in BALB/c mice cured of established AB1

tumours with TIC are tumour specific. ........................................................................... 99

Table 4. 3 No significant improvement in immune cell numbers in partial responders.

....................................................................................................................................... 105

Table 4. 4 Mice cured of established AE17 tumours with mTTI are resistant to re-

challenge. ...................................................................................................................... 113

Page 25: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xxv

PUBLICATIONS AND PROCEEDINGS

Publication arising from Ph.D. candidature

1. Kissick, H. T., Ireland, D. J., Krishnan, S., Madondo, M. & Beilharz, M. W. 2012.

Tumour eradication and induction of memory against murine mesothelioma by

combined immunotherapy. Immunology and Cell Biology, 90, 822-826.

(This paper covers the data described in chapter 3 and represents the early stages of

the development of the successful timed triple immunotherapy (TTI) for the

treatment of subcutaneous murine mesothelioma)

2. Krishnan, S., Bakker, E., Lee, C., Kissick, H. T., Ireland, D. J. & Beilharz, M. W.

2014. Successful combined intra-tumoural immunotherapy of established murine

mesotheliomas requires B cell involvement. Journal of Interferon & Cytokine

Research, (In press).

(This paper covers the data described in chapters 4 and 5 of this thesis which

concerns the development and characterisation of the triple therapy cocktail (TIC)

and also includes the role of B cells in TIC mediated tumour eradication)

Conference proceedings

1. Krishnan, S., Ireland, D. J., Kissick, H. T. & Beilharz, M. W. (2011) Improved

success rate with a triple immunotherapy for the treatment of mesothelioma. The

Australian Society for Medical Research, Western Australian scientific symposium,

Australia. Oral

2. Krishnan, S., Ireland, D. J., Kissick, H. T. & Beilharz, M. W. (2011)

Characterisation of a successful triple immunotherapy for the treatment of

mesothelioma. Combined Biological Science Meeting, Australia. Poster

Page 26: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xxvi

3. Krishnan, S., Ireland, D. J., Kissick, H. T. & Beilharz, M. W. (2012) Tumour

eradication and induction of memory against murine mesothelioma by combined

immunotherapy. Australian Society for Medical Research, Western Australian

scientific symposium, Australia. Oral

4. Krishnan, S., Bakker, E., Ireland, D. J., Kissick, H. T. & Beilharz, M. W. (2012)

Tumour eradication and induction of memory against murine mesothelioma by

combined immunotherapy. Combined Biological Science Meeting, Australia. Oral

5. Krishnan, S., Bakker, E., Lee, C., Kissick, H. T., Ireland, D. J. & Beilharz, M. W.

(2014). Successful combined intra-tumoural immunotherapy of established murine

mesotheliomas requires B cell involvement. International Cytokine and Interferon

Society, Cytokines Down Under in 2014: From Bench to Beyond. Australia. Poster

Page 27: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xxvii

Page 28: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

xxviii

Page 29: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

1

Chapter 1 Literature Review

Page 30: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

2

Page 31: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

3

This literature review was composed subsequent to the completion of the research work

in September 2014.

1.1 Introduction

Malignant mesothelioma is a life-threatening tumour type arising from mesothelial

cells lining the pleura, peritoneum, pericardium and tunica vaginalis (Abe et al., 2002;

Hirano et al., 2002; Berry et al., 2004). The risk of malignant mesothelioma increases in

proportion to the cumulative exposure (3rd or 4th power of time since first exposure) to

asbestos (Reid et al., 2014). The disease has a latency period of 30-40 years with poor

prognosis (with a median survival of 9-12 months) from diagnosis (Robinson and Lake,

2005). The incidence of this disease has been steadily increasing since the late 90s and

is expected to continue rising for another 20 years before declining (Ismail-Khan et al.,

2006). There are currently no effective treatments for mesothelioma and diagnosis tends

to occur quite late due to the long latency period. Despite the ban on asbestos being

introduced in many developed countries; asbestos continues to be a real threat to the

general public, as a) it persists in the community due to previous widespread use and b)

developing countries continue to use asbestos (Bianchi and Bianchi, 2007; Olsen et al.,

2011; Sim, 2013). More than a quarter million deaths due to malignant mesothelioma

are estimated to occur over the next 40 years (Robinson and Lake, 2005). Development

of new treatment approaches is, therefore, necessary to improve outcomes in these

patients. Immunotherapy is one such new treatment and has been shown to be clinically

beneficial for the treatment of other malignancies (Hodi et al., 2010; Kantoff et al.,

2010; Wolchok et al., 2013). The aim of immunotherapy is to boost the patient's

immune response towards the tumour and hence lead to eradication. This treatment

Page 32: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

4

approach can involve either release of immune suppression or direct enhancement of

immune effector activity (Pardoll, 2012).

1.2 Etiology of mesothelioma

The link between asbestos fibres and malignant mesothelioma was first evidenced

by Wagner et al in 1960; with the high incidence of malignant mesothelioma being

reported amongst asbestos miners in South Africa (Wagner et al., 1960). Asbestos was

used extensively in construction and ship building industries due to the fire retardant

and heat insulating properties of asbestos. This widespread usage was most predominant

in the United States and Europe especially from 1940s to 1979 (Ismail-Khan et al.,

2006). With the increase in awareness of carcinogenesis due to asbestos in the 1980s,

restrictions on usage of asbestos were enforced. The increased awareness eventually led

to asbestos being banned in many developed countries including Australia. Despite the

ban, incidence of mesothelioma continues to rise (Kamp, 2009), especially in

developing countries, where the restrictions are not strictly adhered to.

There are two classes of asbestos fibres, the serpentine and the amphibole.

Serpentine includes most commonly used asbestos mineral, chrysotile or white asbestos,

and the amphibole includes several fibres including crocidolite (blue asbestos) and

amosite (brown asbestos) among others (Gibbs, 1990). Of the two different fibre types,

the larger amphibole fibres were thought to be more carcinogenic (Mossman et al.,

1996; Roggli and Sharma, 2014). However, extensive review of literature carried out by

Powers et al (2002) showed that the data was so contradictory; that it was not possible

to determine whether it was the crocidolite or the chrysotile class of asbestos fibres that

were the causative agents of malignant mesothelioma (Powers and Carbone, 2002).

Apart from asbestos fibres, other factors such as erionite (mineral fibres), Simian Virus

40 (SV40), exposure to radiation, and certain genetic predispositions have also been

Page 33: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

5

linked to the development of malignant mesothelioma (Powers and Carbone, 2002;

Carbone et al., 2007; Yang et al., 2008).

The exact mechanisms of asbestos carcinogenicity are not yet fully elucidated. With

the long latency period (34-40 years) of the disease, several proposed mechanisms are

thought to contribute to the development of malignant mesothelioma. One of them, is

thought to be linked to the constant inflammation caused by the persistence of asbestos

fibres in the pleura (Robinson et al., 2005). There is also strong evidence that reactive

oxygen species and reactive nitrogen species catalysed by the high iron content of these

asbestos fibres causes DNA damage (Kamp and Mossman, 2002). Furthermore,

mutations in the p53 gene (tumour suppressor gene) produced by crocidolite fibres have

been studied in mice (Lin et al., 2000). The link between inactivation or loss of the p53

(and other somatic mutations) and tumour development and progression are being

investigated (Altomare et al., 2005). In addition, the role of cytokines and growth

factors that promote mesothelioma proliferation are also being elucidated. These include

factors such as platelet-derived growth factor (PDGF) (Aggarwal et al., 2009),

transforming growth factor-β (TGF-β) and insulin-like growth factor (IGF) (Liu and

Klominek, 2004).

Since the 1980s, Australia has the world’s highest incidence rate of malignant

mesothelioma per capita (Leigh and Driscoll, 2003); followed by Britain (Bianchi and

Bianchi, 2007; Robinson, 2012). In Australia, the incidence rates were 31.8 per million

in 1997 with the highest rate of 47.7 per million per year being reported in Western

Australia in the same year (Leigh and Driscoll, 2003). In 2008, the annual incidence rate

of 29 cases per million was published in Australia and these numbers are predicted to

peak between 2014 and 2021 (Robinson, 2012).

Page 34: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

6

1.3 Importance of early diagnosis of mesothelioma

Early diagnosis, in clinical practice, is important for improving curative

treatment of tumours. However, early diagnosis of mesothelioma can be difficult. Some

of the problems faced include differentiating mesothelioma from other diseases. For

instance, epithelioid mesothelioma needs to be distinguished from benign hyperplasia

(Churg et al., 2000). In the case of pleural mesothelioma, it must be differentiated from

lung adenocarcinoma and for peritoneal mesothelioma, from serous carcinoma (Koss et

al., 1998; Baker et al., 2005). To date, biomarkers such as soluble mesothelin-related

protein (SMRP), tissue polypeptide antigen (TPA), hyaluronan and osteopontin, among

others, are being reported as possible markers for diagnosing and monitoring

progression of mesothelioma (Hedman et al., 2002; Robinson et al., 2003; Grigoriu et

al., 2007). However, their sensitivity and specificity as diagnostic markers are said to be

inadequate (Sato et al., 2014). Given the difficulties of early diagnostics and the

symptoms of the disease not appearing until 20 to 40 years after asbestos exposure;

prognosis for mesothelioma remains unsatisfactory. Current approaches to improving

early diagnosis of mesothelioma are being investigated (Fukuoka et al., 2013; Sato et

al., 2014). However, they are still in their infancy, and no standard diagnostic tool for

detecting mesothelioma exists at the moment.

1.4 Standard treatments for mesothelioma

Standard therapies involved in the management of mesothelioma include surgery,

radiation therapy and chemotherapy.

1.4.1 Surgery

Though procedures like video thoracoscopy and laparoscopy have helped in

establishing a diagnosis for mesothelioma (Waller, 2004); partial pleurectomy,

pleurodesis and pleuro-peritoneal shunting procedures were more palliative in nature

Page 35: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

7

and aided only in relieving pleural effusions (Scherpereel et al., 2010). Only

cytoreduction by either pleurectomy or extra pleural pneumonectomy (EPP) procedures

were recommended as potential curative surgeries for mesothelioma (Stewart et al.,

2004). Such curative surgeries have been shown to reduce the bulk of the tumour

(cytoreduction) and delay disease progression (Stewart et al., 2004; Sugarbaker et al.,

2004). Pleurectomy involves the removal of the whole pleura stretching from the apex

of the lung to the diaphragm. EPP is considered a radical surgical procedure and

involves the removal of a lung along with the majority of the hemidiaphragm, parietal

and visceral pleura and a portion of phrenic nerve along with the pericardium. EPP has

achieved the greatest degree of cytoreduction, with increase in median survival by two

years with good control over local remission of the disease (Rusch, 1999). Even so,

only a few patients are eligible (Karnofsky performance status was required to be

greater than 70%) for the surgeries. The skills of the surgeon for performing such

demanding procedures and the preoperative conditions of the patients are also limiting

factors that hinder this therapeutic approach (Sterman and Albelda, 2005).

1.4.2 Chemotherapy

The current standard first-line systemic chemotherapy combination used for the

treatment of unresectable malignant mesothelioma is the combination of Pemetrexed

(500 mg/m2) and Cisplatin (75 mg/m2) (Jänne, 2003; Vogelzang et al., 2003). In a phase

III study, the median survival time for patients treated with the combination was 12.1

months compared to 9.3 months for patients who received Cisplatin alone. A similarly

weak increase in median survival was also observed for the combination of Cisplatin

(80 mg/m2) and Raltitrexed (3 mg/m2) (van Meerbeeck et al., 2005). Another

combination included Gemcitabine (617 mg/m2) and Carboplatin (80 mg/m2); though

the median survival for this combination was found to be only 66 weeks (Favaretto et

al., 2003). Other chemotherapy combinations are being tested, though none have, as yet,

Page 36: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

8

achieved higher response/survival rates. So far, no standard second-line chemotherapy

regimens exist for the treatment of mesothelioma following the first line.

1.4.3 Radiation therapy

Most studies involving radiation therapy (RT) in the management of malignant

mesothelioma have shown no significant improvement on overall survival in patients

(Aisner, 1995). The main drawback of RT is the diffused nature of the tumour covering

majority of the pleural surface. RT treatment of such large surfaces are known to cause

radiation toxicities in the neighbouring organs (Maasilta, 1991). Nevertheless, RT

treatment studies by Boutin et al (1995) showed that administration of 7 Gy in three

daily sessions given 10-15 days post thoracoscopy, significantly decreased local

recurrence of tumour cells in all 20 treated patients (Boutin et al., 1995). It has also

been shown that it is possible to deliver doses of over 45 Gy with intensity-modulated

radiotherapy (IMRT). This is because the radiation fields are more focussed- by the

placement of markers during surgery, ergo radiation toxicities can be avoided (Stahel et

al., 2010).

Several groups have shown the importance of combining EPP with

administration of radiation or chemotherapy post-operatively to improve outcomes.

Post-operative chemotherapy with doxorubicin and cyclophosphamide, and up to 54 Gy

of RT to the hemithorax (trimodal therapy)- increased survival beyond 5 years in 15%

of the patients treated (Sugarbaker et al., 1999). IMRT doses of 50-60 Gy post-EPP

surgery were shown to annihilate local recurrence of mesothelioma tumours without

severe toxicities (Ahamad et al., 2003a; Ahamad et al., 2003b). Currently, the trimodal

treatment option is undergoing further evaluation in a phase II clinical study. The study

involved four cycles of Cisplatin and Pemetrexed in conjunction with pleurectomy/EPP

followed by IMRT targeted at the pleura (ClinicalTrials.gov Identifier NCT00715611).

Page 37: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

9

Another promising approach involves short high-dose IMRT treatment of the

hemithoracic region followed by EPP surgery and is called SMART (surgery for

mesothelioma after radiation therapy). Results from the initial phase I/II trials showed

that cumulative survival reached 84% without any perioperative morbidity and

mortality (Cho et al., 2014).

Despite interest in the trimodal treatment options and the various combination of

chemotherapy being tested, treatment for malignant mesothelioma remains debatable

(Baud et al., 2014). Currently, novel therapies are being explored for the treatment of

mesothelioma.

1.5 Murine models of mesothelioma

To assist in the development of novel therapies, pre-clinical animal models are

essential. Murine models for mesothelioma have been developed, and these models

have led to a greater understanding of the disease development and in the identification

of potential treatments for mesothelioma. Indeed, murine mesothelioma tumour model

is one of the few models that is homologous to human cancer, with the murine tumours

mimicking the human disease in the manner of the pathology, molecular biology and

clinical behaviour.

Murine mesothelioma models such as AE17, AB1 and AC29 were developed by the

intra-peritoneal inoculation of crocidolite asbestos fibres in mice (Davis et al., 1992;

Jackaman et al., 2003). Development of ascites was observed 29 weeks later and

cytology, histology and ultra-structural analyses verified the presence of mesothelioma

cells within the ascites. A continuous cell line was developed from the serial passaging

of tumour cells in vitro to achieve the immortal cell lines (Davis et al., 1992; Jackaman

et al., 2003). The AE17 cell line induces tumours in C57BL/6J mice, AB1 in BALB/c

mice and the AC29 in CBA mice. These murine models have been used in pre-clinical

Page 38: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

10

trials for determining the efficacy and toxicity of anti-cancer therapies (Needham et al.,

2006; Anraku et al., 2010; Jackaman et al., 2012). The AE17 and AB1 cell lines are

similar, except for the fact that AE17 tumours are associated with higher levels of the

immune suppressive cytokine TGF-β (Davis et al., 1992; Fitzpatrick et al., 1994).

1.6 Immunosurveillance and cancer control

One of the central dogmas proposed by Paul Ehrlich more than a century ago, was

the concept of immunological surveillance against neoplasm. The immune system was

theorised to react to altered self-antigens expressed on cancerous or pre-cancerous cells

and destroyed the neoplasm before they caused a problem (Ehrlich, 1908; Burnet, 1969;

Möller and Möller, 1976). With the improvement in immunological models, the ability

to probe the nature of tumour immunology improved. Over the subsequent years,

vaccination studies that were conducted, have further supported the theory of

immunosurveillance (Van Pel and Boon, 1982; Barth et al., 1990).

The concept of tumour immunosurveillance was first shown by the work on in-bred

Rag2-deficient mice that lacked mature lymphocytes (T, B and NK-T cells). These mice

developed spontaneous cancers by 14-16 months of age (Shankaran et al., 2001). Work

involving the inactivation of perforin, granzyme and interferon signalling (integral part

of the immunosurveillance system) also demonstrated the susceptibility of mice to

tumourigenesis (Dunn et al., 2004a; Bui and Schreiber, 2007). These studies evidenced

the concept of immunosurveillance and the role of the immune system to engage in

tumour killing. Further evidence for immunosurveillance came from the improved

prognosis of patients whose tumours exhibited increased infiltration by lymphocytes

(Dunn et al., 2004a).

Immunosurveillance relies on cells and molecules (cytokines and chemokines)

that constitute the innate and adaptive immunity arms of the immune system. The chief

Page 39: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

11

effector cells of the innate immune system include macrophages, natural killer T (NKT)

cells, dendritic cells (DCs) and neutrophils (de Visser et al., 2006; Ostrand-Rosenberg,

2008). In the adaptive arm, T cells are critical to detection and destruction of tumour

cells (Romagnani et al., 1997; Girardi et al., 2003) and B cells are also implicated to

some extent (DiLillo et al., 2010b; Nelson, 2010). CD4+ T cells including the subsets

Th1 and Th2 lineages are activated by exposure to the cytokines IL-12 and IL-4 and

subsequently, the Th1 cells kill tumour cells directly by secreting high levels of IFN-γ,

cytolytic granules and tumour necrosis factor (TNF)α (Becker, 2006). Girardi et al

(2003) showed that γδ T cells contribute strongly to host protection against papillomas

(Girardi et al., 2003). Destruction of tumour cells by CD8+ T cells is either direct,

through perforin/granzyme-B-mediated pathways, or through further secretion of

cytokines such as IFN-γ and IL-2 (Qin et al., 2003; Sengupta et al., 2010).

Conversely, several mechanisms exist to keep the immune system in check (known

as peripheral self-tolerance), as an excessive response to self-antigens can cause toxicity

and autoimmune reactions. Indeed, a subset of T cells known as regulatory T cells

(Tregs) are known to play a crucial role in maintaining self-tolerance for self-antigens

and preventing cytotoxicity by negatively regulating the immune system (Sakaguchi et

al., 2008). Tregs are known to occur in malignant tumours or ascites (Mougiakakos et al.,

2010). This down-regulation of the immune response by Tregs is one potential

mechanism that enables tumour cells in evading immune surveillance.

The immunoediting concept best explains the interaction between the immune

system and the developing cancer. Immunoediting is regarded as a process composed of

three phases: elimination, equilibrium and escape. The first phase is the elimination,

where immunosurveillance is highly active, and pre-cancerous cells are kept in check by

the immune system. If the elimination phase fails to stop cancer cells, the cells that were

Page 40: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

12

not destroyed by the immune cells enter into the equilibrium phase where the cells are

maintained in a state of dormancy. Escape is when the cancer cells evade the immune

system and grow uncontrollably (Dunn et al., 2004b; Schreiber et al., 2011). The

importance of this evasion of immunosurveillance by growing tumours, is being seen as

one of the new “hallmarks of cancer” (Dunn et al., 2002; Hanahan and Weinberg,

2011).

The mechanisms used by the tumour cells to evade destruction include: reduced

immunogenicity of the tumour cells (Lengauer et al., 1998; Algarra et al., 2000),

recruitment of immunosuppressive cells and release of immunosuppressive cytokines

(Ghiringhelli et al., 2005; Drake et al., 2006; Rabinovich et al., 2007; Schreiber et al.,

2011). Immunotherapy treatments are currently being designed to overcome these

obstacles and enhance the anti-tumour immune responses.

1.7 Immunotherapy

The notion of harnessing the body’s immune system to help eradicate cancer cells is

a logical concept but not a new one. The first systematic study of immunotherapy for

the treatment of malignant tumours was started in the late 1800s by William B Coley, a

bone sarcoma surgeon. He injected cancer patients with Streptococcus pyogenes (that

causes erysipelas) and observed that the tumours disappeared. The bacterial culture

administration was based on the hypothesis that the subsequent infection would trigger

an immune response, not only against the bacteria, but also against the cancer. Over the

next forty years, more than 1000 patients with inoperable bone and soft-tissue sarcomas

were known to be treated with bacteria or bacterial toxins known as “Coley’s toxins”;

with almost half having reportedly undergone near tumour regression (Nauts and

McLaren, 1990; McCarthy, 2006). However, with the lack of reproducibility of the

Page 41: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

13

results along with the development of other modes of cancer treatments such as

chemotherapy and radiation therapy, interest in immunotherapy waned.

With a better understanding of the tumour immunology, the interest in using the

immune system to eradicate cancer cells is once again gaining momentum. The renewed

interest in immunotherapy is especially relevant, as a) the cancer cells are specifically

targeted, b) the duration of anti-tumour response is longer, and c) there is minimal

collateral damage to healthy cells. Current immunotherapy treatment approaches can

accomplish these tasks either by a) enhancing effector activity against the growing

tumours or b) by overcoming immune suppression.

1.7.1 Enhancing anti-tumour effector activity by immunotherapy

One of the initial approaches to immunotherapy primarily revolved around

boosting the anti-cancer immune responses against tumour cells. One of the methods

applied to generating a potent anti-tumour immune response, involved boosting tumour

antigen presentation by dendritic cells (DCs): the essential initiators of adaptive

immunity. This DC priming (illustrated in Figure 1.1a) would in turn enhance cytotoxic

T lymphocyte activity against tumour cells (Banchereau and Steinman, 1998; Nefedova

et al., 2005). One such DC priming approach that has been approved by the FDA for

human treatment is the DC vaccine Sipuleucel-T. Three intra-venous treatments

administered over 2 weeks of the DC vaccine were shown to prolong the overall

survival (25.8 months with minimal toxicities) in patients with metastatic castration-

resistant prostate cancer (ClinicalTrials.gov number, NCT00065442) (Kantoff et al.,

2010).

In the case of malignant mesothelioma, immunotherapy is relevant as a) it has

been found to be sensitive to destruction by immunotherapies and b) patients with

mesothelioma are known to mount an immune response against the growing tumour

Page 42: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

14

(Robinson et al., 2000; Mukherjee and Robinson, 2002). Notwithstanding, these

immune responses are known to be weak and unable to prevent the tumour growth

(Robinson et al., 2000). The first immunotherapeutic approach used for treatment of

mesothelioma involved treating 30 patients with BCG vaccine (Webster et al., 1982).

Though it was a crude treatment, improved survival was observed in the treated patients

and this response led the way for other immunotherapeutic treatments to be tested. The

first use of Bacillus of Calmette and Guerin (BCG) as a cancer vaccine was reported by

Holmgren in 1935 (Holmgren, 1935). Successful intracavitary administration of BCG

vaccine into superficial bladder tumours was conducted by Morales and colleagues in

1976. This paved the way for the BCG vaccine being accepted as an

immunoprophylactic standard treatment for bladder cancer post local surgery (Morales

et al., 1976; Alexandroff et al., 1999).

Intra-pleural treatments with immune-stimulatory cytokines such as IL-2 and

IFN-γ have also been trialled in mesothelioma patients. The treatments improved

outcomes only in patients with early disease and not in those with late-stage tumours

(Robinson et al., 1991; Boutin et al., 1994; Castagneto et al., 2001). Powell et al (2006)

showed that treatment with recombinant GM-CSF (Granulocyte/Macrophage Colony-

Stimulating Factor) combined with autologous whole-cell tumour lysates administered

as a vaccine, induced tumour-specific cellular immunity in 32% of the treated patients

(Powell et al., 2006). GM-CSF is a cytokine that activates APCs and enhances antigen

presentation by these cells to T cells in the tumour draining lymph nodes. Though some

patients were found to have a stable condition throughout the trial, an overall increase in

survival time (11.5 months) compared to historical controls (9.6 months) was observed

(Alberts et al., 1988). Such an example of using tumour vaccine for priming DC is

illustrated in Figure 1.1a.

Page 43: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

15

1.7.1.1 Importance of immune checkpoint blockade

As described above, the immune system is regulated by inhibitory mechanisms

that enable immune evasion by growing tumours. The manipulation of these self-

tolerance pathways could potentially shift the immune response away from tumour

“escape” and move more towards “elimination” of tumour cells. Blocking of such

inhibitory signals or enhancing the activation signals (as illustrated in Figure 1.1b)

could potentially drive the immune system towards the anti-tumour effector

functionality.

Blocking of the primary negative regulatory signals (also known as immune

checkpoints) as potential immunotherapy targets is gaining interest. The first receptor to

be identified as a negative immune regulator was the cytotoxic T lymphocyte antigen-4

(CTLA-4) (Brunet et al., 1987). CTLA-4 is expressed on activated T cells and also on

Tregs and is a negative receptor for CD80/86 that are expressed on antigen presenting

cells (Greenwald et al., 2005; Quezada et al., 2006; Hodi, 2007). CTLA-4 is similar to

CD28 and is capable of binding to CD80 and CD86 (belonging to the B7 family)

present on antigen presenting cells (APCs). Binding of CD28 (which is also expressed

on T cells) to the B7 co-stimulatory molecules efficiently activates T cell responses.

However, following antigen activation of T cells, CTLA-4 expression is upregulated

(Egen et al., 2002) and this is essential for maintaining peripheral tolerance. Their role

in maintaining homeostasis was demonstrated in CTLA-4-/- deficient mice that

developed lethal hypo-lymphoproliferative disorders and multiple organ tissue

destruction (Tivol et al., 1995; Waterhouse et al., 1995).

In the tumour microenvironment, the CTLA-4 receptor binds with higher

affinity to CD80/86 on antigen presenting cells, than the CD28 receptor on T cells and

thereby, essentially shuts down T cell proliferation (Walunas et al., 1994; Schneider et

Page 44: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

16

al., 2006). The up-regulation of CTLA-4 in effect, promotes tolerance and aids in the

immune evasion by the growing tumour cells (Vignali et al., 2008).

Promising clinical response data have been documented with blocking the

CTLA-4 inhibitory signal on T cells using Ipilimumab (Yervoy, Bristol-Myers Squibb,

New York, NY, USA). Treatment with Ipilimumab was shown to enhance patient

survival, with 15-20% of patients with advanced melanoma surviving beyond two years

(Hodi, 2010; Robert et al., 2011). In another study, twenty-nine patients with

chemotherapy-resistant advanced malignant mesothelioma were enrolled into a phase II

clinical study of anti-CTLA-4mAb treatment. Though disease control in 31% (9/29) of

patients was observed, the results seemed disappointing, as only 7% (2/29) achieved

durable partial response to the treatment (Calabrò et al., 2013).

The other well recognised regulator of suppression is the programmed death-1 (PD-1)

pathway (Duraiswamy et al., 2013; Wolchok et al., 2013). While CTLA-4 is expressed

on activated T-cells, including Tregs, PD-1 is expressed more broadly on activated T

cells and haematopoietic cells (Keir et al., 2008). The role of PD-1 blockade on effector

T cell activation is well known and has been found to increase effector T cell responses

(Topalian et al., 2012). Blockade of PD-1 by agonist antibody (BMS-936558) has

already shown varying levels of objective responses in patients with non-small-cell lung

cancer, melanoma and renal-cell carcinomas (Topalian et al., 2012). The results of these

trials strongly support further evaluation of immune checkpoint blockade as future

treatment options. On September 2014, the anti-PD-1mAb Pembrolizumab (Keytryda,

Merck, New Jersey, USA ) was approved by the FDA for the treatment of metastatic

melanoma (Najjar and Kirkwood, 2014).

Page 45: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

17

1.7.2 Overcoming immune suppressive mechanisms employed by growing tumours

A major deterrent to enhancing anti-tumour activity is the immune evasion

mechanisms employed by the growing tumours. This leads to a highly

immunosuppressive environment within the growing tumours (Ganss et al., 2004;

Rabinovich et al., 2007; Whiteside, 2008; Shiao et al., 2011). Recruitment of

immunosuppressive cells such as regulatory T cells (Tregs) and myeloid-derived

suppressor cells (MDSCs) by the growing tumour cells are known to aid in the

suppression of cytotoxic T lymphocytes (Ostrand-Rosenberg and Sinha, 2009;

Mougiakakos et al., 2010). With the secretion of immunosuppressive cytokines or

chemokines such as transforming growth factor-β (TGF-β), vascular endothelial growth

factor (VEGF), interleukin-6 (IL-6) and IL-10; the combined effects are known to

paralyse the activity of infiltrating cytotoxic T lymphocytes (CTL) and natural killer T

(NKT) cells (Shankaran et al., 2001; Lin and Karin, 2007; Rabinovich et al., 2007;

Yang et al., 2010). The down-regulation of the immune suppressive Treg cells are being

investigated as potential cancer treatment strategies.

1.7.2.1 Regulatory T cells

Regulatory T cells (Tregs) are naturally occurring CD4+CD25+FoxP3+ T cells. As

mentioned earlier, these cells play a crucial role in maintaining self-tolerance by

negatively regulating the immune system (Gershon and Kondo, 1970; Sakaguchi, 2004).

Immune tolerance is crucial for maintaining the balance between lack of response and

an excessive response to self-antigens. For a healthy host, immune tolerance is essential

to control the latter where inappropriate responses to self-antigens by auto-reactive T

cells can lead to autoimmune diseases. Indeed, Treg cell depletions have been observed

to induce spontaneous autoimmune diseases such as type I diabetes, gastritis and

thyroiditis in otherwise healthy rodents; their reconstitution preventing these diseases

(Sakaguchi et al., 1985; Sakaguchi, 2000; Singh et al., 2001).

Page 46: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

18

However, in response to tumour growth, Tregs have been found to abrogate

tumour specific T cell immunity. Selective expansion of Tregs in the periphery (tumour

draining lymph nodes) and tumour microenvironment have been demonstrated

previously in different human cancers (Woo et al., 2001; Liyanage et al., 2002; Wolf et

al., 2003; Curiel et al., 2004; Ghiringhelli et al., 2005). These Tregs that occur within the

tumours were found to inhibit the cytotoxic capacity of CD8+ T-cells as well as NKT

cells in a TGF-β dependent manner (Smyth et al., 2006). Suppression of CD4+ T cells

by Tregs has also been found to be through granzyme B dependent cell-to-cell contact

(Wang et al., 2004; Gondek et al., 2005). Glucocorticoid-induced TNF factor (GITR)

and CTLA-4 have also been reported to play critical roles in regulating T cell function

through Tregs (Wing et al., 2008; Friedline et al., 2009).

1.7.2.2 TGF-β, a key immunosuppressive cytokine

TGF-β is a ubiquitous signalling polypeptide and influences multiple processes

such as embryogenesis, carcinogenesis and immune responses (Elliott and Blobe, 2005;

Li and Flavell, 2008). With regards to malignancies, TGF-β is thought to have dual

roles: as a tumour promoter (Kehrl et al., 1986), as well as a suppressor (Markowitz and

Roberts, 1996). But it now seems that the role of TGF-β as a tumour suppressor occurs

earlier on in tumour progression while its role as promoter of tumour growth occurs at

later stages of cancer (Gold, 1998; de Caestecker et al., 2000; Wakefield and Roberts,

2002; Biswas et al., 2011).

This cytokine is produced by some tumours and high levels have also been

shown to contribute to the metastasis of the disease (Friedman et al., 1995; Penafuerte

and Galipeau, 2008; Wang et al., 2013). TGF-β was also reported to be responsible for

the accumulation of the immunosuppressive Tregs within the tumour microenvironment.

Additionally, TGF-β can promote the conversion of naïve CD4+ T cells into Tregs,

Page 47: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

19

thereby contributing further to immunosuppression (Chen et al., 2003; Ghiringhelli et

al., 2005). TGF-β is also known to promote immune tolerance by a) partially mediating

the induction of MDSCs by the growing tumour cells and b) inhibiting the maturation

and production of IFN-γ by APCs (Borkowski et al., 1996; Geissmann et al., 1999;

Xiang et al., 2009).

The implication of TGF-β in progression of cancer has been widely reported

(Elliott and Blobe, 2005). There is growing interest in developing cancer therapeutics

that target the immunosuppressive cytokine TGF-β specifically.

1.7.2.3 Strategies targeting tumour-intrinsic evasion mechanisms

Strategies targeting attenuation of the Tregs and neutralization of the

immunosuppressive cytokines mentioned above are being investigated (as illustrated in

Figure 1.1c). Denileukin Diftitox (DD), a recombinant fusion protein consisting of

peptide sequences of diphtheria toxin and human IL-2; is a FDA approved drug which

targets and kills cells expressing IL-2 receptors (also known as CD25). Phase III clinical

trials of DD for treating patients with late stage cutaneous T-cell lymphoma resulted in

20% of patients with partial response and 10% (7/71) with complete response to the

treatment. Also, reduction in circulating Tregs with the corresponding increase in

cytotoxic capacity of CD8+ T cells was observed in the treated patients (Olsen et al.,

2001). Depletion of Tregs with Denileukin Diftitox (DD) (ONTAK, Ligand

pharmaceuticals Inc., San Diego, CA, USA) has also shown some success in enhancing

immune function in human clinical trials against renal cell carcinoma, T-cell non-

Hodgkin lymphoma, melanoma and non-small cell lung cancer (Dannull et al., 2005;

Dang et al., 2007; Mahnke et al., 2007; Gerena-Lewis et al., 2009).

Therapeutic inhibition of the immune suppressive cytokine TGF-β activity is

also being investigated and includes a) using antisense oligonucleotides to prevent the

Page 48: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

20

translation of TGF-β mRNA, b) inhibition of activity of TGF-β or c) using soluble

receptors or antibodies to neutralize TGF-β (Yingling et al., 2004; Hawinkels and Dijke,

2011). Targeting TGF-β secretion by glioma cells using the antisense oligonucleotide

AP12009 was tested in Phase I/II clinical trials for treating patients with anaplastic

astrocytoma or glioblastoma multiforme. 2/24 (8%) underwent complete tumour

regression, and stable disease was observed in 7/24 (29%) of the treated patients (Hau et

al., 2007).

Immunotherapies involving inhibition of TGF-β have shown great promise in

murine models. IN-1330, a TGF-β type-1 receptor kinase inhibitor was found to inhibit

lung metastasis of breast cancer in mice (Park et al., 2014). Neutralisation of TGF-β

using 1D11 (anti-TGF-βmAb) has been shown to reduce tumour burden in mouse

models of mesothelioma and breast cancer (Saunier and Akhurst, 2006; Kissick et al.,

2010; Biswas et al., 2011; Hawinkels and Dijke, 2011; Kissick et al., 2012).

Similar to the anti-TGF-βmAb, monoclonal antibodies (mAbs) targeting other

immune suppressive cytokines and growth factors, are being actively investigated. A

neutralizing mAb to VEGF, Bevacizumab, was shown to induce tumour regression by

almost 30% in one out of five patients with rectal adenocarcinoma and a significant

prolongation in disease progression was observed in patients with metastatic renal-cell

carcinoma (Yang et al., 2003; Willett et al., 2004). IL-10 is another potent negative

regulator of the immune response. The neutralising anti-IL-10 antibody is currently

being investigated and has shown promise in murine models (Newton et al., 2014).

Page 49: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

21

Figure 1. 1 Major mechanisms by which potent anti-tumour immune responses can be generated a) Priming of dendritic cells to enhance antigen presentation can be achieved by multiple methods. These include i) injecting irradiated whole tumour cells that continue secreting cytokines, ii) re-infusing dendritic cells loaded with tumour cells and matured ex vivo, back into the patients and iii) injecting dead tumour cells (killed by chemotherapy). b) Enhancing cytotoxicity of Effector T cells by i) preventing transduction of negative regulators such as PD-1 and CTLA-4 using antibodies to block these receptors and ii) injecting agonist co-stimulatory antibodies to receptors 4-1BB and GITR to boost effector activity. c) Overcoming immune suppression by i) depleting immunosuppressive cells such as Tregs and MDSCs and ii) neutralizing immunosuppressive cytokines to promote tumour destruction. GITR: Glucocorticoid induced TNF factor, MHC: Major histocompatibility complex and TCR: T cell receptor. Reproduced from Vanneman and Dranoff, 2012.

Page 50: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

22

In order to develop an effective immunotherapy treatment, it is important to

understand the mechanisms employed by the growing tumour to evade destruction by

the immune system. The specific research conducted by our laboratory into the search

for immunotherapeutic targets elucidated the critical roles of Tregs and TGF-β in

immune suppression in murine mesotheliomas.

1.7.2.4 Role of Tregs in mesothelioma

As described above, immune suppression by Tregs is one potential mechanism

that assists tumour cells in evading immune-mediated destruction. Our laboratory was

the first to identify the presence of these Tregs within the AE17 murine mesotheliomas in

C57BL/6J mice (Needham et al., 2006). This model was chosen as it has been shown to

have high levels of Tregs within the tumour microenvironment and represents an

aggressive tumour akin to their human counterpart (Davis et al., 1992; Hegmans et al.,

2006; Needham et al., 2006). These Tregs that were detected within the tumour

microenvironment of the AE17 tumours were found to increase as a percentage of total

CD4+ T cells. as the tumours grew (Needham et al., 2006).

1.7.2.4.1 Impact of Treg cell depletion on survival

With the elucidation of the immune suppression mediated by Tregs in the AE17

murine mesothelioma model, Treg depletion studies were carried out in the same AE17,

as well as other murine mesothelioma tumour models. These studies involved the use of

monotherapies such as anti-CD25 monoclonal antibodies (mAb) and cyclophosphamide

for depleting Tregs (Table 1.1). Work carried out by Anraku et al (2010) showed that a

significantly prolonged survival with no development of tumour could be achieved

when the anti-CD25mAb treatment was administered prior to tumour challenge (Anraku

et al., 2010). However, clinical translation of such data is difficult. In our laboratory,

low dose intra-tumoural (i.t) treatment with anti-CD25mAb was shown to inhibit

Page 51: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

23

tumours successfully in 100% of the cases. However, this inhibition of AE17 tumour

growth was only limited to approximately ten days (Needham et al., 2006).

Page 52: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

24

Table 1. 1 Monotherapies used for the depletion of Tregs in murine mesothelioma models

Mouse strain and tumour

Treg cell depletion strategy Survival Outcome References

C57BL/6J s.c. AE17

Low dose i.t. anti-CD25 mAb (PC61)to established tumours

10 days tumour growth inhibition

Needham et al., 2006

High dose i.t. anti-CD25 mAb to established tumours

No change in tumour growth

Jackaman et al., 2009

BALB/c with i.p. AB1

Cyclophosphamide in drinking water

10 days increase in survival

Veltman et al, 2010

CBA with i.t. AC29

High-dose systemic anti-CD25 mAb prior to tumour challenge

Significantly prolonged survival

Anraku et al., 2010

High-dose systemic anti-CD25 mAb to established tumours

No improvement in survival

Anraku et al., 2010

CBA with s.c. AC29

High dose anti-CD25 mAb to established tumours

Short delay in tumour growth

Wu et al., 2011

(modified from Ireland et al., 2012)

Page 53: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

25

Despite the fact that these immunotherapeutic agents (and many others)

prolonged survival time or caused transient tumour growth inhibition, complete tumour

clearance was never achieved. This lack of clearance of tumours could be due to the

existence of compensatory pathways or other mechanisms of tolerance that may also be

simultaneously at work (Takeda et al., 2010). There is also the possibility that

monotherapies act as selective pressures that further enhance tumour evasion. This led

to the hypothesis that two or more agents used in combination will a) narrow the

window for the tumour to escape immune eradication and b) combination

immunotherapies that target different aspects may improve efficacy over administration

of monotherapies (Takeda et al., 2010).

1.8 Combination therapies are necessary for overcoming immune suppressive

mechanisms

Improved outcomes with combination therapies have now been shown in several

mouse tumour models. The combined treatment of murine B16 melanoma tumours with

recombinant murine IFN-γ and recombinant human IL-2 had improved tumour

eradication in tumour-bearing mice than the mice that received the respective therapies

separately (Silagi et al., 1988). In the same murine melanoma tumour model, treatment

with anti-CTLA-4mAb in combination with GM-CSF was shown to be effective in

eradicating established tumours in 68/85 of the treated mice, whilst the monotherapies

had little or no effect on tumour growth retardation (van Elsas et al., 1999).

A triple therapy combination termed “trimAb” consisting of anti-death receptor

(DR)-5mAb, anti-CD40mAb and anti-CD137mAb was shown to completely eradicate

close to 80% of 4T1 breast carcinoma tumours in mice (Uno et al., 2006). Treatment

with agonist antibodies to CD40 and CD137 (4-1BB) have been shown to activate

antigen presentation by APCs and hence enhance cytotoxic T cell activity against

Page 54: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

26

tumours such as lymphoma, sarcoma and mastocytoma in mice (Melero et al., 1997;

French et al., 1999). In a recent study published by Dai et al (2014), combination of

monoclonal antibodies targeting CD137/CTLA-4/PD-1 or the 4mAb combinations

(CD137/PD-1/CTLA-4/CD19) were trialled in murine models of B16 and SW1

melanoma and the TC1 lung carcinoma (Dai et al., 2014). Addition of CD19 to the

3mAb combination in the above study produced complete tumour regression in 50% of

the mice with large tumours. Increase in long-term memory CD8 T cells and increased

production of IFN-γ and TNF-α was also achieved. Tumour regression was achieved

with the 3mAb, though it was to a lesser extent than the 4mAb combination. The

authors showed that the efficacy of the immunotherapy to treat large tumours could be

improved by combining mAbs that targeted different mechanisms of action. As detailed

previously, anti-CTLA-4mAb and anti-PD-1mAb treatments block the immune

regulatory checkpoints, thereby enhancing tumour activity. Agonist antibodies targeting

CD137 (also known as 4-1BB) expressed on APCs and T cells, enhances the co-

stimulation required for anti-tumour immunity (May et al., 2002). In contrast, the anti-

CD19mAb used in their 4mAb combination was to counteract the B cells and dendritic

cells that were involved in promoting tumour growth and inducing immune tolerance,

respectively (Baban et al., 2005; de Visser et al., 2006).

In the AE17 murine mesothelioma model, complete regression of large tumours

were observed in 87% of the mice that received multiple intra-tumoural treatments with

the combination of the cytokine IL-2 and anti-CD40 antibody delivered intra-

tumourally (Jackaman et al., 2008). More recently, the combination of anti-CTLA-

4mAb and the chemotherapy agent gemcitabine was used to treat BALB/c mice

inoculated with AB1-HA mesothelioma tumour cells. The treatment led to tumour

clearance in close to 60% of treated mice compared to 13% and 8% found in the groups

that received anti-CTLA-4mAb or gemcitabine alone (Lesterhuis et al., 2013).

Page 55: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

27

A clinical trial with a combination of Ipilimumab targeting CTLA-4 and

Nivolumab that targets PD-1 administered intra-venously was tested in patients with

stage III or IV unresectable melanoma tumours. Though adverse events were observed

in 53% of the patients, rapid and deep regression of the melanoma tumours by 80% or

more were observed in 50% of the treated patients (Wolchok et al., 2013). This

evidence shows that combining one or more agents is more effective in mounting an

anti-tumour immune response than single therapies.

1.8.1 Simultaneous targeting of Tregs and TGF-β is more beneficial

Given that combination therapies that target multiple mechanisms were more

beneficial than monotherapies, as reported above, combined depletion of Tregs with

neutralisation of TGF-β (double immunotherapy) was, therefore, examined in our

laboratory. The additional neutralisation of TGF-β was included, as TGF-β was found to

be a master regulator of Treg function within the AE17 murine mesothelioma tumours

(Kissick et al., 2009). The combined depletion of Tregs using anti-CD25mAb and timed

neutralization of TGF-β using anti-TGF-βmAb was tested on the established AE17

tumours in our laboratory. An effective anti-tumour immune response with a transient

regression in tumour growth was generated in the treated mice treated with the timed

double immunotherapy (DI). However, complete tumour eradication was not achieved

(Kissick et al., 2009). This led to the notion that, the therapeutic efficacy of the timed

DI could be improved by perhaps simultaneously boosting the immune response.

1.8.2 Boosting effector cell function

Given that the blockade of the immune checkpoints CTLA-4 and PD-1 on the

surface of Teff cells improved tumour-destructive T cell responses, it was hypothesised

that their addition to the double immunotherapy would improve therapeutic efficacy.

Blockade of CTLA-4 and PD-1 by antibodies have been extensively tested in mouse

Page 56: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

28

tumour models and were shown to improve anti-tumour T cell activity (Hirano et al.,

2005; Zou, 2006). The clinical benefits of using antibodies to block CTLA-4 (Hodi,

2010; Robert et al., 2011), PD-1 (Topalian et al., 2012) and the combination of CTLA-4

and PD-1 (Wolchok et al., 2013) have been proven. More recently, the relationship

between Tregs and PD-1 was elucidated in an article published in JEM, where the studies

involving Treg depletion in mice with chronic lymphocytic chorio-meningitis virus

(LCMV) showed that an up-regulation of PD-L1 on T cells occurred, leading to

inhibition of signals and failure to decrease viral load (Penaloza-MacMaster et al.,

2014). The authors also found that treatment combinations of Treg depletion and PD-L1

blockade resulted in a more significant reduction in viral titres than when the treatments

were administered as monotherapies.

At the time when the research was commenced (March 2011), anti-CTLA-4mAb

(Ipilimumab) was the first among monoclonal antibody treatments, to be approved by

the FDA for clinical treatment of melanoma. The work described in this thesis involved

the addition of anti-CTLA-4mAb to the timed DI regime for the treatment of murine

mesothelioma. In order to differentiate between the previous double immunotherapy

(DI) and the novel combination of using three mAbs against Tregs, TGF-β and CTLA-4

respectively, the new treatment was therefore called timed triple immunotherapy (TTI).

1.9 Role of B cells in immunotherapy for mesothelioma

There is some evidence in the literature that has suggested that B cells can serve as

APCs and thereby enhance cellular immune responses. This enhancement of cellular

responses is initiated by the activation of tumour specific cytotoxic T cells and by the

production of antibodies (antibodies have also been shown to modestly contribute to

anti-tumour activity) (Manson, 1994; Crawford et al., 2006; DiLillo et al., 2010b;

Jackaman et al., 2010; Molnarfi et al., 2013). In the context of pre-clinical

Page 57: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

29

mesothelioma, previous findings by Jackaman et al (2010) suggested a role for B cells

for mounting an effective anti-tumour immune response; in their partially successful

CD40 based immunotherapy against AE17 and AB1 murine mesotheliomas (Jackaman

et al., 2010). The work in my thesis detailing the efficacy of the novel triple

immunotherapy treatment in eradicating murine mesothelioma tumours also included

studies on the role of B cells and whether they play an agonistic or antagonistic role.

1.10 Extending combination therapy to other tumours

The role of Tregs and TGF-β in suppressing immune responses to growing tumours

have been documented not just in pre-clinical models of mesothelioma, but in several

other murine tumour models as well (Onizuka et al., 1999; Shimizu et al., 1999). The

secretion of TGF-β by growing tumours has also been confirmed in other tumour

models such as melanoma and hepatocellular carcinoma among others (Penafuerte and

Galipeau, 2008; Wang et al., 2013). From the literature detailed in the above sections, it

is also clear that the combination immunotherapy being trialled in our laboratory may

have relevance in other tumour models.

1.11 Aims of the thesis

The initial work in this thesis investigated the additional blockade of the negative

regulator CTLA-4 in conjunction with the removal of intra-tumoural immune

suppression by Tregs and TGF-β (as TTI) in the AE17 murine mesothelioma tumour

model. Previously, in our laboratory, the re-accumulation of Tregs 7-10 days post-DI

treatment had been observed (Kissick et al., 2010). With the addition of anti-CTLA-

4mAb to the previous DI treatment protocol, the Treg cell population during the TTI

treatment period was to be investigated. The subsequent success of this approach led to

investigating the efficacy of this triple immunotherapy in a murine mesothelioma that

does not secrete TGF-β (AB1). The importance of timing was also examined by

Page 58: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

30

combining all three agonist antibodies (anti-CD25mAb, anti-TGF-βmAb and anti-

CTLA-4mAb) into a single cocktail for intra-tumoural injection. The previously

reported role of B cells (Jackaman et al., 2010) during tumour clearance in response to

immunotherapy was also investigated in relation to the novel triple immunotherapy

trialled in our laboratory. The goal of the experiments detailed in this thesis was to

develop an effective treatment option to modulate the tumour microenvironment to

overcome immune suppression and enhance tumour destruction. The achievement of

this goal will enable their rapid translation to human trials for the treatment of

mesothelioma.

Page 59: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

31

Chapter 2 Materials and methods

Page 60: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

32

Page 61: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

33

2.1 Murine tumour cell lines

The C57BL/6J tumour cell lines are the AE17 murine mesothelioma, B16

melanoma and EO771 breast carcinoma (Dunham and Stewart, 1953; Fidler, 1973;

Jackaman et al., 2003). The BALB/c cell lines are the AB1 murine mesothelioma and

the 4T1 breast carcinoma (Dexter et al., 1978; Davis et al., 1992).

2.2 Tissue culture

All tissue culture procedures were carried out in a class 2 biological laminar flow

hood using aseptic techniques.

2.2.1 Storage of tumour cell lines

Tumour cells were frozen (-80oC) in 1 ml aliquots containing cell numbers

ranging from 1 × 106 to 1 × 107 in freezing media (Section 2.10). Following freezing,

they were stored in liquid nitrogen.

2.2.2 Resuscitation of frozen stocks

Frozen tumour cells were taken out of the liquid nitrogen storage and thawed at

room temperature and slowly resuspended in 10ml of supplemented RPMI media. Cell

suspension was centrifuged at 350 × g for 5 minutes and supernatant discarded. The

pellet was resuspended in 5 ml of supplemented media and transferred to a 75 cm2

tissue culture flask containing 10 ml of supplemented RPMI medium to bring the total

volume up to 15ml. Cells were incubated at 37°C over-night in 5% CO2 incubator. The

media was replaced the following day with fresh RPMI media, to remove remaining

DMSO and non-adherent dead cells.

2.2.3 Passage of tumour cell lines

Cell lines were passaged when the flasks reached 70-80% confluency. The

RPMI media was discarded, and the cells were washed by adding 10 ml of PBS to the

flask. 1 ml of trypsin was added to the flask to cover the cells completely and then

Page 62: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

34

incubated at 37°C for 2 minutes. The flask was tapped firmly to dislodge cells, and the

trypsin was inactivated by the addition of 10ml of supplemented RPMI media. Cells

were mixed well by pipetting and transferred to a 50 ml centrifuge tube for

centrifugation at 350 × g for 5 minutes. The pelleted cells were resuspended in 10 ml of

supplemented RPMI media. Aliquots of this suspension containing 7.5 × 105 were then

added to 225 cm2 tissue culture flasks containing 30 ml of supplemented RPMI media

and incubated at 37°C.

2.2.4 Harvesting cells for in vivo use

When cells reached 70% confluency, they were considered ready for in vivo use.

The cells for in vivo inoculation were harvested as mentioned above and then washed

twice with PBS by resuspension and centrifugation.

2.2.5 Cell counting

The final cell pellet was resuspended in 10 ml of PBS and a cell count

performed using trypan blue exclusion method by applying a one to one ratio of 0.4%

trypan blue solution to the cell suspension. 10 µl of the trypan blue/cell suspension were

loaded onto a Neubauer haemocytometer by capillary action. Numbers of viable cells

(clear) were counted using a light microscope in the central 5 × 5 square grid as the

dead cells stain blue. The total viable cell number in the sample was determined using

the below equation.

Total live cells= LC × DF × 1 × 104 × ml

LC= Live cells

DF= Dilution factor

Ml= Number of ml of suspended cells

Page 63: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

35

Cell counts were performed in duplicate, and the mean of the two sample counts

was used to determine the cell concentration. Following cell concentration estimation,

cells were pelleted and re-suspended in the required volume of sterile PBS to give the

necessary concentration of tumour cells required for injection.

2.3 In vivo use of tumour cell lines

2.3.1 Inoculation of tumour cells

6-8 weeks old C57/BL6J and BALB/c mice were injected sub-cutaneously (s.c)

on the right flank above the ribcage with the required cell numbers suspended in PBS

using a MICROLITRE Hamilton syringe mounted with a 26g needle. The final

concentrations required were 1 × 107 cells /100µl for AE17 mesothelioma cells, 1 × 106

cells/50µl for AB1 mesothelioma cells and 5 × 105 cells/50µl for B16 melanoma,

EO771 breast carcinoma and 4T1 breast carcinoma cell lines. The cells were injected

into the respective mice within 30 minutes of harvesting.

All animal experiments were approved by The University of Western Australia

Animal Ethics Committee and were documented in the Animal Ethics applications

RA/3/100/498 and RA/3/100/1184 respectively.

2.3.2 Measurement of subcutaneous tumours

Following the inoculation of mice with tumour cells, swelling and redness at the

site of injection was observed, and the mass was soft and was difficult to measure its

size accurately. However, the swelling soon regresses and established hard tumours

could be palpated and measured. The two largest diameters of the palpable tumour were

measured at right angles to each other using Vernier callipers that are multiplied to

obtain tumour area in mm2. Mice bearing tumours were monitored daily for tumour size

measurements, weight loss and changes in overall health.

Page 64: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

36

2.3.3 Euthanasia of tumour bearing mice

Humane culling of the tumour bearing mice were carried out when tumour areas

reached 100mm2 or if there was an overall deterioration in the health of the mice

(symptoms included weight loss, lethargy and seclusion). Cervical dislocation

euthanized mice following anaesthesia with methoxyflurane for 3-5 minutes.

2.3.4 Treatment regimens

Reagents used for the treatment are outlined in Table 2.1. The preparation for

the treatments is outlined below.

Page 65: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

37

Table 2. 1 Mono-clonal antibodies

Compound Source (City,

State, Country)

Catalogue Number

Anti-CD25mAb

(PC61)

eBioscience (San

Diego, CA, USA)

14-0251-86

Anti-CTLA-4mAb

(9H10)

AbSolutions

(Perth, WA,

Australia)

N/A

Anti-TGF-βmAb

(1D11)

R&D Systems

(Minneapolis,

MN, USA)

MAB1835

Page 66: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

38

2.3.1 Preparation of anti-CD25mAb treatments

The anti-CD25mAb stock solution (0.5 mg/ml) was diluted in sterile PBS to

obtain a final concentration of 2 µg/50 µl for AE17 treatments and 2 µg/30 µl for AB1

tumour treatments respectively.

2.3.2 Preparation of anti-CTLA-4mAb treatments

Anti-CTLA-4mAb stock (2mg/ml) was diluted with sterile PBS to have a final

concentration of 1 µg/µl for both AE17 and AB1 treatments.

2.3.3 Preparation of anti-TGF-βmAb treatments

Stock solution of anti-TGF-βmAb (0.5 mg/ml) was diluted in sterile PBS to

obtain a solution of 1 µg/50 µl for AE17 treatment and 1 µg/ 30 µl for AB1 treatments

respectively.

2.3.4 Preparation of triple immunotherapy cocktail (TIC)

Stock solution of the three triple therapy components was diluted with sterile PBS

to make a solution containing 2 µg of anti-CD25mAb, 2 µg of anti-CTLA-4mAb and 2

µg of anti-TGF-βmAb administered at a final volume of 50 µl for AE17 treatments and

30 µl for the other tumour models respectively.

2.4 Treatment administration

The procedures used for the administration of the TTI and TIC treatments are

outlined below

2.4.1 Timed triple immunotherapy (TTI) treatment administration at 9mm2

Murine tumours were allowed to reach an established size of 9mm2 before

treatment was commenced. The TTI protocol as outlined in Figure 2.1 was then

initiated. For intra-peritoneal (i.p) treatments, tumour bearing mice were held securely

with ventral abdomen exposed and head pointed slightly downward. Animals were

Page 67: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

39

injected into the right lower abdominal quadrant with a 0.5 ml insulin syringe mounted

with 29g needle. Intra-tumoural (i.t) treatments were injected into the centre of the

established tumours in mice using a 0.5 ml insulin syringe mounted with 29g needle.

2.4.2 Intra-tumoural (i.t) administration of a single dose of triple immunotherapy

cocktail (TIC)

Mice receiving the triple immunotherapy cocktail (TIC) received 2 µg of each of

the three components as a single i.t injection in 30 µl of PBS into 9mm2 tumours using a

0.5 ml insulin syringe fitted with 29g needle.

Page 68: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

40

Page 69: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

41

Figure 2. 1 Timed Triple Immunotherapy (TTI) experiment protocol. 6-8 week old mice were inoculated s.c with tumour cells and TTI treatment was initiated when the tumours reached 9mm2. The timed triple immunotherapy (TTI) consisted of single intra-tumoural (i.t) injection of anti-CD25mAb with one intra-peritoneal (i.p) injection of anti-CTLA-4mAb on the same day, followed by seven daily i.t injections of anti-TGF-ßmAb. Mice were monitored daily for tumour growth and tumour sizes were measured using microcalipers.

Page 70: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

42

2.5 Sample preparation and analysis by flow cytometry

The procedures used for the preparation and analyses of samples for flow

cytometry are presented below

2.5.1 Preparation of lymph nodes and tumours

Mice were humanely euthanized, and the tumour and tumour draining lymph

nodes (TDLNs) were surgically removed and placed in 1.5 ml microfuge tubes

containing 500 µl of un-supplemented RPMI media. The tissues were homogenised by

mechanically mincing with scissors for 5 minutes and then enzymatically digested by

the addition of 12.5 µl of Liberase blendzyme III (1.4 units/100 µl) and 12.5 µl of

DNase I (2000 kU/100 µl) and mixed on a rotating wheel for 30 minutes. The mixture

was passed through a spoon sieve with the help of a 3 ml syringe plunger. The sieve

was rinsed with additional 1 ml of un-supplemented RPMI media to remove any

remaining cells. This sieved solution containing the single cell suspension was collected

back in the 1.5 ml microfuge tube and centrifuged at 1500 × g for 3 minutes. The

supernatant was discarded and the pellet was washed once with FACS wash buffer and

once in 1 ml of sterile PBS before being suspended in 100 µl of PBS ready to be stained

with the live/dead viability dye.

2.5.2 Preparation of spleen cells

Spleens were surgically removed and placed in 1.5 ml microfuge tubes

containing 500 µl of un-supplemented RPMI media. They were mechanically digested

by chopping them finely with scissors and further homogenised by enzymatically

digesting them with 12.5 µl Liberase blendzyme III and 12.5 µl DNase I on a rotating

wheel for 30 minutes. The mixture was passed through fine wire mesh spoon sieve with

a 3 ml syringe plunger. The sieve was rinsed with 1 ml of un-supplemented RPMI

media to remove any remaining cells, and this solution was collected in a 1.5 ml

Page 71: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

43

microfuge tube and centrifuged at 1500 × g for 3 minutes. The supernatant was

removed, and the pellet was resuspended in 1 ml of red cell lysis buffer for 5 minutes.

The solution was centrifuged, and the supernatant was discarded. The pellet was washed

once with FACS wash buffer and washed once again with 1 ml of sterile PBS and the

pellet was resuspended in 100 µl of PBS and ready to be stained with the live/dead

viability dye.

2.5.3 Live/dead viability dye (eFlour 780) staining

As a single stain control for the live/dead viability dye, a small aliquot (50 µl) of

the single cell suspension was taken in an 1.5 ml microfuge and killed by 3 × freeze-

thaw cycles using liquid nitrogen. These dead cells were mixed with a further 50 µl of

live cells from the single cell suspension. The samples and the single stain live/dead

viability control tube were incubated for 30 minutes at 4°C with the addition of the

live/dead viability dye eFlour780 (1 µl/ml of sample containing 1-10 × 106 cells/ml).

2.5.4 Cell surface staining for flow cytometry

After the live/dead cell staining step, the cells were washed twice with 500 µl of

FACS wash and the pellet was resuspended in 50 µl of master mix containing the

appropriate fluorescently labelled antibodies at the concentrations recommended by the

manufacturers in FACS wash buffer. The antibodies used for flow cytometry are

presented in Table 2.2. The samples were incubated for 30 minutes at 4°C, washed twice

with FACS wash buffer before being resuspended in 500 µl of FACS wash buffer, ready

to be analysed by flow cytometry (FCM).

Page 72: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

44

Table 2. 2 Antibodies used for flow cytometry

Antibody Clone Conjugate Supplier Catalogue #

CD3e 145-2C11 FITC eBioscience 11-0031

CD4 GK1.5 eFlour 450 eBioscience 48-0041

CD4 GK1.5 APC eBioscience 17-0041

CD8a 53-6.7 PE-Cy 7 eBioscience 25-0081

CD8a 53-6.7 PE eBioscience 12-0081

CD25 PC61.5 APC eBioscience 17-0251

CD25 7D4 FITC BD Bioscience 553071

CD25 PC61.5 FITC eBioscience 11-0251

CD44 IM7 V500 BD Bioscience 560780

CD44 IM7 FITC eBioscience 11-0441

FoxP3 FJK-16s PerCP-Cy 5.5 eBioscience 45-5773

FoxP3 FJK-16s PE eBioscience 12-5773

CD80 (B7-1) 16-10A1 PE eBioscience 12-0801

CD11c N418 APC eBioscience 17-0114

MHC II M5/114.15.2 eFlour450 eBioscience 48-5321

CD45R/B220 RA3-6B2 PerCP-Cy 5.5 eBioscience 45-0452

Live/dead - eFlour780 eBioscience 65-0865

Live/dead - DAPI BioLegend 422801

Page 73: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

45

2.5.5 Intracellular Fox P3 and Ki-67 staining

Samples that require intra-cellular staining were prepared by washing the cell

pellet in 500 µl of permeabilisation buffer once and resuspending the cell pellet in

permeabilisation/fixative buffer, which was then incubated for 1 hour at 4°C. Cells were

washed once with permeabilisation buffer and resuspended in 50 µl of master mix

containing FoxP3 and Ki-67 in the recommended concentrations (diluted in

permeabilisation buffer) and incubated at 4°C for 30 minutes. The cells were washed

with permeabilisation buffer and resuspended in FACS buffer for analysis by flow

cytometry.

2.5.6 Preparation of compensation controls for flow cytometry

Single stain controls are required for compensating intrinsic spectral overlaps

that can occur when using multi-colour flow cytometric analyses. If left uncorrected

without using compensation controls, this can lead to misinterpretation of data with

false-positives and artifactual populations when analysing contour plots (Baumgarth and

Roederer, 2000). Compensation beads (positive and negative beads) are thereby used to

set the gates for what proportion of cells is positive and thereby help with interpreting

data (Maecker and Trotter, 2006). Compensation beads were used to create different

single stained controls for all the fluorochromes (Table 2.2) used in the flow cytometry

experiments. Depending on the number of fluorochrome conjugates used in the

experiment, individual tubes were labelled as such for the conjugates. A drop

(approximately 60µl) of positive and negative compensation beads were dispensed into

each tube; following manufacturer’s instructions (BD Bioscience BDTM CompBead cat#

552843). 0.5 µl of the respective fluorochromes (irrespective of the antibody bound to

them) was added and incubated at 4°C for 30 minutes. The controls were then washed

twice with FACS wash buffer before being resuspended in 500 µl of FACS wash buffer,

ready to be used for the compensation step before analysing the samples.

Page 74: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

46

2.5.7 Flow cytometric analysis of samples

Flow cytometry was performed using FACS CantoII bench-top flow cytometer

using the FACSDiva software and the data collected was analysed using the FlowJo

software from Treestar Inc.

2.5.7.1 Analysis of T cells

The gating strategy for live T cells (CD4+ and CD8+) is shown in Figure 2.2.

2.5.7.2 Analysis of Regulatory T cells

Regulatory T cells (Tregs) were defined as CD4+CD25+FoxP3+ T cells and the

gating strategy for Tregs are shown as a percentage of CD4+CD25+ T cells that are also

positive for FoxP3. The gating strategy used for determining Tregs in the TDLNs and

tumours is shown in Figures 2.3A and B.

2.5.7.3 Analysis of DC cells

Dendritic cells (DCs) were defined as CD11c+MHCII+ and their gating is as

shown in Figure 2.4.

2.5.7.4 Determination of relative expression of CD80

Relative expression of CD80 by DCs was determined using Geometric Mean

Fluorescence Intensity (MFI) of the CD11c+MHCII+ cells co-stained with CD80-PE as

shown in Figure 2.4.

Page 75: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

47

Page 76: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

48

Figure 2. 2 Gating strategy for analysing CD4+ and CD8+ T cells. Total lymphocytes population was firstly selected based on their size (forward scatter FSC) and their granularity (side scatter SSC) respectively. Doublets and triplets are eliminated by plotting the size of the lymphocytes based on their area versus height and singlets are selected. The viability dye ef780 (live/dead) was then used to select for live cells that did not fluoresce. Out of these live cells, the population of cells that were positive for CD8 and CD4 T cells were then determined.

Page 77: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

49

Figure 2. 3 Gating strategy for Treg estimation in TDLNs and tumours. CD4+ T cells are selected by plotting against viability dye for TDLNs (A) or against granularity (SSC) when examining within tumours (B). The percentage of cells within this CD4+ population that also express CD25 are then determined. The expression of FoxP3 by the CD4+CD25+ T cells is then plotted as a histogram.

Page 78: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

50

Figure 2. 4 Gating strategy for analysing dendritic cells. For determining total dendritic cell (DC) population, the total live cell population were plotted as MHCII versus CD11c. The gates for positive and negative expression of CD80 were set using the single stained compensation controls. The expression of CD80 by the DCs was then analysed by determining the mean fluorescence intensity (MFI) of the fluorochrome (PE) used for identifying CD80 expression in these cells.

CD80

Page 79: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

51

2.5.7.5 Determination of cell numbers

The number of cells per samples analysed was determined using TruCount tubes

(BD Bioscience) according to the instructions from the manufacturers.

2.5.8 Isolation of immune cells by Fluorescence-activated cell sorting (FACS)

Spleen cells to be sorted by FACS were prepared as mentioned in 2.5.2.

However, instead of the FACS wash, sterile PBS enriched with 5% FCS was used. The

extra-cellular staining of the single cell suspension was the same as the above-

mentioned methodology in 2.5.4. With the one exception that 5% FCS enriched PBS is

used instead of FACS wash in all the steps. Before cells were sorted, they were

resuspended at 5 × 106 cells/ml in 5% FCS enriched PBS.

The FACS sorting was conducted at The Centre for Microscopy,

Characterisation and Analysis (CMCA), UWA, by the CMCA staff using BD Influx

flow cytometer with an 86 µm nozzle and a pressure of 33 psi. The live/dead cell stain

(DAPI at 1µg/ml concentration) was added to the samples prior to sorting and mixed

well, and sorting was initiated. The sorted cells were collected in 50 ml Falcon tubes

containing 5 ml of sterile, cold FCS to maintain the viability of these cells.

2.5.9 Adoptive transfer of immune cells into mice

The immune cells of interest collected in the 50 ml Falcon tubes were centrifuged

at 1500 × g for 3 minutes, and the supernatant was discarded. The pellets were then

resuspended in sterile PBS and counted, reconstituted with sterile cold PBS to the

appropriate concentrations and were injected intra-venously into the recipient mice (tail-

vein injections) using 0.5 ml insulin syringe fitted with 29g needle. The recipient mice

were warmed in an approved warming cabinet before the i.v injection and were

monitored for any sign of overheating which included increased rapid respiration or

panting, red extremities, decreased activity and excessive salivation.

Page 80: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

52

2.6 Tumour specific Immunoglobulin detection analysis

Immunoglobulin G (IgG) in the serum of mice that react specifically against

tumours was determined by enzyme linked immunosorbent assay (ELISA).

2.6.1 Preparation of serum

Blood was collected by cardiac puncture into uncoated blood tubes and was

allowed to clot overnight at 4°C. The serum was transferred into a new microfuge tube

and stored at -20°C. Multiple cycles of freeze/thaw were avoided for serum samples.

2.6.2 Preparation of cell lysate coated ELISA immunosorbent plates

Tumour cells were harvested as mentioned in section 2.2.4 and splenocytes

(from naïve mice) were prepared as single cell suspensions as mentioned in 2.5.2 and

the respective cells were counted and resuspended to a concentration of 2 × 107 cells per

10 ml of sterile PBS. The cells were subjected to 3 cycles of freeze-thaw (liquid

nitrogen followed by thawing at 37°C, 5 minutes each). The resulting lysed cell

suspension was centrifuged at 300×g for 10 mins and the supernatant collected. 100 µl

of the supernatant was added to each well of an immunosorbent 96 well ELISA plate

and incubated overnight at 4°C without shaking. The following day, the supernatant was

removed, and the wells washed thrice with 200 µl of 0.01% Tween-20 in PBS. The

plates were then used immediately or stored at -20°C for later use.

2.6.3 Detection of tumour cell lysate specific IgG in serum

The cell lysate coated wells prepared as mentioned in 3.7.1 were blocked with

200 µl of 5% FCS in PBS for 1 hour at room temperature before running an ELISA.

The blocking solution was removed, and the wells were washed thrice with 200 µl of

0.01% Tween-20 in PBS. After the 3 washes, 100 μl pooled sera diluted in ELISA assay

diluent were added to the plate and incubated without shaking at room temperature for 2

hrs. The wells were washed thrice with 200 µl of 0.01% Tween-20 in PBS once the

Page 81: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

53

serum was removed. Following three washes, 100 µl of biotin conjugated anti-mouse rat

IgG antibody diluted in assay diluent (final concentration of 2 µg/ml) was added and

incubated for 1 hr at room temperature. The wells were washed three times in washing

buffer followed by the addition of 100 µl avidin-horseradish peroxidase (avidin-HRP,

diluted 1:1000 times with assay diluent) and incubated for 30 min at room temperature

and the plate was washed thrice with 200 µl of 0.01% Tween-20 in PBS.

2.6.4 Detection of IgG in serum specific to live tumour cells

For the modified ELISA, tumour cells and splenocytes (from naïve mice) were

harvested as described in sections 2.2.4 and 2.5.2 respectively and used instead of cell

lysates. The cells were counted and diluted, such that there were 2 × 104 cells/100 µl.

The assay was performed in 1.5 ml microcentrifuge tubes with each tube containing 300

µl of the suspended live cells. Washes were performed with sterile PBS and involved

centrifugation and supernatant removal. After incubation with avidin-HRP, the cells

were resuspended in PBS and transferred into pre-blocked (5% FCS in PBS) 96 well

plates (ELISA plate wells were blocked overnight at 4°C with 200 µl of 5% FCS in PBS

and washed twice with 200 µl of 0.01% Tween-20 in PBS before using the plates).

2.6.5 Measurement of optical density of serum IgG specificity

100 µl of TMB substrate solution (substrate for HRP) was added to the

appropriate wells of the 96 well immunosorbent plates and the colour reaction that

develops after 10 minutes; was stopped by the addition of 50 µl of 1M Phosphoric acid

to each well. The optical density was then measured using POLARStar Optima plate

reader at 450nm.

Appropriate controls used with every ELISA run were:

a. NO LYSATE: Wells which were not coated with tumour cell lysates, but

incubated with serum and the rest of the reagents that followed;

Page 82: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

54

b. NO SERUM: Tumour cell lysate coated wells to which no serum was added and

received the subsequent detection antibodies (to detect cross-reactivity); and

c. NO BIOTIN CONJUGATE: Tumour lysate coated wells that were incubated

with serum and all other reagents excluding anti-mouse rat IgG-Biotin, and

d. ONLY LIVE CELLS: An additional control of only live tumour cells (with

nothing else added to them) was used for the whole live cell ELISA analysis to

account for auto-fluorescence of live cells.

2.7 Statistical analysis

All immune cell data collected by flow cytometry are presented as the mean ± SD.

For all tumour size measurements, mean tumour size (± SEM) in mm2 was calculated.

Statistical significance set at p<0.05 were used when only two groups were compared

and were determined by performing student’s two-tailed t-tests. One-way analysis of

variance (ANOVA) with Tukey’s multiple comparison post-hoc tests and linear trend

analysis set at p<0.05 were used for determining differences in immune cell populations

between different groups. All calculations and statistical tests were carried out using

Microsoft Excel (2011) and GraphPad Prism version 5.04 for Windows (GraphPad

Software, San Diego, CA, USA). The Kaplan-Meier survival curves including median

survival calculations were generated using GraphPad Prism version 5.04.

2.8 Materials and Reagents

2.8.1 Mice

Female C57BL/6J and BALB/c mice aged 6-8 weeks were obtained from the

Animal Resource Centre (Perth, Western Australia) and maintained under standard

housing conditions in the M block Animal Care Facility at The University of Western

Australia. The B cell deficient mutant mice (Kitamura et al., 1991) on the BALB/c

background (C.129S- Igh-6tm1Cgn/J; referred to as BKO) aged 4-10 months were bred

Page 83: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

55

in-house at the University of Western Australia animal holding facility and were

provided by Animal Care Services (Perth, Western Australia). All animal work was

carried out in accordance with the guidelines of the National Health and Medical

Research Council of Australia and approval of The University of Western Australia

Animal Ethics committee (RA/3/100/1184).

2.8.2 Reagents

Benzyl Penicillin CSL, Australia

Deoxyribonuclease (DNase) I Sigma, USA

ELISA assay diluent (5x concentrate) eBioscience, USA

Ethanol (96%) Hurst scientific, Australia

Fixation/Permeabilisation concentrate eBioscience, USA

Fixation/Permeabilisation diluent eBioscience, USA

Foetal Calf Serum (FCS) Gibco BRL, USA

Gentamicin Deltawest, Australia

L-glutamine Sigma, USA

Liberase Blendzyme III Roche, USA

Methoxyflurane Medical Developments Australia,

Australia

Permeabilisation buffer eBioscience, USA

Phosphate buffered saline tablets Oxoid, Australia

Phosphoric acid BDH Chemical, Australia

Page 84: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

56

Protease inhibitor cocktail Sigma, USA

RPMI 1640 media Gibco BRL, USA

Sodium azide Sigma Aldrich, USA

Stabilisation fixative (3x concentrate) BD Bioscience, USA

Tris Gibco BRL, USA

Tris-Hydrochloride Sigma, USA

Triton X-100 BDH Chemicals, Australia

Trypan Blue Hopkin and Williams, England

Trypsin/EDTA Sigma, USA

Tween-20 Promega, USA

2-Mercaptoethanol BDH Chemicals, Australia

2.8.3 ELISA reagents

Avidin-HRP 250x concentrate eBioscience, USA

Rat anti-mouse IgG-Biotin eBioscience, USA

3’,3’,5’,5’-Tertramethylbenzidine (TMB) substrate eBioscience, USA

2.9 Equipment

Acrodisc Micropore Syringe Filters Gelman Sciences, USA

BD Influx Becton Dickinson, USA

Blood collection vials

- Non-coated MICROTAINER 2ml Becton Dickinson, USA

Page 85: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

57

Cell strainer (40 µm nylon mesh) BD Falcon, USA

Centrifuge and Eppendorf tubes

- 15 ml and 50 ml Falcon tubes Becton Dickinson Falcon, USA

- 1.5 ml and 2 ml Eppendorf tubes Eppendorf, Germany

Centrifuges

- Eppendorf microfuge 5424 Eppendorf, Germany

- BECKMAN Model TJ-6 centrifuge Beckman Coulter, USA

ELISA plates

- Immunosorbent 96 well plates BD Falcon, USA

FACSCanto II Becton Dickinson, USA

FACS tubes

- Polystyrene round bottom tubes BD Falcon, USA

- Polypropylene round bottom tubes BD Falcon, USA

Flow Cytometry software

- FACSDiva BD Biosciences, USA

- FlowJO Treestar Inc, San Carlos, CA

Incubator

- Forma Series II Water Jacket Thermo Fischer Scientific, USA

Neubauer chamber Hawksley, England

Nunc cryovials 1.8 ml Nalge Nunc International, Denmark

Page 86: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

58

POLARStar Optima BMG Labtech

Rotating wheel Chiltern Scientific, England

Syringes

- 0.5 ml 31g insulin syringe BD Biosciences, USA

- MICROLITRE 100 μl Hamilton, USA

- 50ul syringe (50R-GT) SGE, Australia

Tissue Culture flasks

- 75 cm2 Nalge Nunc International, Denmark

- 225 cm2 Nalge Nunc International, Denmark

Tissue culture hood model BH180 Gelman Sciences

TruCount tubes BD Biosciences, USA

Vernier microcalliper Kaiser, Australia

2.10 Buffers, media and solutions

Antibiotics (4mg/ml Gentamicin and 6mg/ml Benzylpenicillin) (100ml)

Five ampules of Gentamicin and 600mg (1 ampule) of Benzylpenicillin was

dissolved in 100ml of ddH2O, filter sterilised using 0.2 µm filter and stored at -20°C in 5

ml aliquots.

DNAse I (2.3ml)

23mg of DNase I was dissolved in 1.3ml of 10mM Calcium chloride and 1 ml of

ddH2O and filter sterilised through 0.2 µm filter and stored at -20°C in 100 µl aliquots.

Page 87: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

59

ELISA assay diluent

ELISA assay diluent (5X) was diluted in PBS

FACS wash

0.352g of sodium azide (0.005mM NaN3) was dissolved in 100 ml of PBS,

autoclaved and stored at room temperature. Prior to use, 5 ml of FCS was added and

stored thereafter at 4°C.

FCS (Foetal Calf Serum)

Frozen FCS was thawed in water bath at 37°C and heat inactivated for 30

minutes in a 60°C water bath and cooled at room temperature. The heat inactivated FCS

was then aliquoted into 50 ml falcon tubes and stored at -20°C.

Freezing media

10 ml of DMSO was added to 90 ml of FCS and mixed well.

L-glutamine (250ml)

7.3 g of L-glutamine was dissolved in 250 ml of ddH2O, filter sterilised through

0.2 µm filter and stored at -20°C in 5 ml aliquots.

Liberase Blendzyme III

7 mg of Liberase Blendzyme III was dissolved in 2 ml of sterile ddH2O, then

incubated on ice for 30 minutes with intermittent mixing to ensure complete dissolution.

The solution was then filter sterilised using a 0.2 µm filter and stored at -20°C in 100 µl

aliquots.

Page 88: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

60

2-mercaptoethanol (2-ME)

1.74 ml of 2-mercaptoethanol was added to 50 ml of ddH2O and filter sterilised

using a 0.2 µm filter and stored in 500 µl aliquots at -20°C.

Phosphate buffered saline (PBS) (100 ml)

One phosphate buffered saline tablet was dissolved in 100 ml of ddH2O,

autoclaved and stored at room temperature. Typical concentration formula for PBS is

8.0 g/L Sodium chloride, 0.2 g/L Potassium chloride, 1.15 g/L di-sodium hydrogen

phosphate and 0.2 g/L Potassium di-hydrogen phosphate.

Red cell lysis buffer

8.3 g of Ammonium chloride (NH4Cl) was dissolved in 1L of ddH2O to make a

0.83% solution. 450 ml of the 0.83% solution was added to 50 ml of Tris solution

(2.059 g of Tris dissolved in 10 ml of ddH2O and pH adjusted to 7.6) and filter

sterilised.

Supplemented RPMI 1650 media

5 ml of L-glutamine, 5 ml of media antibiotics, 50 ml of FCS and 40 µl of 2-

mercaptoethanol was added to 500 ml of RPMI 1640 media and stored at 4°C for up to a

month.

Page 89: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

61

CHAPTER 3 Development and characterisation of an effective triple

immunotherapy in a TGF-β secreting murine mesothelioma model (AE17 model)

Page 90: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

62

Page 91: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

63

3.1 Introduction

A greater understanding of the interactions between the immune system and

developing tumours has led to immune system modulation being a promising approach

to reducing the morbidity and mortality of cancers. To date, clinical immunotherapy

protocols have predominantly involved only a single agent (O'Brien et al., 2003;

Gerena-Lewis et al., 2009; Hodi et al., 2010; Kantoff et al., 2010; Wolchok et al., 2010;

Wolchok et al., 2013). Such monotherapies have had promising results in animal

models and clinical settings, but their effectiveness has often been limited by peripheral

tolerance induced by tumour growth.

Peripheral tolerance can be induced by regulatory T cells (Tregs) (Sakaguchi,

2004). During tumour growth, increases in Treg numbers have been observed in the

periphery and also within different tumours, including murine and human

mesotheliomas, thereby maintaining T cell anergy and inhibiting the cytotoxicity of

CD8+ T cells and NK cells (Chen et al., 2005; Needham et al., 2006; Smyth et al., 2006;

Zou, 2006; Rabinovich et al., 2007; Hegmans et al., 2010; Whiteside, 2013; 2014).

Danull et al (2005) have shown that elimination of Tregs using Denileukin Diftitox also

enhanced the activity of a DC vaccine that was administered to patients with renal cell

carcinoma (RCC) (Dannull et al., 2005). Elimination of Tregs with corresponding

increase in cytotoxic capacity of CD8+ T cells have also been reported in other human

clinical trials against non-Hodgkin lymphoma, T-cell lymphoma and non-small cell

lung cancer (Olsen et al., 2001; Dang et al., 2007; Gerena-Lewis et al., 2009).

When Treg depletion is carried out systemically, it can lead to undesirable

effects. Work carried out in BALB/c mice by Sakaguchi et al (2006) showed that

adoptive transfer of spleen cells depleted of Tregs into athymic nude BALB/c mice

caused severe autoimmune diseases in multiple organs in these recipient mice

Page 92: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

64

(Sakaguchi et al., 2006). To avoid such systemic problems, we have shown that intra-

tumoural injection of Treg depleting antibodies (as opposed to systemic delivery) can

avoid undesirable autoimmune effects (Needham et al., 2006).

In the work conducted previously in our laboratory on the AE17 murine

mesothelioma (C57BL/6J) model, intra-tumoural (i.t) depletion of Tregs with low dose

anti-CD25mAb (PC61) effectively inhibited growth of tumours for approximately 10

days (Needham et al., 2006). As opposed to high dose anti-CD25mAb therapy which

can in fact enhance tumour growth (Jackaman et al., 2009), we demonstrated that low

dose and local intra-tumoural Tregs depletion using an anti-CD25mAb resulted in

transient tumour growth inhibition and activation of CD4+ helper T cells, CD8+

cytotoxic T cells and dendritic cells (Kissick et al., 2010). A limitation of low dose anti-

CD25mAb therapy, even when administered directly into the tumour, is the transient

nature of tumour growth inhibition, as Treg cells re-accumulated in the tumour

microenvironment ten days post treatment.

We, therefore, hypothesised that multiple mechanisms of peripheral tolerance

are involved in immune suppression within the tumour microenvironment and that;

combinatorial therapies targeting multiple mechanisms will improve anti-tumour

immune responses. This has become a major international research focus; with animal

studies confirming that targeting multiple mechanisms of tumour immunosuppression

result in stronger immune responses against multiple tumour types (van Elsas et al.,

1999; Uno et al., 2006; Kissick et al., 2009; Curran et al., 2010; Jackaman et al., 2012;

vom Berg et al., 2013).

The AE17 model of murine mesothelioma is an excellent model for assessing

pre-clinical efficacy of immune modulation therapies.It is also known to secrete

transforming growth factor-β (TGF-β) (Fitzpatrick et al., 1994; Ireland et al., 2012), an

Page 93: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

65

important immunosuppressive cytokine that has been shown to actively promote

accumulation of Tregs and also assist Tregs in inhibiting cytotoxic capacity of CD8+ T

cells as well as NK cells within the tumour microenvironment (Ghiringhelli et al., 2005;

Smyth et al., 2006). Further investigation of the transient response to Treg cell depletion

in this model found that more than 50% of TGF-β still remained in the tumour area post

Treg depletion; thereby promoting Treg cell re-accumulation or conversion (Kissick et al.,

2010). In order to overcome the immunosuppressive effect of TGF-β, we developed a

combined regime of Treg depletion and timed TGF-β neutralisation. The idea behind this

double immunotherapy regimen was to target two mechanisms of immune suppression

that would potentially overcome Treg based immunosuppression. In this double

immunotherapy (DI) experiment, mice were treated with a single intra-tumoural

injection of anti-CD25mAb when tumours reached 9mm2 followed by daily intra-

tumoural injections of TGF-β soluble receptor (SR) for 7 days. This resulted in around

50% of the tumours being reduced in size such that they were barely palpable; with

significant inhibition of tumour growth in the remaining mice and a significant increase

in the time to death (TTD) (defined as tumours reaching 100mm2) (Kissick et al.,

2009). This combined treatment also resulted in increased intra-tumoural IFN-γ

concentrations- suggesting an increase in activated cytotoxic T cells, when compared

with either treatments administered as monotherapies (Kissick et al., 2010). Though the

DI treatment was clearly an improvement over the anti-CD25mAb monotherapy, long-

term cures of the tumours was not achieved i.e. all the mice eventually grew end-point

tumours.

In order to determine whether the efficacy of this immune modulating therapy

could be further enhanced towards complete eradication of tumours in the murine

model; the promising new anti-CTLA-4mAb therapy was added (Callahan et al., 2010).

Cytotoxic T lymphocyte antigen (CTLA)-4 is a second receptor on T cells for B7 and a

Page 94: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

66

negative receptor involved in immune-regulation (immune check-point). Under normal

circumstances, CTLA-4 is an antagonist for activated T cell proliferation, which is

essential for maintaining self-tolerance and minimising collateral tissue damage

(Korman et al., 2006). However, in the presence of a growing tumour, CTLA-4 binds

with higher affinity to CD80 and CD86 on antigen-presenting cells (APC), out-

competing CD28. A subsequent inhibition of the proliferation of T cells occurs and

ergo, aids in maintaining T cell anergy (Walunas et al., 1994; Krummel and Allison,

1995; Greenwald et al., 2005). CTLA-4 blockade using anti-CTLA-4mAb allows the

interaction between CD28 on T cells with CD80 and CD86 present on dendritic cells,

thereby potentiating T cell activation and consequently assisting in tumour destruction

(Grosso and Jure-Kunkel, 2013). A number of animal studies have been carried out and

have demonstrated enhanced anti-tumour activity following CTLA-4 blockade;

especially when used in conjunction with a tumour vaccine (van Elsas et al., 1999;

Greenwald et al., 2005). The fully humanised monoclonal antibody, Ipilimumab

(Yervoy, Bristol-Myers Squibb, New York, NY, USA) has been approved by the FDA

for treating patients with advanced melanoma and has been found to enhance overall

survival (Hodi, 2010; Robert et al., 2011).

Therefore, the aim of this study was to examine for improved efficacy of the

combination of CTLA-4 blockade with the previously published Treg depletion/TGF-β

neutralisation (double immunotherapy) in the AE17 murine mesothelioma model. We

hypothesised that this triple immunotherapy treatment regime would result in complete

tumour clearance, associated with a sustained depletion of intra-tumoural Treg cells and

activation of cytotoxic T cells. This overall objective of inducing complete tumour

eradication was achieved.

Page 95: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

67

3.2 Results

3.2.1 Intra-tumoural administration of anti-CD25mAb together with intra-

peritoneal anti-CTLA-4mAb and sequential anti-TGF-βmAb, completely

eradicates established AE17 murine mesothelioma tumours

The efficacy of the triple immunotherapy regime by the addition of anti-

CTLA4mAb to the existing double immunotherapy (DI) was examined. A dose of

100µg of anti-CTLA-4mAb delivered intra-peritoneally (i.p) was added to the DI. This

dosage and administration route was based on a previously published protocol (Hurwitz

et al., 1998). This timed triple immunotherapy (TTI) therefore consisted of intra-

tumoural depletion of Tregs using anti-CD24mAb and systemic blocking of CTLA-4 at

treatment initiation (day0), followed by 7 consecutive days of TGF-β neutralisation

within the tumours with anti-TGF-βmAb.

DI or TTI treatments were initiated in sub-cutaneous AE17 tumour bearing mice

when the tumours reached 9mm2. Figure 3.1 shows that 46% of the mice treated with

the TTI (13/28 from 3 independent experiments) underwent complete tumour

regression. Mice receiving the DI of anti-CD25mAb and anti-TGF-βmAb alone had no

complete cures (median survival 24 days compared to untreated mice with a median

survival of 13.5 days, p<0.0001). The remaining 50% of the mice did not completely

respond to the TTI treatment (partial responders), and underwent delayed tumour

growth, reaching end-points later than untreated controls (median survival 25 days;

p<0.0001). Ergo, the TTI treatment was a substantial improvement over previous

treatments tested in our laboratory, where long-term regression of tumours was never

achieved (Kissick et al., 2012).

Page 96: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

68

Figure 3. 1. Improved survival with complete tumour eradication in 46% of mice treated with timed triple immunotherapy (TTI) a significant improvement over double immunotherapy (DI). C57BL/6J mice were implanted s.c with 1x107 AE17 cells. When the tumours reached an established size of 9mm2 (14±1 days), treatments were initiated. DI regime (■) consisted of one 2 µg i.t dose of anti-CD25mAb (given on day 0) followed by 7 daily i.t injections of 1 µg anti-TGF-βmAb. TTI regime (▲) consisted of one 2 µg i.t dose of anti-CD25mAb and one i.p 100 µg dose of anti-CTLA-4mAb concurrently (given on day 0) followed by 7 daily i.t injections of 1 µg anti-TGF-βmAb. Untreated tumours (●) were used as tumour growth controls for the double and triple treatments. Mice were monitored daily and Kaplan-Meier survival curves were plotted using tumour sizes of 100mm2 as end point (n=10 for untreated mice, n=17 for DI treated mice and n=22 for TTI; from three independent experiments).

Page 97: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

69

3.2.2 Administration of timed triple immunotherapy into established AE17 tumours

inhibits re-accumulation of Tregs in tumour draining lymph nodes

The first step towards understanding the anti-tumour immune response elicited by

the TTI was to examine the impact on the immunosuppressive Tregs. Tumours and

tumour draining lymph nodes (TDLNs: pooled brachial, axillary and inguinal) from

C57BL/6J mice with established 9mm2 AE17 tumours treated with either DI or TTI and

were analysed by flow cytometry on days 0, 1, 3 and 10 for changes in Treg

(CD4+CD25+FoxP3+) populations. Tumours and TDLNs collected for determining Treg

cells on day 0 were from untreated tumour bearing mice before the treatment was

initiated. The population of Tregs from these untreated mice (n=6 from two independent

experiments) were used to determine baseline numbers before treatment initiation. The

DI treatment was used as control to determine the added benefit of anti-CTLA-4mAb in

the TTI.

Figure 3.2A shows that Treg numbers within tumours were not significantly different

in mice that received either DI or TTI over a period of 10 days (p=0.45, post-hoc linear

trend analysis). Interestingly, analysis of the TDLNs showed (Figure 3.2B) significant

differences between the two treatment groups. Prior to treatment, the majority of CD4+

T cells present in the TDLNs are Tregs, making up 65% of the total CD4+ T cell

population (Figure 3.2B). On day 1 post treatment with anti-CD25mAb and anti-CTLA-

4mAb, a significant reduction in total Tregs in the TDLNs to 21,824 (±7,839) was

observed in the TTI treated mice as compared to pre-treatment levels of 55,127 (±1,388)

on day 0 (P<0.01, by post hoc Tukey’s multiple comparison testing). This was close to a

60% drop in the total Treg numbers in the TDLNs of TTI treated mice. Even by day 3,

the total Tregs 21,576 (±14,772) in the TTI treated group remained significantly lower

compared to day 0 (p<0.01) and this low Treg levels continued till day 10 with only

23,715 (±12,764) cells present (p< 0.001). In the DI treated mice, though a significant

Page 98: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

70

decrease in Treg numbers (>80% reduction to 8,675 (±3,209)) one day after treatment

initiation was observed (p<0.001), this level of depletion was not maintained. By day 3,

the Treg numbers had increased by 30% to 25,356 (±10,391) and by day 10, the TDLNs

of the DI treated mice were re-populated by Tregs and were close to the pre-treatment

levels (50,290 (±9,759), p=0.97).

Page 99: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

71

Figure 3. 2. Sustained depletion of Treg cells in TDLNs by TTI treatment even 3 days post treatment completion. Tumours (Figure A) and tumour-draining lymph nodes (pooled unilateral brachial, axillary and inguinal) (Figure B) were removed from mice on days 0, 1, 3 and 10 following treatment with either DI (■) or TTI (▲) and the population of Treg cells (CD4+CD25+FoxP3+) were determined by flow cytometry. Treg numbers from untreated tumour-bearing mice were used as baseline numbers on the day of treatment initiation (day 0). (n=6 for untreated only on day 0 from two experiments, n=3 on days 1 and 3 from one experiment and n=8 for day 10 for both DI and TTI treated mice from two experiments. The errors bars represent standard deviations of the mean). Significant differences in Treg cell numbers on day 10 between DI and TTI were determined by unpaired t-test indicated by** p<0.01. Significant differences between all groups were determined by One-way ANOVA.

Page 100: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

72

3.2.2.1 TTI treatment results in greater maturation of DCs and subsequent activation

of effector T cells in the TDLNs

The effect of the TTI treatment on dendritic cells (DCs) in the TDLNs was assessed

by measuring CD80 expression. DCs were defined as CD11c+MHCII+ cells with the

CD80 expression measured in terms of fold changes in mean fluorescence intensity

(MFI) compared to untreated levels on day 0. One day post treatment initiation when

most of the Tregs had been depleted in the TDLNs; a significant (1.70 (± 0.24) fold)

increase in MFI of CD80 expression was observed in the TTI treated mice (p<0.01) as

shown in Figure 3.3 A. TTI treated mice were also found to have a 2 fold higher MFI of

CD80 expression than the DI treated mice on day 1 (p=0.002). By day 3, the level of

CD80 expression in the TTI treatment group slowly decreased (1.23 (±0.07)) and was

close to pre-treatment levels by day 6 (1.02 (±0.12)) (p=0.12). The fold changes in

CD80 expression remained constant on days 1, 3 and 6 and not significantly different to

the pre-treatment levels in the DI treatment mice (p=0.23). It was only 3 days post

treatment completion (day 10), that fold increase in CD80 expression almost doubled in

both DI (1.99 (±0.11)) and TTI (1.80 (±0.18)) treatment groups compared to the pre-

treatment levels on day 0 (1 (±0.21)) (p=0.001).

The number of CD4+ cells and CD8+ T cells in the TDLNs were also examined.

Activated helper T cell (CD4+CD25+FoxP3-) numbers were determined 1, 3, 6 and

10 days following TTI treatment in the TDLNs. With the sustained depletion of Tregs

in the TTI treated mice; Figure 3.3B shows that one day post Treg depletion, the

number of helper T cells was significantly lower than the pre-treatment levels of

73,778 (±19,256) in both DI (12,158 (±4,965)) and TTI (23,231 (±8,939)) treated

mice (p<0.001 and p<0.01, respectively). This number of helper T cells in the DI

treated mice remained significantly lower throughout the treatment period than

Page 101: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

73

untreated levels (p=0.03, post-hoc linear trend analysis) and TTI treated mice

(p=0.001).

However, in the TTI treated mice, the number of helper T cells began to increase

by day 6 to 146,329 (±73,083) to remain higher than that seen in the DI treatment mice

(38,917 (±7645)) on day 6 (p=0.02, post-hoc un-paired student t-test). A similar trend

was observed in the CD8+ T-cell population with a 2.5 fold increase in numbers on day

6 (1,007,951 (±344,234)) compared to untreated controls (438,381 (±162,170))

(p<0.01), figure 3.3C. This increase in CD8+ T cells was more enhanced in the TTI

treated mice when compared to DI over the time-period that they were analysed

(p=0.01, post-hoc linear trend analysis).

Page 102: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

74

Page 103: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

75

Figure 3. 3 Enhanced CD80 expression by dendritic cells and increased effector T cell levels in mice treated with TTI. C57B/6J mice were implanted s.c with 1×107 AE17 cells and the treatment with either DI (■) or TTI (■) was initiated at a tumour size of 9mm2. Fold increases in expression of CD80 mean fluorescence intensity over untreated controls of CD11c+MHCII+ cells (A), number of CD4+CD25+FoxP3- helper T cells (B) and CD8+ T cells (C) present in the TDLNs (pooled unilateral brachial, axillary and inguinal) were removed from mice on days 1, 3, 6 and 10 post treatment and analysed by flow cytometry. Data collected from untreated tumour-bearing mice on day 0 were used as baselines for the representative populations. Error bars represent standard deviation of the mean. (n=6 for untreated only on day 0 from two experiments, n=3 on days 1 and 3 from one experiment and n=8 for day10 for both DI and TTI treated mice from two experiments) Significant differences between all groups were determined by one-way ANOVA. Significant differences between DI and TTI at the same time-points analyzed by un-paired student t-test: *p<0.05, ** p≤0.01 respectively; n.s-not significant.

Page 104: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

76

3.2.3 A specific anti-tumour memory response results from TTI treatment

Mice cured with TTI treatment remained tumour free for up to three months

post-treatment. The next step was to investigate their resistance to tumour re-challenge.

Cured mice re-challenged with a second AE17 inoculum at the original inoculation site

were found to be resistant to tumour re-challenge for a further two month period of

follow-up (Table 3.1). All re-challenged animals were observed to undergo

inflammation at the inoculation site which resolved within 2-3 days of re-challenge and

the mice remained tumour-free. This memory response was systemic as three cured

mice re-challenged on the contralateral flank also resisted tumour re-challenge. Naïve

control mice inoculated at the time of the re-challenge with the same batch of AE17

cells all grew tumours that reached end-points within the expected time frame (median

survival 17 days).

To determine if immunological memory was tumour specific, TTI cured mice

were re-challenged with B16 melanoma cells (syngeneic tumour type). Cured mice were

either re-challenged at the original inoculum site or their distal flank with B16

melanoma cells, and the tumour growths were compared to naïve C57BL/6J mice

challenged at the same time (Table 3.1). In contrast to the AE17 re-challenge, all cured

mice re-challenged with B16 cells developed end-point tumours. The B16 tumours

inoculated in the naïve mice grew within the expected time frame (median survival was

13 days). Compared to naïve mice, the TTI treated cured mice re-challenged on the

same and opposite flanks grew the B16 tumours significantly slower (median survival

of 24 (p= 0.0046) and 31 days (p=0.01) respectively), as summarised in table 3.1.

Page 105: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

77

n-number of mice challenged; Avg-average and SD- Standard deviation.

Naïve mice challenged TTI cured mice re-challenged with original AE17 cells Original site Contralateral site

n Avg tumour size (mm2) ± SD at day 20

n Avg tumour size (mm2) ± SD at day 20

n Avg tumour size (mm2) ± SD at day 20

9 57.84 ± 12.72 17 0 3 0

Naïve mice challenged TTI cured mice re-challenged with B16 melanoma Original site Contralateral site

n Avg tumour size (mm2) ± SD at day 14

n Avg tumour size (mm2) ± SD at day 14

n Avg tumour size (mm2) ± SD at day 14

3 99 ± 9 5 27.66 ± 25.08 3 18.6 ± 0.96

Table 3. 1 TTI cured mice are resistant to re-challenge with original inoculum (AE17) and partially resistant to syngeneic B16 melanoma re-challenge. AE17 tumour-bearing C57BL/6J mice cured by the TTI treatment were re-challenged 1-3 months post treatment completion with either the original AE17 (1 × 107 cells) inoculum or with B16 melanoma (5 × 105 cells) at the same inoculum site or the contralateral site. Naïve mice were challenged with the same batch of AE17 and B16 cells as controls.

Page 106: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

78

3.2.3.1 Sustained depletion of Tregs in the TDLNs of re-challenged AE17 cured mice

In response to the tumour re-challenge, accumulation of Tregs within the TDLNs

of these cured mice was investigated. The TDLNs of cured mice that have been re-

challenged with AE17 cells were analysed for Tregs (CD4+CD25+FoxP3+) by flow

cytometry 3 days post challenge. The naïve mice challenged with the same inoculum 3

days prior to the analysis were used as controls. Compared to the naïve mice with

percentage of Tregs at 50.2 ± 1.6 %, the cured mice were significantly lower (43.7 ± 2.8

%, p<0.007 by post hoc student t-test) as shown in figure 3.6. This sustained depletion

of Tregs induced by the TTI during treatment (as observed earlier in figure 3.2) is

maintained in the TDLNs of cured mice, even 1-3 months post treatment completion.

Page 107: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

79

Figure 3. 4 Sustained depletion of Tregs in the TDLNs of re-challenged cured mice. Cured mice (■) that remained tumour free 1-3 months post treatment completion were re-challenged with 1 × 107 AE17 cells. Naïve untreated mice (●) challenged with the same inoculum were used as controls. Tumour-draining lymph nodes (pooled unilateral brachial, axillary and inguinal) were removed from these mice 3 days post tumour challenge and percentage of CD4+CD25+ T cells that were also Foxp3+, were analysed by flow cytometry. (n=3 for naïve challenged and n=7 for cured re-challenged from one experiment. The errors bars represent standard deviations of the mean). Significant differences in Treg percentages between naïve and cured were determined by unpaired t-test indicated by** p<0.01.

Page 108: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

80

3.2.3.2 Induction of memory T cells by the TTI treatment

A significant increase in expression of the T cell memory marker CD44 was

observed in the cured mice (15.6 ± 4.5%) (figure 3.7A). This increase in CD44+

expression was close to double the percentage of CD44+CD4+ T cells observed in naïve

mice challenged for the first time with AE17 tumour cells (9.09 ± 4.5%, p=0.01

significance determined by post-hoc unpaired t-test). A similar doubling in percentage

of CD8 T cells co-expressing CD44 marker was also found in the cured mice (19.8 ±

6.5%); compared to the naïve challenged mice (10.17 ± 3.9%, p=0.004), as shown in

figure 3.7B. This activation of the memory response in both the CD4+ and CD8+ T cell

compartments determined 3 days post tumour re-challenge was long-lasting and

tumour-specific, as confirmed by the re-challenge experiments carried out and

summarised in table 3.2.

Page 109: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

81

Figure 3. 5 Induction of memory T cells in mice cured of AE17 murine mesothelioma by TTI treatment. Mice cured of AE17 tumours with TTI were re-challenged at the same anatomical site with AE17 cells (1 × 107 cells) 1-3 months post treatment completion. Naïve fully immune-competent C57BL/6J mice were challenged with the same batch of tumour cells and were used as controls (●). 3 days post challenge, the TDLNs (pooled unilateral brachial, axillary and inguinal) were removed from mice and analysed for the expression of the memory CD44 marker within A) CD4+ T cell (▲) and B) CD8+ T cell (♦) compartments (n=6 for naïve challenged mice and n=12 for cured re-challenged mice from two experiments respectively). Significant differences in CD44 marker expression between naïve and cured were determined by unpaired t-test indicated by** p<0.01.

Page 110: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

82

3.3 Discussion

Tumour eradication was achieved in close to 50% of mice bearing established

AE17 mesothelioma tumours treated with the TTI. This was a significant improvement

over any of the single or double immunotherapy treatments previously trialled in our

laboratory. Depletion of Tregs alone by a single anti-CD25mAb treatment only delayed

the growth of AE17 tumours, though the treatment did not confer any significant

improvement in tumour growth inhibition. Monotherapies exclusively involving just

TGF-β neutralisation or CTLA-blockade, both resulted in only a marginal improvement

in survival time (median survival of 17 and 20 days respectively) but ultimately, all the

mice grew end-point tumours (Kissick et al., 2012). Complete tumour eradication or

long-term survival of mice bearing murine mesotheliomas (AC29 and AB12) have been

previously achieved by treatment with anti-CD25mAb by other groups. However, it is

important to note that, these experiments involved systemic depletion of Tregs with anti-

CD25mAb before tumour challenge (Anraku et al., 2010). Though single treatment

approaches are ideal for certain diseases, the factors involved in immune suppression in

the presence of a growing tumour are multivariate and more complex. This supports our

hypothesis that combinatorial immunotherapy approaches improved the efficacy to

overcome tumour immune evasion.

Different double immunotherapy combinations trialled in our laboratory on

C57BL/6J mice bearing AE17 tumours showed that the increased survival time due to

significant tumour growth inhibition was greatest with the combination of TGF-β

neutralisation and Treg depletion (median survival improved to 24 days). Neither the

combinations TGF-β neutralisation/CTLA-4 blockade nor Treg depletion/CTLA-4

blockade conferred any improvement in survival (median survival of 19 days and 21

days respectively) when compared to mice that received CTLA blockade alone (median

Page 111: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

83

survival 20 days). Ultimately, long-term regression of tumours was never achieved

(Kissick et al., 2012).

Combining all three components (anti-CD25mAb, anti-TGF-βmAb and anti-

CTLA-4mAb) conferred a significant survival advantage over the best DI combination

of anti-CD25mAb and anti-TGF-βmAb. Most importantly, for the first time in our

laboratory, complete eradication of tumours in 46% of mice bearing established AE17

tumours was achieved with the novel TTI treatment (Figure 3.1). The remaining 50% of

mice that did not completely respond to the treatment (partial responders) did undergo

delayed tumour growth with increased survival time when compared to untreated mice.

This question of partial responders to the TTI could be a result of a sub-optimal

response to the treatment, and perhaps experiments involving recovery of these mice

with additional top-ups or by titrating the treatment components could enhance the cure

rate (see chapter 4 section 4.2.4).

From the previous work conducted in our lab, we knew that the DI treatment

only achieved transient tumour regression. This observation was in conjunction with the

re-accumulation of Tregs 7-10 days post depletion within the tumour microenvironment

(Kissick et al., 2010). Despite the addition of anti-CTLA-4mAb to the combination

therapy, Treg numbers were not significantly different between the DI and TTI treated

mice in the tumours as analysed by flow cytometry (Figure 3.2A). This could be

because, the treatment components that are injected intra-tumourally (anti-CD25mAB

and anti-TGF-βmAb) are not different between the DI and TTI (anti-CTLA-4mAb in

the TTI treatment is administered i.p). However, it was found that even 3 days post TTI

treatment completion; the Treg levels remained significantly lower in the TDLNs when

compared to Treg levels reaching pre-treatment levels in the TDLNs of mice that

received the DI (Figure 3.2B). This was a significant finding, as this suggested that the

Page 112: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

84

TTI treatment had a more profound impact on inhibiting the re-accumulation of Tregs

within the TDLNs (Kissick et al., 2012). Selective recruitment and expansion of Tregs in

the periphery and within other tumours has been reported in different types of cancers

(Woo et al., 2001; Liyanage et al., 2002; Wolf et al., 2003; Curiel et al., 2004). It is,

therefore, possible that the TTI approach developed here may be useful in treating other

tumours (see chapter 6).

Although it is well recognised that Treg depleting therapies can inhibit tumour

development in several murine models and human cancers; a common cause for concern

is the long-term side effects of autoimmune diseases observed with the administration

of Treg cell depletions systemically or for prolonged periods of time (Takeuchi et al.,

2004; Ruddle and Prince, 2007). Another cause for concern is the production of

neutralizing antibodies by the patient, as observed by Sterman et al (2010) with their

phase I trial of repeated IFN-γ gene transfer treatment for treating mesothelioma and

metastatic pleural effusions (Sterman et al., 2010). In relation to the novel TTI

treatment, the following observations can be made: a) by employing intra-tumoural

administration, the risk of systemic depletion of Tregs is substantially reduced. Indeed, no

auto-immune based ill-effects (such as weight loss, lethargy, reduced appetite, bloating

or diarrhoea) were observed in any of the treated mice, b) the complete cures without

the need for repeated administration suggests that neutralising antibody problems will

be similarly reduced, and c) the anti-CTLA-4mAb was systemically administered in

these experiments and hence could cause auto-immune issues. It would be appropriate

to examine the possibility of incorporating this reagent into the intra-tumoural

administration (see chapter 4).

Tregs are known to regulate the proliferation and maturation of DCs resulting in a

shift of the DC phenotype in an IL-10/TGF-β dependent manner (Kim et al., 2006;

Page 113: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

85

Onishi et al., 2008). Concurrent to the maintenance of low Tregs in the TDLNs over the

time-period of treatment with TTI, increased expression of CD80 by DCs was also

found in the TDLNs of TTI treated mice (Figure 3.3A). The increased CD80 expression

observed on day 1 in the TTI treated mice could suggest the removal of brakes on the

proliferation of DCs. This activation of DCs correlated with the expansion of helper T

cells (CD4+CD25+FoxP3-) and CD8+ T cells within the TDLNs by day 6 (Figures 3.3 B

and C respectively). The decrease in helper CD4+ T cells and CD8+ T cells observed on

day 10 could indicate the possible migration of DC primed CD4+ and CD8+ T cells into

the tumour microenvironment, as the TTI treatment induced tumour clearance in the

tumour-bearing mice. Flow cytometric analysis for the detection of these subsets of T

cells within the tumours would aid in confirming the migration of these anti-tumour

effector cells into growing tumours. Also, it is important to note that CD80 is just one of

many markers that are expressed on DCs. Future experiments that examine other

maturation and proliferation markers such as CD86, CD83 and CD40 would further

strengthen the phenomenon observed. Another option is to analyse concentrations of IL-

12 that are secreted by DCs and are necessary for the differentiation of activated T cells

into T helper cells.

The TTI treated mice that underwent complete tumour regression were kept for

between 1-3 months with no re-emergence of tumours. Development of immunological

memory in these mice was substantiated with these long-term cured mice failing to

develop tumours when re-challenged with the same inoculum of AE17 tumour cells

(Table 3.1). These re-challenged mice remained tumour free for a further two months

post re-challenge, irrespective of site of inoculum, thereby suggesting systemic immune

memory. This emergence of resistance to re-challenge in the TTI treated long-term

cured mice denotes the first reversal of dominant tolerance within the tumour

microenvironment.

Page 114: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

86

In response to tumour re-challenge, a significant doubling in the percentage of

CD4+ and CD8+ T cells co-expressing the memory CD44 marker was seen in the

TDLNs of TTI-cured mice, just 3 days after tumour re-challenge, when compared to

untreated, naive mice challenged for the first time with a single inoculum of AE17

tumour cells (Figure 3.7 A and B). Preliminary analysis of the tumour implantation site

of three of the TTI cured and re-challenged mice showed a rapid accumulation of CD4+

T cells at the re-challenge site just 3 days after tumour challenge. This was not observed

in the untreated naive mice implanted for the first time with tumour cells (data not

shown).

The systemic immunity elicited in the cured mice was found to be tumour-

specific, as re-challenge with B16 melanoma (syngeneic tumour model for the

C57BL/6J) resulted in tumour growth. Though the B16 tumours grew in these AE17

cured mice, their growth to end-point was significantly slower compared to their growth

in untreated naïve mice. This could be indicative of some partial immunity generated in

the cured mice towards the melanoma. Melanoma and mesothelioma are both cancers

arising from squamous epithelial cells, and perhaps share common tumour antigens

(Rivera et al., 2012). This could be the reason for the slow growth of melanoma

tumours in the cured mice. Perhaps re-challenge with a different syngeneic tumour cell

line such as EO771 breast carcinoma could help in understanding this notion of partial

immunity in the future.

Together, these data support the hypothesis that targeting multiple mechanisms

of immune suppression induces a superior anti-tumour immune response. Data from

these experiments have provided evidence that long-term tumour eradication can be

achieved, with continued resistance to tumour re-challenge when various aspects of

immune suppressive mechanisms are targeted. More importantly, all treatments used in

Page 115: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

87

this triple combination approach have human equivalents, that have been approved for

treating cancer patients. Denileukin Diftitox (DD), a recombinant fusion protein that

consists of peptide sequences of diphtheria toxin and human IL-2, is a FDA approved

drug for depleting Tregs in different cancers (Dannull et al., 2005; Dang et al., 2007;

Mahnke et al., 2007; Gerena-Lewis et al., 2009). Moreover, as mentioned earlier,

Ipilimumab (anti-CTLA-4mAb) has been approved for treating patients with advanced

melanoma (Hodi, 2010; Robert et al., 2011).

The concept of targeting multiple mechanisms of immune suppression is

evidenced in the literature, with combination therapies involving more than two

components being tested in various other laboratories. For example, combination

therapy involving 4-1BB (CD137) activation with anti-CTLA-4mAb treatment given as

a triple therapy in the setting of radiation therapy was found to improve survival of mice

with glioblastomas (Belcaid et al., 2014). More recently, a phase I clinical trial was

reported, that involved administering castration-resistant prostate cancer patients with

Ipilimumab, vaccine containing transgenes for prostate-specific antigen (PSA) and a

triad of co-stimulatory molecules (PROSTVAC). 30 patients were trialled, and these

patients were found to have increased overall survival (OS) (Jochems et al., 2014).

Combinatorial therapies are leading the way into the future of cancer treatments.

Coupled with the fact that current mesothelioma treatments are of low efficacy and

provide minimal improvements in survival rates, makes the TTI a promising new

development. Testing the efficacy of the TTI in a different murine mesothelioma model

would assist in translation of this novel TTI for treating human patients with

mesothelioma. Based on this, the following chapter will investigate the efficacy of the

TTI in the AB1 (BALB/c) murine mesothelioma model.

Page 116: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

88

Page 117: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

89

CHAPTER 4 Improved efficacy of the triple immunotherapy in the non-TGF-β

secreting murine mesothelioma model (AB1 model)

Page 118: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

90

Page 119: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

91

4.1 Introduction

The previous research in the AE17 murine mesothelioma model showed that the

timed triple immunotherapy (TTI) completely eradicated tumours in close to 50% of the

treated C57BL/6J mice. This was a significant improvement, as tumour eradication has

never been achieved with single or double immunotherapies (DI) trialled in our

laboratory. Long-term survival of treated mice was achieved with tumours failing to re-

grow post cessation of treatment. Development of immunological memory was

confirmed by flow cytometric analyses of memory T cells in TDLNs, and with the

continued resistance of the cured mice to AE17 tumour re-challenge. This denotes a

reversal of the dominant tolerogenic immune-surveillance present in the tumour

microenvironment and is particularly impressive, given the ability of the AE17 tumour

cells to secrete TGF-β that is known to recruit and/or convert Tregs (Chen et al., 2003).

In this chapter the TTI approach was applied to the BALB/c murine mesothelioma

(MM) tumour model, AB1. AB1 tumours were derived from the peritoneal cavity of H-

2d BALB/c mice injected with crocidolite asbestos fibres. Ascites, solid tumours or

peritoneal flushing were collected from the mice and grown in-vitro, where they were

serially passaged to derive the MM tumour cell line (Davis et al., 1992).

Immunohistological staining of intra-peritoneal AB1 tumour biopsies have already

provided phenotypic evidence for the increased accumulation of regulatory T cells

(CD4+CD25+FoxP3+) within growing tumours (Veltman et al., 2010). The AB1 MM

has also shown sensitivity to immunotherapy (Caminschi et al., 1998; Hegmans et al.,

2005; Mahaweni et al., 2013). Moreover, the tumour cells themselves are known to not

secrete the immunosuppressive cytokine TGF-β (Fitzpatrick et al., 1994). It was,

therefore, hypothesised that the TTI approach would induce significantly greater

percentage of AB1 tumour bearing mice to undergo complete tumour clearance as these

tumours are non-TGF-β secreting.

Page 120: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

92

Experiments described in this chapter aimed a) to study the efficacy of the triple

immunotherapy treatment in a murine mesothelioma that does not secrete TGF-β (AB1)

b) to examine the importance of timing in the delivery of the three immunotherapeutic

agents and c) to improve the 50% cure rate achieved in the AE17 murine mesothelioma

model.

4.2 Results

4.2.1 Timed administration of antibodies targeting CD25, TGF-β and CTLA-4

completely cleared established AB1 sub-cutaneous tumours in 100% of

BALB/c mice

AB1 mesothelioma cells were subcutaneously injected into BALB/c mice and

the TTI treatment consisting of anti-CD25mAb, anti-TGF-βmAb and anti-CTLA-4mAb

was initiated when tumours reached 9mm2; as described for AE17 tumours in the

previous chapter and in Kissick et al, 2012. Immediately post treatment, a period of

local inflammation at the tumour site was observed with tumours swelling to a

maximum size of 15.4 (±4.8) mm2 in the TTI treated mice on day 3 (Figure 4.1). By

day 10 (3 days post treatment completion), no palpable tumour could be felt at the

inoculation site of the TTI treated mice. This was significantly different to the tumours

growing in the untreated controls which had an average size of 28.6 (±11.6) mm2

(p=0.001 significance determined by un-paired student t-test). These TTI treated mice

remained free of tumours for at least 3 months post-treatment completion.

This timed treatment of established AB1 mesotheliomas was trialled in 16 mice,

and figure 4.2 shows that 100% of animals (16/16, from three independent experiments)

underwent complete tumour clearance, compared to untreated controls (median

untreated survival 19 days; p<0.0001).

Page 121: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

93

Figure 4.1 Complete tumour eradication in 100% of AB1 tumour bearing BALB/c mice treated with TTI. BALB/c mice were implanted s.c with 1 × 106 AB1 tumour cells. TTI was initiated at a tumour size of 9mm2 (7 ± 1 days). TTI regime (▲) consisted of one 2 µg i.t dose of anti-CD25mAb and one i.p 100 µg dose of anti-CTLA-4mAb concurrently (given on day 0) followed by 7 daily i.t injections of 1 µg anti-TGF-βmAb. Untreated tumours (●) were used as tumour growth controls. (n=3 for untreated mice and n=5 for TTI; from one experiment). The data are shown as mean ± SD.

Page 122: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

94

4.2.2 A combined, single administration of the triple treatment as a cocktail (TIC) is

sufficient to induce complete clearance of AB1 tumours in close to 90% of

animals

Clinically, a single immunotherapy treatment regime would be easier to

implement than a timed daily dose regimen. A combined triple immunotherapy cocktail

(TIC) delivered directly into the established 9mm2 tumours via a single injection was,

therefore, tested. The TIC for intra-tumoural administration combined the anti-

CD25mAb at the same total concentration employed in TTI, the anti-TGF-βmAb at 2

µg instead of 1 µg and the anti-CTLA-4mAb at 2 µg reduced from the 100 µg

administered intra-peritoneally. The TIC was developed to test the importance of timed

component administration relative to the simplicity of a single injection. Figures 4.2A

and B show that a single intra-tumoural injection of the TIC was as efficient as the TTI

(p=0.1636), with complete tumour clearance seen in 88% of treated mice (15/17 mice

over three independent experiments) compared to untreated controls (p<0.0001)

(Krishnan et al., 2014).

Page 123: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

95

Figure 4. 2 Tumour growth kinetics and survival of AB1 tumour bearing BALB/c mice following treatment with the timed triple immunotherapy (TTI) or the triple immunotherapy cocktail (TIC). Mice were implanted s.c. with 1 × 106 AB1 tumour cells. The triple immunotherapy, either TTI (▲) or TIC (■), was initiated at a tumour size of 9 mm² with tumour growth compared to untreated mice (●). Mice were monitored daily for (A) survival and plotted as Kaplan-Meier survival curve with 100mm2 tumour as endpoint and (B) tumour growth (mean ± SD) was calculated with measurements made using microcalipers. The pooled data are from three independent experiments (n=16 for untreated mice and TTI treated mice and n=17 for TIC treated mice).

Page 124: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

96

4.2.3 Induction of systemic immune response in cured mice

Mice treated successfully with TTI were kept 1-3 months post treatment

completion, and all tumours failed to regrow. These long-term cured mice also failed to

re-grow tumours when re-challenged with the same inoculum of AB1 cells at either the

original or contralateral inoculation sites (Table 4.1). In comparison, 100% of control

naïve BALB/c mice inoculated with the same batch of AB1 cells grew tumours to

endpoint (100mm2) (Table 4.1).

Similar to mice cured by the TTI; Table 4.1 shows that BALB/c mice cured by

the TIC failed to regrow tumours when challenged on the same flank with AB1 tumour

cells one month after complete tumour clearance. Naïve BALB/c mice were inoculated

with AB1 cells at the same time as the re-challenge, and all grew end-point tumours.

Page 125: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

97

n-number of mice challenged; Avg-average; SD- Standard deviation; and ND-not done.

Naïve mice challenged TTI cured mice re-challenged Original site Contralateral site

n Avg tumour size (mm2) ± SD at day 20

n Avg tumour size (mm2) ± SD at day 20

n Avg tumour size (mm2) ± SD at day 20

9 57.84 ± 12.72 11 0 3 0

Naïve mice challenged TIC cured mice re-challenged Original site Contralateral site

n Avg tumour size (mm2) ± SD at day 20

n Avg tumour size (mm2) ± SD at day 20

n Avg tumour size (mm2) ± SD at day 20

6 54.54 ± 6.91 5 0 ND ND

Table 4. 1 BALB/c mice cured of established AB1 tumours with TTI or TIC are resistant to re-challenge. AB1 tumour-bearing mice cured by TTI or TIC treatment were re-challenged 1-3 months after complete tumour clearance with 1 × 106 AB1 cells at the same inoculum site or the contralateral site. Naïve mice were challenged with the same batch of AB1 cells as controls.

Page 126: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

98

4.2.3.1 Susceptibility of cured mice to re-challenge with syngeneic alternate tumour

type-4T1 breast carcinoma

To establish whether this immunological memory in the cured mice was tumour

specific, mice cured of AB1 tumours by TIC treatment were re-challenged with

syngeneic 4T1 breast carcinoma cells. Similar to the re-challenge experiment described

above, the TIC cured mice were either re-challenged at the original inoculum site or the

contralateral site with 4T1 breast carcinoma cells. Naïve BALB/c mice were also

inoculated at the same time as the re-challenge (Table 4.2). All TIC cured mice re-

challenged with syngeneic 4T1 cells, developed tumours irrespective of whether they

were challenged on the original or contralateral flanks. 20 days post 4T1 tumour cell

inoculation; compared with tumour growth in naïve BALB/c mice challenged with 4T1

cell, no significant difference in tumour growth was observed between mice injected on

the original site or contralateral sites (p=0.23 and p=0.208, respectively).

Page 127: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

99

n-number of mice challenged; Avg-average; SD- Standard deviation.

Naïve mice challenged TIC cured mice re-challenged with 4T1 cells

Original site Contralateral site

n Avg tumour size (mm2) ± SD at day 20

n Avg tumour size (mm2) ± SD at day

20

n Avg tumour size (mm2) ± SD at

day 20

3

40.33 ± 9.60

3

62.66 ± 26.02

2

52.50 ± 4.94

Table 4. 2 Immunological memory generated in BALB/c mice cured of established AB1 tumours with TIC are tumour specific. AB1 tumour-bearing mice cured by TIC treatment were re-challenged 1 month after complete tumour clearance with 1 × 105 4T1 cells at the same inoculum site or the contralateral site. Naïve mice were challenged with the same batch of 4T1 cells as controls.

Page 128: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

100

4.2.4 Incomplete neutralisation of TGF-β within the AE17 tumour

microenvironment is integral to the suboptimal response generated in AE17

tumour bearing mice treated with TTI.

Compared to only 50% survival in the TGF-β secreting AE17 mesothelioma

model (Chapter 3 and Kissick et al., 2012), complete tumour clearance in 100% of mice

treated with TTI in the AB1 murine mesothelioma model was achieved (Krishnan et al.,

2014). The improved outcome seemed most likely associated with the lack of TGF-β

secretion from the AB1 murine mesothelioma cells (Fitzpatrick et al., 1994). This

formed the basis for the next set of experiments; that aimed at improving the 50% cure

rate of AE17 tumour bearing C57BL/6J mice treated with TTI. The 50% of TTI treated

mice that grew end-point tumours were called partial responders, as they had a median

survival of 25 days and had a significantly delayed time to death (TTD) when compared

to UT controls (median survival 13.5 days, p<0.0001)).

The time-point at which TTI treated mice, based on the response to the TTI

treatment, differentiated into responders and partial responders, was investigated. Once

the treatment regime of 8 days was completed, due to the local inflammation at the

tumour site, tumours swelled to a maximum size of 8 (±2) mm2 in the TTI treated mice

on day 7. By day 10, the swelling continued and reached a maximum of 10 (±3) mm2 as

shown in figure 4.3A. At this point, the tumour sizes in both the TTI and DI treated

mice were similar and were significantly different to the untreated controls (p=0.001

and p=0.01 respectively, significance determined by un-paired student t-test,

respectively).

The threshold, wherein the separation within the TTI treated mice into a)

responders that undergo complete tumour clearance and b) the partial responders, took

place 3 days post-treatment completion (day 10). It is at this point in the TTI treated

Page 129: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

101

mice, that the tumours either gradually decreased in size (3/5, 60%) and no palpable

tumours were detectable (responders); or increased in size (2/5, 40%) and eventually

reached end-point tumours (partial-responders) as shown in figure 4.3B. Though the

partial-responders reached end-points significantly slower than untreated controls as

mentioned earlier, their TTD was not significantly different from DI treated mice

(median survival 25 days, p=0.27 significance determined by log-rank test).

Page 130: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

102

Page 131: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

103

Figure 4. 3 Tumour growth of partial responders to the TTI treatment is not significantly different from the DI treated mice. C57BL/6J mice were inoculated s.c with 1 × 107 AE17 cells and treatments were initiated when the tumours reached 9mm2 (14 ±1 days). A) Growth of AE17 tumours in TTI (■) (n=5) and DI (▲) (n=5) treated mice post treatment initiation compared to UT controls (●) (n=3). B) Growth of AE17 tumours in TTI diverged into responders (▼) (n=3) and TTI partial responders (♦) (n=2) compared to DI and UT controls. Treatment with DI consisted of one i.t dose of anti-CD25mAb (given on day 0) followed by 7 daily i.t injections of anti-TGF-βmAb. TTI regime consisted of one i.t dose of anti-CD25mAb and one i.p dose of anti-CTLA-4mAb concurrently (given on day 0) followed by 7 daily i.t injections of anti-TGF-βmAb. Tumour growths were monitored daily with a tumour size of 100 mm2 considered as end-point. The data are shown as mean ± SD.

Page 132: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

104

In a separate experiment, the immunomodulatory environment within the

TDLNs of partial responders (average tumour size of 28 (±8) mm2) was investigated, in

comparison to the untreated end-point tumour bearing controls (average tumour size of

68 (±21) mm2). The immune responses elicited in the partial responders were not

significantly different from the untreated controls (Table 3.1). These results demonstrate

that despite the increased TTD in the partial responders, the activation of immune cells

in these mice is nevertheless sub-optimal.

Page 133: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

105

Immune cells in TDLNs Total number of cells (Avg ± SD) Significance

p<0.05 Partial

responders

(n=3)

UT controls (n=3)

CD4+CD25+ T cells 38,242 ± 7,458

28,773 ± 5, 938 0.16 (n.s)

CD8+ T cells 263,913±

144,896

387,378 ± 46,135 0.27 (n.s)

CD11c+MHCII+ cells 64,745 ± 29,761 89,386 ± 95,675 0.69 (n.s)

n, number of mice analysed; Avg, average; SD, Standard deviation; n.s-not significant

Table 4. 3 No significant improvement in immune cell numbers in partial responders. TDLNs (pooled unilateral brachial, axillary and inguinal) were removed from TTI treated partial responders and total number of CD4+CD25+ T cells, CD8+ T cells and CD11c+MHCII+ dendritic cells were analysed by flow cytometry and compared with the respective populations present in untreated end-point tumour bearing controls. Significant differences in immune cell numbers between partial responders and untreated controls were determined by t-test.

Page 134: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

106

4.2.4.1 Sub-optimal response observed in partial responders despite attempts at

recovering them with additional top-up of immunotherapy treatment

With the TTD of partial responders being similar to DI treated mice, the idea

that perhaps this was due to a sub-optimal treatment needed to be investigated. In order

to do that, an experimental protocol involving treatment of tumour bearing mice at

9mm2 with the normal TTI treatment followed by either a single i.p shot of anti-CTLA-

4mAb on day 10 (figure 3.5A) or secondary regimen of i.p anti-CTLA-4mAb and

sequential TGF-βmAb dosages (i.t) starting from day 10 (figure 3.5B) was assessed. A

second dose of anti-CD25mAb was not included in the secondary treatment protocol; as

previous work in our laboratory has shown that excess depletion with anti-CD25mAb

within the tumour microenvironment accelerated tumour growth (Kissick et al., 2009).

Figures 3.5A and C show that the mice that received the additional top-up of a

single i.p injection of anti-CTLA-4mAb had a median survival of 33 days which was

significantly longer than UT controls (median survival 17 days, p=0.01). Though their

survival was longer (by 16 days) than the untreated mice, only 50% (3/6 from one

experiment) of the treated mice underwent complete tumour regression.

A similar trend was observed in the mice treated with TTI coupled to the

secondary regimen of single i.p treatment with anti-CTLA-4mAb on day 10 followed by

7 daily i.t injections of anti-TGF-βmAb. Though their median survival (40.5 days) was

significantly longer than vehicle (median survival 16 days, p<0.01) or UT controls

(p<0.01); only 50% of the treated mice (3/6 from one experiment) underwent complete

tumour regression (Figures 3.5 B and C). Also, the survival of the secondary

administration group was not significantly different from the group that received just a

single additional top-up dose of anti-CTLA-4mAb (p=0.82). With regards to the vehicle

control group, the additional i.t injections of PBS within the tumours made no

Page 135: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

107

difference to tumour growth as their median survival was not significantly different

from UT controls (p=0.68).

Page 136: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

108

Page 137: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

109

Figure 4. 4 Failed recovery of partial responders despite attempts with secondary round of immunotherapy. C57BL/6J mice were inoculated s.c with 1 × 107 AE17 cells and primary treatments were initiated when the tumours reached 9mm2 (14 ±1 days). (A) TTI treated mice received secondary single i.p dose of anti-CTLA-4mAb alone (▲) (n=6) on day 10 and their tumour growths were compared with UT controls (●) (n=3). (B) TTI treated mice received secondary regime consisting of one i.p dose of anti-CTLA-4mAb (given on day 10) followed by 7 daily i.t injections of anti-TGF-βmAb (▲) (n=6). Their tumour growths were compared with UT controls and vehicle control mice (■) (n=3). Vehicle control mice received 8 100 µl i.t injections of PBS from day 0-day 8, followed by only 5 100 µl i.t injections of PBS from day10-14 as the tumour sites were too inflamed to complete the remaining 3 i.t injections. C) Tumour growth was monitored daily and Kaplan-Meier survival curves were plotted using tumours with an end-point of 100mm2. The data are from one experiment and are shown as mean ± SD. Unpaired t-test were used to determine significant differences in tumour growth on day 18 with *p<0.05 between UT controls and TTI+1 anti-CTLA-4mAb treated group and * p<0.05 between vehicle controls and TTI +1 anti-CTLA-4mAb+ 7 anti-TGF-βmAb treated group.

Page 138: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

110

4.2.4.2 Increased anti-TGF-βmAb dosage in the original TTI results in complete

tumour eradication in AE17 tumour bearing mice

Based on the conclusions of the previous experiment, it was hypothesised that

targeting the TGF-β early with double the dosage could have a more profound effect on

neutralisation than a second round of late treatments.

C57BL/6J mice with established 9mm2 AE17 tumours were treated with the

modified TTI (mTTI); that consisted of anti-CD25mAb and anti-CTLA-4mAb at the

same total concentrations as before, but anti-TGF-βmAb was increased from 1 µg to 2

µg over the initial 7 day treatment period. After treatment initiation, tumours swelled to

a maximum size of 17.50 (±5.58) mm2 in the mTTI treated mice on day 4 (Figure 4.5).

By day 10 (3 days post treatment completion), tumours had reduced to an average size

of 4 (±0.70) mm2 and were barely palpable at the inoculation site of the treated mice.

This was found to be significantly different to the untreated controls which had an

average tumour size of 97.2 (±4.5) mm2 (end-point tumour size) (p<0.0001 significance

determined by un-paired student t-test).

Page 139: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

111

Figure 4. 5 Complete tumour eradication in 100% of AE17 tumour bearing C57BL/6J mice treated with mTTI. C57BL/6J mice were implanted s.c with 1 × 107 AE17 tumour cells. Modified TTI (mTTI) was initiated at a tumour size of 9mm2. mTTI regime (▲) consisted of one 2 µg i.t dose of anti-CD25mAb and one i.p 100 µg dose of anti-CTLA-4mAb concurrently (given on day 0) followed by 7 daily i.t injections of 2 µg anti-TGF-βmAb. Mice with untreated tumours (●) were used as tumour growth controls. (n=7 for untreated mice and n=11 for mTTI; from two experiment). The data are shown as mean ± SD.

Page 140: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

112

4.2.4.2.1 Induction of systemic immune response in the mice cured using mTTI

Mice that were cured successfully of AE17 tumours with mTTI were kept for

four weeks post-treatment completion, and all mice failed to regrow tumours. These

cured mice also failed to re-grow tumours when re-challenged with the same inoculum

of AE17 cells at either the original or contralateral inoculation sites (Table 4.4). In

comparison, 100% of control naïve C57B/6J mice inoculated with the same batch of

AE17 cells grew tumours to endpoint (100mm2) (Table 4.4).

Page 141: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

113

n-number of mice challenged; Avg-average; SD- Standard deviation.

Naïve mice challenged mTTI cured mice re-challenged with AE17 cells

Original site Contralateral site

n Avg tumour size (mm2) ± SD at day 9

n Avg tumour size (mm2) ± SD at

day 9

n Avg tumour size (mm2) ± SD at

day 9

6

79.70 ± 13.90

8

0

3

0

Table 4. 4 Mice cured of established AE17 tumours with mTTI are resistant to re-challenge. AE17 tumour-bearing C57BL/6J mice cured by mTTI treatment were re-challenged 4 weeks after complete tumour clearance with 1 × 107 AE17 cells at the same inoculum site or the contralateral site. Naïve mice were challenged with the same batch of AE17 cells as controls.

Page 142: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

114

4.3 Discussion

The work described in this chapter has demonstrated complete tumour clearance in

100% of mice treated with the TTI in the non-TGF-β secreting AB1 murine

mesothelioma model (Figure 4.1), compared to the 50% cures observed in the TGF-β

secreting AE17 mesothelioma model (Chapter 3 and Kissick et al., 2012). This supports

the hypothesis, as the TTI approach induced significantly greater percentage of tumour

bearing BALB/c mice to undergo complete tumour clearance in the non-TGF-β

secreting AB1 mesothelioma model. The improved outcome is most likely associated

with the lack of TGF-β secretion from the AB1 murine mesothelioma cells (Fitzpatrick

et al., 1994). Increasing the anti-TGF-βmAb dosage used in the mTTI (modified TTI)

improved the outcome in the AE17 model to 100% (Figure 4.5). The reduced levels of

TGF-β in the AB1 tumour microenvironment probably also resulted in lower levels of

Treg accumulation within these tumours (see Chapter 6). This may also be a factor

contributing to the 100% tumour clearance rate when using far less anti-TGF-βmAb in

the TTI.

It was initially thought that the efficacy of the TTI was dependent on the precise

timing of the TGF-β neutralisation injections, which consisted of 7 separate injections

given consecutively for 7 days. However, with the lack of TGF-β secretions by the

AB1 tumours themselves, the TIC treatment was devised where all three components

were administered directly into the AB1 MM tumours as a single injection. The TIC

was as effective as the TTI in the AB1 model with 88% of mice undergoing complete

tumour eradication (Figures 4.2 A and B). This suggested that timing based

administration of immunotherapy was not critical in the AB1 MM model and that, this

single intra-tumoural administration of the TIC was sufficient for overcoming immune

suppression and reactivating anti-tumour immunity within the tumour

microenvironment (Krishnan et al., 2014). It should be noted that the high effectiveness

Page 143: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

115

of TIC in the AB1 MM model resulted in no control studies. Specifically, it cannot be

excluded that a double immunotherapy cocktail might not be as effective as the triple

immunotherapy cocktail.

In order to develop the triple immunotherapy cocktail (TIC), anti-TGF-βmAb

dosage was increased to allow for the single intra-tumoural administration and their

sustained presence within the tumour microenvironment. The concentration of anti-

CTLA-4mAb for the TIC was reduced significantly (as it was modified from systemic

to local delivery) from the dosage used previously in the timed triple immunotherapy

(TTI) (Kissick et al., 2012). Changing the route of administration of anti-CTLA-4mAb

from i.p to i.t was to overcome the systemic toxicities that have been reported and

particularly associated with anti-CTLA-4mAb treatment in humans. Severe systemic

toxicities associated with the intravenous administrations that have been reported

include neutropenia, diarrhoea, rash, colitis and transaminitis in over 60% of the

melanoma patients in different clinical trials (Hodi et al., 2010; Graziani et al., 2012;

Madan et al., 2012). A single intra-tumoural administration of this mAb, either alone or

in combination with other immune modifiers as shown here, may reduce the risk of

these systemic side effects. The mice were routinely monitored for side effects to the

therapy including weight loss, lethargy, decreased appetite, bloating and diarrhoea. To

date, no ill-effects to either the TTI or TIC have been observed in the treated mice.

Intra-tumoural administration of a combined immunotherapy has also been recently

shown in a mouse brain tumour model (vom Berg et al., 2013). In this work, the serious

adverse events seen with systemic IL-12 delivery were avoided by intra-tumoural

delivery. Furthermore, when combined with systemic anti-CTLA-4mAb, the glioma

rejection was so efficient, that the rapid translation to human clinical trials was called

for. Clinical combined immunotherapies for cancer are, however, in their infancy. The

single published example at the time of writing (Wolchok et al., 2013) used fully

Page 144: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

116

humanised antibodies against PD-1 and CTLA-4. These antibodies have complementary

roles, in that, anti-PD-1mAb contributes to T cells avoiding programmed cell death and

anti-CTLA-4mAb blocks attenuation of effector T cell activity. Rapid and substantial

tumour regression was seen in advanced melanoma patients, and these responses were

deemed superior to either agent used in monotherapy.

Long-term clearance of AB1 tumours was also achieved in the mice that were

treated with either the TTI or TIC treatments. Cured mice were also protected from

further re-challenges with the original inoculum (Table 4.1). Notably, although these

cured mice were resistant to tumour relapse or re-challenge with the same tumour, they

were found to be susceptible to challenges with a different syngeneic tumour type, as

demonstrated with the growth of 4T1 breast carcinoma tumours (Table 4.2). This

indicates specificity of the immunological memory. Previous work by Twitty et al

(2011) reported that mice vaccinated with short-lived tumour-specific antigens elicited

an effective cross-protection against sarcoma tumour challenge in these mice (Twitty et

al., 2011). However, in this study, the TIC treatment could not induce effective cross-

protection against 4T1 tumour challenge in the AB1 cured mice. This absence of cross-

protection could be explained by the fact that mammary carcinomas arise from

glandular epithelial cells, whereas mesotheliomas arise from squamous epithelial cells

and the tumour associated antigens (TAA) could be distinct between the two tumour

types. This question of whether there are any common TAAs between the two groups

can be confirmed by detecting tumour antigen-specific antibodies in the TTI or TIC

cured mice sera by ELISA (see chapter 5, section 5.2.4.1).

Despite the depletion of Tregs by anti-CD25mAb, neutralization of existing TGF-β

by anti-TGF-βmAb and the boost to T cell activity by anti-CTLA-4mAb, the TTI

treatment only effectively eradicated tumours in 50% of the TGF-β secreting AE17

Page 145: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

117

mesothelioma model at the initial anti-TGF-βmAb concentration. Day 10 (3 days post

first round of TTI treatment) formed a threshold at which point the differentiation

between mice responding to the treatment and partial responders occurred (Figure 4.3

B). The recovery of these partial responders was not successful even with the additional

supplementation of top-up treatments at day 10 (Figure 4.4 A and B). This research

demonstrated that it was hard to recover the mice that did not respond to the treatment,

once the tumours grew past the threshold. A preliminary analysis was conducted on

mice bearing AE17 tumours, where the treatment was initiated at a tumour size of

25mm2 and at this size, only 20% survival was achieved (1/5 from one experiment)

(data not shown). This inability to reject larger tumour burden (tumour size of 25mm2)

further supports the idea that once the tumours grow beyond a threshold point, it is

harder to achieve tumour eradication. The inability to reject significant tumour burden is

supported by work undertaken by other groups, including pre-clinical trials on B16-

OVA melanomas lung metastases, CMS5 fibrosarcomas and AE17 mesotheliomas

model. Therapies and/or adoptive transfer studies conducted on the above mentioned

tumour models were found to be less effective against large tumour burdens

(Dobrzanski et al., 2000; Hanson et al., 2000; Jackaman et al., 2008).

This led to testing the efficacy of the TTI by doubling the dosage of anti-TGF-

βmAb for sustained presence within the tumour microenvironment. This would also

help avoid the second round of consecutive injections as translation of such a

complicated treatment protocol into the clinic will be difficult. Also, the prolonged

treatment could likewise lead to the production of neutralising antibodies, which could

make the treatment ineffective. One such example, for the production of neutralizing

antibodies against repeated treatments, making them less potent, was reported in the

phase I trial of IFN-γ gene transfer therapy for patients with mesothelioma and

metastatic pleural effusions(Sterman et al., 2010).

Page 146: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

118

Treatment of established AE17 tumour-bearing mice with the modified TTI (mTTI)

with double the anti-TGF-βmAb dosage proved to be highly effective, with 100% of the

mice undergoing complete tumour eradication. This data suggested that the increased

dosage of anti-TGF-βmAb in the mTTI treatment aided in sustained neutralisation of

TGF-β within the tumour microenvironment. This also suggests that the rejection of

MM tumours is anti-TGF-βmAb dose dependent, and ergo, complete tumour

eradication of established AE17 tumours was achieved.

Patients presenting to the clinic would be in various stages of the disease

progression with varying tumour mass. With the ability of TTI, TIC and mTTI to treat

9mm2 tumours raises questions such as a) what about larger tumours, b) the TGF-β

present within these larger tumours and c) ability of the injected material to diffuse

through a larger tumour? This leads to future experiments that could involve titration

studies of anti-TGF-βmAb in larger tumours, increase in treatment volume which could

aid in better diffusion of the treatment within the large tumours and perhaps even

multiple injections at different angles for the efficient delivery of the treatment intra-

tumourally.

Another option for the future testing of the triple immunotherapy include testing

their efficacy in a more clinically relevant model. There are multiple studies involving

sub-cutaneous injections of the murine mesothelioma tumour cells, including the work

performed in our laboratory (Jackaman et al., 2003; Needham et al., 2006; Jackaman

and Nelson, 2012; Kissick et al., 2012). This is advantageous because sub-cutaneous

growth allows the direct monitoring of tumour size and easier delivery of intra-tumoural

therapy treatments. However, this does not approximate the patterns of tumour growth

or sensitivity to the treatments of intra-thoracic tumours (Nakataki et al., 2006). Using a

mouse model that mimics natural anatomical location and a tumour microenvironment

Page 147: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

119

similar to the human disease would allow for better translation of the triple

immunotherapy for human clinical trials. All the experiments that were performed on

the s.c model of mesothelioma can also be conducted on the MM tumour cell lines that

are grown i.p (intra-peritoneally) in the same mice backgrounds. The i.p model is

regarded as being closer to the human pleural cavity area of mesotheliomas and several

such studies have been done on the i.p MM AE17 murine models previously (van

Bruggen et al., 2005; Andujar et al., 2007). Intra-peritoneal tumours produce ascites with

local growth and invasion without metastases, and are associated with cachexia reminiscent

of human MM disease progression, which can commonly occur in the peritoneum

(Bielefeldt-Ohmann et al., 1994). The i.p model of murine mesothelioma allows for the

systemic study of the efficacy of new therapies in a model with more nodular and diffused

tumours reminiscent of the clinically defined disease. One such i.p model that can be used is

the AB1-DsRed MM model. In this i.p model, the AB1 tumour cell line is transfected with

pDsRed2-1 (Clontech) which encodes for DsRed2. DsRed is a variant engineered for faster

maturation, lower non-specific aggregation and higher expression in mammalian cells. An

investigation of efficacy of the triple immunotherapy in this murine i.p model of

mesothelioma (a more clinically relevant model) could aid in the clinical translatability of

this preclinical research.

Chapter 3 detailed the role of T cells (both CD4+ and CD8+) in the tumour

eradication mediated by TTI treatment in the AE17 murine mesothelioma model. The

role of B cells in tumour eradication has not been previously investigated in our

laboratory. A small amount of literature suggests that B cells positively enhance cellular

immune responses by serving as APCs, which can lead to tumour-specific cytotoxic T

cell activation and aid in the subsequent production of antibodies that contribute

modestly to anti-tumour immunity (Manson, 1994; Crawford et al., 2006; DiLillo et al.,

2010b; Jackaman et al., 2010). Antibodies have been reported (in both animal tumour

Page 148: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

120

models and in cancer patients) to recognise tumour antigens present on the surface of

cells, as well as intracellular antigens (Canevari et al., 1996; Disis et al., 1997). More

specifically, previous findings by Jackaman et al (2010) suggested a role for B cells in a

partially successful CD40 based immunotherapy against the two MM models used in

the present study (AE17 and AB1 murine mesotheliomas) (Jackaman et al., 2010). The

next step as described in the subsequent chapter dealt with the analysis of tumour

specific IgG antibody responses investigated in both the AE17 and AB1 tumour models

and the importance of these IgG antibodies during tumour eradication. The involvement

of B cells in tumour eradication as previously reported by Jackaman et al (2010) was

also investigated in the AB1 tumour model.

Page 149: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

121

Chapter 5 Evidence of B cell involvement in triple immunotherapy

Page 150: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

122

Page 151: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

123

5.1 Introduction

The role of B cells in anti-tumour activity is not well understood. B lymphocytes are

effector cells of the humoral immunity, a subsystem of the acquired immune response.

The primary functions of B cells include performing the role of antigen presenting cells

(APCs), production of cytokines, differentiation into antibody-secreting plasma cells or

developing into memory B cells post-antigen interaction (Mauri and Bosma, 2012).

Analysis of existing literature has shown conflicting data on the role of B cells in

tumour immunity.

Several studies have shown that B cells hamper the anti-tumour immune response.

Qin et al (1998) showed that the immunisation with irradiated tumour cells in B cell

deficient mice (µMT) provided protection against spontaneous mammary

adenocarcinoma development. Moreover, B cell deficiency in these µMT mice was

found to enhance T cell (CD4+ and CD8+) mediated tumour immunity (Qin et al.,

1998). Similarly, µMT mice were shown to have enhanced resistance to thymoma and

colon carcinoma tumour challenges when compared to their wild-type counterparts.

Delayed growth of melanoma tumours was also observed in these µMT mice in the

same study (Shah et al., 2005). Regulatory B cells (Bregs) have also been described in

some studies; whose functions have been found to be similar to Tregs, wherein these cells

negatively regulated the immune system and hence promoted tumour growth (Yanaba et

al., 2008; DiLillo et al., 2010a; Olkhanud et al., 2011). Antibody production has also

been implicated in enhancing tumour development due to chronic inflammation

(Houghton et al., 2005; de Visser et al., 2006).

In contrast to the preceding paragraph, a positive significance of B cells in anti-

tumour immunity has also been reported. Impaired T cell activation with enhanced

melanoma tumour growth have been demonstrated in mice that were depleted of mature

Page 152: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

124

B cells (DiLillo et al., 2010b). Adoptive transfer studies involving transfer of activated

or primed B cells into mice have shown a) to enhance anti-tumour T cell activity, b)

slow tumour growth and also c) production of high titres of tumour specific IgG

antibodies (Ritchie et al., 2004; Li et al., 2011). Jackaman et al. (2010) had suggested a

role for B cells in a partially successful CD40 based immunotherapy against AE17 and

AB1 murine mesotheliomas. Specifically, they found high levels of tumour reactive

IgG and IgM antibodies in the sera during agonist anti-CD40 Ab treatment, with

significantly high serum IgG levels detected by the 6th dose using enzyme-linked

immunosorbent assays (ELISAs). In the same study, sera from C57Bl6/J mice that had

been cured of their AE17 tumours by anti-CD40 Ab treatment were collected; adjusted

to contain 25 µg of IgG, and then were transferred into naïve mice (i.v) and these mice

were challenged 24 hours later with AE17 cells. 2/5 of these recipient mice that

received the IgG antibodies from the cured mice had significantly retarded tumour

growth (Jackaman et al., 2010).

The work in this chapter was designed to further investigate the immune responses

involved in the complete tumour eradication achieved by the triple immunotherapy, but

especially focuses on the involvement of B cells. In light of the significantly high IgG

levels observed in the AE17 mesothelioma model treated with the agonist anti-CD40

Ab treatment (Jackaman et al., 2010), experiments were designed to systematically

examine whether the successful tumour eradication achieved with the triple

immunotherapy (TTI and TIC) elicited such high IgG levels in the mice cured of AE17

and AB1 tumours respectively.

The primary aim of the studies described in this chapter; was to determine whether

high IgG levels could be detected in the TTI and TIC cured mice and also to determine

Page 153: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

125

whether B cells played a positive role in the anti-tumour immunity in our successful

triple immunotherapy for the treatment of murine mesothelioma.

5.2 Result

5.2.1 Detection of tumour specific IgG antibodies in the serum of AE17 tumour-

bearing mice by ELISA

In order to measure IgG antibodies specific to tumours, an ELISA that used

tumour cell lysates as the antigenic target (Dols et al., 2003) was developed in our

laboratory (see Chapter 2, section 2.6). Initial experiments to detect IgG antibodies were

carried out against the AE17 murine mesothelioma tumour cell lysates. Plate controls

were run on each experiment to account for any non-specific binding of antibodies, and

these included a) no serum control, b) no tumour lysate control and c) no IgG-Biotin

antibody conjugate control. Sera collected from mice with AE17 end-point (100mm2)

tumours were pooled and initially used to determine if reactivity to the tumour lysates

could be detected. The OD absorbance noted in the duplicate wells that contained the

neat sera pooled from untreated mice were significantly higher (2.68 (±0.34)) when

compared to the plate controls, with the absorbance being 24 fold higher than the no

serum control (0.115 (±0.02)), 55 fold higher than no tumour control (0.04 (±0.007))

and 51 fold higher than the no biotin control (0.05 (±0.0007)), as shown in Figure 5.1.

Page 154: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

126

Figure 5. 1 Elevated AE17 cell lysate specific IgG antibodies detected in the sera of AE17 end-point tumour bearing untreated mice. Pooled sera (neat) were collected from untreated end-point (100mm2) tumour bearing C57BL/6J mice (n=9) and were incubated, in duplicate, on an ELISA plate coated with AE17 cell lysates along with the appropriate plate controls. Neat sera from untreated mice were used for the no biotin and no tumour plate controls. No serum was added to the wells used as no serum control. Tumour specific IgG was detected using a biotinylated anti-mouse IgG antibody, measuring optical density at 450 nm. OD absorbances are shown as means across the duplicates ± standard deviations from one experiment. Significant differences in IgG levels are indicated: ** p≤0.01 as shown. OD indicates optical density.

Page 155: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

127

5.2.2 Elevated levels of tumour specific IgG antibodies in the sera of AE17 tumour-

bearing mice cured by TTI

With the high IgG levels observed in the sera from untreated AE17 end-point

tumour bearing mice, the next step was to investigate whether high IgG levels could be

detected in the mice cured of AE17 tumours by the TTI treatment. During the

standardisation step of ELISA tests; when neat sera from TTI cured mice were used,

maximal OD readings of 3.5 were noted (data not shown). This resulted in the dilution

of sera by 20 times (in assay diluent) to be used for all consecutive assays across the

different C57BL/6J groups.

IgG reactivity in the pooled sera collected from mice cured of AE17 tumours

more than 2 months post treatment with TTI, gave 4.1 fold and 3 fold higher absorbance

readings compared to pooled sera from naïve C57BL/6J mice and end-point AE17

tumour bearing untreated mice respectively (Figure 5.2). Though the IgG reactivity in

the sera of untreated tumour bearing mice was significantly higher than the reactivity

observed in sera from naïve C57BL/6J mice (p<0.003, significance determined by un-

paired student t-test), the fold increase was only marginal (1.3 fold).

Page 156: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

128

Figure 5. 2 Elevated levels of tumour specific IgG observed in sera of TTI cured mice compared to untreated controls. Pooled sera (1:20 dilution) were collected from naïve C57BL/6J mice ( ), untreated endpoint (100mm2) tumour bearing mice ( ) and TTI cured mice (■) (>2 months post treatment). Pooled sera from each group were incubated, in duplicate, on an ELISA plate coated with AE17 cell lysates along with the appropriate plate controls (■). Tumour specific IgG was detected using a biotinylated anti-mouse IgG antibody, measuring optical density at 450 nm. Mean absorbance are shown as means ± standard deviations for 5-9 mice per group over 3 independent experiments. Significant differences in IgG absorbance when comparing indicated columns by t-test are: **p<0.01 and ***p<0.001 respectively. OD indicates optical density.

Page 157: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

129

5.2.2.1 Partial cross-reactivity of IgG antibodies in TTI cured mice to B16 melanoma

cell lysates and spleen cell lysates

In order to determine the antibody specificity in the TTI cured mice, pooled sera

from the different groups were tested against a) syngeneic B16 melanoma cell lysates

and b) non-syngeneic AB1 murine mesothelioma tumour lysates and c) lysed self-cells

(splenocytes).

In terms of reactivity against AE17 cell lysates, the reactivity of IgG antibodies in

the pooled sera from TTI long-term cured mice were 5 fold and 3.6 fold higher than

reactivity found in naïve and untreated tumour bearing mice respectively (p<0.00001

and p<0.0001 respectively, Figure 5.3). In the case of reactivity to syngeneic B16

melanoma tumour cell lysates, a similar pattern was observed; wherein the reactivity of

the IgG antibodies was higher in the TTI cured sera compared to naïve sera and

untreated tumour controls (3 fold and 2.7 fold, respectively). When comparing the fold

change in IgG reactivity in the TTI cure mice to AE17 and B16 cell lysates (tumours

that are syngeneic to C57BL/6J mice), the reactivity against the non-syngeneic AB1

was the lowest (p=0.0003 and p=0.01, respectively).

In order to determine whether this IgG reactivity in the TTI cured mice was

auto-reactive, the pooled sera from the different C57BL/6J groups were incubated

against lysed splenocytes (self-cells) taken from naïve mice. Relative to the reactivity

against lysed splenocytes observed in the naïve group, no significantly different reaction

was seen in the untreated tumour group (0.30). However, the reactivity of IgG

antibodies against splenocytes were 2.4 fold and 2.13 fold higher in the TTI cured mice

group relative to both naïve and untreated groups (p=0.0001 and p=0.0003

respectively). Overall, though reactivity of the IgG from TTI cured mice group was

Page 158: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

130

present against lysed B16 tumour cells and naïve splenocytes, reactivity nevertheless

remained much higher against the AE17 tumour cell lysates.

Page 159: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

131

Figure 5. 3 Partial cross-reactivity of IgG antibodies in the serum of TTI long-term cured mice to syngeneic B16 melanoma tumour cell lysates. Pooled sera (1:20 dilution) were collected from naïve C57BL/6J mice ( ), untreated endpoint (100mm2) tumour bearing mice ( ) and TTI cured mice ( ) (>2 months post treatment). Pooled sera from each group were incubated, in duplicate, on an ELISA plate coated with cell lysates of AE17, B16, AB1 and naïve lysed splenocytes. Tumour specific IgG was detected using a biotinylated anti-mouse IgG antibody, measuring optical density at 450 nm. Fold increases are shown as means ± standard deviations for 5-9 mice per group over 2 independent experiments. Significant differences in fold increases of IgG absorbance when comparing indicated columns by t-test are: *p<0.05, ** p≤0.01 and ***p<0.001 respectively; n.s-not significant. OD indicates optical density.

Page 160: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

132

5.2.3 Development of a living whole cell ELISA for detecting serum IgG levels

against AE17 tumour cells

The next aim was to determine whether IgG reactivity could be detected in the

different groups against whole live in vitro cultured AE17 tumour cells. Technical

difficulties of losing cells during the washing steps in the 96 well plates were overcome

by performing the assay in 1.5ml microcentrifuge tubes as detailed in chapter 2 section

2.6.4.

The preliminary ELISA test conducted against whole live in vitro cultured AE17

cells: showed 7.9 fold and 6 fold higher IgG reactivity in sera from TTI cured mice and

untreated mice relative to naïve mice sera (p<0.001 and p<0.001, respectively). When

comparing the relative difference in reactivity between the untreated and TTI long-term

cured mice, the IgG levels were only 1.3 fold higher in the TTI than the untreated

control group (p<0.004). Overall, though the IgG reactivity in TTI cured mice against

whole live AE17 was only marginally higher than untreated tumour controls; both

reacted very strongly to live AE17 cells.

Page 161: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

133

Figure 5. 4 Detection of increased reactivity of serum IgG in TTI cured mice to whole live AE17 cells. Pooled sera (1:20 dilution) were collected from naïve C57BL/6J mice ( ), untreated endpoint (100mm2) tumour bearing mice ( ) and TTI cured mice ( ) (>2 months post treatment). Pooled sera from each group were incubated with live in vitro cultured AE17 cells along with the appropriate assay controls (■). Tumour specific IgG was detected using a biotinylated anti-mouse IgG antibody, measuring optical density at 450 nm. Mean absorbances are shown as means ± standard deviations for 4-8 mice per group from one experiment. Significant differences in IgG absorbance when comparing indicated columns by t-test are: **p<0.01 and ***p<0.001 respectively. OD indicates optical density.

Page 162: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

134

5.2.3.1 Increased reactivity of serum IgG to whole AE17 live cells in TTI long-term

cured mice

Compared to the IgG reactivity in the pooled serum collected from naïve and

untreated end-point tumour bearing mice, the IgG reactivity against whole live in vitro

cultured AE17 cells were 7.6 fold and 1.3 fold higher in the TTI cured mice sera

(p=0.003 and p=0.04 respectively, Figure 5.5). A similar pattern of reactivity was

observed against AE17 tumour cell lysates with IgG reactivity being 3.4 fold and 1.2

fold higher in the TTI cured mice pooled sera compared to pooled sera from naïve and

untreated groups (p<0.01 and p=0.003 respectively).

Page 163: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

135

Figure 5. 5 Increased reactivity of serum IgG to whole AE17 live cells in TTI cured mice compared to AE17 cell lysates. Pooled sera (1:20 dilution) were collected from naïve C57BL/6J mice ( ), untreated endpoint (100mm2) tumour bearing mice ( ) and TTI cured mice ( ) (>2 months post treatment). Pooled sera from each group were incubated, in duplicate, on an ELISA plate coated with AE17 cell lysates or incubated with live in vitro cultured AE17 cells. Tumour specific IgG was detected using a biotinylated anti-mouse IgG antibody, measuring optical density at 450 nm. The vertical axis shows the OD fold increases in tumour specific IgG over naïve mice sera. Fold increases are shown as means ± standard deviations for 4-8 mice per group from one experiment. Significant differences in fold increases of IgG absorbance when comparing indicated columns by t-test are: *p<0.05 and ** p≤0.01 respectively; n.s-not significant. OD indicates optical density.

Page 164: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

136

5.2.4 Increased levels of AB1 tumour specific IgG in sera of mice 95 days post

treatment with the TTI or TIC

The previous section determined that a higher IgG reactivity was observed in

C57BL/6J mice cured of AE17 tumours with TTI and these IgG antibodies had greater

reactivity to whole live AE17 cells than the tumour cell lysates. The next aim was to

determine if a similar high IgG reactivity could be detected in the sera from BALB/c

mice treated with TTI or the triple immunotherapy cocktail (TIC).

Serum IgG levels reactive with AB1 tumour cell lysates and whole live in vitro

cultured AB1 cells were compared in TTI and TIC treated mice; relative to IgG levels in

the sera of both naïve and untreated AB1 tumour bearing mice. Preliminary ELISA tests

conducted with sera diluted 20 times from TTI cured mice showed maximal OD

readings of 3.5 (data not shown). This resulted in the dilution of pooled sera by 50 times

(in assay diluent) being used for all consecutive analyses across all different BALB/c

groups.

IgG reactivity in the pooled serum collected from mice cured of AB1 tumours 95

days post treatment with the TTI or TIC gave 8.7 fold and 3.6 fold higher absorbance

readings respectively against whole live in vitro cultured AB1 cells, compared to pooled

sera from end-point tumour bearing untreated mice (p<0.001 and p<0.0001 respectively,

Figure 5.6). A similar pattern of reactivity was observed against AB1 tumour cell

lysates, though the IgG reactivity was only marginally higher (̴ 1.3 fold) in the pooled

sera from TTI or TIC cured mice compared to pooled sera from untreated mice with

end-point tumours (p<0.001 and p<0.001 respectively, Figure 5.6). Interestingly,

looking at the sera IgG reactivity within the TTI and TIC cured mice themselves,

though their reactivity to AB1 cell lysates are not significantly different from each other

(p=0.66); against whole live AB1 cells, the BALB/c mice that were cured with TTI had

Page 165: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

137

significantly higher reactivity (1.7 fold) than the TIC cured mice (p<0.001) (Krishnan et

al., 2014).

Page 166: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

138

Figure 5. 6 Elevated levels of tumour specific IgG antibodies detected in combined immunotherapy (TTI and TIC) treated mice. Pooled sera (1:50 dilution) were collected from naïve BALB/c mice ( ), untreated endpoint (100mm2) tumour bearing mice ( ), TTI cured ( ) and TIC cured mice ( ) (95 days post treatment). Pooled sera from each group were incubated, in triplicate, on an ELISA plate coated with AB1 cell lysates or incubated with live in vitro cultured AB1 cells. Tumour specific IgG was detected using a biotinylated anti-mouse IgG antibody, measuring optical density at 450 nm. The vertical axis shows the OD fold increases in tumour specific IgG over naïve mice sera. Fold increases are shown as means ± standard deviations for 5-8 mice per group over 3 independent experiments. Significant differences in fold increases of IgG absorbance when comparing indicated columns by t-test are: ***p<0.001; n.s-not significant. OD indicates optical density.

Page 167: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

139

5.2.4.1 Partial cross-reactivity of IgG antibodies in TTI and TIC cured mice to

syngeneic 4T1breast carcinoma cells

In order to determine the tumour specificity of the IgG antibodies, the next

experiment was designed to test IgG reactivity to other live tumour cells. The tumour

types used were, live in vitro cultured 4T1 BALB/c syngeneic breast carcinoma, the

allogeneic (specific to C57BL/6J) AE17 murine mesothelioma and B16 melanoma cells.

Partial cross-reactivity in the sera of TTI and TIC cured mice were higher against

their respective syngeneic live 4T1 breast carcinoma tumour cells (Figure 5.7). The IgG

reactivity in the TTI and TIC cured mice were 5.5 fold and 4.4 fold higher against 4T1

live tumour cells, relative to the fold increase in IgG reactivity observed in the untreated

end-point tumour bearing mice serum (p<0.0001 and p=0.002 respectively).

The IgG reactivity in TTI cured mice were not significantly different from naïve, or

untreated mice sera when incubated against live AE17 tumour cells (p=0.53 and p=0.75

respectively). The TIC cured mice sera were not tested against live AE17 tumour cells,

so no data is available for their IgG reactivity. In the case of allogeneic B16 live

tumours; there is some cross-reactivity in IgG present in the TTI and TIC long-term

cured mice compared to untreated controls (4.2 fold and 2.7 fold higher), though the

fold increases are not as high as the reactivity to syngeneic 4T1 tumour cells.

Page 168: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

140

Figure 5. 7 Partial cross-reactivity high against live syngeneic 4T1 tumour cells in TTI and TIC cured mice. Pooled sera (1:50 dilution) were collected from naïve BALB/c mice ( ), untreated endpoint (100mm2) tumour bearing mice ( ), TTI cured ( ) and TIC cured mice ( ) (95 days post treatment). Pooled sera from each group were incubated, in triplicate with live in vitro cultured AB1, 4T1, AE17 and B16 tumour cells. Tumour specific IgG was detected using a biotinylated anti-mouse IgG antibody, measuring optical density at 450 nm. The vertical axis shows the OD fold increases in tumour specific IgG over naïve mice sera. Fold increases are shown as means ± standard deviations for 5-8 mice per group over 3 independent experiments. Significant differences in fold increases of IgG absorbance when comparing indicated columns by t-test are: *p<0.05, ** p≤0.01 and ***p<0.001 respectively; n.s-not significant. OD indicates optical density.

Page 169: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

141

5.2.4.2 IgG antibodies present in the TTI and TIC cured BALB/c mice are tumour

specific and not auto-reactive

Compared to the high specificity of the reactivity of IgG antibodies to live AB1

tumour cells in both the TTI and TIC treated mice, marginal reactivity was observed

against live splenocytes taken from a naïve BALB/c mouse. Interestingly, though the

reactivity in TTI cured mice sera was higher than sera from the untreated group (1.5

fold higher, p=0.03), the reactivity in TIC cured mice sera was lower than the untreated

end-point tumour bearing control sera (p=0.03). Overall, this experiment determined

that the treatment with TTI or TIC did not elevate auto-reactive antibodies in these long-

term cured mice.

Page 170: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

142

Figure 5. 8 Low levels of auto-reactive antibodies in TTI and TIC cured mice. Pooled sera (1:50 dilution) were collected from naïve BALB/c mice ( ), untreated endpoint (100mm2) tumour bearing mice ( ), TTI cured ( ) and TIC cured mice ( ) (95 days post treatment). Pooled sera from each group were incubated, in triplicate with live in vitro cultured AB1 cells and live splenocytes from a naïve mouse. Tumour specific IgG was detected using a biotinylated anti-mouse IgG antibody, measuring optical density at 450 nm. The vertical axis shows the OD fold increases in tumour specific IgG over naïve mice sera. Fold increases are shown as means ± standard deviations for 5-8 mice per group from one experiment. Significant differences in fold increases of IgG absorbance when comparing indicated columns by t-test are: *p<0.05, ** p≤0.01 and ***p<0.001 respectively; n.s-not significant. OD indicates optical density.

Page 171: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

143

5.2.5 Tumour specific antibodies are not elevated during tumour eradication

The time to complete clearance of established 9mm2 AB1 tumours was between 10-

12 days post treatment with the TIC (Chapter 4). Sera collected on the day of TIC

injection (Day 0) and at time-points immediately post treatment were analysed by whole

live cell ELISA to determine the relative levels of anti-tumour IgG during the period of

tumour clearance.

Relative to IgG levels on day 0 (against whole AB1 cells), no significant increases

in IgG levels were observed within the period of maximal tumour regression (days 2- 8

post TIC treatment, Figure 5.9). The absorbance readings were also constant at days 16

and 20 post treatment, when no palpable tumours remained. Relative to pre-treatment

IgG levels measured on day 0, a significantly higher absorbance reading (5 fold

increase, p<0.001) was found for pooled sera collected on day 95 post TIC treatment

(Day 95, Figure 5.9) (Krishnan et al., 2014).

Page 172: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

144

Figure 5. 9 No significant change in serum IgG levels in TIC treated mice up to 20 days post treatment. BALB/c mice were implanted s.c with 1 × 106 AB1 tumour cells and treated intra-tumourally with TIC at a tumour size of 9 mm² (day 0). Pooled sera (1:50 dilution) were collected before TIC treatment on day 0 and at time-points post TIC treatment (days 1, 2, 4, 8, 12, 16, 20 and 95). Pooled sera from each time-point were incubated, in triplicate, with live in vitro cultured AB1 cells. Tumour specific IgG was detected using a biotinylated anti-mouse IgG antibody, measuring optical density at 450 nm. The left vertical axis shows the tumour areas (■) at each time-point (n=14) measured over three independent experiments. The right vertical axis shows the fold increases (■) in tumour specific IgG over day 0 sera. Fold increases are shown as means ± standard deviations for each time-point. OD indicates optical density.

Page 173: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

145

5.2.5.1 Elevated B cell numbers during TIC induced tumour eradication

In the same time course experiments described above, the percentages of B220+

cells within the tumour draining lymph nodes (TDLNs) and the spleens of TIC treated

mice were determined.

Flow cytometric analysis showed some minor fluctuations in B cell percentages on

days 1 and 2. However, the percentage of B cells in both spleen and TDLNs was

greatest on day 4 for the TIC treated mice compared to untreated mice at the same time

points (Figures 5.10 A and B). The percentage of B cells in the spleen increased from

46.4% on day 1 to 72.4 % on day 4. The percentage of B cells in TDLNs increased from

30.1% on day 1 to 42.8% on day 4. Day 4 post TIC treatment is central to the period of

maximal tumour shrinkage. By day 8, when the tumours were barely palpable under the

skin, the B cell percentages had reduced from the day 4 high in both the spleen and

TDLNs, but were still higher than the day 0 values (Krishnan et al., 2014).

Page 174: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

146

Figure 5. 10 Changes in B cell percentage post TIC treatment. Spleen (A) and tumour-draining lymph nodes (TDLNs: pooled unilateral brachial, axillary and inguinal) (B) were removed from mice on days 1, 2, 4, 8 and 12 days after treatment with TIC (■) and the population of B cells (B220+) was determined by flow cytometry and compared with cell populations in untreated mice (●) at the same time-points. Baseline percentages of B cells before treatment initiation were determined from untreated 9mm2 tumour bearing mice (day 0). (n=6 for untreated mice only on day 0 and n=2 for untreated and TIC treated mice for all other time-points, from one experiment. The error bars represent standard deviations of the mean). Significant differences in B cell percentages between untreated controls and TIC treated mice at different time-points determined by t-test are indicated: *p<0.05 and ** p=0.01 respectively.

Page 175: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

147

5.2.5.2 B cells are vital to the efficacy of the combined triple immunotherapy

The efficacy of the TIC in B-cell knock-out (BKO) mice were used to

substantiate the suggested role of B cells in the anti-tumour response. BKO mice

(C.129S- Igh-6tm1Cgn/J) on the BALB/c background were injected s.c with AB1 cells

and treated with the TIC when tumours reached 9 mm2. Figure 5.11 shows that BKO

mice left untreated grew tumours faster (median survival 16 days) than their wild-type

counterparts (median survival 19 days, p=0.0001 significance determined by log-rank

test). More importantly, Figure 5.11 also shows a complete failure of the TIC to

eradicate the AB1 tumours in these B-cell knock-out mice. This contrasts strongly with

the 88% clearance (15/17 mice over three independent experiments) seen with B cell

positive BALB/c wild-type mice. Note that 19/19 untreated BALB/c wild-type mice

developed tumours to end-point (Figure 5.11). The BKO treated mice instead had only a

partial response to the treatment with a transient delay in tumour growth compared to

the BKO mice left untreated (median survival 16 days, p=0.38) (Krishnan et al., 2014).

Page 176: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

148

Page 177: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

149

Figure 5. 11 Successful tumour eradication with TIC requires B cells. B cell knock-out mice (BKO) and wild-type BALB/c mice were implanted s.c. with 1 × 106 AB1 tumour cells and TIC was administered intra-tumourally at a tumour size of 9 mm² (day 0). Mice were monitored daily for (A) survival and plotted as Kaplan-Meier survival curve with 100 mm2 tumour as endpoint and (B) tumour growth (mean ± standard deviation) was calculated with measurements made using microcalipers. (n=5 for BKO untreated (■) and n=5 for BKO TIC treated (◊). n=19 for wild-type BALB/c untreated (●) and n=17 for wild-type TIC treated controls (▼) have been included for reference).

Page 178: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

150

5.2.5.2.1 B cells are required for successful treatment of AB1 tumours by TIC

The previous section determined that in the absence of B cells in these BKO mice,

TIC was not successful in eradicating established AB1 tumours. Thus, the next

experiment was designed to assess the anti-tumour role of B cells at the time of intra-

tumoural treatment with TIC by re-constituting the B cells in these BKO mice. Long-

term cured wild-type (WT) BALB/c mice were re-challenged with the original 1 × 106

AB1 tumour cells, and their spleens were harvested 3 days later and made into single

cell suspensions as mentioned in chapter 2 section 2.5.2. These spleen cells were then

either (a) left unfractionated (from 3 re-challenged cured mice) or (b) positively selected

for B220+ B cells (from 2 re-challenged cured mice, purities of >90% were determined

by FACS) and were then transferred through the tail-veil into 9mm2 AB1 tumour-

bearing BKO mice simultaneous to TIC treatment given intra-tumourally.

Figure 5.12 shows that 25% (1/4, from one experiment) of BKO mice that received

the TIC treatment at the same time as positively selected B220+ B cells underwent

complete tumour clearance. This was a significant advantage over (a) the BKO mice

that received TIC plus unfractionated spleen cells (p<0.05) as well as (b) the BKO mice

left untreated (median survival 14 days, p=0.02). The BKO mice that received TIC plus

unfractionated spleen cells did not survive longer than either the untreated BKO

controls (median survival 16 days, p=0.11) or the WT BALB/c untreated control mice

(median survival 18 days, p=0.14) and all 100% of mice (4/4) grew AB1 tumours to

end-point.

Page 179: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

151

Figure 5. 12 Anti-tumour efficacy of TIC treatment is augmented by B cells. B cell knock-out mice (BKO) and wild-type BALB/c mice were implanted s.c. with 1 × 106 AB1 tumour cells and i.t TIC and i.v transfer of spleen cells were initiated at a tumour size of 9 mm² (day 0). Spleens were harvested from long-term cured mice (n=5) that were challenged with 1 × 106 AB1 tumour cells 3 days prior to day 0. The spleens from 3 mice were pooled and made into single cell suspensions and left unfractionated while the pooled splenocytes from the remaining 2 mice were sorted by FACS into B220+ B cells. On day 0, the BKO treatment groups along with TIC administered intra-tumourally, also received i.v transfers of either 1 × 107 unfractionated splenocytes (♦) or positively selected 1 × 106 B220+ B cells (▼). Tumour growth (mean ± standard deviation) was calculated with measurements made using microcalipers and the tumour growth in the treated mice were compared with AB1 tumours growing in untreated wild-type BALB/c mice (●) and BKO mice (■). (n=4 for the treated mice and n=2 for the untreated mice groups respectively).

Page 180: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

152

5.2.5.2.2 Overcoming the immunosuppressive tumour microenvironment is critical

to tumour clearance

The above data suggested that i.t TIC treatment was more effective post-B cell

reconstitution in BKO mice. However, this raised the question of what happens in WT

BALB/c mice that received only the primed immune cells from re-challenged cured

mice; in the absence of TIC treatment.

In the next experiment, WT BALB/c mice were inoculated s.c with AB1 tumour

cells and the tumours were allowed to grow to an established size of 9 mm2. Spleens

from long-term cured mice (re-challenged 3 days prior with 1 × 106 AB1 tumour cells)

were harvested and made into single cell suspensions, and positively selected for B220+

B cells or CD3+ T cells by FACS with purities > 98% for both T and B cells. These

positively selected immune cells were transferred through the tail vein into these 9mm2

AB1 tumour bearing BALB/c mice as either (a) B220+ B cells only, (b) CD3+ T cells

only or (c) a mixture of both B and T cells and the tumour growth in the respective

groups were monitored daily.

Figure 5.13 shows that irrespective of whether the naïve AB1 tumour bearing mice

received only B cells, T cells or both T and B cells, tumours grew in 100% of the mice

in all 3 groups (3/3, one experiment). Transfer of only B cells or mixture of T and B

cells into these tumour-bearing mice made no difference in delaying tumour growth, as

their median survival of 14 and 15 days were not significantly different to untreated

mice (median survival of 14 days, p=0.19 and p=0.45). Only the mice that received just

T cells survived longer than untreated controls (median survival 17 days, p=0.02).

Page 181: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

153

Figure 5. 13 Manipulation of tumour microenvironment is important for successful clearance of established tumours. Wild-type BALB/c mice were implanted s.c. with 1 × 106 AB1 tumour cells and antigen primed immune cells from re-challenged cured mice were transferred intra-venously at a tumour size of 9 mm² (day 0). Spleens were harvested from long-term cured mice (n=2) that were challenged with 1 × 106 AB1 tumour cells 3 days prior to day 0. The spleens were pooled and made into single cell suspensions and sorted by FACS into B220+ B cells and CD3+ T cells. These positively selected cells were then transferred either as (a) 1 × 106 B220+ B cells only (■) or (b) 1 × 106 CD3+ T cells only (▼) or as (c) mixture of 1 × 106 both T and B cells (♦). Tumour growth (mean ± standard deviation) was calculated with measurements made using microcalipers and the tumour growth in the treated mice were compared with AB1 tumours growing in untreated wild-type BALB/c mice (●). (n=3 for the both treated mice and untreated mice groups, from one experiment).

Page 182: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

154

5.3 Discussion

The work described in this chapter has demonstrated strong initial evidence that

rapid elimination of tumours caused by the TTI and the TIC treatments are B cell

dependent. There is some literature that indicated the role of B cells in positively

enhancing cellular immune responses by serving as antigen presenting cells (APCs).

The APC function of B cells subsequently led to the activation of tumour specific

cytotoxic T cells and production of antibodies (which may contribute modestly to anti-

tumour immunity) (Manson, 1994; Ritchie et al., 2004; Crawford et al., 2006; DiLillo et

al., 2010b; Jackaman et al., 2010; Li et al., 2011; Molnarfi et al., 2013).

More specifically, the interest in B cells was initiated with the publication of the

work conducted by Jackaman et al., 2010, wherein the involvement of B cells in the

same two- AE17 and AB1 murine mesothelioma (MM) cell lines was investigated using

intra-tumoural anti-CD40 Ab treatment. This paper was relevant at two levels: (a) the

treatment involved timed intra-tumoural administrations of their anti-CD40 Ab therapy

(similar to the TTI) and (b) tumour reactive IgG and IgM antibodies were determined in

the sera during agonist anti-CD40 Ab treatment, with significantly high serum IgG level

detected by the 6th dose (Jackaman et al., 2010).

Therefore, investigations of the IgG reactivity to AE17 tumour cell lysates in the

sera of C57BL/6J mice were initiated. The result from the first test using neat sera

showed that IgG reactivity against the AE17 tumour cell lysates could be detected in the

sera from untreated end-point tumour bearing mice (Figure 5.1). It was important to use

sera from untreated mice in the initial analysis, as these mice have had AE17 tumour

cells implanted in them sub-cutaneously, which grew to end-point. It was expected that

these mice would naturally have antibodies capable of recognising tumour antigens.

This initial experiment with neat sera was also crucial to show that the low absorbance

Page 183: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

155

reading noted in the plate controls account for the absence of cross-reactivity. Thereby,

the reactivity observed in the wells containing the neat pooled sera from untreated mice

was in fact, cross-reacting to the AE17 tumour cell lysates. It could be concluded from

this initial assay, that a) tumour reactive IgG antibodies were present in mice that have

been exposed to the AE17 tumour cells and also, b) reactivity to FCS (integral to the

RPMI media used for culturing the cells in vitro) is highly unlikely.

The following experiments showed that relative to these untreated mice, IgG

reactivity in the TTI long-term cured C57BL/6J mice were highest against AE17 cell

lysates (Figure 5.2). It is clear that the TTI treatment induces higher levels of circulating

IgG in these long-term (>2 months post TTI treatment) cured mice specific to the

antigens over-expressed by the MM tumours, with no adverse effects noted.

Numerous studies have identified common tumour antigens such as MUC1, CEA,

EMA and CSPG4 that are expressed on multiple human tumour types such as lung

cancer, breast cancer, mesothelioma, and melanoma among others (Pinkus and Kurtin,

1985; Pinto et al., 1986; Schumacher et al., 1993; Rivera et al., 2012). The work

conducted by Rivera et al., 2012 showed that Chondroitin Sulphate Proteoglycan 4

(CSPG4), a melanoma-associated antigen, was also found to be highly expressed in

malignant mesothelioma cells in humans (Rivera et al., 2012). This could explain the

partial cross-reactivity of the IgG present in the sera collected from the TTI cured mice

to the syngeneic B16 melanoma cell lysates (Figure 5.3). However, low reactivity to the

non-syngeneic AB1 (raised in BALB/c mice) cell lysates was noted. This raises the

possibility that perhaps tumour antigen recognition is mouse strain (on which the

tumours were raised against) specific, as the better reactivity to B16 tumour cell lysate

was due to AE17 and B16 both being raised against C57BL/6J mice. Moreover, the low

Page 184: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

156

reactivity to AB1cell lysates, despite it being another MM tumour model, could be due

to their background being the allogeneic BALB/c mice (Figure 5.3).

In the future, perhaps testing the C57BL/6J cured mice sera IgG specificity against

several different tumour cell lines could aid in further understanding the notion of cross-

reactivity. The syngeneic EO771 breast carcinoma cell line (in C57BL/6J mice) and the

non-syngeneic 4T1 breast carcinoma cell line (in BALB/c mice) would be ideal

candidates to cross-check IgG reactivity.

There is evidence in the literature that antibodies that recognise intracellular as well

as surface antigens have been reported in both animal cancer models and cancer patients

(Canevari et al., 1996; Disis et al., 1997; Dao et al., 2013; Grandjean et al., 2013; Wei

et al., 2013). The IgG reactivity in the sera from TTI cured mice was found to be

greatest against whole live AE17 tumour cells and lesser against AE17 cell lysates

(Figure 5.5). A similar reactivity was observed in the sera of BALB/c mice cured of

AB1 tumours by TTI and TIC treatments. Reactivity of the IgG antibodies in these

long-term (greater than 95 days post treatment) cured mice were greatest against whole

live AB1 tumour cells and lesser against AB1 tumour cell lysates (Figure 5.6 and

Krishnan et al, 2014) and very low against wild-type BALB/c whole live naïve

splenocytes (self-cells) suggesting tumour specificity (Figure 5.8). Similar to the cured

C57BL/6J mice, the IgG reactivity of the BALB/c mice cured mice was found to be

more cross-reactive against live syngeneic 4T1 breast carcinoma cells (that were also

raised in BALB/c mice) and very low against the other non-syngeneic AE17 MM cells

(Figure 5.7).

Antibody dependent cell-mediated cytotoxicity (ADCC) is an important mechanism

by which antibodies recognise tumour antigen receptors present on the target cells by

their Fab domains. The Fc portions of these antibodies bind to the corresponding Fc

Page 185: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

157

receptors present on immune cells such as monocytes, macrophages and natural killer T

(NKT) cells. This in turn leads to the activation and crosslinking of the Fc receptors and

ultimately, lysis of the target cells by the release of cytokines and formation of cytotoxic

granules that contain granzyme and perforin (Leibson, 1997; Lyubchenko et al., 2001).

ADCC has been reported as an important mechanism of action for monoclonal antibody

(mAb) mediated therapies for cancer, such as rituximab (anti-CD20mAb), cetuximab

(EGFR inhibitor) and trastuzumab (Her-2/neu inhibitor) (Carter et al., 1992; Reff et al.,

1994; Vermorken et al., 2008).

However, the time course experiments showed that the tumour specific IgG levels

remained stable and low during the tumour shrinkage post TIC treatment; compared to

the long-term cured mice (Krishnan et al., 2014). As elevated IgG levels were only

detected in long-term cured mice, it could mean that TIC mediated tumour eradication

was unlikely to involve IgG antibodies as low IgG levels were detected at this stage.

This suggested that perhaps tumour eradication in the AB1 BALB/c model by TIC is

not dependent on an ADCC mechanism as previously recorded for other tumour models

(Herlyn et al., 1980; Jasinska et al., 2003).

The high levels of IgG detected only in the long-term cured mice tended to indicate

that possibly an amplification of the adaptive immune response occurs in response to

endogenous adjuvants released during immunotherapy-induced tumour cell death.

Activation of dendritic cells by apoptotic cells and/or release of damage associated

molecular patterns have been previously shown to result in an increased tumour antigen

availability, which then leads to an increase in tumour antigen-specific antibodies (Shi

and Rock, 2002; Apetoh et al., 2007; Kono and Rock, 2008). It is also possible that high

levels of IgG antibodies in the cured mice played a more robust role in the prevention of

tumour relapse and are not actively involved in primary tumour eradication. Further

Page 186: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

158

studies to elucidate the exact function of antibodies, and their involvement in tumour

eradication could include immune-histochemical sectioning of receding tumours in

response to TIC from days 0 to day 12 for detection of IgG antibodies present in the

tumour microenvironment. Assessing the tumour microenvironment for NK cells,

macrophages and monocytes could also shed light on whether ADCC mediated tumour

lysis is prevalent in the triple mAb therapy. In the future, enzyme-linked immunospot

(ELISPOT) assays can also be used to study the secreted antibodies, but at a more

cellular level, as described previously (Sedgwick and Holt, 1983; Moody and Haynes,

2008).

Elevated numbers of B cells in the spleens and TDLNs of TIC treated mice at the

precise time of maximal tumour shrinkage (with low unchanged antibody levels)

suggested that B cells may be playing another role such as antigen presentation in

tumour immunity (Manson, 1994; Ritchie et al., 2004; Crawford et al., 2006; DiLillo et

al., 2010b; Jackaman et al., 2010; Li et al., 2011; Molnarfi et al., 2013). The minor

fluctuations in B cell percentages on days 1 and 2 may represent migration of B cells

but may also be within the sample size error range (Figure 5.10). Technical difficulties

of accurately determining B cell percentage in the rapidly shrinking tumours precluded

reliable, detailed analyses. Tumour-specific effector T cell percentages of both CD4+

and CD8+ remained unchanged in the spleens and TDLNs at day 4 of the treated mice

(data not shown) and this could be due to the de-repression of the existing effectors cells

being modulated by the TIC treatment. This observation, in conjunction with the

increase in B cells in both spleen and TDLNs of TIC treated mice on day 4, suggests

that B cells were rapidly stimulated by the agonist TIC treatment and perhaps B cells act

as antigen-presenting cells (APCs) and aid in optimal activation of CD4+ and CD8+

tumour immunity in the TIC induced tumour clearance. This anti-tumour activity

contributed by B cells, by playing the role of antigen presentation in tumour immunity

Page 187: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

159

is supported by the work conducted by other research groups (Manson, 1994; Crawford

et al., 2006; DiLillo et al., 2010b; Jackaman et al., 2012; Molnarfi et al., 2013). One

caveat on this interpretation is that the B220+ marker is not entirely specific to B cells.

Dendritic cells also can carry this marker and may be contributing to the day 4

elevations noted in the TDLNs and spleen. Additional work using extra lineage specific

antibodies is required to define the situation.

This notion of B cell involvement in tumour eradication was further supported by

the experiments in B cell knock-out mice. In the first experiment, it was noted that

untreated BKO mice grew tumours quicker than in BALB/c wild-type, and the TIC

failed to induce complete tumour clearance in any BKO mice treated, in comparison to

88% of BALB/c wild-type mice undergoing complete tumour eradication (Figure 5.11A

and B). This is supported by findings of impaired T cell activity in B cell deficient mice

reported with diminution in tumour specific effector T cell proliferation (Gordon et al.,

1982; Schultz et al., 1990; DiLillo et al., 2010b).

The preliminary data from the adoptive transfer experiment in the BKO mice was

also consistent with the hypothesised APC role of B cells for T cell-mediated tumour

eradication by TIC treatment (see Figure 5.12). The data from this experiment suggested

that i.t treatment with TIC was effective only in the presence of B cells (that was

adoptively transferred from cured mice) in the BKO mice, as BKO mice that received

only the TIC treatment alone, were not successful in eradicating AB1 tumours.

Though the simultaneous administration of TIC (i.t) and B220+ B cells (i.v) were

successful in tumour clearance in at least one (out of four) AB1 tumour-bearing BKO

mice, B220+ B cells from cured mice when administered in the absence of TIC- was not

sufficient to mount an effective anti-tumour immune response and clear established

AB1 tumours in WT mice (see Figure 5.13). In this adoptive transfer experiment, mice

Page 188: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

160

that received only T cells transferred through tail-vein from re-challenged mice showed

marginal improvement in survival compared to the other groups. The above mentioned

experiment showed that, though B cells involvement seemed to be critical in the TIC

mediated tumour clearance, in the absence of manipulation of the tumour micro-

environment, the tumours grew un-hindered.

Though the data from the adoptive transfer experiments mentioned above looks

promising; and supports the APC role of B cells, the factor that needs to be taken into

consideration is that, these were data from one experiment each. There is the possibility

of the adoptive transfers being sub-optimal as tail-vein transfers are difficult, and there

is always the chance that the transfers are not always 100% successful. One way to

confirm proper transfers would be to perhaps collect blood from the recipient mice one

hour post-transfer and look for the fluorescent tagged immune cell sub-sets. Also,

increasing the number of recipient mice could also aid with data validation. A repeat

examination of the adoptive transfer experiments would also help validate the results to

a greater extent. In the future, further differentiation of B cell sub-sets, such as memory

B cells (CD27+) and plasma cells (CD138+) and their adoptive transfers into tumour-

bearing mice could also help in determining which of the sub-sets are more important in

tumour eradication.

Although the exact mechanism of B cell involvement in a direct effector capacity is

not yet known; the work with the TIC in eradicating AB1 tumours; and the work on

BKO mice augment previous works that reported the requirement of B cells as APC for

T cell activation- in the context of tumour immunity. With the current mesothelioma

treatments being of low efficacy, the TIC looks particularly promising for clinical trials.

Also, the three components that constitute the triple therapy have been approved for

clinical use (as detailed in chapter 3).

Page 189: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

161

With more clinical trials at present focussing on combined therapy for treatment of

cancer in humans (Madan et al., 2012; Wolchok et al., 2013), we are on the right path

and testing the efficacy of this TIC treatment in other tumour models such as B16

melanoma, EO771 and 4T1 breast carcinoma; could perhaps facilitate their approval for

clinical trials. The next chapter details the preliminary work conducted on testing the

efficacy of the TIC against these non-mesothelioma tumour models.

Page 190: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

162

Page 191: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

163

Chapter 6 Preliminary investigations of the intra-tumoural triple

immunotherapy in other tumour models and its effects on distal tumours

Page 192: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

164

Page 193: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

165

6.1 Introduction

The effective triple immunotherapy (TIC) described in detail in chapter 4 and other

combination therapies examined in animal models; have resulted in stronger immune

responses against multiple tumour types (van Elsas et al., 1999; Uno et al., 2006;

Kissick et al., 2009; Curran et al., 2010; Jackaman and Nelson, 2012; vom Berg et al.,

2013). Clinical combined immunotherapies for cancer are, however, in their infancy.

The published example (Wolchok et al., 2013) used fully humanised antibodies against

PD-1 (Nivolumab) and CTLA-4 (Iipilimumab) for the treatment of advanced

melanoma. These antibodies have complementary roles, in that, anti-PD1mAb

contributes to T cells avoiding programmed cell death and anti-CTLA-4mAb blocks the

attenuation of effector T cell activity. Rapid and substantial tumour regression was seen

in 80% of the patients with advanced melanoma. Though adverse events related to the

systemic delivery of this treatment were observed, they were generally reversible and

this improvement in tumour reduction with the combined immunotherapy was greater

than when the treatments were administered as monotherapies (Wolchok et al., 2013).

More recently, different permutations of anti-CTLA-4mAb, anti-PD-L1mAb and

IDO inhibitor INCB23843 were tested in the murine B16-SIY melanoma model

(C57BL/6J mice). B16-SIY are engineered to express the antigen SIYRYYGL (SIY), a

green fluorescent fusion protein (GFP) (Blank et al., 2004). The B16-SIY is a useful

cell line, as it aids in monitoring T cell responses that are specific to the SIY antigen

(Kline et al., 2012). All three components that were used in the aforementioned study,

target the immunosuppressive mechanisms that operate within the tumour

microenvironment. In this murine study, the tumours were allowed to grow to an

established size and timed i.p injections of anti-CTLA-4mAb and anti-PD-L1mAb were

administered alternatively for 12 days and the IDO (indoleamine -2,3-dioxygenase)

inhibitor was given by oral gavage. For the combination of anti-CTLA-4/anti-PD-

Page 194: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

166

L1mAb, 55% of mice were shown to undergo complete tumour rejection and a lower

percentage of tumour clearance was observed with anti-CTLA-4mAb/IDOi (18%) and

anti-PD-L1mAb/IDOi (13%) and the triple combination. The doublet combinations

were also tested on the parent cell line B16-F10 tumours, but the authors mention that

though the tumour growth was significantly delayed, tumour clearance was never

achieved with the parent B16-F10 cell line (Spranger et al., 2014).

One of the main features of the TIC treatment is the depletion of Tregs by anti-

CD25mAb within the tumour microenvironment. Anti-TGF-βmAb is also partly

responsible for the depletion of Tregs, as is anti-CTLA-4mAb. From literature, it is

known that Treg associated immune suppression and maintenance of T cell anergy have

been previously determined in several murine and human tumours (Liyanage et al.,

2002; Curiel et al., 2004; Needham et al., 2006; Zhou et al., 2006; Zou, 2006; Hegmans

et al., 2010; Özdemir et al., 2014; Tang et al., 2014; Weiss et al., 2014). Most recently,

recruitment of Tregs and the mRNA expression of FoxP3 was found to be significantly

higher within the tumours than in the normal skin of patients suffering from oral and

cutaneous squamous cell carcinoma (SCC) (Schipmann et al., 2014). This led to the

hypothesis, that the newly developed TIC treatment that targets multiple mechanisms of

immune suppression would also be effective in eradicating other non-mesothelioma

murine tumours such as B16 melanoma, EO771 and 4T1 breast carcinomas.

The main aims of the experiments presented in this chapter was to a) firstly

determine the levels of Tregs within these alternate tumours to give an indication that

they may (at least theoretically) be sensitive to TIC and b) secondly, test the efficacy of

the TIC treatment i.t in the B16 melanoma, EO771 and 4T1 breast carcinoma tumour

types. Additionally, in a clinical setting, patients report to the clinic with varying

degrees of disease progression (from varied tumour sizes to wide-ranging levels of

Page 195: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

167

metastases). In order to replicate such conditions in the murine model, effectiveness of

the triple immunotherapy was also examined in BALB/c mice bearing large AB1

tumours (16-25mm2 tumour sizes). With metastases being another major factor to

decreasing survival time in patients, it was important to determine whether i.t treatment

of one tumour was sufficient to generate enough anti-tumour immune response to

eradicate distal tumours. Therefore, a final experiment to test the effectiveness of a

single treatment with TIC on a distal tumour was also undertaken.

6.2 Results

6.2.1 Detection of high levels of Tregs within the tumour microenvironment of non-

mesothelioma tumours by flow cytometry

The intra-tumoural (i.t) TIC treatment focuses directly (anti-CD25mAb) or

indirectly (anti-TGF-βmAb and anti-CLA-4mAb) on the importance of overcoming the

immune suppression maintained by Tregs. The presence of Tregs in these non-

mesothelioma tumours was, therefore, estimated prior to attempting TIC treatment.

The accumulation of Tregs within the growing tumours were analysed by flow

cytometry, and the tumours at the time of harvesting for the analysis were 26.5 (±6.75)

mm2 for AE17 tumours, 23 (±7.7) mm2 for B16 melanoma tumours and 15.7 (±5.23)

mm2 for EO771 breast carcinoma tumours in the C57BL/6J mice. For the tumours

examined in the BALB/c mice, the tumours sizes were 25.5 (±1.8) mm2 for AB1

mesothelioma and 22.4 (±7.9) mm2 for 4T1 breast carcinoma tumours respectively.

Figures 6.1A and B show that high levels of Tregs were detected in all the three

non-mesothelioma tumour models, with the highest percentage detected in the 4T1

breast tumours (52.86 (±5.73)), closely followed by the other breast model EO771 at

47.54 (±11.21) and B16 lastly with 36 (±6.72). However, when comparing the levels

within the tumours developed in C57BL/6J mice (Figure 6.1A), the Treg levels in B16

Page 196: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

168

and EO771 tumours were not significantly different from the Tregs percentage of total

CD4+ T cells examined within AE17 tumours (43.57 (±14.99), p=0.39 against B16 and

p=0.66 against EO771 tumours respectively, by post hoc student t-test). For the tumours

grown in BALB/c mice, the AB1 tumours had the lowest Treg percentage of total CD4+

T cells at 13.53 (±4.72) (Figure 6.1 B) but in contrast, the Treg percentage within 4T1

breast carcinoma tumours were ~4 fold higher than within AB1 tumours (p<0.0001).

Page 197: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

169

Figure 6. 1 Detection of Tregs within non-mesothelioma tumours grown in both C57BL/6J and BALB/c mice. C57BL/6J mice were inoculated s.c with either 1 × 107 AE17 mesothelioma cells (●), 5 × 105 B16 melanoma (■) or 5 × 105 EO771 breast carcinoma cells (▲). BALB/c mice were inoculated s.c with either 1 × 106 AB1 mesothelioma cells (●) or with 5 × 105 4T1 breast carcinoma cells (■). Tumour growth was monitored daily. Once the tumours sizes were between 16-25 mm2, the mice were humanely sacrificed and their tumours were harvested and the population of Treg cells was determined by flow cytometry. (n=4-5 for all tumour types, from one experiment). The errors bars represent standard deviations of the mean. Significant differences in fold increases of IgG absorbance when comparing indicated columns by t-test: ***p<0.001as shown; n.s-not significant.

Page 198: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

170

6.2.2 TIC treatment is not effective in eradicating B16 melanoma tumours in

C57BL/6J mice.

B16-F10 melanoma tumours are aggressive tumours with high metastatic abilities that

are syngeneic to the C57BL/6J mouse and are widely regarded as poorly immunogenic

in nature. B16 cells were originally derived from tumours that were chemically induced

in C57BL/6J mice, more than 60 years ago. A system of tissue culture and animal

transplantation was used to develop the B16-F10 cell line (Fidler, 1973). With the

detection of Tregs in the B16 melanoma tumours as shown in the previous section, the

next step was to investigate the efficacy of the TIC treatment in this tumour model.

B16 tumour cells were injected subcutaneously into C57BL/6J mice, and the TIC was

initiated when tumours reached 9mm2. When compared to the untreated mice (median

survival of 9.5 days), there was no significant difference in the tumour growth in the

TIC treated mice (median survival 10 days, p=0.96, significance determined by log-rank

test), as all 100% (9/9, from two independent experiments) grew tumours to end-point

size of 100mm2 (Figure 6.2).

Page 199: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

171

Figure 6. 2 Growth of melanoma tumours in C57BL/6J mice is unhindered by TIC treatment. C57BL/6J mice were inoculated s.c with 5 × 105 B16 cells and TIC treatment was initiated when the tumours reached 9mm2 (4 ± 1 days). TIC (■) consisted of anti-CD25mAb, anti-CTLA-4mAb and anti-TGF-βmAb given simultaneously at a dosage of 2 µg of all three components. Mice were monitored daily for tumour growth and tumour sizes were measured using microcalipers. Untreated mice (●) were used as tumour growth controls. (n=7 for untreated mice and n=9 for TIC; from two independent experiments). The data are shown as mean ± SD.

Page 200: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

172

6.2.2.1 Increased dosage of all three components of TIC has no effect on tumour

retardation.

Having established that the TIC treatment had no effect on melanoma growth,

let alone eradicating them entirely; in the next experiment, the dosage of all three

components of the TIC was doubled.

Mice were inoculated with B16 melanoma cells and the treatment with double

the dosage of all components of the TIC, called dTIC, was administered when the

tumours reached a size of 9mm2. Figures 6.3A and B show that despite the increased

dosage of all three cocktail components, the treatment had no effect on the melanoma

growth in the treated mice when compared to mice left untreated (p=0.78, significance

determined by log-rank test). Even 2 days post treatment with dTIC, the average

tumour size was 11.75 (±3.97) mm2 and this was found to be not significantly different

from the untreated controls with average tumour size of 11.83 (±3.68) mm2 (p=0.97

significance determined by un-paired student t-test). All dTIC treated mice reached end-

point tumour sizes of 100mm2 at the same time as the untreated tumour group (median

survival for both being 10 days).

Page 201: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

173

Figure 6. 3 Double the dosage of all three components in the TIC has no effect on melanoma growth in C57BL/6J mice. C57BL/6J mice were inoculated s.c with 5 × 105 B16 cells and dTIC treatment was initiated when the tumours reached 9mm2. dTIC (▲) consisted of anti-CD25mAb, anti-CTLA-4mAb and anti-TGF-βmAb given simultaneously at a dosage of 4 µg of all three components. Mice were monitored daily for (A) survival and plotted as Kaplan-Meier survival curve with 100 mm2 tumour as endpoint and (B) tumour growth (mean ± standard deviation) was calculated with measurements made using microcalipers. Untreated mice (●) were used as tumour growth controls. (n=3 for untreated mice and n=5 for dTIC; from one experiment).

Page 202: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

174

6.2.3 Transient period of EO771 breast carcinoma tumour growth retardation in

C57BL/6J mice treated with TIC.

Simultaneously, the efficacy of the TIC and dTIC were examined against EO771

breast carcinoma tumours that are also syngeneic to C57BL/6J mice. EO771 tumours

are adenocarcinoma of the mammary gland that are of spontaneous origin (Dunham and

Stewart, 1953). The tumour cells were inoculated s.c and treatment with either TIC or

dTIC was initiated when the tumours reached a size of 9mm2.

Figure 6.4 shows, compared to the tumours growing in the untreated control

group, a transient delay in tumour growth was observed in the TIC and dTIC treated

mice groups. Even by day 8, while the average tumour size of untreated controls was at

53 (±15.3) mm2, the tumour sizes were significantly smaller in the TIC treated with an

average of 22.5 (±6.4) mm2 and in the dTIC treated with a tumour size of 20.3 (±10.01)

respectively (p=0.01 and p=0.03 respectively, significance determined by un-paired

student t-test). The tumour sizes were not significantly different between the TIC and

dTIC treated groups (p=0.73). Despite the fact that the tumours grew slower in the

treatment groups, the time to end-point (median survival) was inconclusive, as the i.t

treatments lead to ulceration with exudation from within the tumours and the mice had

to be culled before end-point tumour sizes could be reached. 2/4 mice in the TIC group

and 2/3 mice in the dTIC group developed ulcers (a health concern under animal ethics

guidelines), and the mice had to be humanely culled.

Page 203: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

175

Figure 6. 4 Treatment with TIC or dTIC did not significantly improve survival of mice bearing EO771 tumours. C57BL/6J mice were inoculated s.c with 5 × 10

5 EO771 breast carcinoma cells and treatment with either TIC (■) or

dTIC (▲) was initiated when the tumours reached 9mm2 (5 ± 2 days). Mice were

monitored daily for tumour growth (mean ± standard deviation), calculated with measurements made using microcalipers. Untreated mice (●) were used as tumour growth controls. (n=3 for untreated and dTIC treated mice and n=4 for TIC treated groups, from one experiment). Significant differences in tumour growth between untreated controls and treated mice determined by t-test are indicated: *p<0.05 and ** p=0.01 respectively.

Page 204: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

176

6.2.4 Increased survival time in 4T1 tumour-bearing BALB/c mice treated with

either TIC or dTIC.

The efficacy of the TIC and the dTIC were then trialled in the BALB/c mice

bearing s.c 4T1 breast carcinoma tumours. The 4T1 breast carcinoma originates from

410.4 tumours that were in turn isolated from a spontaneously arising mammary tumour

of BALB/c mice (Dexter et al., 1978).

Figure 6.5 shows that TIC (median survival of 14 days) was as effective as dTIC

(median survival of 15 days, p=0.6073) in increasing the survival time of treated mice

compared to untreated controls (median survival of 11 days, p= 0.01 and p=0.0042,

respectively). Nevertheless, 100% of TIC (5/5) and dTIC (5/5) treated mice developed

tumours to end-point of 100 mm2. In the mice treated with dTIC, hollow depressions

were seen with some ulceration within the tumours, 6 ± 2 days post treatment. However,

these ulcerations were exudate free and by day 9, had healed completely as shown in

figure 6.6, leaving a centralised zone of tumour clearance, with raised periphery. The

tumours in the periphery eventually grew to end-point tumour sizes (Figure 6.6).

Overall, a significant improvement in survival with TIC or dTIC was only observed in

the 4T1 breast cancer cells syngeneic to BALB/c mice.

Page 205: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

177

Figure 6. 5 Improved survival with TIC or dTIC in BALB/c mice bearing 4T1 tumours, compared to mice left untreated. BALB/c mice were implanted sub-cutaneously with 5 × 105 4T1 breast carcinoma cells and treatment with either TIC (■) or dTIC (▲) was initiated when the tumours reached 9mm2 (4 ± 1 days). Mice were monitored daily and the pooled data from two experiments were plotted as Kaplan-Meier survival curve with 100 mm2 tumour as endpoint. Untreated mice (●) were used as tumour growth controls. (n=7 for untreated and n=5 for TIC and dTIC treatment groups).

Page 206: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

178

Figure 6. 6 Central zone of tumour clearance observed in mice post treatment with dTIC. BALB/c mice treated with the dTIC when the 4T1 tumours reached a size of 9mm2 were monitored daily and photographs were taken on day 6 (A), day 9 (B) and day 13 (C) post treatment initiation. The ulceration and the eventual development of central zone of hollow tumour clearance shown in the pictures were from the same mouse, and representative of all mice in the dTIC treatment group.

A B C

Page 207: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

179

6.2.5 Efficacy of the triple immunotherapy is diminished with an increase in tumour

burden.

Efficacy of immunotherapy against varying tumour burdens is clinically

relevant; as patients report to the clinic at varied stages of disease progression. The

effectiveness of the triple immunotherapy against a larger tumour size was, therefore,

trialled.

In this experiment, AB1 tumour cells inoculated sub-cutaneously were allowed

to grow to a size of 25mm2 before the treatment with TTI was started. Figure 6.7 shows

that compared to the untreated controls (median survival of 14 days), the mice with

large tumours, treated with the TTI had a significant improvement in their survival

(median survival 24 days, p=0.006). However, only 20% of mice (1/5, from one

experiment) receiving the TTI completely cleared the large AB1 tumours. This was a

significantly diminished efficacy in tumour eradication from the 100% achieved when

the TTI treatment was initiated when the tumours were 9mm2 (p<0.001).

Page 208: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

180

Figure 6. 7 Tumour eradication efficacy of TTI treatment lowered with increase in tumour burden in AB1 tumour bearing BALB/c mice. Mice were implanted s.c. with 1 × 106 AB1 tumour cells. Treatment with TTI (■) was initiated at a tumour size of 25 mm² (17 ± 3 days) with tumour growth compared to untreated mice (●). Mice were monitored daily and survival was plotted as Kaplan-Meier survival curve with 100 mm2 tumour as endpoint (n=3 for untreated mice and n=5 for TTI treated mice, from one experiment).

Page 209: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

181

6.2.5.1 TIC treatment was effective in generating partial concomitant immunity– a

preliminary study on the effect of TIC on distal tumours.

Mesothelioma is a metastatic tumour, and whilst they are slow growing and have

a latency period of 30-40 years, secondary metastases are always a feature of the

disease. In order to completely eradicate mesothelioma, an ideal immunotherapy would

be able to cause complete regression in not only the primary tumour that is directly

treated, but also initiate immune activation, to result in secondary tumour regression at

distal sites.

In this experiment, BALB/c mice were simultaneously co-challenged sub-

cutaneously with AB1 cells on both flanks. Tumour growths on both right and left

flanks were monitored daily, and the TIC treatment into the primary tumour (growing

on the right flank) was initiated when these tumours reached a size of 9mm2. The

growths of the primary and secondary tumours are split into two separate tumour

growth graphs (Figures 6.8A and 6.8B respectively) for better understanding of the

growth kinetics in these mice that were challenged on both flanks.

Figure 6.8A shows the tumour growth of the TIC treated primary tumours in

comparison to untreated controls. Growth of the primary tumours post TIC (the red line

in the graph) were split into cured and non-responders as only 40% of the mice (2/5,

from one experiment) that received the TIC treatment underwent complete tumour

regression of their primary tumours. The other 60% of the mice (3/5) did not respond to

the treatment (non-responders) and grew end-point tumours of 100 mm2. By day 18, the

average sizes of these primary tumours in the mice that did not respond to the TIC

treatment were 70.66 (±25.40) and were not significantly different to the untreated mice

that had an average end-point tumour size of 100.33 (±16.74) mm2 (p=0.16).

Page 210: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

182

Figure 6.8B shows the tumour growth of only the untreated secondary tumours

(on the contralateral flank) in the mice that were cured of the primary tumours and the

mice that did not respond to the TIC treatment. When it came to these secondary

tumours, all 100% of the tumours developed. However, the tumour growth of the

secondary tumours in the mice that were cured of their primary tumours (2/5) was

significantly retarded when compared to the sizes of secondary tumours in the mice that

did not respond to the TIC treatment. By day 20, the secondary tumour sizes of the TIC

non-responders were 83.08 (±13) mm2, while the secondary tumours in the TIC cured

mice were 32.5 (±10.60) mm2 (p=0.02). This shows that there is some partial

concomitant immunity generated in the mice that were cured of their primary tumours,

as the secondary tumours in these mice grew significantly slower. However, the

immunity was not sufficient to completely eradicate tumours.

Overall, though TIC treatment mediated tumour eradication was only 40% (2/5)

efficient in the mice with distal tumours, their secondary tumours continued to grow

progressively in the same mice, but at a much lower rate compared to the 60% that did

not respond to the treatment at all.

Page 211: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

183

Figure 6. 8 TIC treatment of single primary tumour in mice co-challenged simultaneously with AB1 tumours was effective in generating partial concomitant immunity to secondary tumours. BALB/c mice were implanted sub-cutaneously with 1 × 106 AB1 mesothelioma on both right and their left flanks and treatment with TIC was initiated when the tumours on the right flank reached 9mm2. Mice were monitored daily and tumour growths on (A) right flank and (B) left flank were calculated with measurements made using microcalipers. n=2 for the TIC mediated cured mice (♦) and n=3 for TIC non-responders (■). n=3 for untreated mice (●) that were only implanted with AB1 cells on their right flanks. Data are shown as mean ± standard deviation, from one experiment.

Page 212: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

184

6.3 Discussion

Despite the high level of Tregs detected within the non-mesothelioma tumours, the

TIC treatment was ineffective, as 100% of the mice that were treated with the TIC grew

end-point tumours; thereby rejecting the hypothesis. The primary work described in this

chapter focussed on testing the efficacy of the triple immunotherapy cocktail (TIC) in

different non-mesothelioma tumours that were syngeneic to both C57BL/6J and

BALB/c mice. All three components which constitute the triple immunotherapy directly

or indirectly involved removal of immune suppression within the tumour

microenvironment mediated by Tregs. In this study, the intention was to examine Treg

levels at the time of TIC treatment initiation (which was an established size of 9mm2).

However, the technical difficulties of losing cells during the preparation of these small

tumours for flow cytometry required the use of larger tumours. From the previous work

performed on AE17 tumours at various sizes by Needham et al, 2006, it is known that

percentage of Tregs accumulating within tumours do no significantly increase between

9mm2-30mm2 and it is only in tumours greater than 30mm2 that a difference is observed

(Needham et al., 2006). In the light of this historic data on Treg accumulation in AE17

tumours, these non-mesothelioma tumours were inoculated sub-cutaneously into their

respective mice strains and allowed to grow to sizes between 16 - 25mm2.

The role of Tregs in immune responses to growing tumours has been documented in

several studies that looked into depletion/inhibition of these cells by anti-CD25mAb;

which subsequently led to increased Teff activation and a potent antitumour immune

response. (Onizuka et al., 1999; Shimizu et al., 1999). The success of Treg depletion on

tumour eradication has been varied towards different tumour types raised against

different murine strain backgrounds (Ostrand-Rosenberg et al., 2002; Chaput et al.,

2007; Bergot et al., 2010; Kline et al., 2012); including 80-100% in the AB1 and 50-

Page 213: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

185

100% in the AE17 mesothelioma studies; detailed in this thesis (and published in

Kissick et al., 2012 and Krishnan et al., 2014).

However, in the experiments conducted on the non-mesothelioma tumour models,

despite the Treg populations being determined within these tumours, the TIC and dTIC

treatments did not predict the effectiveness in eradicating established tumours. Testing

the TIC at 4µg of all three components (dTIC) was based on the Treg contents

determined within these non-mesothelioma tumours. With the dTIC treatment,

improved survival of the tumour-bearing mice was found in the 4T1 breast model and a

transient delay in tumour growth with the EO771 breast model. In the low immunogenic

B16 melanoma, the dTIC treatment made no impact on tumour growth when compared

to the normal growth kinetics observed in the untreated control group (see Figures 6.3A

and B). In future, experiments testing the efficacy of the TIC at a much larger range of

concentrations (titration studies) can be carried out. These future studies can include (a)

dosage titration of anti-CD25mAb alone in the TIC, to better deplete all Tregs, or (b)

different dosages of all three components can be tested and/or (c) existence of other

immunosuppressive mechanisms operating within these tumours also need to be taken

into consideration for formulating future treatments. With such titration studies, there is

the possibility of spillage into the systemic immunomodulation at higher concentrations.

So these factors need to be taken into account, and the mice need to be monitored for

systemic auto-immune symptoms such as weight loss, lethargy, reduced appetite,

bloating and diarrhoea.

From the previous experiments detailed in chapter 4, it was known that TIC

treatment was effective in eradicating AB1 tumours-where less than 15% of the CD4+ T

cells present within the tumours were Tregs (Figure 6.1B). However, in the case of the

B16 melanoma tumours, the average percentage of CD4+ T cells that are FoxP3+ was

Page 214: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

186

~2.7 fold higher (40%) than the levels present in the AB1. Despite the high Treg content

within B16 tumours, targeting immune suppression by Tregs is clearly not enough in the

B16 melanoma model. The role of Tregs as the primary regulators of immune

suppression has been elucidated in previous studies carried out on B16 tumours (Turk et

al., 2004). Perhaps increasing the dosage of anti-CD25mAb (that depletes Tregs) alone in

the TIC could in fact, overcome the immune suppression and eradicate the B16 tumours

in tumour-bearing mice. So in the future, titration studies involving anti-CD25mAb in

the TIC could potentially enable the eradication of B16 tumours.

In the case of the EO771 breast carcinoma tumours, a transient period of growth

retardation was observed in both TIC and dTIC treatment groups (Figure 6.4). However,

2/4 in the TIC and 2/3 in the dTIC treatment groups had to be humanely culled due to

their development of ulceration with exudates at the tumour site and thereby, calculation

of median survival for the treatment groups was skewered (inconclusive). Also, it has to

be mentioned that the data presented was from only one experiment with small

treatment groups. It would have been beneficial to repeat the experiment with bigger

TIC and dTIC treatment groups. This would have also assisted in determining whether

the ulceration at the tumour site was (a) a rare incidence, or (b) if the intra-tumoural

delivery of all three components was highly effective in abrogating the immune

suppression, leading to increased tumour necrosis, which in turn led to ulceration at the

tumour site. In the case of the other breast cancer model, the 4T1 in BALB/c mice,

compared to untreated mice group, a significant increase in overall survival when

treated with TIC or the dTIC was observed.

Ulcerations were also observed in these 4T1 tumour bearing mice treated with the

dTIC (similar to EO771). However, in this case, the ulcerations were observed in all 5

mice that received the dTIC treatment (Figure 6.6A). Nevertheless, these ulcerations

Page 215: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

187

healed quickly wherein the centre had collapsed onto itself and presented with a zone of

clearing in the centre of the tumours, with no exudates; leaving the impression of a ring

with hollowed out centre (Figure 6.6B). This eventually led to a central zone of

clearance with an raised edge, as the tumour cells in the periphery kept growing, and

these tumours with the hollowed out centre reached end-point sizes of 100mm2 (Figure

6.6C).

Treatment associated ulceration is not necessarily a negative response and one such

study that supports this concept was the adjuvant interferon (IFN) therapy used for the

treatment of melanoma by McMasters et al (2010). In their study, ulceration was found

to be a predictive marker for response in the stage III melanoma patients to IFN therapy

(McMasters et al., 2010). The authors also mentioned that perhaps the ulceration is

related to a greater inclination to vascular invasion of the tumours. A similar conclusion

of ulceration being a factor of good response to adoptive immunotherapy of tumour

infiltrating lymphocytes (TIL), was also reached with the treatment of melanoma stage

III patients (Peuvrel et al., 2011).

This active role of ulceration could be true in the ring-shaped tumour growth found

in the 4T1-dTIC treatment experiment. It is important to note that 4T1 breast carcinoma

tumours were found to have the highest Treg percentage (Figure 6.1B) amongst the non-

mesothelioma tumour models. This ring-shaped growth could mean that the single i.t

administration did have a response in these 4T1 tumours, with this zone of clearance

observed in the centre of the solid tumour. In the future, time course experiments,

wherein the analysis by flow cytometry or immunohistological sectioning of tumours at

various time-points immediately post treatment with TIC; would help in understanding

whether ulceration is beneficial to tumour eradication.

Page 216: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

188

However, the central zone of clearance observed post dTIC treatment, also raises the

question that perhaps the volume (30µl) injected into the solid tumours was not

sufficient to permeate the treatment throughout the solid tumour, with the corresponding

inaccessibility of Teff cell infiltration, and this eventually leading to the ring-shaped

growth of the 4T1 tumours. In the future, varied volumes could be investigated, wherein

one treatment group receives 30µl as used in the earlier experiment, but the other

receiving 50µl (the volume that was originally used for i.t injections in the TTI) of the

dTIC intra-tumourally. This will be based on the hypothesis that increased volume of

the treatment would correspond with greater diffusion within the solid tumours, leading

to improved survival.

Results detailed in chapter 4 showed that a 100% and close to a 90% cure rate

were obtained with TTI and TIC treatment respectively in the AB1 BALB/c model.

However, this efficacy was achieved when the treatment was initiated at a tumour size

of 9mm2. Greenberg (1991) suggested that the effectiveness of any immunotherapy

could be hindered qualitatively and/or quantitatively by significant tumour cell burden

(Greenberg, 1991). The TTI treatment injected into 25mm2 AB1 tumours showed that

the tumour eradication was diminished to 20% (1/5 mice undergoing complete tumour

clearance) from the 100% against 9mm2 AB1 tumours. This is clear evidence that

bigger the tumour burden (125mm3, in terms of volume), lower the effectiveness of the

treatment (Figure 6.7). This is also supported by the work conducted by other groups on

different tumours models, wherein therapies and/or adoptive transfer studies using

effector T cells were not effective against large tumour burdens (Dobrzanski et al.,

2000; Hanson et al., 2000; Jackaman et al., 2008).

At the time of increase in tumour burden, several factors have been proposed to

account for the lowered effectiveness of the treatment. They are a) inaccessibility of

Page 217: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

189

tumour tissue as the tumour grows; b) decrease in antigen specific T cell priming and c)

induction of antigen specific tolerance (Hanson et al., 2000).

With regards to inaccessibility to tumour tissue; as the tumour increases in size, the

complexity of the tissue microenvironment also increases correspondingly. The tissue

microenvironment of a developing tumour consists of extracellular matrix (ECM)

proteins, fibroblasts, signalling molecules such as cytokines/chemokines, endothelial

cells and immune cells. Immune cells infiltrating the growing tumour, together with the

crosstalk with the surrounding tissue, fibroblasts and the extracellular matrix from the

scaffold, ultimately contributes to tumour growth expansion (Whiteside, 2008; Levental

et al., 2009). This could also help explain the need for greater volumes to be tested, for

greater diffusion of the treatments within the solid tumours.

In certain cases, a decrease in T cell priming is also known to occur, where tumour-

specific T cells are activated, however undergo anergy (unresponsiveness) and are

unable to affect tumour growth (Staveley-O’Carroll et al., 1998; Shrikant and Mescher,

1999). Immune tolerance to growing tumours are mediated by regulatory cells such as

Tregs, myeloid-derived suppressor cells (MDSC) and Th17 cells, and also by

cytokines/chemokines such as TGF-β, IL-6 and IL-10 just to name a few (Shankaran et

al., 2001; Curiel et al., 2004; Willimsky and Blankenstein, 2005; Lin and Karin, 2007;

Rabinovich et al., 2007; Gabrilovich and Nagaraj, 2009).

In the AE17 murine mesothelioma model, complete tumour eradication was

achieved due to the timed neutralization (for 7 consecutive days) of excess TGF-β

secreted by the growing tumour with the timed triple immunotherapy (chapter 3 and

Kissick et al, 2012). This 50% was improved to a 100% with the increased dosage of

anti-TGF-βmAb alone in the modified TTI, detailed in chapter 4. In the non TGF-β

secreting AB1 mesothelioma model, the TIC treatment formulation was developed

Page 218: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

190

where a single treatment was sufficient to eradicate as effectively as the TTI (Krishnan

et al., 2014). With the focus now shifting more towards the neutralization of TGF-β in

the tumour microenvironment being pertinent to tumour eradication, perhaps the first

step for future studies on tumour burden should involve analysing TGF-β concentrations

as the tumours grow (by ELISA), and this could perhaps point towards dose titration

studies involving anti-TGF-βmAb and keep the other two components constant in the

TIC treatment protocol.

Removal of the immune suppression within the tumour microenvironment by i.t

treatments with avoidance of systemic toxicities have also been the focus of this thesis

so far. However, with the lowered effectiveness in the treatment of larger tumour

burdens, it is clear that the tumour burden is also an important factor that affects

efficacy of immunotherapy. This importance of both tumour burden and overcoming

intra-tumoural immune suppression were confirmed in the experiment where mice were

challenged on both flanks with AB1 cells and only one of the 9 mm2 (27 mm3 in terms

of volume) established tumours were treated. Intra-tumoural TIC treatment into the

primary tumours only aided in clearing these tumours in 40% (2/5) of the treated mice

(as opposed to the 5/5 cures usually noted) and no cures with the non-injected distal

tumour were observed, although this second tumour did grow significantly more slowly

(Figure 6.8B). This delayed growth of an already established secondary tumour could

be due to the partial activity of concomitant immunity. Concomitant immunity is a

process in which tumour-bearing hosts develop resistance to growth of secondary

inoculations of the same tumour. Tumour metastases (dissemination) are considered as

secondary tumours that develop concurrent to primary tumour growth and concomitant

immunity is considered as a means to control the development of these metastases

(Ruggiero et al., 1988).

Page 219: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

191

The importance of eliciting concomitant immunity to combat secondary tumours

is further supported by a recent study conducted by Korrer and Routes (2014). In this

study, mice inoculated with MCA-205 fibrosarcomas transfected with ovalbumin

(OVA) on one flank, rejected a subsequent secondary inoculum of the MCA-205-OVA

tumours on the contralateral side in a OVA-specific T cell-mediated response (Korrer

and Routes, 2014). In another study, B-1 cells (subset of B lymphocytes) were

selectively obtained from BALB/c mice bearing Ehlrich tumours (derived from

mammary adenocarcinoma) developing in their footpad. These B-1 cells that were

adoptively transferred; were able to protect naïve BALB/c and BALB/c xid (B-1 cell

knockout) mice against primary tumour challenge with the same tumour type. This was

an example of concomitant tumour immunity being generated in a different mice post-

adoptive transfer of tumour primed B-1 cells (Azevedo et al., 2014).

Such examples are relevant to understanding the importance of concomitant

immunity. However, the preliminary distal tumour experiment detailed in this thesis is

significantly different, as the mice were co-challenged simultaneously and were not

challenged a few days apart. The tumour burden in this experiment, in toto, was 54 mm3

(2 × 27 mm3 tumours, based on volume). The tentative impression is that having two

tumours on a single mouse may be the same as a single 54 mm3 tumour, making the

overall treatment less effective. This analysis showed that the successful tumour

eradication and the corresponding anti-tumour immunity raised were not sufficient to

eradicate distal tumours due to an increase in tumour burden. Further investigation is

required, where mice with tumour on both flanks would be used as untreated controls

and the treatment groups that can be tested are:

a) dTIC treatment into only the right flank,

b) Target only right flank with dTIC at a higher volume (50 µ1),

Page 220: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

192

c) Target both right and left flank tumours with TIC at 9mm2,

d) Increase only the dosage of anti-TGF-β (preliminary study before testing in the

non-mesothelioma models) and

e) Administer i.t TIC into one tumour in conjunction with i.v lysed tumour

antigens (in vitro cultured AB1 cell lysates) to boost antigenic exposure of the

immune system.

These follow-up experiments could help in understanding the correlation between

tumour burden and the tolerance maintained within these growing tumours.

Furthermore, the data from the above experiment in addition to the TGF-β

concentrations assessed within the growing tumours (both mesothelioma and the non-

mesothelioma) could aid in making TIC as effective against the non-mesothelioma

tumours, as it is against mesothelioma tumours.

6.4 Conclusion and future perspectives

Current mesothelioma treatments are of low efficacy and provide minimal

improvement in survival rates. Immunotherapy is of particular interest for cancer

treatment, as it aids in overcoming immune evasion mechanisms employed by the

growing tumours and boosts the host’s anti-tumour immune response. The combined

triple immunotherapy combination using three agonist antibodies (anti-CD25mAb, anti-

TGF-βmAb and anti-CTLA-4mAb) produced complete tumour clearance in both the

C57BL/6J and BALB/c murine mesothelioma tumour models. The immunity generated

in these cured mice protected these mice against further re-challenges. All the treatment

components that constitute the triple immunotherapy have human equivalents and are

approved for use or are undergoing clinical trials. There is clinical potential for the

triple immunotherapy treatment to be used in clinical trials for treating mesothelioma

patients.

Page 221: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

193

6.4.1 Future of the triple immunotherapy

Future investigation into the efficacy of the triple immunotherapy in more highly

relevant pre-clinical models such as intra-thoracic mesothelioma model and mammary

carcinomas grown in the mammary pads of mice for the breast cancer models are

warranted. However, there are many factors that could affect the delivery of the

treatments intra-tumourally in these tumour models. Perhaps strategies such as CT,

endoscopy or ultrasound guided deliveries could enable the precisions with which the

treatments are delivered intra-tumourally. With the setback of treating larger tumours

with the triple immunotherapy, perhaps combination of chemotherapy with

immunotherapy can also be tested in the near future. While chemotherapy induces

lymphopenia in recipients, there is much research that indicates that a number of

chemotherapeutic agents provide some benefit to the anti-tumour function of the

immune system. Several mechanisms have been proposed for how this occurs, including

increase in antigen availability due to tumour cell death, with activation of dendritic

cells by apoptotic cells or the release of damage associated molecular patterns (Apetoh

et al., 2007), lymphablation of suppressive cells (North, 1982; Ghiringhelli et al., 2007)

and increased immunogenicity of tumour cells (Galetto et al., 2003). With this in mind,

certain chemotherapy combinations have been used with immunotherapy to good effect,

in both human and animal models, including gemcitabine/cisplatin and 5-

fluorouracil/cisplatin (Tanaka et al., 2002). Another possibility is a combination of

debulking surgery followed by immunotherapy. Surgery to remove mesothelioma

tumours, followed by radio- and/or chemotherapy forms part of the current treatment for

MM in the clinic (Sugarbaker et al., 1999). Resection of the tumour to decrease tumour

load with the corresponding increase in tumour antigen exposure will aid in effective

anti-tumour immunity being generated following immunotherapy.

Page 222: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

194

6.4.1.1 Addition of more agonist antibodies to the TIC treatment to improve tumour

regression

In a recent article published in JEM, studies involving Treg depletion in mice

with chronic lymphocytic choriomeningitis virus (LCMV) showed that an up-regulation

of PD-L1 on T cells occurred, leading to inhibition of signals (Penaloza-MacMaster et

al., 2014). The authors also found that treatment combination of Treg depletion and PD-

L1 blockade resulted in a significant reduction in viral titres than when the treatments

were administered as monotherapies. Blockade of PD-1 by agonist antibody (BMS-

936558) have already been shown to have varying levels of objective responses in

patients with non-small-cell lung cancer, melanoma and renal-cell carcinomas (Topalian

et al., 2012).

In a more recent article, intra-tumoural treatment of 3mAb (CD137/CTLA-

4/PD-1) and 4mAb (CD137/PD-1/CTLA-4/CD19) combinations were tested by Dai et

al (2014) for treating sub-cutaneous murine models of B16 and SW1 melanoma and

TC1 lung carcinoma (Dai et al., 2014). The 4mAb combination treatment was found to

eradicate large tumours (~80mm2) in 50% of the treated mice. The treatment involved

injecting 0.25 mg of each of the monoclonal antibodies intra-tumourally into these large

tumours. Systemic toxicities associated with the aforementioned high dose intra-

tumoural treatment included hepatitis in liver, hair loss and depigmentation.

With the successful eradication of large tumours being achieved as described

above, perhaps the addition of other agonist antibodies such as anti-PD-1mAb could

further enhance the efficacy of the TIC. It is also important to note that the maximum

amount of the monoclonal antibodies that we have tested in our laboratory has been 4

µg. With mAb amounts that are 60 times more, being tested in other laboratories for

treating cancer, perhaps titrated dosages of 4mAb or even 5mAb combinations in the

Page 223: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

195

future could be most ideal to eradicate not just large mesothelioma tumours, but other

non-mesothelioma tumours as well.

The significance of this work will aid in the development of a treatment

approach that will improve the poor outcomes for patients diagnosed with different

aggressive and incurable malignant cancers.

Page 224: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

196

Page 225: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

197

Chapter 7 Bibliography

Page 226: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

198

Page 227: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

199

Abe, K., Kato, N., Miki, K., Nimura, S., Suzuki, M., Kiyota, H., Onodera, S. & Oishi,

Y. 2002. Malignant mesothelioma of testicular tunica vaginalis. International

Journal of Urology, 9(10), 602-603.

Aggarwal, B. B., Vijayalekshmi, R. & Sung, B. 2009. Targeting inflammatory pathways

for prevention and therapy of cancer: short-term friend, long-term foe. Clinical

Cancer Research, 15(2), 425-430.

Ahamad, A., Stevens, C. W., Smythe, W. R., Liao, Z., Vaporciyan, A. A., Rice, D.,

Walsh, G., Guerrero, T., Chang, J. & Bell, B. 2003a. Promising early local

control of malignant pleural mesothelioma following postoperative intensity

modulated radiotherapy (IMRT) to the chest. The Cancer Journal, 9(6), 476-

484.

Ahamad, A., Stevens, C. W., Smythe, W. R., Vaporciyan, A. A., Komaki, R., Kelly, J.

F., Liao, Z., Starkschall, G. & Forster, K. M. 2003b. Intensity-modulated

radiation therapy: a novel approach to the management of malignant pleural

mesothelioma. International Journal of Radiation Oncology* Biology* Physics,

55(3), 768-775.

Aisner, J. 1995. Current approach to malignant mesothelioma of the pleura. CHEST

Journal, 107(6_Supplement), 332S-344S.

Alberts, A. S., Falkson, G., Goedhals, L., Vorobiof, D. & Van der Merwe, C. 1988.

Malignant pleural mesothelioma: a disease unaffected by current therapeutic

maneuvers. Journal of Clinical Oncology, 6(3), 527-535.

Alexandroff, A. B., Jackson, A. M., O'Donnell, M. A. & James, K. 1999. BCG

immunotherapy of bladder cancer: 20 years on. The Lancet, 353(9165), 1689-

1694.

Algarra, I., Cabrera, T. & Garrido, F. 2000. The HLA crossroad in tumor immunology.

Human immunology, 61(1), 65-73.

Page 228: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

200

Altomare, D. A., Vaslet, C. A., Skele, K. L., De Rienzo, A., Devarajan, K., Jhanwar, S.

C., McClatchey, A. I., Kane, A. B. & Testa, J. R. 2005. A mouse model

recapitulating molecular features of human mesothelioma. Cancer Research,

65(18), 8090-8095.

Andujar, P., Lecomte, C., Renier, A., Fleury-Feith, J., Kheuang, L., Daubriac, J., Janin,

A. & Jaurand, M.-C. 2007. Clinico-pathological features and somatic gene

alterations in refractory ceramic fibre-induced murine mesothelioma reveal

mineral fibre-induced mesothelioma identities. Carcinogenesis, 28(7), 1599-

1605.

Anraku, M., Tagawa, T., Wu, L., Yun, Z., Keshavjee, S., Zhang, L., Johnston, M. R. &

de Perrot, M. 2010. Synergistic Antitumor Effects of Regulatory T Cell

Blockade Combined with Pemetrexed in Murine Malignant Mesothelioma. The

Journal of Immunology, 185(2), 956-966.

Apetoh, L., Ghiringhelli, F., Tesniere, A., Obeid, M., Ortiz, C., Criollo, A., Mignot, G.,

Maiuri, M. C., Ullrich, E., Saulnier, P., Yang, H., Amigorena, S., Ryffel, B.,

Barrat, F. J., Saftig, P., Levi, F., Lidereau, R., Nogues, C., Mira, J.-P.,

Chompret, A., Joulin, V., Clavel-Chapelon, F., Bourhis, J., Andre, F., Delaloge,

S., Tursz, T., Kroemer, G. & Zitvogel, L. 2007. Toll-like receptor 4-dependent

contribution of the immune system to anticancer chemotherapy and

radiotherapy. Nat Med, 13(9), 1050-1059.

Azevedo, M., Palos, M., Osugui, L., Laurindo, M., Masutani, D., Nonogaki, S., Bachi,

A., Melo, F. & Mariano, M. 2014. B-1 cells and concomitant immunity in

Ehrlich tumour progression. Immunobiology, 219(5), 357-366.

Baban, B., Hansen, A. M., Chandler, P. R., Manlapat, A., Bingaman, A., Kahler, D. J.,

Munn, D. H. & Mellor, A. L. 2005. A minor population of splenic dendritic cells

Page 229: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

201

expressing CD19 mediates IDO-dependent T cell suppression via type I IFN

signaling following B7 ligation. International Immunology, 17(7), 909-919.

Baker, P. M., Clement, P. B. & Young, R. H. 2005. Malignant Peritoneal Mesothelioma

in Women A Study of 75 Cases With Emphasis on Their Morphologic Spectrum

and Differential Diagnosis. American journal of clinical pathology, 123(5), 724-

737.

Banchereau, J. & Steinman, R. M. 1998. Dendritic cells and the control of immunity.

Nature, 392(6673), 245-252.

Barth, R. J., Bock, S. N., Mulé, J. J. & Rosenberg, S. A. 1990. Unique murine tumor-

associated antigens identified by tumor infiltrating lymphocytes. The Journal of

Immunology, 144(4), 1531-1537.

Baud, M., Bobbio, A., Lococo, F., Regnard, J.-F. & Alifano, M. 2014. Should We

Continue to Offer Extrapleural Pneumonectomy to Selected Mesothelioma

Patients? A Single Center Experience Comparing Surgical and Non-surgical

Management. Japanese Journal of Clinical Oncology, hyu134.

Baumgarth, N. & Roederer, M. 2000. A practical approach to multicolor flow cytometry

for immunophenotyping. Journal of immunological methods, 243(1), 77-97.

Becker, Y. 2006. Molecular Immunological Approaches to Biotherapy of Human

Cancers - A Review, Hypothesis and Implications. Anticancer research, 26(2A),

1113-1134.

Belcaid, Z., Phallen, J. A., Zeng, J., See, A. P., Mathios, D., Gottschalk, C., Nicholas,

S., Kellett, M., Ruzevick, J. & Jackson, C. 2014. Focal Radiation Therapy

Combined with 4-1BB Activation and CTLA-4 Blockade Yields Long-Term

Survival and a Protective Antigen-Specific Memory Response in a Murine

Glioma Model. PLoS ONE, 9(7), e101764.

Page 230: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

202

Bergot, A., Durgeau, A., Levacher, B., Colombo, B., Cohen, J. & Klatzmann, D. 2010.

Antigen quality determines the efficiency of antitumor immune responses

generated in the absence of regulatory T cells. Cancer gene therapy, 17(9), 645-

654.

Berry, G., de Klerk, N. H., Reid, A., Ambrosini, G. L., Fritschi, L., Olsen, N. J., Merler,

E. & Musk, A. W. 2004. Malignant pleural and peritoneal mesotheliomas in

former miners and millers of crocidolite at Wittenoom, Western Australia.

Occupational and Environmental Medicine, 61(4), e14.

Bianchi, C. & Bianchi, T. 2007. Malignant Mesothelioma: Global Incidence and

Relationship with Asbestos. Industrial Health, 45(3), 379-387.

Bielefeldt-Ohmann, H., Fitzpatrick, D., Marzo, A., Jarnicki, A., Himbeck, R., Davis,

M., Manning, L. & Robinson, B. 1994. Patho-and immunobiology of malignant

mesothelioma: characterisation of tumour infiltrating leucocytes and cytokine

production in a murine model. Cancer Immunology, Immunotherapy, 39(6), 347-

359.

Biswas, S., Nyman, J. S., Alvarez, J., Chakrabarti, A., Ayres, A., Sterling, J., Edwards,

J., Rana, T., Johnson, R. & Perrien, D. S. 2011. Anti-transforming growth factor

ss antibody treatment rescues bone loss and prevents breast cancer metastasis to

bone. PLoS ONE, 6(11), e27090.

Blank, C., Brown, I., Peterson, A. C., Spiotto, M., Iwai, Y., Honjo, T. & Gajewski, T. F.

2004. PD-L1/B7H-1 Inhibits the Effector Phase of Tumor Rejection by T Cell

Receptor (TCR) Transgenic CD8+ T Cells. Cancer Research, 64(3), 1140-1145.

Borkowski, T. A., Letterio, J. J., Farr, A. G. & Udey, M. C. 1996. A role for

endogenous transforming growth factor β1 in Langerhans cell biology: the skin

of transforming growth factor β1 null mice is devoid of epidermal Langerhans

cells. The Journal of Experimental Medicine, 184(6), 2417-2422.

Page 231: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

203

Boutin, C., Nussbaum, E., Monnet, I., Bignon, J., Vanderschueren, R., Guerin, J. C.,

Menard, O., Mignot, P., Dabouis, G. & Douillard, J. Y. 1994. Intrapleural

treatment with recombinant gamma‐interferon in early stage malignant pleural

mesothelioma. Cancer, 74(9), 2460-2467.

Boutin, C., Rey, F. & Viallat, J.-R. 1995. Prevention of malignant seeding after invasive

diagnostic procedures in patients with pleural mesothelioma A randomized trial

of local radiotherapy. CHEST Journal, 108(3), 754-758.

Brunet, J.-F., Denizot, F., Luciani, M.-F., Roux-Dosseto, M., Suzan, M., Mattei, M.-G.

& Golstein, P. 1987. A new member of the immunoglobulin superfamily-CTLA-

4. Nature, 328, 267-270.

Bui, J. D. & Schreiber, R. D. 2007. Cancer immunosurveillance, immunoediting and

inflammation: independent or interdependent processes? Current Opinion in

Immunology, 19(2), 203-208.

Burnet, F. 1969. The concept of immunological surveillance. Progress in experimental

tumor research, 13, 1-27.

Calabrò, L., Morra, A., Fonsatti, E., Cutaia, O., Amato, G., Giannarelli, D., Di

Giacomo, A. M., Danielli, R., Altomonte, M. & Mutti, L. 2013. Tremelimumab

for patients with chemotherapy-resistant advanced malignant mesothelioma: an

open-label, single-arm, phase 2 trial. The lancet oncology, 14(11), 1104-1111.

Callahan, M. K., Wolchok, J. D. & Allison, J. P. 2010. Anti-CTLA-4 Antibody

Therapy: Immune Monitoring During Clinical Development of a Novel

Immunotherapy. Seminars in Oncology, 37(5), 473-484.

Caminschi, I., Venetsanakos, E., Leong, C. C., Garlepp, M. J., Scott, B. & Robinson, B.

W. S. 1998. Interleukin-12 Induces an Effective Antitumor Response in

Malignant Mesothelioma. Am. J. Respir. Cell Mol. Biol., 19(5), 738-746.

Page 232: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

204

Canevari, S., Pupa, S. & Menard, S. 1996. 1975–1995 revised anti-cancer serological

response: biological significance and clinical implications. Annals of Oncology,

7(3), 227-232.

Carbone, M., Emri, S., Dogan, A. U., Steele, I., Tuncer, M., Pass, H. I. & Baris, Y. I.

2007. A mesothelioma epidemic in Cappadocia: scientific developments and

unexpected social outcomes. Nature Reviews Cancer, 7(2), 147-154.

Carter, P., Presta, L., Gorman, C. M., Ridgway, J., Henner, D., Wong, W., Rowland, A.

M., Kotts, C., Carver, M. E. & Shepard, H. M. 1992. Humanization of an anti-

p185HER2 antibody for human cancer therapy. Proceedings of the National

Academy of Sciences, 89(10), 4285-4289.

Castagneto, B., Zai, S., Mutti, L., Lazzaro, A., Ridolfi, R., Piccolini, E., Ardizzoni, A.,

Fumagalli, L., Valsuani, G. & Botta, M. 2001. Palliative and therapeutic activity

of IL-2 immunotherapy in unresectable malignant pleural mesothelioma with

pleural effusion: results of a phase II study on 31 consecutive patients. Lung

Cancer, 31(2), 303-310.

Chaput, N., Darrasse-Jèze, G., Bergot, A.-S., Cordier, C., Ngo-Abdalla, S., Klatzmann,

D. & Azogui, O. 2007. Regulatory T cells prevent CD8 T cell maturation by

inhibiting CD4 Th cells at tumor sites. The Journal of Immunology, 179(8),

4969-4978.

Chen, M.-L., Pittet, M. J., Gorelik, L., Flavell, R. A., Weissleder, R., von Boehmer, H.

& Khazaie, K. 2005. Regulatory T cells suppress tumor-specific CD8 T cell

cytotoxicity through TGF-β signals in vivo. Proceedings of the National

Academy of Sciences of the United States of America, 102(2), 419-424.

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+

Page 233: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

205

CD25+ regulatory T cells by TGF-β induction of transcription factor Foxp3. The

Journal of Experimental Medicine, 198(12), 1875-1886.

Cho, B. J., Feld, R., Leighl, N., Opitz, I., Anraku, M., Tsao, M.-S., Hwang, D. M.,

Hope, A. & de Perrot, M. 2014. A feasibility study evaluating Surgery for

Mesothelioma After Radiation Therapy: the “SMART” approach for resectable

malignant pleural mesothelioma. Journal of Thoracic Oncology, 9(3), 397-402.

Churg, A., Colby, T., Cagle, P., Corson, J., Gibbs, A., Gilks, B., Grimes, M., Hammar,

S., Roggli, V. & Travis, W. 2000. The separation of benign and malignant

mesothelial proliferations. American Journal of Surgical Pathology, 24(9),

1183-1200.

Crawford, A., MacLeod, M., Schumacher, T., Corlett, L. & Gray, D. 2006. Primary T

cell expansion and differentiation in vivo requires antigen presentation by B

cells. The Journal of Immunology, 176(6), 3498-3506.

Curiel, T. J., Coukos, G., Zou, L., Alvarez, X., Cheng, P., Mottram, P., Evdemon-

Hogan, M., Conejo-Garcia, J. R., Zhang, L. & Burow, M. 2004. Specific

recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege

and predicts reduced survival. Nature Medicine, 10(9), 942-949.

Curran, M. A., Montalvo, W., Yagita, H. & Allison, J. P. 2010. PD-1 and CTLA-4

combination blockade expands infiltrating T cells and reduces regulatory T and

myeloid cells within B16 melanoma tumors. Proceedings of the National

Academy of Sciences, 107(9), 4275-4280.

Dai, M., Yip, Y. Y., Hellstrom, I. & Hellstrom, K. E. 2014. Curing mice with large

tumors by locally delivering combinations of immunomodulatory antibodies.

Clinical Cancer Research, clincanres. 1339.2014.

Dang, N. H., Pro, B., Hagemeister, F. B., Samaniego, F., Jones, D., Samuels, B. I.,

Rodriguez, M. A., Goy, A., Romaguera, J. E. & McLaughlin, P. 2007. Phase II

Page 234: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

206

trial of denileukin diftitox for relapsed/refractory T-cell non-Hodgkin

lymphoma. British journal of haematology, 136(3), 439-447.

Dannull, J., Su, Z., Rizzieri, D., Yang, B. K., Coleman, D., Yancey, D., Zhang, A.,

Dahm, P., Chao, N. & Gilboa, E. 2005. Enhancement of vaccine-mediated

antitumor immunity in cancer patients after depletion of regulatory T cells.

Journal of Clinical Investigation, 115(12), 3623-3633.

Dao, T., Liu, C. & Scheinberg, D. A. 2013. Approaching untargetable tumor-associated

antigens with antibodies. OncoImmunology, 2(7), e24678.

Davis, M., Manning, L., Whitaker, D., Garlepp, M. & Robinson, B. 1992.

Establishment of a murine model of malignant mesothelioma. International

journal of cancer, 52(6), 881-886.

de Caestecker, M. P., Piek, E. & Roberts, A. B. 2000. Role of transforming growth

factor-β signaling in cancer. Journal of the National Cancer Institute, 92(17),

1388-1402.

de Visser, K. E., Eichten, A. & Coussens, L. M. 2006. Paradoxical roles of the immune

system during cancer development. Nat Rev Cancer, 6(1), 24-37.

Dexter, D. L., Kowalski, H. M., Blazar, B. A., Fligiel, Z., Vogel, R. & Heppner, G. H.

1978. Heterogeneity of tumor cells from a single mouse mammary tumor.

Cancer Research, 38(10), 3174-3181.

DiLillo, D. J., Matsushita, T. & Tedder, T. F. 2010a. B10 cells and regulatory B cells

balance immune responses during inflammation, autoimmunity, and cancer.

Annals of the New York Academy of Sciences, 1183(1), 38-57.

DiLillo, D. J., Yanaba, K. & Tedder, T. F. 2010b. B cells are required for optimal CD4+

and CD8+ T cell tumor immunity: therapeutic B cell depletion enhances B16

melanoma growth in mice. The Journal of Immunology, 184(7), 4006-4016.

Page 235: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

207

Disis, M. L., Pupa, S. M., Gralow, J. R., Dittadi, R., Menard, S. & Cheever, M. A. 1997.

High-titer HER-2/neu protein-specific antibody can be detected in patients with

early-stage breast cancer. Journal of Clinical Oncology, 15(11), 3363-3367.

Dobrzanski, M. J., Reome, J. B. & Dutton, R. W. 2000. Type 1 and type 2 CD8+

effector T cell subpopulations promote long-term tumor immunity and

protection to progressively growing tumor. The Journal of Immunology, 164(2),

916-925.

Dols, A., Meijer, S. L., Hu, H.-M., Goodell, V., Disis, M. L., von Mensdorff-Pouilly, S.,

Verheijen, R., Alvord, W. G., Smith, J. W. & Urba, W. J. 2003. Identification of

tumor-specific antibodies in patients with breast cancer vaccinated with gene-

modified allogeneic tumor cells. Journal of Immunotherapy, 26(2), 163-170.

Drake, C. G., Jaffee, E. & Pardoll, D. M. 2006. Mechanisms of Immune Evasion by

Tumors. In: JAMES P. ALLISON, G. D. & FREDERICK, W. A. (eds.)

Advances in Immunology. Academic Press,Volume 90, 51-81.

Dunham, L. J. & Stewart, H. L. 1953. A survey of transplantable and transmissible

animal tumors. J Natl Cancer Inst, 13(5), 1299-377.

Dunn, G. P., Bruce, A. T., Ikeda, H., Old, L. J. & Schreiber, R. D. 2002. Cancer

immunoediting: from immunosurveillance to tumor escape. Nat Immunol, 3(11),

991-998.

Dunn, G. P., Old, L. J. & Schreiber, R. D. 2004a. The Immunobiology of Cancer

Immunosurveillance and Immunoediting. Immunity, 21(2), 137-148.

Dunn, G. P., Old, L. J. & Schreiber, R. D. 2004b. The Three Es of Cancer

Immunoediting. Annual Review of Immunology, 22(1), 329-360.

Duraiswamy, J., Kaluza, K. M., Freeman, G. J. & Coukos, G. 2013. Dual blockade of

PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell

rejection function in tumors. Cancer Research, 73(12), 3591-3603.

Page 236: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

208

Egen, J. G., Kuhns, M. S. & Allison, J. P. 2002. CTLA-4: new insights into its

biological function and use in tumor immunotherapy. Nature Immunology, 3(7),

611-618.

Ehrlich, P. 1908. Ueber den jetzigen Stand der Karzinomforschung, Ned Tijdschr

Geneeskd,1, 273-290.

Elliott, R. L. & Blobe, G. C. 2005. Role of transforming growth factor Beta in human

cancer. Journal of Clinical Oncology, 23(9), 2078-2093.

Favaretto, A. G., Aversa, S. M., Paccagnella, A., Manzini, V. D. P., Palmisano, V.,

Oniga, F., Stefani, M., Rea, F., Bortolotti, L. & Loreggian, L. 2003.

Gemcitabine combined with carboplatin in patients with malignant pleural

mesothelioma. Cancer, 97(11), 2791-2797.

Fidler, I. 1973. Selection of successive tumour lines for metastasis. Nature, 242(118),

148-149.

Fitzpatrick, D. R., Bielefeldt-Ohmann, H., Himbeck, R. P., Jarnicki, A. G., Marzo, A. L.

& Robinson, B. W. 1994. Transforming growth factor-beta: antisense RNA-

mediated inhibition affects anchorage-independent growth, tumorigenicity and

tumor-infiltrating T-cells in malignant mesothelioma. Growth Factors, 11(1),

29-44.

French, R. R., Chan, H. C., Tutt, A. L. & Glennie, M. J. 1999. CD40 antibody evokes a

cytotoxic T-cell response that eradicates lymphoma and bypasses T-cell help.

Nature Medicine, 5(5), 548-553.

Friedline, R. H., Brown, D. S., Nguyen, H., Kornfeld, H., Lee, J., Zhang, Y., Appleby,

M., Der, S. D., Kang, J. & Chambers, C. A. 2009. CD4+ regulatory T cells

require CTLA-4 for the maintenance of systemic tolerance. The Journal of

Experimental Medicine, 206(2), 421-434.

Page 237: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

209

Friedman, E., Gold, L. I., Klimstra, D., Zeng, Z., Winawer, S. & Cohen, A. 1995. High

levels of transforming growth factor beta 1 correlate with disease progression in

human colon cancer. Cancer Epidemiology Biomarkers & Prevention, 4(5), 549-

554.

Fukuoka, K., Kuribayashi, K., Yamada, S., Tamura, K., Tabata, C. & Nakano, T. 2013.

Combined serum mesothelin and carcinoembryonic antigen measurement in the

diagnosis of malignant mesothelioma. Molecular and clinical oncology, 1(6),

942-948.

Gabrilovich, D. I. & Nagaraj, S. 2009. Myeloid-derived suppressor cells as regulators of

the immune system. Nature Reviews Immunology, 9(3), 162-174.

Galetto, A., Buttiglieri, S., Forno, S., Moro, F., Mussa, A. & Matera, L. 2003. Drug-and

cell-mediated antitumor cytotoxicities modulate cross-presentation of tumor

antigens by myeloid dendritic cells. Anti-cancer drugs, 14(10), 833-843.

Ganss, R., Arnold, B. & Hämmerling, G. J. 2004. Mini‐review: Overcoming tumor‐

intrinsic resistance to immune effector function. European journal of

immunology, 34(10), 2635-2641.

Geissmann, F., Revy, P., Regnault, A., Lepelletier, Y., Dy, M., Brousse, N., Amigorena,

S., Hermine, O. & Durandy, A. 1999. TGF-β1 Prevents the Noncognate

Maturation of Human Dendritic Langerhans Cells. The Journal of Immunology,

162(8), 4567-4575.

Gerena-Lewis, M., Crawford, J., Bonomi, P., Maddox, A. M., Hainsworth, J., McCune,

D. E., Shukla, R., Zeigler, H., Hurtubise, P. & Chowdhury, T. R. 2009. A Phase

II trial of Denileukin Diftitox in patients with previously treated advanced non-

small cell lung cancer. American journal of clinical oncology, 32(3), 269-273.

Gershon, R. K. & Kondo, K. 1970. Cell interactions in the induction of tolerance: the

role of thymic lymphocytes. Immunology, 18(5), 723-737.

Page 238: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

210

Ghiringhelli, F., Menard, C., Puig, P., Ladoire, S., Roux, S., Martin, F., Solary, E., Le

Cesne, A., Zitvogel, L. & Chauffert, B. 2007. Metronomic cyclophosphamide

regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and

NK effector functions in end stage cancer patients. Cancer Immunology,

Immunotherapy, 56(5), 641-648.

Ghiringhelli, F., Puig, P. E., Roux, S., Parcellier, A., Schmitt, E., Solary, E., Kroemer,

G., Martin, F., Chauffert, B. & Zitvogel, L. 2005. Tumor cells convert immature

myeloid dendritic cells into TGF-β–secreting cells inducing CD4+CD25+

regulatory T cell proliferation. The Journal of Experimental Medicine, 202(7),

919-929.

Gibbs, A. 1990. Role of asbestos and other fibres in the development of diffuse

malignant mesothelioma. Thorax, 45(9), 649-654.

Girardi, M., Glusac, E., Filler, R. B., Roberts, S. J., Propperova, I., Lewis, J., Tigelaar,

R. E. & Hayday, A. C. 2003. The Distinct Contributions of Murine T Cell

Receptor (TCR)γδ+ and TCRαβ+ T Cells to Different Stages of Chemically

Induced Skin Cancer. The Journal of Experimental Medicine, 198(5), 747-755.

Gold, L. I. 1998. The role for transforming growth factor-beta (TGF-beta) in human

cancer. Critical reviews in oncogenesis, 10(4), 303-360.

Gondek, D. C., Lu, L.-F., Quezada, S. A., Sakaguchi, S. & Noelle, R. J. 2005. Cutting

edge: contact-mediated suppression by CD4+ CD25+ regulatory cells involves a

granzyme B-dependent, perforin-independent mechanism. The Journal of

Immunology, 174(4), 1783-1786.

Gordon, J., Holden, H. T., Segal, S. & Feldman, M. 1982. Anti‐tumor immunity in b‐

lymphocyte‐deprived mice. iii. immunity to primary moloney sarcoma virus‐

induced tumors. International journal of cancer, 29(3), 351-357.

Page 239: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

211

Grandjean, M., Sermeus, A., Branders, S., Defresne, F., Dieu, M., Dupont, P., Raes, M.,

De Ridder, M. & Feron, O. 2013. Hypoxia integration in the serological

proteome analysis unmasks tumor antigens and fosters the identification of anti-

phospho-eEF2 antibodies as potential cancer biomarkers. PLoS ONE, 8(10),

e76508.

Graziani, G., Tentori, L. & Navarra, P. 2012. Ipilimumab: a novel immunostimulatory

monoclonal antibody for the treatment of cancer. Pharmacological Research,

65(1), 9-22.

Greenberg, P. D. 1991. Adoptive T cell therapy of tumors: mechanisms operative in the

recognition and elimination of tumor cells. Adv Immunol, 49, 281-355.

Greenwald, R. J., Freeman, G. J. & Sharpe, A. H. 2005. THE B7 FAMILY

REVISITED. Annual Review of Immunology, 23(1), 515-548.

Grigoriu, B.-D., Scherpereel, A., Devos, P., Chahine, B., Letourneux, M., Lebailly, P.,

Grégoire, M., Porte, H., Copin, M.-C. & Lassalle, P. 2007. Utility of osteopontin

and serum mesothelin in malignant pleural mesothelioma diagnosis and

prognosis assessment. Clinical Cancer Research, 13(10), 2928-2935.

Grosso, J. F. & Jure-Kunkel, M. N. 2013. CTLA-4 blockade in tumor models: an

overview of preclinical and translational research. Cancer immunity, 13, 5.

Hanahan, D. & Weinberg, Robert A. 2011. Hallmarks of Cancer: The Next Generation.

Cell, 144(5), 646-674.

Hanson, H. L., Donermeyer, D. L., Ikeda, H., White, J. M., Shankaran, V., Old, L. J.,

Shiku, H., Schreiber, R. D. & Allen, P. M. 2000. Eradication of Established

Tumors by CD8+ T Cell Adoptive Immunotherapy. Immunity, 13(2), 265-276.

Hau, P., Jachimczak, P., Schlingensiepen, R., Schulmeyer, F., Jauch, T., Steinbrecher,

A., Brawanski, A., Proescholdt, M., Schlaier, J. & Buchroithner, J. 2007.

Page 240: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

212

Inhibition of TGF-β 2 with ap 12009 in recurrent malignant gliomas: from

preclinical to phase I/II studies. Oligonucleotides, 17(2), 201-212.

Hawinkels, L. J. & Dijke, P. 2011. Exploring anti-TGF-β therapies in cancer and

fibrosis. Growth Factors, 29(4), 140-152.

Hedman, M., Arnberg, H., Wernlund, J., Riska, H. & Brodin, O. 2002. Tissue

polypeptide antigen (TPA), hyaluronan and CA 125 as serum markers in

malignant mesothelioma. Anticancer research, 23(1B), 531-536.

Hegmans, J. P., Hemmes, A., Hammad, H., Boon, L., Hoogsteden, H. C. & Lambrecht,

B. N. 2006. Mesothelioma environment comprises cytokines and T-regulatory

cells that suppress immune responses. European Respiratory Journal, 27(6),

1086-1095.

Hegmans, J. P., Veltman, J. D., Lambers, M. E., de Vries, I. J. M., Figdor, C. G., W.

Hendriks, R., Hoogsteden, H. C., Lambrecht, B. N. & Aerts, J. G. 2010.

Consolidative Dendritic Cell-based Immunotherapy Elicits Cytotoxicity against

Malignant Mesothelioma. Am. J. Respir. Crit. Care Med., 181(12), 1383-1390.

Hegmans, J. P. J. J., Hemmes, A., Aerts, J. G., Hoogsteden, H. C. & Lambrecht, B. N.

2005. Immunotherapy of Murine Malignant Mesothelioma Using Tumor Lysate-

pulsed Dendritic Cells. Am. J. Respir. Crit. Care Med., 171(10), 1168-1177.

Herlyn, D. M., Steplewski, Z., Herlyn, M. F. & Koprowski, H. 1980. Inhibition of

growth of colorectal carcinoma in nude mice by monoclonal antibody. Cancer

Research, 40(3), 717-721.

Hirano, F., Kaneko, K., Tamura, H., Dong, H., Wang, S., Ichikawa, M., Rietz, C., Flies,

D. B., Lau, J. S. & Zhu, G. 2005. Blockade of B7-H1 and PD-1 by monoclonal

antibodies potentiates cancer therapeutic immunity. Cancer Research, 65(3),

1089-1096.

Page 241: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

213

Hirano, H., Maeda, T., Tsuji, M., Ito, Y., Kizaki, T., Yoshii, Y. & Sashikata, T. 2002.

Malignant mesothelioma of the pericardium: Case reports and

immunohistochemical studies including Ki-67 expression. Pathology

International, 52(10), 669-676.

Hodi, F. S. 2007. Cytotoxic T-Lymphocyte–Associated Antigen-4. Clinical Cancer

Research, 13(18), 5238-5242.

Hodi, F. S. 2010. Overcoming immunological tolerance to melanoma: Targeting CTLA-

4. Asia-Pacific Journal of Clinical Oncology, 6, S16-S23.

Hodi, F. S., O'Day, S. J., McDermott, D. F., Weber, R. W., Sosman, J. A., Haanen, J.

B., Gonzalez, R., Robert, C., Schadendorf, D., Hassel, J. C., Akerley, W., van

den Eertwegh, A. J. M., Lutzky, J., Lorigan, P., Vaubel, J. M., Linette, G. P.,

Hogg, D., Ottensmeier, C. H., Lebbé, C., Peschel, C., Quirt, I., Clark, J. I.,

Wolchok, J. D., Weber, J. S., Tian, J., Yellin, M. J., Nichol, G. M., Hoos, A. &

Urba, W. J. 2010. Improved Survival with Ipilimumab in Patients with

Metastatic Melanoma. New England Journal of Medicine, 363(8), 711-723.

Holmgren, I. 1935. La tuberculine et le BCG chez les cancéreux. Schweiz Med

Wochenschr, 65(120), 1206.

Houghton, A. N., Uchi, H. & Wolchok, J. D. 2005. The role of the immune system in

early epithelial carcinogenesis: B-ware the double-edged sword. Cancer Cell,

7(5), 403-405.

Hurwitz, A. A., Yu, T. F.-Y., Leach, D. R. & Allison, J. P. 1998. CTLA-4 blockade

synergizes with tumor-derived granulocyte- macrophage colony-stimulating

factor for treatment of an experimental mammary carcinoma. Proceedings of the

National Academy of Sciences, 95(17), 10067-10071.

Ireland, D., Kissick, H. & Beilharz, M. 2012. The Role of Regulatory T Cells in

Mesothelioma. Cancer Microenvironment, 5(2), 165-172.

Page 242: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

214

Ismail-Khan, R., Robinson, L. A., Williams, C. C., Garrett, C. R., Bepler, G. & Simon,

G. R. 2006. Malignant pleural mesothelioma: a comprehensive review. Cancer

control, 13(4), 255.

Jackaman, C., Bundell, C. S., Kinnear, B. F., Smith, A. M., Filion, P., van Hagen, D.,

Robinson, B. W. S. & Nelson, D. J. 2003. IL-2 Intratumoral Immunotherapy

Enhances CD8+ T Cells That Mediate Destruction of Tumor Cells and Tumor-

Associated Vasculature: A Novel Mechanism for IL-2. The Journal of

Immunology, 171(10), 5051-5063.

Jackaman, C., Cornwall, S., Graham, P. T. & Nelson, D. J. 2010. CD40-activated B

cells contribute to mesothelioma tumor regression. Immunology and cell

biology, 89(2), 255-267.

Jackaman, C., Cornwall, S., Lew, A. M., Zhan, Y., Robinson, B. W. & Nelson, D. J.

2009. Local effector failure in mesothelioma is not mediated by CD4+ CD25+

T-regulator cells. European Respiratory Journal, 34(1), 162-175.

Jackaman, C., Lansley, S., Allan, J. E., Robinson, B. W. S. & Nelson, D. J. 2012. IL-

2/CD40-driven NK cells install and maintain potency in the anti-mesothelioma

effector/memory phase. International Immunology, 24(6), 357-368.

Jackaman, C., Lew, A. M., Zhan, Y., Allan, J. E., Koloska, B., Graham, P. T.,

Robinson, B. W. S. & Nelson, D. J. 2008. Deliberately provoking local

inflammation drives tumors to become their own protective vaccine site.

International Immunology, 20(11), 1467-1479.

Jackaman, C. & Nelson, D. 2012. Intratumoral interleukin-2/agonist CD40 antibody

drives CD4+-independent resolution of treated-tumors and CD4+-dependent

systemic and memory responses. Cancer Immunology, Immunotherapy, 61(4),

549-560.

Page 243: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

215

Jänne, P. A. 2003. Chemotherapy for malignant pleural mesothelioma. Clinical lung

cancer, 5(2), 98-106.

Jasinska, J., Wagner, S., Radauer, C., Sedivy, R., Brodowicz, T., Wiltschke, C.,

Breiteneder, H., Pehamberger, H., Scheiner, O. & Wiedermann, U. 2003.

Inhibition of tumor cell growth by antibodies induced after vaccination with

peptides derived from the extracellular domain of Her‐2/neu. International

journal of cancer, 107(6), 976-983.

Jochems, C., Tucker, J. A., Tsang, K.-Y., Madan, R. A., Dahut, W. L., Liewehr, D. J.,

Steinberg, S. M., Gulley, J. L. & Schlom, J. 2014. A combination trial of

vaccine plus ipilimumab in metastatic castration-resistant prostate cancer

patients: immune correlates. Cancer Immunology, Immunotherapy, 63(4), 407-

418.

Kamp, D. W. 2009. Asbestos-induced lung diseases: an update. Translational Research,

153(4), 143-152.

Kamp, D. W. & Mossman, B. T. 2002. Asbestos-associated cancers: clinical spectrum

and pathogenic mechanisms. Clinics in Occupational and Environmental

Medicine, 2(4), 753-777.

Kantoff, P. W., Higano, C. S., Shore, N. D., Berger, E. R., Small, E. J., Penson, D. F.,

Redfern, C. H., Ferrari, A. C., Dreicer, R. & Sims, R. B. 2010. Sipuleucel-T

immunotherapy for castration-resistant prostate cancer. New England Journal of

Medicine, 363(5), 411-422.

Kehrl, J. H., Wakefield, L. M., Roberts, A. B., Jakowlew, S., Alvarez-Mon, M.,

Derynck, R., Sporn, M. B. & Fauci, A. S. 1986. Production of transforming

growth factor beta by human T lymphocytes and its potential role in the

regulation of T cell growth. The Journal of Experimental Medicine, 163(5),

1037-1050.

Page 244: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

216

Keir, M. E., Butte, M. J., Freeman, G. J. & Sharpe, A. H. 2008. PD-1 and its ligands in

tolerance and immunity. Annu. Rev. Immunol., 26, 677-704.

Kim, J. M., Rasmussen, J. P. & Rudensky, A. Y. 2006. Regulatory T cells prevent

catastrophic autoimmunity throughout the lifespan of mice. Nature Immunology,

8(2), 191-197.

Kissick, H. T., Ireland, D. J. & Beilharz, M. W. 2009. Combined Intratumoral

Regulatory T-Cell Depletion and Transforming Growth Factor- Neutralization

Induces Regression of Established AE17 Murine Mesothelioma Tumors.

Journal of Interferon and Cytokine Research, 29(4), 209-216.

Kissick, H. T., Ireland, D. J., Greay, S. J. & Beilharz, M. W. 2010. Mechanisms of

Immune Suppression Exerted by Regulatory T-Cells in Subcutaneous AE17

Murine Mesothelioma. Journal of Interferon & Cytokine Research, 30(11), 829-

834.

Kissick, H. T., Ireland, D. J., Krishnan, S., Madondo, M. & Beilharz, M. W. 2012.

Tumour eradication and induction of memory against murine mesothelioma by

combined immunotherapy. Immunol Cell Biol, 90(8), 822-826.

Kitamura, D., Roes, J., Kühn, R. & Rajewsky, K. 1991. AB cell-deficient mouse by

targeted disruption of the membrane exon of the immunoglobulin μ chain gene.

Kline, J., Zhang, L., Battaglia, L., Cohen, K. S. & Gajewski, T. F. 2012. Cellular and

molecular requirements for rejection of B16 melanoma in the setting of

regulatory T cell depletion and homeostatic proliferation. The Journal of

Immunology, 188(6), 2630-2642.

Kono, H. & Rock, K. L. 2008. How dying cells alert the immune system to danger.

Nature Reviews Immunology, 8(4), 279-289.

Korman, A. J., Peggs, K. S. & Allison, J. P. 2006. Checkpoint blockade in cancer

immunotherapy. Advances in immunology, 90, 297-339.

Page 245: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

217

Korrer, M. J. & Routes, J. M. 2014. Possible Role of Arginase-1 in Concomitant Tumor

Immunity. PLoS ONE, 9(3), e91370.

Koss, M. N., Fleming, M., Przygodzki, R. M., Sherrod, A., Travis, W. & Hochholzer, L.

1998. Adenocarcinoma simulating mesothelioma: a clinicopathologic and

immunohistochemical study of 29 cases. Annals of diagnostic pathology, 2(2),

93-102.

Krishnan, S., Bakker, E., Lee, C., Kissick, H. T., Ireland, D. J. & Beilharz, M. W. 2014.

Successful combined intra-tumoural immunotherapy of established murine

mesotheliomas requires B cell involvement. Journal of Interferon & Cytokine

Research, In press.

Krummel, M. F. & Allison, J. P. 1995. CD28 and CTLA-4 have opposing effects on the

response of T cells to stimulation. The Journal of Experimental Medicine,

182(2), 459-465.

Leibson, P. J. 1997. Signal transduction during natural killer cell activation: inside the

mind of a killer. Immunity, 6(6), 655-661.

Leigh, J. & Driscoll, T. 2003. Malignant mesothelioma in Australia, 1945–2002.

International Journal of Occupational and Environmental Health, 9(3), 206-

217.

Lengauer, C., Kinzler, K. W. & Vogelstein, B. 1998. Genetic instabilities in human

cancers. Nature, 396(6712), 643-649.

Lesterhuis, W. J., Salmons, J., Nowak, A. K., Rozali, E. N., Khong, A., Dick, I. M.,

Harken, J. A., Robinson, B. W. & Lake, R. A. 2013. Synergistic effect of

CTLA-4 blockade and cancer chemotherapy in the induction of anti-tumor

immunity. PLoS ONE, 8(4), e61895.

Page 246: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

218

Levental, K. R., Yu, H., Kass, L., Lakins, J. N., Egeblad, M., Erler, J. T., Fong, S. F.,

Csiszar, K., Giaccia, A. & Weninger, W. 2009. Matrix crosslinking forces tumor

progression by enhancing integrin signaling. Cell, 139(5), 891-906.

Li, M. O. & Flavell, R. A. 2008. TGF-β: a master of all T cell trades. Cell, 134(3), 392-

404.

Li, Q., Lao, X., Pan, Q., Ning, N., Yet, J., Xu, Y., Li, S. & Chang, A. E. 2011. Adoptive

Transfer of Tumor Reactive B Cells Confers Host T-Cell Immunity and Tumor

Regression. Clinical Cancer Research, 17(15), 4987-4995.

Lin, F., Liu, Y., Liu, Y., Keshava, N. & Li, S. 2000. Crocidolite induces cell

transformation and p53 gene mutation in BALB/c‐3T3 cells. Teratogenesis,

carcinogenesis, and mutagenesis, 20(5), 273-281.

Lin, W.-W. & Karin, M. 2007. A cytokine-mediated link between innate immunity,

inflammation, and cancer. The Journal of Clinical Investigation, 117(5), 1175-

1183.

Liu, Z. & Klominek, J. 2004. Chemotaxis and chemokinesis of malignant mesothelioma

cells to multiple growth factors. Anticancer research, 24(3A), 1625-1630.

Liyanage, U. K., Moore, T. T., Joo, H.-G., Tanaka, Y., Herrmann, V., Doherty, G.,

Drebin, J. A., Strasberg, S. M., Eberlein, T. J. & Goedegebuure, P. S. 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(5), 2756-2761.

Lyubchenko, T. A., Wurth, G. A. & Zweifach, A. 2001. Role of calcium influx in

cytotoxic T lymphocyte lytic granule exocytosis during target cell killing.

Immunity, 15(5), 847-859.

Page 247: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

219

Maasilta, P. 1991. Deterioration in lung function following hemithorax irradiation for

pleural mesothelioma. International Journal of Radiation Oncology* Biology*

Physics, 20(3), 433-438.

Madan, R. A., Mohebtash, M., Arlen, P. M., Vergati, M., Rauckhorst, M., Steinberg, S.

M., Tsang, K. Y., Poole, D. J., Parnes, H. L. & Wright, J. J. 2012. Ipilimumab

and a poxviral vaccine targeting prostate-specific antigen in metastatic

castration-resistant prostate cancer: a phase 1 dose-escalation trial. The lancet

oncology, 13(5), 501-508.

Maecker, H. T. & Trotter, J. 2006. Flow cytometry controls, instrument setup, and the

determination of positivity. Cytometry Part A, 69(9), 1037-1042.

Mahaweni, N. M., Kaijen-Lambers, M. E., Dekkers, J., Aerts, J. G. & Hegmans, J. P.

2013. Tumour-derived exosomes as antigen delivery carriers in dendritic cell-

based immunotherapy for malignant mesothelioma. Journal of extracellular

vesicles, 2, 10.

Mahnke, K., Schönfeld, K., Fondel, S., Ring, S., Karakhanova, S., Wiedemeyer, K.,

Bedke, T., Johnson, T. S., Storn, V. & Schallenberg, S. 2007. Depletion of

CD4+ CD25+ human regulatory T cells in vivo: kinetics of Treg depletion and

alterations in immune functions in vivo and in vitro. International journal of

cancer, 120(12), 2723-2733.

Manson, L. A. 1994. Anti-tumor immune responses of the tumor-bearing host: the case

for antibody-mediated immunologic enhancement. Clinical immunology and

immunopathology, 72(1), 1-8.

Markowitz, S. D. & Roberts, A. B. 1996. Tumor suppressor activity of the TGF-β

pathway in human cancers. Cytokine & growth factor reviews, 7(1), 93-102.

Mauri, C. & Bosma, A. 2012. Immune Regulatory Function of B Cells. Annual Review

of Immunology, 30(1), 221-241.

Page 248: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

220

May, K. F., Chen, L., Zheng, P. & Liu, Y. 2002. Anti-4-1BB monoclonal antibody

enhances rejection of large tumor burden by promoting survival but not clonal

expansion of tumor-specific CD8+ T cells. Cancer Research, 62(12), 3459-

3465.

McCarthy, E. F. 2006. The toxins of William B. Coley and the treatment of bone and

soft-tissue sarcomas. The Iowa orthopaedic journal, 26, 154-158.

McMasters, K. M., Edwards, M. J., Ross, M. I., Reintgen, D. S., Martin, R. C., Urist, M.

M., Noyes, R. D., Sussman, J. J., Stromberg, A. J. & Scoggins, C. R. 2010.

Ulceration as a predictive marker for response to adjuvant interferon therapy in

melanoma. Annals of surgery, 252(3), 460-466.

Melero, I., Shuford, W. W., Newby, S. A., Aruffo, A., Ledbetter, J. A., Hellström, K.

E., Mittler, R. S. & Chen, L. 1997. Monoclonal antibodies against the 4-1BB T-

cell activation molecule eradicate established tumors. Nature Medicine, 3(6),

682-685.

Möller, G. & Möller, E. 1976. The concept of immunological surveillance against

neoplasia. Immunological Reviews, 28(1), 3-17.

Molnarfi, N., Schulze-Topphoff, U., Weber, M. S., Patarroyo, J. C., Prod’homme, T.,

Varrin-Doyer, M., Shetty, A., Linington, C., Slavin, A. J., Hidalgo, J., Jenne, D.

E., Wekerle, H., Sobel, R. A., Bernard, C. C. A., Shlomchik, M. J. & Zamvil, S.

S. 2013. MHC class II–dependent B cell APC function is required for induction

of CNS autoimmunity independent of myelin-specific antibodies. The Journal of

Experimental Medicine, 210(13), 2921-2937.

Moody, M. A. & Haynes, B. F. 2008. Antigen‐specific B cell detection reagents: Use

and quality control. Cytometry Part A, 73(11), 1086-1092.

Page 249: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

221

Morales, A., Eidinger, D. & Bruce, A. 1976. Intracavitary Bacillus Calmette-Guerin in

the treatment of superficial bladder tumors. The Journal of urology, 116(2), 180-

183.

Mossman, B. T., Kamp, D. W. & Weitzman, S. A. 1996. Mechanisms of carcinogenesis

and clinical features of asbestos-associated cancers. Cancer investigation, 14(5),

466-480.

Mougiakakos, D., Choudhury, A., Lladser, A., Kiessling, R. & Johansson, C. C. 2010.

Regulatory T cells in cancer. Advances in cancer research, 107, 57-117.

Mukherjee, S. & Robinson, B. W. 2002. Immunotherapy of malignant mesothelioma.

Mesothelioma. London: Martin Dunitz, 325-38.

Najjar, Y. G. & Kirkwood, J. M. 2014. Melanoma and Skin Cancer Pembrolizumab:

Pharmacology and Therapeutics Review. CME, 2014.

Nakataki, E., Yano, S., Matsumori, Y., Goto, H., Kakiuchi, S., Muguruma, H., Bando,

Y., Uehara, H., Hamada, H. & Kito, K. 2006. Novel orthotopic implantation

model of human malignant pleural mesothelioma (EHMES‐10 cells) highly

expressing vascular endothelial growth factor and its receptor. Cancer science,

97(3), 183-191.

Nauts, H. C. & McLaren, J. R. 1990. Coley toxins—the first century. In: BICHER, H.,

MCLAREN, J. R. & PIGLIUCCI, G. M. (eds.) Consensus on Hyperthermia for

the 1990s. Springer US,267, 483-500.

Needham, D. J., Lee, J. X. & Beilharz, M. W. 2006. Intra-tumoural regulatory T cells:

A potential new target in cancer immunotherapy. Biochemical and Biophysical

Research Communications, 343(3), 684-691.

Nefedova, Y., Cheng, P., Gilkes, D., Blaskovich, M., Beg, A. A., Sebti, S. M. &

Gabrilovich, D. I. 2005. Activation of dendritic cells via inhibition of

Jak2/STAT3 signaling. The Journal of Immunology, 175(7), 4338-4346.

Page 250: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

222

Nelson, B. H. 2010. CD20+ B cells: the other tumor-infiltrating lymphocytes. The

Journal of Immunology, 185(9), 4977-4982.

Newton, M. R., Askeland, E. J., Andresen, E. D., Chehval, V. A., Wang, X., Askeland,

R. W., O'Donnell, M. A. & Luo, Y. 2014. Anti‐interleukin‐10R1 monoclonal

antibody in combination with bacillus Calmette–Guérin is protective against

bladder cancer metastasis in a murine orthotopic tumour model and

demonstrates systemic specific anti‐tumour immunity. Clinical & Experimental

Immunology, 177(1), 261-268.

North, R. J. 1982. Cyclophosphamide-facilitated adoptive immunotherapy of an

established tumor depends on elimination of tumor-induced suppressor T cells.

The Journal of Experimental Medicine, 155(4), 1063.

O'Brien, S. G., Guilhot, F., Larson, R. A., Gathmann, I., Baccarani, M., Cervantes, F.,

Cornelissen, J. J., Fischer, T., Hochhaus, A. & Hughes, T. 2003. Imatinib

compared with interferon and low-dose cytarabine for newly diagnosed chronic-

phase chronic myeloid leukemia. New England Journal of Medicine, 348(11),

994-1004.

Olkhanud, P. B., Damdinsuren, B., Bodogai, M., Gress, R. E., Sen, R., Wejksza, K.,

Malchinkhuu, E., Wersto, R. P. & Biragyn, A. 2011. Tumor-Evoked Regulatory

B Cells Promote Breast Cancer Metastasis by Converting Resting CD4+ T Cells

to T-Regulatory Cells. Cancer Research, 71(10), 3505-3515.

Olsen, E., Duvic, M., Frankel, A., Kim, Y., Martin, A., Vonderheid, E., Jegasothy, B.,

Wood, G., Gordon, M. & Heald, P. 2001. Pivotal phase III trial of two dose

levels of denileukin diftitox for the treatment of cutaneous T-cell lymphoma.

Journal of Clinical Oncology, 19(2), 376-388.

Olsen, N. J., Franklin, P. J., Reid, A., de Klerk, N. H., Threlfall, T. J., Shilkin, K. &

Musk, B. 2011. Increasing incidence of malignant mesothelioma after exposure

Page 251: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

223

to asbestos during home maintenance and renovation. Med J Aust, 195(5), 271-

274.

Onishi, Y., Fehervari, Z., Yamaguchi, T. & Sakaguchi, S. 2008. Foxp3+ natural

regulatory T cells preferentially form aggregates on dendritic cells in vitro and

actively inhibit their maturation. Proceedings of the National Academy of

Sciences, 105(29), 10113-10118.

Onizuka, S., Tawara, I., Shimizu, J., Sakaguchi, S., Fujita, T. & Nakayama, E. 1999.

Tumor Rejection by in Vivo Administration of Anti-CD25 (Interleukin-2

Receptor α) Monoclonal Antibody. Cancer Research, 59(13), 3128-3133.

Ostrand-Rosenberg, S. 2008. Immune surveillance: a balance between protumor and

antitumor immunity. Current opinion in genetics & development, 18(1), 11-18.

Ostrand-Rosenberg, S., Clements, V. K., Terabe, M., Park, J. M., Berzofsky, J. A. &

Dissanayake, S. K. 2002. Resistance to metastatic disease in STAT6-deficient

mice requires hemopoietic and nonhemopoietic cells and is IFN-γ dependent.

The Journal of Immunology, 169(10), 5796-5804.

Ostrand-Rosenberg, S. & Sinha, P. 2009. Myeloid-derived suppressor cells: linking

inflammation and cancer. The Journal of Immunology, 182(8), 4499-4506.

Özdemir, B. C., Pentcheva-Hoang, T., Carstens, J. L., Zheng, X., Wu, C.-C., Simpson,

T. R., Laklai, H., Sugimoto, H., Kahlert, C. & Novitskiy, S. V. 2014. Depletion

of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and

accelerates pancreas cancer with reduced survival. Cancer Cell, 25(6), 719-734.

Pardoll, D. M. 2012. The blockade of immune checkpoints in cancer immunotherapy.

Nature Reviews Cancer, 12(4), 252-264.

Park, C.-Y., Min, K. N., Son, J.-Y., Park, S.-Y., Nam, J.-S., Kim, D.-K. & Sheen, Y. Y.

2014. An novel inhibitor of TGF-β type I receptor, IN-1130, blocks breast

Page 252: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

224

cancer lung metastasis through inhibition of epithelial-mesenchymal transition.

Cancer letters, 351(1), 72-80.

Penafuerte, C. & Galipeau, J. 2008. TGFβ secreted by B16 melanoma antagonizes

cancer gene immunotherapy bystander effect. Cancer Immunology,

Immunotherapy, 57(8), 1197-1206.

Penaloza-MacMaster, P., Kamphorst, A. O., Wieland, A., Araki, K., Iyer, S. S., West,

E. E., O’Mara, L., Yang, S., Konieczny, B. T., Sharpe, A. H., Freeman, G. J.,

Rudensky, A. Y. & Ahmed, R. 2014. Interplay between regulatory T cells and

PD-1 in modulating T cell exhaustion and viral control during chronic LCMV

infection. The Journal of Experimental Medicine, 211(9), 1905-1918.

Peuvrel, L., Nguyen, J., Khammari, A., Quereux, G., Brocard, A. & Dreno, B. 2011. Is

primary melanoma ulceration a factor of good response to adoptive

immunotherapy? Journal of the European Academy of Dermatology and

Venereology, 25(11), 1311-1317.

Pinkus, G. S. & Kurtin, P. J. 1985. Epithelial membrane antigen-a diagnostic

discriminant in surgical pathology: immunohistochemical profile in epithelial,

mesenchymal, and hematopoietic neoplasms using paraffin sections and

monoclonal antibodies. Human pathology, 16(9), 929-940.

Pinto, M., Bernstein, L., Brogan, D. & Criscuolo, E. 1986. Carcinoembryonic antigen in

effusions. A diagnostic adjunct to cytology. Acta cytologica, 31(2), 113-118.

Powell, A., Creaney, J., Broomfield, S., Van Bruggen, I. & Robinson, B. 2006.

Recombinant GM-CSF plus autologous tumor cells as a vaccine for patients

with mesothelioma. Lung Cancer, 52(2), 189-197.

Powers, A. & Carbone, M. 2002. The role of environmental carcinogens, viruses and

genetic predisposition in the pathogenesis of mesothelioma. Cancer biology &

therapy, 1(4), 348-353.

Page 253: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

225

Qin, Z., Richter, G., Schüler, T., Ibe, S., Cao, X. & Blankenstein, T. 1998. B cells

inhibit induction of T cell-dependent tumor immunity. Nature Medicine, 4(5),

627-630.

Qin, Z., Schwartzkopff, J., Pradera, F., Kammertœns, T., Seliger, B., Pircher, H. &

Blankenstein, T. 2003. A critical requirement of interferon γ-mediated

angiostasis for tumor rejection by CD8+ T cells. Cancer Research, 63(14),

4095-4100.

Quezada, S. A., Peggs, K. S., Curran, M. A. & Allison, J. P. 2006. CTLA4 blockade and

GM-CSF combination immunotherapy alters the intratumor balance of effector

and regulatory T cells. The Journal of Clinical Investigation, 116(7), 1935-1945.

Rabinovich, G. A., Gabrilovich, D. & Sotomayor, E. M. 2007. Immunosuppressive

Strategies that are Mediated by Tumor Cells. Annual Review of Immunology,

25(1), 267-96.

Reff, M. E., Carner, K., Chambers, K., Chinn, P., Leonard, J., Raab, R., Newman, R.,

Hanna, N. & Anderson, D. 1994. Depletion of B cells in vivo by a chimeric

mouse human monoclonal antibody to CD20. Blood, 83(2), 435-445.

Reid, A., de Klerk, N. H., Magnani, C., Ferrante, D., Berry, G., Musk, A. W. & Merler,

E. 2014. Mesothelioma risk after 40 years since first exposure to asbestos: a

pooled analysis. Thorax.

Ritchie, D. S., Yang, J., Hermans, I. F. & Ronchese, F. 2004. B-Lymphocytes Activated

by CD40 Ligand Induce an Antigen-Specific Anti-Tumour Immune Response

by Direct and Indirect Activation of CD8+ T-cells. Scandinavian Journal of

Immunology, 60(6), 543-551.

Rivera, Z., Ferrone, S., Wang, X., Jube, S., Yang, H., Pass, H. I., Kanodia, S., Gaudino,

G. & Carbone, M. 2012. CSPG4 as a target of antibody-based immunotherapy

for malignant mesothelioma. Clinical Cancer Research, 18(19), 5352-5363.

Page 254: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

226

Robert, C., Thomas, L., Bondarenko, I., O'Day, S., Weber, J., Garbe, C., Lebbe, C.,

Baurain, J.-F., Testori, A. & Grob, J.-J. 2011. Ipilimumab plus dacarbazine for

previously untreated metastatic melanoma. New England Journal of Medicine,

364(26), 2517-2526.

Robinson, B., Bowman, R., Christmas, T., Musk, A. & Manning, L. 1991.

Immunotherapy for malignant mesothelioma: use of interleukin-2 and interferon

alpha. Interferons Cytokines, 18, 5-7.

Robinson, B. M. 2012. Malignant pleural mesothelioma: an epidemiological

perspective. Annals of Cardiothoracic Surgery, 1(4), 491-496.

Robinson, B. W. S., Creaney, J., Lake, R., Nowak, A., Musk, A. W., de Klerk, N.,

Winzell, P., Hellstrom, K. E. & Hellstrom, I. 2003. Mesothelin-family proteins

and diagnosis of mesothelioma. The Lancet, 362(9396), 1612-1616.

Robinson, B. W. S. & Lake, R. A. 2005. Advances in Malignant Mesothelioma. New

England Journal of Medicine, 353(15), 1591-1603.

Robinson, B. W. S., Musk, A. W. & Lake, R. A. 2005. Malignant mesothelioma. The

Lancet, 366(9483), 397-408.

Robinson, C., Callow, M., Stevenson, S., Scott, B., Robinson, B. W. & Lake, R. A.

2000. Serologic responses in patients with malignant mesothelioma: evidence

for both public and private specificities. American journal of respiratory cell

and molecular biology, 22(5), 550-556.

Roggli, V. L. & Sharma, A. 2014. Analysis of tissue mineral fiber content. Pathology of

asbestos-associated diseases. Springer, 253-292.

Romagnani, S., Parronchi, P., D’elios, M., Romagnani, P., Annunziato, F., Piccinni, M.,

Manetti, R., Sampognaro, S., Mavilia, C. & De Carli, M. 1997. An update on

human Th1 and Th2 cells. International archives of allergy and immunology,

113(1-3), 153-156.

Page 255: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

227

Ruddle, J. & Prince, H. M. 2007. Denileukin diftitox and vision loss. Leukemia &

lymphoma, 48(4), 655-656.

Ruggiero, R., Bustuoadad, O., Bonfil, R., Sordelli, D., Fontan, P., Meiss, R. &

Pasqualini, C. 1988. [Antitumor concomitant immunity: a possible metastasis

control mechanism]. Medicina, 49(3), 277-281.

Rusch, V. W. 1999. Indications for pneumonectomy. Extrapleural pneumonectomy.

Chest surgery clinics of North America, 9(2), 327-338.

Sakaguchi, S. 2000. Animal models of autoimmunity and their relevance to human

diseases. Current Opinion in Immunology, 12(6), 684-690.

Sakaguchi, S. 2004. Naturally Arising CD4+ Regulatory T Cells for Immunologic Self-

Tolerance and Negative Control of Immune Responses. Annual Review of

Immunology, 22(1), 531-562.

Sakaguchi, S., Fukuma, K., Kuribayashi, K. & Masuda, T. 1985. Organ-specific

autoimmune diseases induced in mice by elimination of T cell subset. I.

Evidence for the active participation of T cells in natural self-tolerance; deficit

of a T cell subset as a possible cause of autoimmune disease. The Journal of

Experimental Medicine, 161(1), 72-87.

Sakaguchi, S., Ono, M., Setoguchi, R., Yagi, H., Hori, S., Fehervari, Z., Shimizu, J.,

Takahashi, T. & Nomura, T. 2006. Foxp3+CD25+CD4+ natural regulatory T

cells in dominant self-tolerance and autoimmune disease. Immunological

Reviews, 212(1), 8-27.

Sakaguchi, S., Yamaguchi, T., Nomura, T. & Ono, M. 2008. Regulatory T cells and

immune tolerance. Cell, 133(5), 775-787.

Sato, T., Suzuki, Y., Mori, T., Maeda, M., Abe, M., Hino, O. & Takahashi, K. 2014.

Newly established ELISA for N‐ERC/mesothelin improves diagnostic accuracy

in patients with suspected pleural mesothelioma. Cancer medicine.

Page 256: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

228

Saunier, E. F. & Akhurst, R. J. 2006. TGF beta inhibition for cancer therapy. Current

cancer drug targets, 6(7), 565-578.

Scherpereel, A., Astoul, P., Baas, P., Berghmans, T., Clayson, H., De Vuyst, P.,

Dienemann, H., Galateau-Salle, F., Hennequin, C. & Hillerdal, G. 2010.

Guidelines of the European Respiratory Society and the European Society of

Thoracic Surgeons for the management of malignant pleural mesothelioma.

European Respiratory Journal, 35(3), 479-495.

Schipmann, S., Wermker, K., Schulze, H.-J., Kleinheinz, J. & Brunner, G. 2014.

Cutaneous and oral squamous cell carcinoma–dual immunosuppression via

recruitment of FOXP3+ regulatory T cells and endogenous tumour FOXP3

expression? Journal of Cranio-Maxillofacial Surgery.

Schneider, H., Downey, J., Smith, A., Zinselmeyer, B. H., Rush, C., Brewer, J. M., Wei,

B., Hogg, N., Garside, P. & Rudd, C. E. 2006. Reversal of the TCR stop signal

by CTLA-4. Science, 313(5795), 1972-1975.

Schreiber, R. D., Old, L. J. & Smyth, M. J. 2011. Cancer immunoediting: integrating

immunity's roles in cancer suppression and promotion. Science Signaling,

331(6024), 1565-1570.

Schultz, K. R., Klarnet, J. P., Gieni, R. S., HAYGLAss, K. T. & Greenberg, P. D. 1990.

The role of B cells for in vivo T cell responses to a Friend virus-induced

leukemia. Science, 249(4971), 921-923.

Schumacher, U., Mukthar, D. & Schenker, T. 1993. Reactivity of monoclonal

antibodies directed against lung cancer antigens with human lung, breast and

colon cancer cell lines. Disease markers, 11(5-6), 225-237.

Sedgwick, J. & Holt, P. 1983. A solid-phase immunoenzymatic technique for the

enumeration of specific antibody-secreting cells. Journal of immunological

methods, 57(1), 301-309.

Page 257: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

229

Sengupta, N., MacFie, T. S., MacDonald, T. T., Pennington, D. & Silver, A. R. 2010.

Cancer immunoediting and “spontaneous” tumor regression. Pathology -

Research and Practice, 206(1), 1-8.

Shah, S., Divekar, A. A., Hilchey, S. P., Cho, H. M., Newman, C. L., Shin, S. U.,

Nechustan, H., Challita‐Eid, P. M., Segal, B. M. & Yi, K. H. 2005. Increased

rejection of primary tumors in mice lacking B cells: Inhibition of anti‐tumor

CTL and TH1 cytokine responses by B cells. International journal of cancer,

117(4), 574-586.

Shankaran, V., Ikeda, H., Bruce, A. T., White, J. M., Swanson, P. E., Old, L. J. &

Schreiber, R. D. 2001. IFNγ and lymphocytes prevent primary tumour

development and shape tumour immunogenicity. Nature, 410(6832), 1107-1111.

Shi, Y. & Rock, K. L. 2002. Cell death releases endogenous adjuvants that selectively

enhance immune surveillance of particulate antigens. European journal of

immunology, 32(1), 155-162.

Shiao, S. L., Ganesan, A. P., Rugo, H. S. & Coussens, L. M. 2011. Immune

microenvironments in solid tumors: new targets for therapy. Genes &

Development, 25(24), 2559-2572.

Shimizu, J., Yamazaki, S. & Sakaguchi, S. 1999. Induction of tumor immunity by

removing CD25+ CD4+ T cells: a common basis between tumor immunity and

autoimmunity. The Journal of Immunology, 163(10), 5211-5218.

Shrikant, P. & Mescher, M. F. 1999. Control of syngeneic tumor growth by activation

of CD8+ T cells: efficacy is limited by migration away from the site and

induction of nonresponsiveness. The Journal of Immunology, 162(5), 2858-

2866.

Page 258: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

230

Silagi, S., Dutkowski, R. & Schaefer, A. 1988. Eradication of mouse melanoma by

combined treatment with recombinant human interleukin 2 and recombinant

murine interferon‐gamma. International journal of cancer, 41(2), 315-322.

Sim, M. R. 2013. A worldwide ban on asbestos production and use: some recent

progress, but more still to be done. Occupational and Environmental Medicine,

70(1), 1-2.

Singh, B., Read, S., Asseman, C., Malmström, V., Mottet, C., Stephens, L. A.,

Stepankova, R., Tlaskalova, H. & Powrie, F. 2001. Control of intestinal

inflammation by regulatory T cells. Immunological Reviews, 182(1), 190-200.

Smyth, M. J., Teng, M. W. L., Swann, J., Kyparissoudis, K., Godfrey, D. I. &

Hayakawa, Y. 2006. CD4+CD25+ T Regulatory Cells Suppress NK Cell-

Mediated Immunotherapy of Cancer. The Journal of Immunology, 176(3), 1582-

1587.

Spranger, S., Koblish, H. K., Horton, B., Scherle, P. A., Newton, R. & Gajewski, T. F.

2014. Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or

IDO blockade involves restored IL-2 production and proliferation of CD8+ T

cells directly within the tumor microenvironment. Journal for immunotherapy of

cancer, 2(1), 3.

Stahel, R., Weder, W., Lievens, Y. & Felip, E. 2010. Malignant pleural mesothelioma:

ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann

Oncol, 21(Suppl 5), v126-v128.

Staveley-O’Carroll, K., Sotomayor, E., Montgomery, J., Borrello, I., Hwang, L., Fein,

S., Pardoll, D. & Levitsky, H. 1998. Induction of antigen-specific T cell anergy:

an early event in the course of tumor progression. Proceedings of the National

Academy of Sciences, 95(3), 1178-1183.

Page 259: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

231

Sterman, D. H. & Albelda, S. M. 2005. Advances in the diagnosis, evaluation, and

management of malignant pleural mesothelioma. Respirology, 10(3), 266-283.

Sterman, D. H., Recio, A., Haas, A. R., Vachani, A., Katz, S. I., Gillespie, C. T., Cheng,

G., Sun, J., Moon, E. & Pereira, L. 2010. A phase I trial of repeated intrapleural

adenoviral-mediated interferon-β gene transfer for mesothelioma and metastatic

pleural effusions. Molecular therapy, 18(4), 852-860.

Stewart, D. J., Martin-Ucar, A., Pilling, J. E., Edwards, J. G., O'Byrne, K. J. & Waller,

D. A. 2004. The effect of extent of local resection on patterns of disease

progression in malignant pleural mesothelioma. The Annals of thoracic surgery,

78(1), 245-252.

Sugarbaker, D. J., Flores, R. M., Jaklitsch, M. T., Richards, W. G., Strauss, G. M.,

Corson, J. M., DeCamp Jr, M. M., Swanson, S. J., Bueno, R. & Lukanich, J. M.

1999. Resection margins, extrapleural nodal status, and cell type determine

postoperative long-term survival in trimodality therapy of malignant pleural

mesothelioma: results in 183 patients. The Journal of thoracic and

cardiovascular surgery, 117(1), 54-65.

Sugarbaker, D. J., Jaklitsch, M. T., Bueno, R., Richards, W., Lukanich, J., Mentzer, S.

J., Colson, Y., Linden, P., Chang, M. & Capalbo, L. 2004. Prevention, early

detection, and management of complications after 328 consecutive extrapleural

pneumonectomies. The Journal of thoracic and cardiovascular surgery, 128(1),

138-146.

Takeda, K., Kojima, Y., Uno, T., Hayakawa, Y., Teng, M. W. L., Yoshizawa, H.,

Yagita, H., Gejyo, F., Okumura, K. & Smyth, M. J. 2010. Combination Therapy

of Established Tumors by Antibodies Targeting Immune Activating and

Suppressing Molecules. The Journal of Immunology, 184(10), 5493-5501.

Page 260: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

232

Takeuchi, M., Keino, H., Kezuka, T., Usui, M. & Taguchi, O. 2004. Immune Responses

to Retinal Self-Antigens in CD25+ CD4+ Regulatory T-Cell–Depleted Mice.

Investigative ophthalmology & visual science, 45(6), 1879-1886.

Tanaka, F., Yamaguchi, H., Ohta, M., Mashino, K., Sonoda, H., Sadanaga, N., Inoue, H.

& Mori, M. 2002. Intratumoral injection of dendritic cells after treatment of

anticancer drugs induces tumor-specific antitumor effect in vivo. International

Journal of Cancer, 101(3), 265-269.

Tang, Y., Xu, X., Guo, S., Zhang, C., Tang, Y., Tian, Y., Ni, B., Lu, B. & Wang, H.

2014. An Increased Abundance of Tumor-Infiltrating Regulatory T Cells Is

Correlated with the Progression and Prognosis of Pancreatic Ductal

Adenocarcinoma. PLoS ONE, 9(3), e91551.

Tivol, E. A., Borriello, F., Schweitzer, A. N., Lynch, W. P., Bluestone, J. A. & Sharpe,

A. H. 1995. Loss of CTLA-4 leads to massive lymphoproliferation and fatal

multiorgan tissue destruction, revealing a critical negative regulatory role of

CTLA-4. Immunity, 3(5), 541-547.

Topalian, S. L., Hodi, F. S., Brahmer, J. R., Gettinger, S. N., Smith, D. C., McDermott,

D. F., Powderly, J. D., Carvajal, R. D., Sosman, J. A., Atkins, M. B., Leming, P.

D., Spigel, D. R., Antonia, S. J., Horn, L., Drake, C. G., Pardoll, D. M., Chen,

L., Sharfman, W. H., Anders, R. A., Taube, J. M., McMiller, T. L., Xu, H.,

Korman, A. J., Jure-Kunkel, M., Agrawal, S., McDonald, D., Kollia, G. D.,

Gupta, A., Wigginton, J. M. & Sznol, M. 2012. Safety, Activity, and Immune

Correlates of Anti–PD-1 Antibody in Cancer. New England Journal of

Medicine, 366(26), 2443-2454.

Turk, M. J., Guevara-Patiño, J. A., Rizzuto, G. A., Engelhorn, M. E. & Houghton, A. N.

2004. Concomitant tumor immunity to a poorly immunogenic melanoma is

Page 261: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

233

prevented by regulatory T cells. The Journal of Experimental Medicine, 200(6),

771-782.

Twitty, C. G., Jensen, S. M., Hu, H.-M. & Fox, B. A. 2011. Tumor-derived

autophagosome vaccine: induction of cross-protective immune responses against

short-lived proteins through a p62-dependent mechanism. Clinical Cancer

Research, 17(20), 6467-6481.

Uno, T., Takeda, K., Kojima, Y., Yoshizawa, H., Akiba, H., Mittler, R. S., Gejyo, F.,

Okumura, K., Yagita, H. & Smyth, M. J. 2006. Eradication of established

tumors in mice by a combination antibody-based therapy. Nature Medicine,

12(6), 693-698.

van Bruggen, I., Nelson, D. J., Currie, A. J., Jackaman, C. & Robinson, B. W. S. 2005.

Intratumoral poly-N-acetyl glucosamine-based polymer matrix provokes a

prolonged local inflammatory response that, when combined with IL-2, induces

regression of malignant mesothelioma in a murine model. Journal of

Immunotherapy, 28(4), 359-367.

van Elsas, A., Hurwitz, A. A. & Allison, J. P. 1999. Combination Immunotherapy of

B16 Melanoma Using Anti–Cytotoxic T Lymphocyte–Associated Antigen 4

(CTLA-4) and Granulocyte/Macrophage Colony-Stimulating Factor (GM-CSF)-

Producing Vaccines Induces Rejection of Subcutaneous and Metastatic Tumors

Accompanied by Autoimmune Depigmentation. The Journal of Experimental

Medicine, 190(3), 355-366.

van Meerbeeck, J. P., Gaafar, R., Manegold, C., Van Klaveren, R. J., Van Marck, E. A.,

Vincent, M., Legrand, C., Bottomley, A., Debruyne, C. & Giaccone, G. 2005.

Randomized phase III study of cisplatin with or without raltitrexed in patients

with malignant pleural mesothelioma: an intergroup study of the European

Organisation for Research and Treatment of Cancer Lung Cancer Group and the

Page 262: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

234

National Cancer Institute of Canada. Journal of Clinical Oncology, 23(28),

6881-6889.

Van Pel, A. & Boon, T. 1982. Protection against a nonimmunogenic mouse leukemia by

an immunogenic variant obtained by mutagenesis. Proceedings of the National

Academy of Sciences, 79(15), 4718-4722.

Vanneman, M. & Dranoff, G. 2012. Combining immunotherapy and targeted therapies

in cancer treatment. Nature Reviews Cancer, 12(4), 237-251.

Veltman, J. D., Lambers, M. E., van Nimwegen, M., de Jong, S., Hendriks, R. W.,

Hoogsteden, H. C., Aerts, J. G. & Hegmans, J. P. 2010. Low-dose

cyclophosphamide synergizes with dendritic cell-based immunotherapy in

antitumor activity. BioMed Research International, 2010, 10.

Vermorken, J. B., Mesia, R., Rivera, F., Remenar, E., Kawecki, A., Rottey, S., Erfan, J.,

Zabolotnyy, D., Kienzer, H.-R. & Cupissol, D. 2008. Platinum-based

chemotherapy plus cetuximab in head and neck cancer. New England Journal of

Medicine, 359(11), 1116-1127.

Vignali, D. A., Collison, L. W. & Workman, C. J. 2008. How regulatory T cells work.

Nature Reviews Immunology, 8(7), 523-532.

Vogelzang, N. J., Rusthoven, J. J., Symanowski, J., Denham, C., Kaukel, E., Ruffie, P.,

Gatzemeier, U., Boyer, M., Emri, S. & Manegold, C. 2003. Phase III study of

pemetrexed in combination with cisplatin versus cisplatin alone in patients with

malignant pleural mesothelioma. Journal of Clinical Oncology, 21(14), 2636-

2644.

vom Berg, J., Vrohlings, M., Haller, S., Haimovici, A., Kulig, P., Sledzinska, A.,

Weller, M. & Becher, B. 2013. Intratumoral IL-12 combined with CTLA-4

blockade elicits T cell–mediated glioma rejection. The Journal of Experimental

Medicine, 210(13), 2803-2811.

Page 263: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

235

Wagner, J. C., Sleggs, C. A. & Marchand, P. 1960. Diffuse Pleural Mesothelioma and

Asbestos Exposure in the North Western Cape Province. British Journal of

Industrial Medicine, 17(4), 260-271.

Wakefield, L. M. & Roberts, A. B. 2002. TGF-β signaling: positive and negative effects

on tumorigenesis. Current opinion in genetics & development, 12(1), 22-29.

Waller, D. A. 2004. Malignant mesothelioma—British surgical strategies. Lung Cancer,

45, S81-S84.

Walunas, T. L., Lenschow, D. J., Bakker, C. Y., Linsley, P. S., Freeman, G. J., Green, J.

M., Thompson, C. B. & Bluestone, J. A. 1994. CTLA-4 can function as a

negative regulator of T cell activation. Immunity, 1(5), 405-413.

Wang, H. Y., Lee, D. A., Peng, G., Guo, Z., Li, Y., Kiniwa, Y., Shevach, E. M. &

Wang, R.-F. 2004. Tumor-Specific Human CD4+ Regulatory T Cells and Their

Ligands: Implications for Immunotherapy. Immunity, 20(1), 107-118.

Wang, Y., Deng, B., Tang, W., Liu, T. & Shen, X. 2013. TGF-β1 secreted by

hepatocellular carcinoma induces the expression of the Foxp3 gene and

suppresses antitumor immunity in the tumor microenvironment. Digestive

diseases and sciences, 58(6), 1644-1652.

Waterhouse, P., Penninger, J. M., Timms, E., Wakeham, A., Shahinian, A., Lee, K. P.,

Thompson, C. B., Griesser, H. & Mak, T. W. 1995. Lymphoproliferative

disorders with early lethality in mice deficient in CTLA-4. Science, 270(5238),

985-988.

Webster, I., Cochrane, J. & Burkhardt, K. 1982. Immunotherapy with BCG vaccine in

30 cases of mesothelioma. South African medical journal= Suid-Afrikaanse

tydskrif vir geneeskunde, 61(8), 277-278.

Page 264: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

236

Wei, P., Zhang, W., Yang, L.-S., Zhang, H.-S., Xu, X.-E., Jiang, Y.-H., Huang, F.-P. &

Shi, Q. 2013. Serum GFAP autoantibody as an ELISA-detectable glioma

marker. Tumor Biology, 34(4), 2283-2292.

Weiss, J. M., Subleski, J. J., Back, T., Chen, X., Watkins, S. K., Yagita, H., Sayers, T.

J., Murphy, W. J. & Wiltrout, R. H. 2014. Regulatory T Cells and Myeloid-

Derived Suppressor Cells in the Tumor Microenvironment Undergo Fas-

Dependent Cell Death during IL-2/αCD40 Therapy. The Journal of

Immunology, 192(12), 5821-5829.

Whiteside, T. 2008. The tumor microenvironment and its role in promoting tumor

growth. Oncogene, 27(45), 5904-5912.

Whiteside, T. L. 2013. Immune modulation of T-cell and NK (natural killer) cell

activities by TEXs (tumour-derived exosomes). Biochemical Society

Transactions, 41(1), 245-251.

Whiteside, T. L. 2014. Regulatory T cell subsets in human cancer: are they regulating

for or against tumor progression? Cancer Immunology, Immunotherapy, 63(1),

67-72.

Willett, C. G., Boucher, Y., Di Tomaso, E., Duda, D. G., Munn, L. L., Tong, R. T.,

Chung, D. C., Sahani, D. V., Kalva, S. P. & Kozin, S. V. 2004. Direct evidence

that the VEGF-specific antibody bevacizumab has antivascular effects in human

rectal cancer. Nature Medicine, 10(2), 145-147.

Willimsky, G. & Blankenstein, T. 2005. Sporadic immunogenic tumours avoid

destruction by inducing T-cell tolerance. Nature, 437(7055), 141-146.

Wing, K., Onishi, Y., Prieto-Martin, P., Yamaguchi, T., Miyara, M., Fehervari, Z.,

Nomura, T. & Sakaguchi, S. 2008. CTLA-4 control over Foxp3+ regulatory T

cell function. Science, 322(5899), 271-275.

Page 265: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

237

Wolchok, J. D., Kluger, H., Callahan, M. K., Postow, M. A., Rizvi, N. A., Lesokhin, A.

M., Segal, N. H., Ariyan, C. E., Gordon, R.-A. & Reed, K. 2013. Nivolumab

plus ipilimumab in advanced melanoma. New England Journal of Medicine,

369, 122-133.

Wolchok, J. D., Neyns, B., Linette, G., Negrier, S., Lutzky, J., Thomas, L., Waterfield,

W., Schadendorf, D., Smylie, M. & Guthrie, T. 2010. Ipilimumab monotherapy

in patients with pretreated advanced melanoma: a randomised, double-blind,

multicentre, phase 2, dose-ranging study. The lancet oncology, 11(2), 155-164.

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(2), 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(12), 4766-4772.

Wu, L., Yun, Z., Tagawa, T., Rey-McIntyre, K., Anraku, M. & de Perrot, M. 2011.

Tumor cell repopulation between cycles of chemotherapy is inhibited by

regulatory T-cell depletion in a murine mesothelioma model. Journal of

Thoracic Oncology, 6(9), 1578-1586.

Xiang, X., Poliakov, A., Liu, C., Liu, Y., Deng, Z. b., Wang, J., Cheng, Z., Shah, S. V.,

Wang, G. J. & Zhang, L. 2009. Induction of myeloid‐derived suppressor cells by

tumor exosomes. International journal of cancer, 124(11), 2621-2633.

Yanaba, K., Bouaziz, J.-D., Haas, K. M., Poe, J. C., Fujimoto, M. & Tedder, T. F. 2008.

A Regulatory B Cell Subset with a Unique CD1dhiCD5+ Phenotype Controls T

Cell-Dependent Inflammatory Responses. Immunity, 28(5), 639-650.

Page 266: Eradication of established murine mesothelioma tumours by … › files › 4904871 › ... · Eradication of established murine mesothelioma tumours by combined immunotherapy . Shruti

238

Yang, H., Testa, J. R. & Carbone, M. 2008. Mesothelioma epidemiology,

carcinogenesis, and pathogenesis. Current treatment options in oncology, 9(2-3),

147-157.

Yang, J. C., Haworth, L., Sherry, R. M., Hwu, P., Schwartzentruber, D. J., Topalian, S.

L., Steinberg, S. M., Chen, H. X. & Rosenberg, S. A. 2003. A randomized trial

of bevacizumab, an anti–vascular endothelial growth factor antibody, for

metastatic renal cancer. New England Journal of Medicine, 349(5), 427-434.

Yang, L., Pang, Y. & Moses, H. L. 2010. TGF-β and immune cells: an important

regulatory axis in the tumor microenvironment and progression. Trends in

Immunology, 31(6), 220-227.

Yingling, J. M., Blanchard, K. L. & Sawyer, J. S. 2004. Development of TGF-β

signalling inhibitors for cancer therapy. Nature reviews Drug discovery, 3(12),

1011-1022.

Zhou, G., Drake, C. G. & Levitsky, H. I. 2006. Amplification of tumor-specific

regulatory T cells following therapeutic cancer vaccines. Blood, 107(2), 628-

636.

Zou, W. 2006. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev

Immunol, 6(4), 295-307.