autologous hematopoietic stem cell …...ii autologous hematopoietic stem cell transplantation for...

177
Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation By Hassan Sadozai A thesis submitted in conformity with the requirements for the degree of Master of Science Graduate Department of the Institute of Medical Science University of Toronto © Copyright by Hassan Sadozai 2016

Upload: others

Post on 31-May-2020

17 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

Autologous Hematopoietic Stem Cell Transplantation for Tolerance

Induction in a Mouse Model of Solid Organ Transplantation

By

Hassan Sadozai

A thesis submitted in conformity with the requirements

for the degree of Master of Science

Graduate Department of the Institute of Medical Science

University of Toronto

© Copyright by Hassan Sadozai 2016

Page 2: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

ii

Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a

Mouse Model of Solid Organ Transplantation

Hassan Sadozai

Masters of Science, 2016

Institute of Medical Science

University of Toronto

ABSTRACT

The need for long term immunosuppression negatively impacts long term survival

and quality of life for solid organ transplant patients. One solution for this would be

establishment of immune tolerance. Autologous HSCT has been used clinically for the

restoration of self-tolerance in autoimmune disease. We tested whether autologous

HSCT and a short treatment with rapamycin could result in tolerance to fully MHC-

mismatched allografts. Mice that received HSCT and a short course of the mTORi

rapamycin demonstrated significantly prolonged allograft survival compared to

untreated and rapamycin-only treated controls. Immunologic studies showed that

HSCT-treated mice displayed active immune regulation as demonstrated by a primary

MLR response, markedly diminished donor specific antibody levels and significantly

higher frequencies of CD4+CD25+FOXP3+ Tregs. These data provide a rationale for

human clinical trials to examine the ability of autologous stem cell transplantation to

induce tolerance in liver transplant patients (ASCOTT) which are now ongoing.

Page 3: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

iii

Acknowledgments

I would like to express sincere gratitude for everyone who has helped me

throughout my Master’s degree. Their invaluable contributions have made this work

possible and enriched my graduate experience.

I would like to humbly thank my supervisor Dr. Gary Levy for his support and

guidance. His input has greatly honed my ability to think critically and design well-

thought out experiments. My experience in his lab has also imbued me with a love for

immunology, a subject I hope to pursue in my future academic career. I would also like

to thank members of my advisory committee, Drs. Reginald Gorczynski, Li Zhang and

Mark Minden. Their support and scientific input has been indispensable to the progress

and completion of this work. In particular, I would like to thank Reg Gorczynski for his

rapier wit, invaluable assistance and profound insights into not only my work but also

the entire field of immunology. I would also like to thank our collaborators for providing

their expertise and assistance: Dr. Harold Atkins (Ottawa Hospital Research Institute),

Dr. Oyedele Adeyi and Dr. Clinton Robbins. Dr. Adeyi’s input and direction has

tremendously enriched this work.

I wish to express my sincere gratitude to all members of the Levy Lab. I am very

thankful to Dr. Andrzej Chruscinski, whose tireless efforts and excellent scientific

guidance have significantly improved this thesis. I am also sincerely indebted to Dr. Wei

He, whose helpful demeanour and superb surgical expertise, made this work possible. I

am also very thankful to Jianhua Zhang for her technical assistance and cheerful

disposition. I am very grateful to my peers and cherished friends, Vanessa Rojas

Page 4: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

iv

Luengas, Kaveh Farrokhi, Mani Mian and Angela Li for their encouragement, support

and technical assistance without which, this work would not have been possible. I am

also thankful to all current and past members of the Levy Lab including Dr. Nazia

Selzner, Dr. Peter Urbanellis, Dr. Agata Bartczak, Anna Cocco, Kai Yu, Charmaine

Beal, Olga Luft, Albert Nguyen and Justin Manuel. I am very thankful to Andre Siegel for

his assistance and his advice. I also wish to express gratitude to members of the

Gorczynski Lab, Dr. Ismat Khatri, Camila Balgobin, Fang Zhu and Anna Curry, whose

help and kindness significantly enriched my graduate experience. I am also indebted to

all the talented summer students I have had the opportunity to train and befriend; Celine

Yoo, Dario Ferri, Conan Chua and Nancy Qin. I would also like to thank my friends Alya

Bhimji, Sherine Ensan, Natalie Simard and Arian Khandani for their scientific insights,

encouragement and invaluable support. I am very thankful to my friend Novina Wong for

her continual support and camaraderie. Finally, my sincerest gratitude goes out to the

IMS department for ensuring that this graduate degree will be a most memorable one.

In particular, I would like to thank Dr. Howard Mount for his wise counsel and helpful

advice over the years.

Finally, I would like to thank my friends and extended family for their inveterate

support of my career aspirations and academic pursuits. I would like to thank my

parents and my sister who continue to inspire me with excellence in their respective

fields. I am also thankful to other members of my family who have encouraged me in my

career. In particular, I am thankful to my bosom friend Dorsa Saeidi, whose loving

support and profound insights push me to work diligently and be a better person. Merci

à tous.

Page 5: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

v

Contributions

I would like to thank the following individuals for their contributions towards this thesis

Dr. Andrzej Chruscinski – Experiment design and data collection

Dr. Reginald Gorczynski – Experiment design and scientific input

Dr. Oyedele Adeyi – Selection of pictures for histology, pathology scores and

morphometric analyses

Dr. William (Wei) He – Murine heterotopic heart transplants and data collection

UHN Flow Cytometry Facility – Sorting of labelled LSK cells

UHN STTARR Facility – Morphometric analyses and slide scanning

UHN Animal Resource Center – Tail vein injections, murine handling and housing,

animal ordering

Page 6: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

vi

Table of Contents

ACKNOWLEDGMENTS ........................................................................................................................................... III

CONTRIBUTIONS .................................................................................................................................................... V

TABLE OF CONTENTS ............................................................................................................................................. VI

LIST OF TABLES ..................................................................................................................................................... IX

LIST OF FIGURES ..................................................................................................................................................... X

ABBREVIATIONS ................................................................................................................................................... XI

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

1.1 SOLID ORGAN TRANSPLANTATION .......................................................................................................................... 1

1.1.1 Current Status of Solid Organ Transplantation .................................................................................... 1

1.2 THE IMMUNE SYSTEM .......................................................................................................................................... 3

1.2.1 Immunology of graft rejection .............................................................................................................. 5

1.2.1.1 Overview ..................................................................................................................................................... 5

1.2.1.2 Major and Minor Histocompatibility Antigens ........................................................................................... 6

1.2.1.3 Mechanisms of Allo-Recognition ................................................................................................................ 7

1.2.1.4 Types of Graft Rejection .............................................................................................................................. 9

1.2.1.4.1 Hyperacute rejection ........................................................................................................................... 10

1.2.1.4.2 Acute Rejection .................................................................................................................................... 11

1.2.1.4.3 Chronic Rejection ................................................................................................................................. 17

1.2.2 Prevention of Rejection ....................................................................................................................... 19

1.2.2.1 Immunosuppression .................................................................................................................................. 19

1.2.2.2 Induction Agents ....................................................................................................................................... 20

1.2.2.3 De-sensitization therapies ........................................................................................................................ 21

1.2.2.4 Anti-metabolites ....................................................................................................................................... 22

1.2.2.5 Corticosteroids .......................................................................................................................................... 24

1.2.2.6 Calcineurin Inhibitors (CNI) ....................................................................................................................... 25

1.2.2.7 mTOR inhibitors (mTORi) .......................................................................................................................... 26

1.2.2.8 Novel therapies in the pipeline ................................................................................................................. 30

1.3 IMMUNE TOLERANCE .......................................................................................................................................... 31

1.3.1 Overview.............................................................................................................................................. 31

1.3.2 B cell tolerance .................................................................................................................................... 33

1.3.3 Central T cell tolerance ....................................................................................................................... 34

1.3.4 Peripheral T cell tolerance .................................................................................................................. 36

Page 7: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

vii

1.3.4.1 Ignorance ................................................................................................................................................... 37

1.3.4.2 Anergy ....................................................................................................................................................... 38

1.3.4.3 Activation-induced cell death ................................................................................................................... 40

1.3.5 Suppression by immunomodulatory cells ........................................................................................... 41

1.3.5.1 Dendritic cells ............................................................................................................................................ 42

1.3.5.2 Regulatory B cells ...................................................................................................................................... 43

1.3.5.3 Regulatory T cells ...................................................................................................................................... 45

1.3.5.3.1 CD4+CD25+FOXP3+ Tregs ...................................................................................................................... 46

1.3.5.3.2 Mechanisms of Treg suppression ........................................................................................................ 48

1.4 STRATEGIES TO INDUCE TRANSPLANT TOLERANCE ................................................................................................... 51

1.4.1 Overview.............................................................................................................................................. 51

1.4.2 Co-stimulatory blockade ..................................................................................................................... 52

1.4.3 Induction or administration of Tregs .................................................................................................. 54

1.4.4 Hematopoietic stem cell transplantation ........................................................................................... 56

1.4.4.1 Hematopoietic stem cells .......................................................................................................................... 57

1.4.4.2 Allogeneic HSCT and mixed chimerism ..................................................................................................... 61

1.4.4.3 Autologous HSCT ....................................................................................................................................... 64

HYPOTHESES AND AIMS ............................................................................................................................. 69

MATERIALS AND METHODS ........................................................................................................................ 70

3.1 MICE ............................................................................................................................................................... 70

3.2 HETEROTOPIC CARDIAC TRANSPLANTATION .............................................................................................................. 70

3.3 FLOW CYTOMETRY .............................................................................................................................................. 72

3.4 PURIFICATION OF LSK CELLS ................................................................................................................................. 73

3.5 HEMATOPOIETIC STEM CELL TRANSPLANTATION ........................................................................................................ 74

3.6 TREATMENT GROUPS........................................................................................................................................... 75

3.7 HISTOLOGY AND IMMUNOHISTOCHEMISTRY ............................................................................................................. 77

3.8 MIXED LYMPHOCYTE REACTION ............................................................................................................................. 77

3.9 FLOW CYTOMETRY FOR DONOR-SPECIFIC ANTIBODIES (DSA) ....................................................................................... 78

3.10 STATISTICS ................................................................................................................................................... 79

RESULTS ...................................................................................................................................................... 80

4.1 PURIFICATION OF LSK CELLS AND DOSE SELECTION .................................................................................................... 80

4.2 HSCT WITH LSK CELLS RESULTS IN FULL HEMATOPOIETIC RECONSTITUTION AND IS NOT IMPAIRED BY RAPAMYCIN TREATMENT. 82

4.3 HSCT PROMOTES LONG-TERM CARDIAC ALLOGRAFT SURVIVAL .................................................................................... 86

4.4 HSCT-TREATED MICE MAINTAIN PRIMARY IMMUNE RESPONSE IN VITRO ........................................................................ 94

4.5 HSCT TREATMENT MARKEDLY DIMINISHES DSA AND EXPANDS TREGS .......................................................................... 96

Page 8: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

viii

DISCUSSION ................................................................................................................................................ 98

CONCLUSIONS .......................................................................................................................................... 113

FUTURE DIRECTIONS ................................................................................................................................. 114

REFERENCES ............................................................................................................................................. 117

COPYRIGHT ACKNOWLEDGEMENTS ................................................................................................................... 163

Page 9: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

ix

List of Tables

Table 1-1 General Comparison of Innate and Adaptive Immunity ....................................... 5

Table 1-2 Treg effector molecules ........................................................................................... 50

Table 1-3 Overview of Autologous versus Allogeneic HSCT .............................................. 57

Table 3-1 Heterotopic heart transplantation treatment groups ........................................... 75

Table 4-1 Complete blood counts at Day 100 post-HSCT (data are shown as mean

±SEM) .......................................................................................................................................... 85

Page 10: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

x

List of Figures

Figure 1-1 Schematic of co-stimulatory and co-inhibitory receptors .................................. 14

Figure 1-2 Treg-mediated mechanisms of immunosuppression ........................................ 49

Figure 1-3 Schematic of mouse and human hematopoeitic development ........................ 60

Figure 3-1 Schematic of HSCT treatment for tolerance induction in cardiac

allotransplant model ................................................................................................................... 76

Figure 4-1 Isolation of LSK cells .............................................................................................. 81

Figure 4-2 LSK cell dose selection .......................................................................................... 82

Figure 4-3 Rapamycin does not impair long-term hematopoietic reconstitution after

HSCT with LSK cells .................................................................................................................. 84

Figure 4-4 HSCT prolongs cardiac allograft survival ............................................................ 90

Figure 4-5 HSCT treatment preserves cardiac allograft morphology ................................ 91

Figure 4-6 Representative immunoperoxidase staining of graft infiltrating cells.............. 92

Figure 4-7 Morphometric analyses of immunoperoxidase staining .................................... 93

Figure 4-8 Lymphocytes from HSCT treated mice maintain primary immune response in

vitro ............................................................................................................................................... 95

Figure 4-9 Splenic Treg and DSA quantitation ...................................................................... 97

Page 11: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

xi

Abbreviations

Ab Antibody

ACR Acute cellular rejection

Ag Antigen

AHR Acute humoural rejection

AICD Activation induced cell death

ALPS Autoimmune lymphoproliferative syndrome

AP-1 Activator protein 1

APC Allophycocyanin

APC Antigen presenting cell

APECED Autoimmune polyendocrinopathy candidiasis ectodermal dystrophy

BCR B cell receptor

CAV Chronic allograft vasculopathy

CD Cluster of differentiation

cDC conventional DC

CNI Calcineurin inhibitor

CR Chronic rejection

CsA Cyclosporine A

CTL Cytotoxic T lymphocyte

CTLA-4 Cytotoxic T lymphocyte antigen-4

DAMP Damage associated molecular pattern

DC Dendritic cell

DN Double negative

DP Double positive

DSA Donor specific antibody

Page 12: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

xii

Fc Constant region fragment

FcR Fc receptor

FITC Fluoroscein isothiocyanate

FKBP12 FK-506 binding protein 1A, 12 kDa

FOXP3 Forkhead box P3

H&E Hematoxylin and eosin

HAR Hyperacute rejection

Hct Hematocrit

HSCT Hematopoietic stem cell transplantation

IDO Indoleamine 2,3-dioxygenase

IFNγ Interferon gamma

IL Interleukin

IPEX Immune dysfunction, polyendocrinopathy, enteropathy X-linked syndrome

iTreg Induced (in vitro) regulatory T cells

LFA-1 Lymphocyte function associated antigen-1

LFA-3 Lymphocyte function associated antigen-3

LLPC Long-lived plasma cell

LSK Lin-Sca1+c-kit+ stem cells

mAb Monoclonal antibody

MLR Mixed lymphocyte reaction

MS Multiple sclerosis

mTOR Mammalian target of rapamycin

mTORi mTOR inhibitor

NFAT Nuclear factor of activated T cells

NF-κB Nuclear factor kappa B

Page 13: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

xiii

NK Natural killer cells

NKT Natural killer T cells

PAMP Pathogen associated molecular pattern

pDC plasmacytoid DC

PE Phycoerythrin

PMN Polymorphonuclear cells (neutrophils)

pTreg Peripheral (induced) regulatory T cells

Rapa Rapamycin

SLE Systemic lupus erythematosus

SOT Solid organ transplantation

TCR T cell receptor

TEC Thymic epithelial cell

Teff Effector T cells

TGF-β Transforming growth factor beta

TNF Tissue necrosis factor

TolDC Tolerogenic DC

Treg T regulatory cells

tTreg Thymic derived (natural) regulatory T cells

TxHSCT HSCT group - BALB/C allograft in C57BL/6 recipient with HSCT and rapamycin

TxRapa Rapamycin group – BALB/C allograft in C57BL/6 recipients with rapamycin

TxRej Rejecting group – BALB/C allograft in C57BL/6 recipients with no treatment

TxSyn Syngeneic group – C57BL/6 isograft in C57BL/6 recipients with no treatment

WBC White blood cell

Page 14: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

1

Introduction

1.1 Solid Organ Transplantation

1.1.1 Current Status of Solid Organ Transplantation

Currently, solid organ transplantation (SOT) is the most effective therapy for

patients with end-stage organ failure.1 The first successful solid organ transplant was a

kidney transplant from one identical twin to another by Joseph Murray in 1954.2 This

was followed shortly by the first liver transplant by Thomas Starzl in 1963 and the first

heart transplant in 1967.3,4 The first successful immunosuppressive drug regime

consisting of the anti-proliferative azathioprine, the corticosteroid prednisone and anti-

lymphocyte globulin (ALG) managed to improve 1 year graft survival to between 40 and

50% in the early 1960s.1,5 In the decades that followed there were significant advances

both in immunosuppressive drugs, as well as, surgical and organ preservation

techniques that allowed for improved graft survival. The modern era of

immunosuppression (IS) was ushered in by the discovery of the calcineurin inhibitor

(CNI), cyclosporine (CsA), which increased 1-year graft survival rates to between 70-

80%.6,7 Currently transplantation of kidney, liver, heart, lung and pancreas and small

bowel are routinely performed in clinical medicine.8,9 In past decade, pancreatic islet

cell transplantation has also being studied for treating a subset of Type-1 diabetes

mellitus patients.10 The primary success of SOT may be demonstrated by increased

survival benefit in terms of life-years over alternative therapies. In a recent study by

Page 15: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

2

Rana et al., it was demonstrated that over 2 million life-years were saved by SOT over

a 25-year study period from 1987 to 2012.11 Furthermore, for certain end-stage organ

diseases, transplantation is far more cost-effective than existing alternative therapies. In

the context of end-stage renal disease, transplantation can result in $300,000 direct

savings per patient over 5 years compared with dialysis.12

In Canada, the number of transplants increased from 2,093 in 2009 to 2,356 in

2014.13 The majority (1,430) of these were kidney transplants followed by liver (537) but

over 4,514 patients were still on the waiting list at the end of 2014.13 While the gap

between donated organs and potential recipients continues to increase, the long-term

maintenance of graft function is also a fundamental obstacle in transplantation

medicine.14 In a US cohort of organ recipients studied from 1989 to 2008, it was shown

that while 1-year graft survival rates were above 80% for nearly all organ types, graft

survival falls below 60% for heart, lung, liver and intestine graft survival at the 10-year

mark.15 Notwithstanding, age-related differences in graft outcomes, much of the chronic

graft loss currently observed can be attributed to the adverse effects of long-term

immunosuppression (further discussed in 1.2.3). These include nephrotoxicity,

cardiovascular disease, de novo diabetes, opportunistic infections, recurrence of

original disease (e.g.hepatitis C virus) and cancer.15–19 Thus, the concept of

immunosuppression-free “immune tolerance” remains the holy grail of transplantation

medicine and an important area of research. Immune tolerance denotes an

immunological state whereby organ-specific unresponsiveness may be achieved

without the need for chronic immunosuppression while retaining a functional immune

system for protective immunity against pathogens.14,20 In the clinic, limited numbers of

Page 16: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

3

patients have achieved “operational tolerance”, whereby the graft displays no signs of

immune rejection for at least 1 year in the absence of any immunosuppressive drugs.21

In order to understand how immune tolerance may be achieved, knowledge of the

complex immunobiology of graft rejection is necessary.

1.2 The Immune System

A detailed description of the immune system will not be presented as this is not

the main focus of this thesis. I refer readers to the following for a more detailed

description of advances of our understanding of the immune system.22–25 The immune

system is a heterogeneous network of organs, cells and molecules that functions to

distinguish self from non-self.22 This allows for recognition of foreign antigens while

retaining tolerance to self-tissue. The immune system is classically partitioned into two

separate components based primarily, on the pace and specificity of the response

(Table 1-1).24,26 The innate immune system comprises of physical and mechanical

barriers, such as the skin, as well as cells expressing a limited number of pattern-

recognition receptors (PRRs) that are encoded in the germ-line, and recognize

pathogen-associated molecular patterns (PAMPs). These PAMPs include viral nucleic

acids as well as, molecules found on bacterial and fungal cell walls.27–29 Thus, the

innate immune system furnishes the immediate host response to an invading pathogen.

There is continuous cross-talk between the adaptive and innate arms of the immune

response and research over the past two decades has demonstrated a vital role for the

innate immune system in the modulation of the adaptive immune response.30,31

Page 17: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

4

The adaptive immune system is capable of a protracted response and eventual

clearance of a pathogen. It is able to do so by generating an immense variety of cell

surface receptors that can recognize and react to a plethora of antigens.32 The adaptive

immune system mediates its primary functions through two types of immune cells, T

and B lymphocytes.33 Both T and B cells have evolved the capacity to generate a

diverse repertoire of antigen receptor genes (> 108) through recombination of a limited

number of gene segments.34 While most self-reactive T and B cells are eliminated

through central and peripheral mechanisms reviewed in Chapter 1.3, some cells

manage to escape these mechanisms. Unchecked self-reactivity results in the wide

range of autoimmune pathologies observed in humans. Conversely, the large repertoire

of antigens recognized by the immune system poses a significant barrier to

transplantation of foreign tissue such as in allogeneic bone marrow or solid organ

transplantation.

Page 18: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

5

Table 1-1 General Comparison of Innate and Adaptive Immunity

1.2.1 Immunology of graft rejection

1.2.1.1 Overview

A transplanted organ elicits a broad acting immune response involving

components of both the innate and adaptive immune system. Tissue damage as a

result of surgery and ischemia-reperfusion injury generate pro-inflammatory signals

such as damage-associated molecular patterns (DAMPs), that are recognized by the

INNATE IMMUNITY ADAPTIVE IMMUNITY

Encoding Receptors Germ-line Somatic

Receptor Recombination No Yes

Receptor Target Conserved (Invariable) Large variety of antigen specific receptors

Memory No Yes

Onset of Response Fast Slow

Cellular Components

Neutrophils Eosinophils Basophils Mast Cells

Macrophages Natural Killer Cells

T cells B cells

Soluble Components Complement

Cytokines Antibodies Cytokines

Page 19: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

6

PRRs of innate immune cells as well as receptors of the complement system.35–37 The

innate immune response is non-specific but it serves to prime the adaptive arm of the

immune system which, if left unchecked, will ultimately result in graft loss. The antigen-

specific receptors of T and B cells can recognize various antigens in a transplanted

organ such as minor histocompatibility antigens, ABO blood group antigens and

endothelial cell antigens.36,38 However, the primary response is targeted to the major

histocompatibility complex (MHC) group of antigens.39,40

1.2.1.2 Major and Minor Histocompatibility Antigens

The MHC class I and class II antigens are structurally similar molecules that are

involved in antigen presentation to CD4+ (TH – helper T) and CD8+ (Tc – cytotoxic T )

cells.41,42 T cell activation is dependent upon recognition of antigen in the context of the

peptide:MHC (p:MHC) complex.41,43 MHC class I molecules are expressed in the

surface of all nucleated cells and present peptides of endogenous cytosolic origin

including viral and bacterial peptides in infected cells, to CD8+ T cells.44 MHC class II

molecules are expressed by cells of the thymic epithelium (TECs) and by professional

antigen presenting cells (APCs) such as dendritic cells (DCs), macrophages and B cells

and express peptides of extracellular origin which are degraded via the endocytic

pathway.45. Activated human T cells have also been shown to express MHC class II and

a majority of cell types can be induced to express MHC class II by interferon γ (IFNγ)

through the class II transactivator gene (CIITA).42,46

MHC antigens were discovered in mice by Peter Gorer and later it was George

Snell who described the multi-locus nature of MHC genes.47,48 Both MHC Class I and

Page 20: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

7

Class II are encoded for by a set of closely related genes that are located on

chromosome 6 in humans and chromosome 17 in mice.49 The human MHC is termed

HLA (human leukocyte antigen) and is extremely polymorphic. HLA Class I molecules

are primarily encoded by the HLA-A, HLA-B and HLA-C loci. Additional loci HLA-E,

HLA-F, HLA-G have also been recently described and their exact functions are not well

described.50 The 3 primary HLA Class II loci are HLA-DQ, HLA-DP and HLA-DR.50 In

mice, the MHC genes are termed H-2 genes. In mice the three Class I loci are termed

K, D and L while Class II genes are located in the I region and are usually sub-classified

as I-A and I-E.48 In placental mammals, a third region of high gene-density termed MHC

Class III is present but it codes for molecules which are involved in the inflammatory

response and structurally different from the Class I and II molecules.51 HLA genes

display Mendelian inheritance and are expressed co-dominantly with the entire

complement of HLA genes derived from one parents termed the “haplotype”.52 In

laboratory strains of inbred mice, all mice are homozygous for the same haplotype

represented by superscript letters such as H-2b, H-2d etc.48 Finally, over 50 minor

histocompatibility (MiH) antigens have been described. These are T cell epitopes

derived from several different proteins encoded on various chromosomes and are

implicated in anti-donor immune responses in organ transplantation as well as HLA-

matched hematopoietic stem cell transplantation.53,54

1.2.1.3 Mechanisms of Allo-Recognition

Due to the central role of T cells in orchestrating graft rejection, this section will

focus on the mechanisms through which T cells recognize alloantigen. T cell allo-

Page 21: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

8

recognition is proposed to occur through 2 well-described pathways. In the “direct”

pathway, recipient T cells are capable of directly binding to non self-peptide:MHC

complexes expressed on donor APCs that are transferred along with the allograft.55,56

As APCs express both MHC class I and II, both CD4+ and CD8+ can be activated by the

direct pathway. This observation is unique to transplantation and defies the canonical

understanding that T cells are restricted to recognition of only self-MHC during thymic

education.49 However, based on the principle of molecular mimicry, memory T cells

reactive for particular microbes may cross-react with allogeneic MHC directly, a concept

known as “heterologous immunity”.57 In a recent study by Macedo et al., it was shown

that virus-specific T cells contributed significantly to the alloreactive response.58 This

serves to expand the pool of potentially alloreactive T cells and in fact, studies have

shown that up to 10% of the T cell repertoire can is involved in the alloresponse to a

transplanted organ.59 In mice, the direct pathway of allo-recognition has been

extensively studied.60,61 In particular, the study by Pietra et al. demonstrated that even

in class II deficient, and recombination activating gene (RAG) deficient mice, injection of

wild-type CD4+ cells could mediate acute rejection in the absence of host MHC class

II.60 Hence, the direct allo-recognition pathway is a crucial mediator of the alloresponse,

and is believed to predominate early after transplantation prior to the depletion of donor

APCs by maintenance immunosuppression and recipient natural killer (NK) cells.62,63

The “indirect” pathway of allo-recognition occurs when alloantigen (mostly MHC

alloantigen) is processed and presented on recipient MHC complexes expressed host

APCs. This pathway alone was demonstrated to mediate rejection in preclinical studies

where using MHC class II deficient mice as donors resulted in graft rejection.64,65

Page 22: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

9

However, graft rejection mediated through the indirect pathway is believed to be slower

and is deemed more relevant for chronic allograft rejection.66 The indirect pathway is

important for the production of alloantibody responses, in particular, the generation of

long-lived plasma cells (LLPCs) that reside in the bone marrow.62 Moreover, studies in

murine models of regulatory T cell (Treg)-mediated transplant tolerance have also

suggested that these Treg are activated through the indirect pathway.67,68 Finally, a third

pathway of “semi-direct” allo-recognition has also been proposed.69 This concept,

known as cross-dressing or cross-priming, may occur when recipient APC acquire and

present intact donor MHC and bound peptides on their surface , thereby activating both

CD4+ and CD8+ immunity.70 A recent study has demonstrated strong evidence for this

pathway in a murine model where recipient DCs were able to present intact donor MHC

class I to directly recruit CD8+ cells and mediate an acute rejection response.71

However, study is required to further delineate this model.

1.2.1.4 Types of Graft Rejection

Currently, histological analysis of the allograft biopsy serves as the best evidence

of rejection.72 The clinical stages of rejection are classified mostly on the

pathophysiological changes, and the pace of rejection. The three major classifications

are hyperacute, acute and chronic rejection.73 Various immune mechanisms are

involved in each stage of rejection.

Page 23: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

10

1.2.1.4.1 Hyperacute rejection

Hyperacute rejection occurs within minutes to hours of the transplant and is a

consequence of pre-formed donor-specific alloantibodies (DSA) towards donor MHC,

graft endothelium or ABO blood groups as a result of a previously rejected graft,

pregnancy or blood transfusion.74 The DSA bind immediately to the graft and result in

activation and binding of complement subunits resulting in cell destruction via the

membrane attack complex.75 Additional complement byproducts such as C3a and C5a

are also potent chemokines.76 Antibody-binding can also result in injury by NK cells and

macrophages through antibody-dependent cell-mediated cytotoxicity (ADCC).77

Antibody deposition on endothelial cells also causes them to increase expression of

Von Willebrand factor (vWF) and the adhesion molecule p-selectin (CD62P). The

transplanted organ therefore suffers from endothelial necrosis, thrombosis and

increased local coagulation with some cases even resulting in hemorrhage as a result of

severely compromised vascular integrity.72,78 The organ ceases to function and nearly

all cases require excision of the organ.78

Due to the advent of HLA antibody flow crossmatch (FXM) platforms and

selection of ABO compatible donors, hyperacute rejection is a rare occurrence in the

clinic (<1%).73,74 Several treatments have been used in an attempt to reduce DSA in

pre-sensitized transplant patients. These include IVIG, plasmapheresis, the B-cell

depleting mAb rituximab, and the proteasome inhibitor Bortezomib but their success is

limited.78–81 In experimental models, hyperacute rejection is observed primarily in

xenograft models (transplants between different species) as all mammalian species

have pre-formed xenoreactive antibodies.82,83 Xenograft studies are of particular interest

Page 24: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

11

as successful xenografting could provide a limitless supply of organs for clinical use.

Hence, research is being conducted on the use of genetically modified pigs lacking

epitopes (such as α-1,3-Gal) that are recognized by human xenoreactive antibodies.84

1.2.1.4.2 Acute Rejection

In the absence of predisposing factors for hyperacute rejection, the allograft is

still at risk of acute rejection which can occur anywhere in the range of 1 week to 1 year

post-transplantation and has an incidence of between 10 to 20% depending on organ

type and therapeutic regimens.74,85 Since acute allograft rejection is mediated by the

humoural and the cellular components of the immune system, each of these now

constitute a distinct sub-classification of acute rejection and warrant further discussion.

Acute humoural rejection

Acute humoural rejection-AHR (also known as acute vascular rejection), is

mediated through the emergence of de novo or pre-formed DSA.86 Occurring within

days to weeks post-transplantation, it contributes significantly to clinical episodes of

acute rejection.86 DSA are primarily formed towards HLA class I, and to a lesser extent

class II, but can also be directed towards autoantigens, endothelium, ABO antigens and

miH.86,87 In the context of transplantation, the interaction of alloantigen with the B cell

receptor (BCR) results in the activation of B cells and the production of IgM as the initial

anti-donor immunoglobulin (Ig).88 B cells can also receive activation signals from binding

of complement subunits C3b and C3d to co-receptors (CD21) on their surface.89 The

Page 25: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

12

internalized alloantigen is then presented in the context of MHC class II of activated B

cells. In the secondary lymphoid organs, B cells undergo somatic hypermutation and

class-switch recombination which results in the production of higher affinity IgG

alloantibodies.88 As mentioned previously, CD4+ cells activated through the indirect

pathway of allo-recognition play an important role in activating and expanding the B cell

alloresponse through cognate TCR:BCR interactions and co-stimulation through T cell

bound CD40L (CD154) interacting with CD40 on B cells.90,91 In particular, T follicular

helper (TFH) cells are associated with B cells in the germinal center and play an

important role in the formation of long-lived plasma cells (LLPC) as described

previously.92 As a result of these interactions, AHR is often clinically associated with

acute cellular rejection. When AHR alone is implicated, the histopathological findings

are interstitial edema, thrombosis and neutrophil infiltration with absence of

mononuclear cell infiltrates.86,93 C4d, a byproduct of complement C4b, is also found

associated with acute humoural rejection, particularly in kidney transplantation.94,95

Treatment of AHR involves the previously described therapeutics (IVIG,

plasmapheresis) as well as B cell-targeted therapies (rituximab, bortezomib). Recently

eculizumab, a mAb against complement subunit C5, has been shown to reverse some

cases of AHR in kidney patients but further clinical study of its utility is required. In a few

patients who are refractory to other forms of treatment, splenectomy has been utilized

as a successful therapy although this renders them susceptible to life-long

immunodeficiency.96

Page 26: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

13

Acute cellular rejection

Acute cellular rejection (ACR) results primarily from the T cell-mediated immune

response to alloantigen. In the early days of organ transplantation, ACR was the major

cause of graft loss.1,14 As previously described, during acute allograft rejection, both

CD4+ and CD8+ allorecognition occurs through either a semi-direct or direct pathway.

However, binding of TCR to MHC:peptide complexes is not sufficient for activation.

Currently, T cell activation is purported to occur through a 3 signal mechanism.97,98 The

first signal for activation is provided through binding of the TCR. The second signal is

relayed by interactions between molecules expressed on APCs and “positive” and

“negative” co-stimulatory molecules on the T cell surface.99 These are molecules that

belong to either the Ig family (e.g. CD28, CTLA-4, PD1), the TIM family (T cell Ig and

mucin domain), the TNF-TNFR family (e.g. CD40, OX40) or are cell surface adhesion

molecules (e.g. LFA-1).99 A schematic representation of the major co-stimulatory and

co-inhibitory receptor engagement is depicted in Figure 1-2. The best understood of

these pathways is the T-cell bound CD28, which is expressed on 50% of CD8+ and 95%

of CD4+ cells in humans (all T cell subsets in mice).100 CD28 comes into contact with

APCs (in a region known as the immunological synapse) and binds to B7-1 (CD80) and

B7-2 (CD86) to provide a co-stimulatory signal for T cell survival, proliferation and

cytokine production through its immunoreceptor tyrosine-based activation motif

(ITAM).101 If TCR binding occurs without CD28 co-stimulation, it results in the T cell

becoming anergic. Upon CD28 binding to B7-1/2, T cells also upregulate the expression

of CTLA-4, which shares homology with CD28 and can bind to B7-1/2 with greater

affinity than CD28 resulting in inhibition of Akt signalling and cell cycle arrest..102 Thus,

Page 27: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

14

negative feedback loops such as CTLA-4 act as immune checkpoints to regulate T cell

activation (Fig 1-2).

Figure 1-1 Schematic of co-stimulatory and co-inhibitory receptors

This figure depicts the key co-stimulatory and co-inhibitory receptors involved in

T cell activation and inhibition. The first signal for T cell activation is provided by

TCR binding to a peptide:MHC complex. The second signal is mediated by

CD28 binding to CD80/86 on APCs. CTLA-4 which is upregulated upon T cell

activation can bind CD80/86 with higher affinity than CD28. T cells can also

license APCs by CD40-CD40L interactions. The PD-PD1L axis is important for

negatively regulating T cell activation and is crucial for self-tolerance.

Page 28: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

15

The third signal for T cell activation is provided by cytokines, most importantly

interleukin (IL)-2.103 IL-2 binds to IL-2 receptor (IL-2R) to activate downstream

transcription through various signalling pathways such as the PI3K-Akt pathway which

lies upstream of the mammalian target of rapamycin (mTOR).104 mTOR signalling is

important for cellular metabolism, downstream DNA synthesis and clonal proliferation.73

Activation of T cells also leads to the formation of memory T cells which contribute to a

more vigorous secondary response upon re-exposure to alloantigen (second-set

rejection).105 Central memory (CD44+CD62Lhi) circulate in the blood and secondary

lymphoid organs but upon differentiation to effector memory cells and entering tissues,

these cells lose expression of the adhesion molecules CCR7 and CD62L (L-selectin)

preventing re-entry into peripheral lymph nodes.105

Once activated, CD4+ and CD8+ T cells mediate acute graft rejection through

various mechanisms. CD4+ cells are divided into functional subsets based on the types

of cytokines they secrete and their function in vivo (e.g TH1, TH2, TH17, TH9 etc).106 In

the context of organ transplantation, TH1 and TH2 are the best characterized subsets.

TH1 cells produce IL-2 and IFNγ causing activation of CD8+ cytotoxic T lymphocytes

(CTL) and NK cells, which also produce IFNγ once activated, thereby acting as a

positive feedback loop to TH1 cells.106 TH1 also prime B cells for alloantibody production

and recruit macrophages through the induction of delayed-typed hypersensitivity (DTH)

responses.106 Clinical studies in rejecting kidney patients have demonstrated a strong

role for TH1 cells expressing IFNγ.107 However, the observation that IFNγ expression

was required in a murine model for alloreactive Treg function, complicates its classical

perception as solely a rejection-associated biomarker.108 TH2 cells are marked by the

Page 29: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

16

production of immunomodulatory cytokines such as IL-4 and IL-10 and are purported to

play an important role in B cell alloresponse. In mouse models, TH2 cells can cause

graft rejection in the absence of TH1 cells, possibly through the involvement of

eosinophils.109,110 Recent research has also outlined an important role for the TH17

subset of CD4+ cells through IL-17 mediated inflammation which depends on the

infiltration and function of neutrophils.106,111 As mentioned previously, TFH cells play an

important role in promoting the production of graft-specific alloantibodies.112 Finally,

CD4+ cells are believed to activate APCs through CD40-CD40L interacts, “licensing”

them to activate CD8+ CTLs.113,114 CTLs release cytotoxic granules containing perforin,

which perforates the target cell membrane and granzyme A and B, which induce

caspase-dependent apoptosis.36,57 CTLs can also upregulate the expression of Fas

ligand (FasL) which binds to Fas on target cells to induce apoptosis through the action

of caspases.115 In cardiac transplant rejection, Fas and FasL can be detected

histologically, with increased FasL expression in rejecting hearts.116 CTLs also release

cytokines such as IFNγ and tumour necrosis factor (TNF)-α to mediate graft damage.57

Finally, the roles of NK cells in acute cellular rejection are the being investigated. NK

cells can detect self/non-self by binding of their inhibitory receptors (KIR) to self-MHC

class I.36 NK cells also produce IFNγ and TNFα and possess the capacity for cell-

mediated cytotoxicity.57 A murine study blocking the NK cell NKG2D receptor, in CD28

deficient mice, led to graft acceptance.117 NK cells were also found to target donor DCs

in lymph nodes thereby promoting the indirect allorecognition pathway for T cells.63

Through the actions of these various cell types, ACR is characterized histologically by

the infiltration of macrophages and T cells in the allograft interstitium, widespread tissue

Page 30: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

17

necrosis and edema.74 Due to the central role played by T cells in this process, most

immunosuppressive therapies for acute rejection target T cell proliferation and activation

(reviewed in Section 1.2.2). As a result of the success of these treatments, acute

cellular rejection rates are only observed in under 15% of all non-sensitized transplant

recipients.118

1.2.1.4.3 Chronic Rejection

Despite the tremendous advances in the prevention and treatment of hyperacute

and acute rejection, there has been very little improvement in the rates of chronic

rejection (CR) in the past 20 years.74,119 CR is a multifactorial, gradual process and

neither the etiology nor the pathophysiology of this late-term graft dysfunction are fully

understood. The time to onset for chronic rejection is highly variable ranging from

several weeks to several years post-transplant.74 In cardiac transplantation, cardiac

allograft vasculopathy (CAV) along with malignancy, are the most common causes of

patient mortality after 3 years post-transplantation.120 At 5 years post-transplant, CAV is

detectable in over 30% of heart transplant recipients.121 Similarly, chronic allograft

nephropathy is present in over 50% of kidney transplant recipients at 10 years post-

transplant.122 CR is mediated by both immune and non-immune mechanisms which

warrant further discussion.

As previously described, long-term immune damage of allografts is purported to

occur through a process involving the indirect pathway of T cell allorecognition and

involves the production of alloantibody.123 Clinically, the incidence of acute cellular

rejection is a known risk factor for later onset of CAV.124 However, the incidence of

Page 31: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

18

acute humoural or antibody-mediated rejection (AMR), is seen as a far more potent

prognostic factor for CAV. The risk of CR has been shown to increase incrementally

with each episode of AMR and overall, patients with AMR have a 9-fold higher

incidence of CAV versus patients with ACR alone.125 Even in the absence of AMR, the

presence of anti-donor HLA antibodies is a risk factor for CR of heart, kidney, liver and

lung transplants.126,127 Moreover, heart transplant patients can also develop CAV in the

absence of anti-HLA antibodies through autoimmune mechanisms that produce

antibodies against self-antigens such as myosin and vimentin.124,128,129 The heterotopic

cardiac transplantation model in mice, first described by Corry et al., has been used to

study the immune mechanisms involved in CR.130 Studies using this model have

demonstrated that chronic rejection in mice is strongly associated with the presence of

DSA even in mice treated with CD4+ and CD8+ depleting mAbs.131 In contrast, male

cardiac grafts are rejected in female recipients even in the absence of DSA and C4d

deposition, signaling the role for a T-cell mediated mechanism.132 These studies have

also signaled an important role for NK cells in CAV. NK cells have been demonstrated

to mediate CAV through an Fc-dependent mechanism involving the deposition of

alloantibody.133 NK cells were also implicated in causing CAV in a model of RAG

deficient mice infected with lymphocytic choriomeningitis virus (LCMV).134 These

findings can explain why infection with cytomegalovirus (CMV) in the clinical setting, is

associated with a higher incidence of CAV.135 A recent study demonstrated that in

antibody-deficient (AID/µS KO) mice, B cells can contribute to CR through antigen

presentation and supporting T cell infiltration.136 Finally, the non-immunological factors

Page 32: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

19

associated with CAV are hyperlipidemia, hyperglycemia and pre-existing coronary

artery disease.124

Histologically, chronic rejection manifests as intimal thickening and fibrosis with

collagen deposition in the graft parenchyma and blood vessels.74 Further, endothelial

cell proliferation is observed resulting in the formation of neointima and vessel stenosis

as well as result in coronary atherosclerosis.105,124 Both the interstitium and

parenchyma may also be invaded by T cells and macrophages.72 The results from

studies conducted in both humans and mice using C4d to distinguish CR have been

inconsistent suggesting that C4d alone cannot be utilized as a histological marker of

CR.132,137 Despite, the continued emergence of novel immunosuppressive therapies,

there is currently no effective treatment for treatment of CR and most cases result in

graft loss. Furthermore, certain predisposing factors for chronic allograft dysfunction are

often the result of immunosuppression toxicity such as hyperlipidemia and recurrence of

CMV (discussed further in 1.2.2).124 Therefore, the ultimate goal of transplantation

medicine is to induce donor-specific immune tolerance without the need for long-term

immunosuppression.

1.2.2 Prevention of Rejection

1.2.2.1 Immunosuppression

The goal of current immunosuppressive therapy in the fields of autoimmune

disease and organ transplantation is to attenuate the immune response to self-antigen

or alloantigen respectively.138,139 Therefore, immunosuppressive agents used in

transplantation medicine are also used to treat autoimmunity. Treatment guidelines and

immunosuppressive drugs currently in use for organ transplantation and autoimmune

Page 33: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

20

disease are reviewed elsewhere.140–143 Nevertheless, the major classes of

immunosuppressive drugs and their mechanisms of action warrant further discussion

here. In the context of organ transplantation, immunosuppressive drugs can be

classified as i) induction agents; which serve to broadly eliminate alloreactive

lymphocytes immediately post-transplant thus limiting the incidence of acute rejection

and ii) maintenance immunosuppression; monotherapy or drug cocktails that aim to

provide prophylaxis against rejection.144 Maintenance immunosuppressive drugs fall into

the following major classes, anti-metabolites, corticosteroids, calcineurin inhibitors,

mTOR inhibitors. Due to the use of rapamycin in our studies, mTOR inhibitors and their

mechanisms of action will be the primary focus of this section. Finally, due to their

relevance to transplantation, treatments for desensitization as well as emerging targeted

therapies will be discussed.

1.2.2.2 Induction Agents

ATG (Antithymocyte Globulin), is a cocktail of polyclonal antibodies, generated in

rabbits and horses against human thymocytes. Rabbit ATG has been demonstrated to

be superior to horse ATG in the prevention of acute rejection.145 ATG depletes T cells in

peripheral blood and lymphoid tissues as well as induces B cell apoptosis and interferes

with dendritic cell function.146 The most common side effect of polyclonal antibodies is

excessive cytokine release resulting in flulike symptoms of fever, malaise, nausea and

in rare cases, anaphylaxis.144

There has also been a drive towards development of mAbs to target T cells as

induction therapy. Orthoclone OKT3 is a mouse IgG2a antibody to CD3ε, that has been

Page 34: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

21

used as an induction therapy in combination with other drugs for kidney and liver

transplantation, often to delay use of cyclosporine and its toxic side effects.147

Basiliximab, is a chimeric mouse/human mAb targeted to the α-chain of the IL-2

receptor (CD25). It was demonstrated to have similar efficacy to ATG and OKT3 in

reducing the incidence of acute rejection but fewer side effects in renal transplant

patients.148 Another humanized mAb, Alemtuzumab (Campath-1H) targets the

glycoprotein CD52, found on the surfaces of T and B cells as well as macrophages, NK

cells and granulocytes.149 In a recent trail, alemtuzumab was compared to ATG and

basiliximab and was found to be associated with a lower risk for acute rejection at 6 and

12-months post-transplantation.150 However, in the same trail, no differences were

observed between alemtuzumab and ATG for high-risk patients.

1.2.2.3 De-sensitization therapies

Sensitization to donor-antigen prior to transplantation is a major barrier to

successful transplantation, particularly in kidney transplantation.151 Current approaches

to de-sensitization of these patients involve plasmapheresis in addition to IVIG and B-

cell targeted therapeutics.152 IVIG is a polyclonal antibody blood product prepared from

the serum of about 1000 to 15000 patients and was developed as a therapy for patients

with antibody deficiencies and immune thrombocytopenic purpura (ITP).153 The major

mechanisms of IVIG are believed to be mediated through anti-idiotypic antibodies that

neutralize recipient antibodies.153 IVIG can also mediate its effects by inhibiting

complement and by binding to Fc receptors (such as the inhibitory FcγRIIB). IVIG has

been shown to inhibit T cell proliferation, induce apoptosis in B cells, reduce cytokine

Page 35: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

22

synthesis and promote the expansion of Tregs.153,154 Clinically, IVIG alone has not been

proven to be very efficient at de-sensitization and requires additional therapies. 151

Rituximab is a mAb targeted to the B-cell surface antigen CD20, which plays a role in

regulating BCR-induced calcium influxes. CD20 is expressed on pre-B and mature B

lymphocytes but its expression is lost after differentiation to plasma cells.154 Although

originally approved for B-cell lymphomas, rituximab has been used in combination with

plasmapheresis and IVIG to desensitize kidney and heart transplant patients with high

DSA titres.155 Kohei et al. have shown that rituximab is capable of reducing DSA and

risk of chronic AMR episode in ABO incompatible kidney transplant recipients.156

Another study demonstrated the efficacy of IVIG in combination with rituximab versus

IVIG alone.157 Recently, novel therapies targeting plasma cells have been developed.

Bortezomib, is a first-in-class proteasome inhibitor, which binds reversibly to the 26S

proteasome in plasma cells resulting in apoptosis by counteracting survival signals.158

Tocilizumab is an antagonistic mAb to IL-6R and is designed to interfere with the IL-6

mediated progression of B cells to plasma cells.151 These novel therapies have

undergone clinical trials but results are mixed.151,159 Further research is warranted as

currently there is no FDA-approved therapy for the treatment of pre-sensitized

transplant patients.151

1.2.2.4 Anti-metabolites

Azathioprine, a prodrug of 6-mercaptopurine (6-MP) inhibits purine synthesis and

DNA replication thereby inhibiting cell proliferation.160 Several decades after its first

clinical use, research also demonstrated that azathioprine blocked Rac1 activation

Page 36: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

23

downstream of the CD28 co-stimulatory signal, resulting in apoptosis.161 The

development of azathioprine and 6-MP allowed for the implementation of organ

transplantation as a viable therapeutic option for end-stage organ failure and resulted in

a Nobel Prize for the pharmaceutical pioneers who had developed these drugs,

Gertrude Elion and George Hitchings.162 From the 1950s onwards, 6-MP, and later,

azathioprine were incorporated for use in kidney and liver transplantation but were not

potent or specific enough to hinder acute allograft rejection.163,164 The major side

effects associated with azathioprine are myeloid suppression and hepatotoxicity.165,166

Mycophenolate Mofetil (MMF), a prodrug of the bioactive substance

mycophenolic acid (MPA), exerts its action through blocking inosine monophosphate

dehydrogenase which is required for guanosine synthesis.167 In 1995, the FDA

approved MMF after clinical trials had demonstrated its efficacy over azathioprine.168

MMF preferentially affects T and B lymphocytes, due to their lack of a purine salvage

pathway, and is therefore more selective than azathioprine. It also has been shown to

decrease nitric oxide (NO) production thereby counteracting the effects of classically

activated macrophages, which produce NO.169 The most common side effects of MMF

are symptoms of gastrointestinal distress, nausea and hematological cytopenias.170

Enteric-coated mycophenolate sodium (EC-MPS) has been developed to avoid the

gastrointestinal side effects associated with MMF, but its clinical utility in reducing these

side effects has not been demonstrated compared to MMF.144

Page 37: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

24

1.2.2.5 Corticosteroids

The inclusion of the corticosteroid prednisone, as part of the first

immunosuppressive cocktail was pioneered by Thomas Starzl and significantly reduced

the incidence of acute rejection in renal transplantation.171 Prednisone belongs to the

glucocorticoid class (GC) of corticosteroids. Synthetic GCs such as prednisone,

prednisolone, methylprednisolone and hydrocortisone are the main corticosteroids used

in transplantation.73 GCs are currently not widely used for long-term maintenance

immunosuppression but pulse doses are still utilized as the first-line treatment for acute

rejection.155 Like other steroids, GCs mediate their effect through the GC receptor

(GCR), which upon activation, translocates to the nucleus and activate or repress gene

transcription. GCs are believed to counteract the pro-inflammatory transcriptional

activities of the molecules nuclear factor – kappa B (NF-κB) and activator protein 1 (AP-

1) as well as interfere with IL-2 signalling.144 GCs can also promote the transcription of

anti-inflammatory genes such as lipocortin-1.155 Thus GCs are highly potent

immunosuppressants and affect the immune system in many ways. These include

suppression of macrophage function, inhibition of T cell proliferation and expansion,

inhibiting the formation of cytokines such as IL-1, IL-8 and TNFα.73,172 There have also

been studies demonstrating the ability of GCs to reprogram DCs to a tolerogenic, IL-10

producing phenotype.173,174 As a result of their wide-ranging activity, there are several

side-effects associated with GC use in transplantation such as the risk for new-onset

diabetes mellitus (NODM), hyperlipidemia and osteoporosis.

Page 38: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

25

1.2.2.6 Calcineurin Inhibitors (CNI)

Currently, the calcineurin inhibitors (CNIs) cyclosporine A (CsA) and Tacrolimus

(FK506) are the most widely used maintenance immunosuppression in solid and bone

marrow transplantation.155 CsA is a cyclic, 11-amino acid macrolide antibiotic isolated

from the fungus Tolypocladium inflatum and its application in the 1980s, significantly

improved short-term allograft survival.144 CsA functions by binding to the cytoplasmic

protein cyclophilin (an immunophilin), and together this heterodimer inactivates the

calcium-dependent serine phosphatase, calcineurin.175 Calcineurin is responsible for

dephosphorylating the cytoplasmic nuclear factor of activated T cells (NFAT), which

translocates to the nucleus and activates key cytokines required for T cell activation and

function such as IL-2, IL-4 and IFNγ.73,176 Tacrolimus, is a macrolide lactone isolated

from the bacterium Streptomyces tsukubaenis, and binds to the immunophilin FKB1A,

which then inhibits calcineurin with a higher potency than CsA.155 Both CsA and

tacrolimus are also observed to increase expression of transforming growth factor beta

(TGF-β).177 The major side effects associated with CsA are hirsutism, nephrotoxicity,

fibrosis and dyslipidemia. Comparatively, tacrolimus has a better safety profile but

significantly increases the risk for de novo diabetes mellitus.178 Although tacrolimus is

not believed to be less nephrotoxic than CsA, it is utilized more widely after trials that

showed that it resulted in a lower incidence of acute rejection compared to CsA.179

Alternately, patients with a high risk for diabetes, are still preferentially treated with

CsA.73

Page 39: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

26

1.2.2.7 mTOR inhibitors (mTORi)

In 1999, the FDA approved sirolimus, or rapamycin for use in kidney

transplantation.180 Rapamycin, is a macrocylic lactone that was isolated in 1975 from

the bacterium Streptomyces hygroscopicus and initially investigated as an antibiotic.180

Further research showed that rapamycin bound to FKBP12 and inhibited the

serine/threonine protein kinase mTOR, also known as the mechanistic and in mammals,

mammalian target of rapamycin.181 mTOR is a crucial regulator of cell metabolism and

proliferation downstream of growth factor or cytokine signaling. Its role in the immune

system is the subject of much investigation.182–184 In T and B cells, mTOR lies

downstream of phosphoinositide 3-kinase (PI3K), which is induced by TCR/BCR

activation, co-stimulatory molecules (CD28) as well as cytokine (IL-2) signalling.183,185,186

In mammals, mTOR exists as two functionally and structurally distinct complexes known

as mTORC1 and mTORC2. These complexes are distinguished primarily by the

proteins associated with each compex.183 Whereas mTORC1 binds with the rapamycin-

sensitive, regulatory-protein of mTOR (RAPTOR), mTORC2 binds with the rapamycin-

insensitive companion of mTOR (RICTOR).181,187,188 Despite the evidence that only

mTORC1 is affected by rapamycin, recent studies have shown that rapamycin

treatment can also inhibit mTORC2 in certain cell lines and particularly, in naive T

cells.189,190 Downstream of mTORC1, S6 kinase 1 (S6K1) and eukaryotic initiation factor

4E-binding protein 1 (EIF4EBP1) regulate mRNA synthesis and translation. mTORC1

also negatively modulates autophagy and is involved in lipid metabolism.181,183 The

downstream effects of mTORC2 are not well-described but it is purported to rely on

downstream signaling by Akt, glucocorticoid regulated kinase 1 (SGK1) and protein

Page 40: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

27

kinase C alpha (PKCα) to modulate cell shape and growth.191 Finally, rapamycin can

also inactivate ribosome synthesis and delay cell cycle progression by binding to p70S6

kinase.192 The multiple roles of mTORC1 and mTORC2 in cell biology have made them

an attractive target for cancer research.193

Early studies into the effects of rapamycin showed that it had potent in vitro

effects on inhibiting proliferation of human T cells, inducing anergy in TH1 cells and

blocking IL-2-dependent and independent proliferation of human B cells.194,195 Studies

have also shown its effects in reducing the production of harmful autoantibodies in

murine models of SLE (systemic lupus erythematosus).186 Studies have also shown that

rapamycin inhibits DC antigen uptake and maturation.196 The full role of mTOR inhibition

in innate immune function is not well described and is a highly researched area in

immunobiology.184 In contrast to its role as an immunosuppressant, research has also

demonstrated that rapamycin can improve CD8+ memory formation versus LCMV

infection in mice.197 Furthermore, studies performed in mice containing a CD4-specific

mTOR knockout (as full mTOR knockout is embryonic lethal), have demonstrated a

more nuanced role for the role of mTOR inhibition in T helper cells.198 It was

demonstrated that mTOR deficiency did not significantly alter T cell proliferation or IL-2

production but inhibited the differentiation of CD4+ cells into TH1, TH2 and TH17

subsets.198

Arguably, one of the most intriguing function of rapamycin is its ability to promote

the differentiation and expansion of CD4+CD25+ regulatory T cells (Tregs) that also

express the transcription factor Forkhead box P3 (FOXP3) .199–201 The mechanisms of

Page 41: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

28

this induction and preferential expansion are not yet fully understood. FOXP3

expression is believed to be regulated in part by TGF-β signaling where TGF-β-induced

SMAD3 (mothers against decapentaplegic homologue 3) and NFAT act together to

increase FOXP3 transcription.196 Constitutively active mTOR counteracts this pathway

and thus rapamycin-mediated mTOR inhibition induces FOXP3 expression.196 As

expected, CD4+-specific mTOR deficiency results in preferential expansion of Treg cells

and constitutive SMAD3 phosphorylation.198 This increased SMAD3 signaling is

believed to be a result of increased sensitivity to TGF-β and not an increase in TGF-β

expression. In fact, high levels of TGF-β can induce FOXP3 expression even in the

presence of mTOR activity.202 Interestingly, TH17 could be converted to Tregs in the

presence of high TGF-β activity due to its ability to counteract the transcription factor

RORγt.202 The TGF-β-independent pathway of rapamycin-dependent Treg induction is

purported to be a result of PI3K-Akt-mTOR dependent chromosomal rearrangement

that favours FOXP3 expression when TCR and CD28 signalling is interrupted early (<18

hours).196,203 Using rapamycin in this setting greatly enhanced the formation of FOXP3+

Tregs and mAbs to TGF-β did not affect Treg promotion indicating a TGF-β-

independent pathway.203 Finally, it is also important to note that FOXP3+ Tregs have a

survival advantage over conventional T cells in the absence of mTOR activity due to

their ability to upregulate Pim-2 kinase, a serine/threonine kinase with characteristics

similar to Akt.204

Rapamycin, due to its multiple immunological effects, is therefore an ideal

candidate for an immunosuppressant in the context of organ transplantation. Several

studies, including previous work from our lab, have demonstrated the ability of

Page 42: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

29

rapamycin to prolong cardiac allograft survival in murine models.205–208 Clinically,

sirolimus and its analogue, everolimus are used in CNI-sparing regimens to reduce the

risk of CNI-induced nephrotoxicity.180 Although they have not replaced CNIs as the

predominant drug for maintenance immunosuppression, several studies have shown

that using an mTOR inhibitor while minimizing tacrolimus preserved renal function in

heart, lung and liver transplant recipients while not increasing the risk of acute rejection

over standard therapy.209 In particular, everolimus has shown its efficacy as a CNI-

sparing regimen in liver transplant patients, who are at high risk for end-stage renal

disease.155 Recent data from the PROTECT trial has demonstrated that replacement of

CNIs with everolimus was just as efficacious as CNIs in terms of preventing acute

rejection, while preserving better renal function in these patients.210 Furthermore,

tacrolimus to sirolimus conversion in liver transplant recipients was also demonstrated

to lead to increased Tregs in peripheral blood mononuclear cells (PBMCs) and the

expression of immunoregulatory genes.211 Similarly, in cardiac transplantation,

everolimus with reduced-dose CNI has been demonstrated to be just as potent as MMF

in preserving graft survival and preventing acute rejection.212 Inhibiting mTOR is also

known to inhibit vascular endothelial growth factor (VEGF) signaling which is important

for neoangiogenesis and endothelial remodelling.183 Hence, data from recent trials using

both sirolimus and everolimus have demonstrated reduced incidence and severity of

CAV in patients treated with mTOR inhibitors compared with CNI and MMF treatment

respectively.213,214 Treatment with mTOR inhibitors is associated with reduced incidence

of CMV and BK virus recurrence as well as, reduced malignancy in kidney

transplantation.181 However, both sirolimus and everolimus have potent side effects

Page 43: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

30

including hyperlipidemia, increased proteinuria, thrombocytopenia and reduced

spermatogenesis.181,209 The complete role and utility of mTOR inhibitors in organ

transplantation are still being investigated. Their ability to promote expansion of Tregs

warrants their inclusion in further studies designed to induce immune tolerance in the

clinic, which is the “holy grail” of transplantation immunology research.14

1.2.2.8 Novel therapies in the pipeline

As the scientific literature continues to further the understanding of mechanisms

of immune activation, novel therapies continue to emerge that are specifically targeted

to cell surface receptors or intracellular kinases in an attempt to selectively inhibit

alloreactive T and B cells.144 The importance of the role of co-stimulatory molecules

(signal 2) for T cell activation has generated interest in targeting them to suppress the

immune system (discussed further in 1.4.2). Belatacept (LEA29Y), is a fusion protein

comprised of the extracellular domain of the co-inhibitory molecule CTLA-4 and the Fc

portion of human IgG1.215 Belatacept was approved by the FDA in 2011, and early

results demonstrated that it had safer toxicity profiles compared to CNIs for kidney

transplantation but not improved efficacy.216 However, a recently published study by

Vincenti et al., demonstrated for the first time, the superiority of belatacept over CsA, in

terms of patient and graft survival for a follow-up period of 7 years.217 Both belatacept

(low-dosage and high-dosage) and CsA patients received MMF, basiliximab induction

and glucocorticoids. The belatacept (high and low-dosage) groups demonstrated

improved graft survival, lower DSA and fewer deaths associated with cardiovascular

Page 44: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

31

complications versus the CsA group.217 Another targeted therapeutic that has been

recently studied is the oral Janus Kinase-3 (JAK3) inhibitor, Tofacitinib.218 JAK3 is

expressed predominantly in hematopoietic cells and is required for lymphocyte

activation.219 In a phase II clinical trial in renal transplant patients, high-intensity

tofacitinib exposure resulted in a lower incidence of acute rejection as well as lower

symptoms of chronic allograft nephropathy at 1-year post-transplant. However, this

treatment also significantly enhanced the risk for infections and PTLD.218,219 Another

related kinase inhibitor currently being investigated is Sotrastaurin.220 Sotrastaurin

targets protein-kinase C (PKC) which is required for early T cell activation and mediates

its effects by downstream NF-κB, NFAT and AP-1 signalling.220 However, phase II

studies in both kidney and liver transplant patients demonstrated that sotrastaurin had

either lower or equivalent efficacy and more serious adverse effects compared to

tacrolimus.221,222 The drive towards selective therapeutics in organ transplantation has

resulted in better targeted therapies and offers the potential to minimize and/or replace

current immunosuppressants. However, these novel therapies are also associated with

adverse effects and have not resulted in significant increases in long-term graft survival.

1.3 Immune Tolerance

1.3.1 Overview

A healthy immune system is defined by the ability to mount protective immune

responses against pathogens and foreign antigens while maintaining tolerance to self-

tissue. Immune tolerance therefore, can be defined as the lack of immune response to a

Page 45: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

32

specific antigen in an otherwise functional immune system.14,223,224 Transplant tolerance

therefore denotes a situation where an organ is accepted without the need for long-term

immunosuppression thereby preserving protective immunity.14 The induction of

transplant tolerance remains the ultimate therapeutic goal of transplantation research.

As mentioned previously, limited numbers of patients, particularly liver transplant

recipients, achieve “operational tolerance” whereby they are able to retain graft function

in the absence of immunosuppressive drugs.20,21 In contrast to the clinic, there have

been several demonstrations of tolerance induction in experimental murine

models.225,226 In 1953, the concept of “acquired immune tolerance” was demonstrated

by Billingham, Brent and Medawar.227 This was performed by intra-uterine injections of

donor spleen cells thus exposing neonatal mice to donor antigen. These mice were then

capable of permanently accepting donor skin grafts.227 In 1956, Frank MacFarlane

Burnet proposed the theory of clonal selection as the mechanism through which self-

reactive lymphocytes are deleted to prevent immune recognition of self. In the decades

that followed, the process of intra-thymic clonal deletion was studied and classified as

central T cell tolerance (i.e. deletional tolerance).228 Similarly, the mechanisms for

controlling autoreactive mature T cells in the peripheral tissues i.e. peripheral tolerance,

have also been identified.229 In addition to T cells, B cells are subject to central and

peripheral mechanisms of tolerance.230,231 Due to their central role in transplantation,

this section will focus primarily on mechanisms underlying T cell tolerance.

Page 46: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

33

1.3.2 B cell tolerance

Studies performed in transgenic mice have shed light on the mechanisms

underlying B cell self-tolerance, but the exact mechanisms in humans are not well

defined.232 In adult mammals, B cells arise from common lymphoid progenitors (CLPs)

in the bone marrow and undergo sequential developmental stages.231,233 Re-

arrangements of immunoglobulin heavy and light chains along with the expression of Ig-

α and Ig-β (which are critical for signal transduction), results in the formation of a fully

functional IgM BCR on immature B cells.233 These immature B cells begin to express

IgD as transitional B cells and migrate to the spleen to become fully functional mature B

cells.232 Early studies of both humans and mice demonstrated that a large proportion of

B cells (50-75%) expressed receptors reactive to self-antigens while in the bone

marrow. This proportion was significantly smaller (20%-40%) in B cell repertoires in the

spleen and peripheral blood.234,235 Using transgenic mouse models, three mechanisms

of self-education of B cells in the bone marrow have been identified. The dominant

mechanism is now known to be receptor editing, whereby self-reactive B cells undergo

persistent re-arrangement of their Ig light-chain genes replacing autoreactive BCRs with

non-autoreactive BCRs.236,237 It is estimated that nearly 35% of autoreactive immature

B cells undergo receptor editing while the remaining cells undergo clonal anergy or

deletion.232,238 Further clonal selection is posited to occur in the spleen between the

transition from transitional B cells to naive mature B cells. An important cytokine

modulating the emergence of mature B cells is B cell activating factor (BAFF).239 Self-

reactive B cells are demonstrated to express lower levels of BAFF receptor and as

such, BAFF preferentially selects for non-autoreactive B cells.238 Elevated BAFF is

Page 47: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

34

associated with various autoimmune diseases in humans such Sjogrens syndrome, SLE

and rheumatoid arthritis.235 The mechanisms underlying B cell tolerance warrant further

study. Finally, recent studies have begun to unravel the roles of suppressive B cell

populations, i.e. B regulatory cells in maintaining peripheral immune tolerance

(discussed further in 1.3.5.2).240

1.3.3 Central T cell tolerance

The thymus is a rigorous environment for T cell development with only 1-2% of

total thymocytes emerging as mature T cells in mice.241 In the thymus, recognition of

self has contrasting effects on T cell development. It is essential for thymocyte survival

and maturation during the stage of positive selection. In the next stage, negative

selection results in “clonal deletion” of thymocytes that bind too strongly to self-MHC.242

Positive and negative selection occur primarily in different compartments of the thymus;

the cortex and the medulla respectively.24 In mice, multiple precursor cell types possess

the capacity for giving rise to thymocytes including common lymphoid progenitors

(CLPs), early thymic precursors (ETPs) and early lymphoid precursors.243,244 In the

initial stage of thymocyte maturation, they lack either CD4 or CD8 expression and are

termed double negative (DN). At this stage, VDJ recombination of the TCRβ chain in

combination with the CD3 and TCRα chain results in the formation of the pre-TCR.245

For cells that do not undergo TCRβ rearrangement, the re-arrangement of the γ chain

results in the formation of γδ T cells that are not self-MHC restricted and play an

important role in anti-microbial as well as anti-tumor immunity.246 Subsequently, the αβ

Page 48: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

35

T cells upregulate both CD4 and CD8 expression to become double positive (DP)

thymocytes and their TCRα chain undergoes rearrangement to form a fully functional

TCR. The DP thymocytes then interact with MHC class I or II to become single positive

(SP) CD8+ or CD4+ cells respectively.243 Cells that do not bind to any MHC undergo cell

death through neglect.243 The differentiation of the CD4 lineage is dependent on the

transcription factors ThPOK and Gata3 whereas the Runx proteins Runx1 and Runx3

are involved in CD8 differentiation.247

SP thymocytes subsequently undergo negative selection (clonal deletion) of

autoreactive cells primarily in the thymic medulla.242 The medulla contains medullary

thymic epithelial cells (mTECs) that display tissue restricted antigens (TRA) to SP cells.

TRA (defined as antigens expressed in less than 5 body tissues) expression is

regulated by the transcription factor, autoimmune regulator (AIRE).242,248 In accordance

with the crucial role of AIRE, mutation in the AIRE gene in humans results in a multi-

organ disease called autoimmune polyendocrinopathy-candidiasis-ectodermal

dystrophy (APECED).249 For several years, AIRE was known to be the only regulator of

TRA expression even though TRA expression was observed to in AIRE-deficient

mice.250 In 2015, however, Takaba et al. identified Fezf2 as another transcription

involved in TRA expression.250 Fezf2 deficient mice also displayed signs of autoimmune

organ damage and production of autoantibody.250

The thymus is also populated by a small population of resident and migratory

DCs that play important roles in the presentation of thymic and peripheral antigens with

some evidence pointing to their role in protection from thymic viral infection.242,251 A

Page 49: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

36

recent study in mice has also outlined the important role of circulating B cells that

migrate to the thymus and express AIRE to display endogenous self-antigens to CD4+

SP thymocytes.252 Thymocytes that bind self-MHC with high affinity are deleted through

apoptosis. Alternately, SP thymocytes with low affinity for self-MHC undergo maturation

to the next step.243 Co-stimulatory molecules are purported to play an important role in

this process. Buhlmann et al. also demonstrated using B7-1/2 double knock-out mice

that negative selection is predicated on co-stimulatory signaling through B7-1/2 and

CD28.253 Similarly, the study on aforementioned thymic B cells by Yamano et al.

showed that B cells required CD40-CD40L licensing to mediate their APC functions.252

Finally, the thymus also gives rise to Treg, NKT and CD8αα regulatory cells.254

Although the exact mechanisms are not fully understood, it is posited that this process

relies on TCR avidity for self-antigen where low avidity results in maturation, high avidity

in clonal deletion and intermediate avidity in the generation of Treg.255 In recent years,

the importance of this clonal diversion to Treg has been elucidated and is now

purported to be as important as clonal deletion in the maintenance of self-tolerance.229

1.3.4 Peripheral T cell tolerance

The thymus is stringent in its selection for functional and non-autoreactive T

cells. However, T cells with the potential for autoreactivity manage to escape to the

periphery where they necessitate control by the mechanisms of peripheral

tolerance.256,257 These suppressive mechanisms are either intrinsic such as ignorance,

Page 50: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

37

anergy and activation-induced cell death (AICD), or due to the extrinsic modulation by

regulatory cell populations.229

1.3.4.1 Ignorance

Ignorance denotes a state whereby cells with self-reactive potential remain

inactivated and unable to respond to self.258 In 1991, Ohashi et al., demonstrated that

in the absence of LCMV infection, naïve transgenic (P14) CD8+ T cells specific for

LCMV glycoprotein (GP), did not attack islet cells that were expressing the LCMV-GP

under the control of a rat insulin promoter (RIP-GP mice).259 Infection of these mice with

LCMV resulted in induction of CD8+ islet cell destruction and diabetes indicating that the

T cells had become activated in presence of high levels of antigen.259 Thus, T cells

bearing TCRs with low avidity for self-antigens (TRA) may escape thymic deletion but

remain inactivated in the absence of high-antigen exposure. This is further achieved by

the physical separation of naive circulating lymphocytes that are restricted to the blood,

efferent lymph and secondary lymphoid organs.229,258 Conversely, antigen-experienced

T cells are able to enter peripheral tissues by downregulating CD62L and CCR7, where

they may be exposed to TRA.229 However, even in this setting, a stimulus may be

required to liberate self-antigens and cause inflammation. This was shown recently by

injecting OVA-specific transgenic OTI (CD8+) and OTII (CD4+) cells into transgenic mice

bearing the OVA protein in their skin. Even though antigen-experienced cells were

observed, a further inflammatory stimulus, in this case tape stripping, was required for T

cell homing to the skin and the induction of inflammatory skin disease.260

Page 51: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

38

1.3.4.2 Anergy

T cell anergy is defined as a state of prolonged hyporesponsiveness towards an

antigen and is marked by cell cycle arrest as well as reduced TCR signaling and IL-2

expression.261 In CD4+ and CD8+ T cells, anergy also results in impaired production of

IFNγ and TNFα.262 There are multiple mechanisms that underlie the induction of anergy

in mature functional T cells.243,257,262 Several years ago, experimental studies

demonstrated that T cell activation by TCR binding in the absence of CD28 signaling

resulted in anergy.263,264 Subsequent studies have begun to unravel the molecular

pathways involved in the induction of anergy.262 Complete activation of T cells is

dependent on TCR and CD28 mediated recruitment of the transcription factors NFAT,

AP-1 and NF-κB for IL-2 expression. IL-2 expression and autocrine signaling through

the IL-2R, results in a complete activation of the PI3K-Akt-mTOR pathway which among

several other targets, degrades the cyclin-dependent kinase inhibitor p27kip1.265 In

contrast, TCR stimulation alone results in calcineurin/NFAT activation in the absence of

AP-1. This results in the formation of NFAT homodimers which induces a gene profile

characteristic of anergic T cells.261 These genes include E3 ubiquitin ligases such as

Cbl-b, GRAIL, Itch and Deltex-1 which target the molecular pathways downstream of

TCR and CD28 signaling.261,265 The anergic state also induces the activation of

transcription factors such as Erg1/2, cAMP response element modulator (CREM) and

Ikaros, which mediate direct repression of the IL-2 gene locus.265

Page 52: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

39

The anergic state can also be induced by engaging co-inhibitory molecules on T

cells. CTLA-4 is an important inhibitory pathway as described previously. CTLA-4 is

induced after TCR and CD28 activation and binds with higher affinity to B7-1/2 on

APCs.266 The importance of CTLA-4 in maintaining peripheral tolerance is evident from

studies that showed that CTLA-4 deficient mice develop autoimmunity and CTLA-4

knockout CD4+ T cells are resistant to induction of anergy.267,268 Another crucial

inhibitory pathway is that of the immune checkpoint, programmed death-1 (PD-1)

(CD279), a receptor from the immunoglobulin family expressed on T cells, B cells and

myeloid cells.269 The ligands for PD-1 are PD-L1 (CD274) and PD-L2 (CD273), which

are expressed on T cells, B cells, APCs, endothelial cells and tumours.243 TCR

activation in addition to PD-1 ligation through its downstream immunoreceptor tyrosine-

switch motif (ITSM), results in activation of SHP-1 and SHP-2 phosphatases which

counteract the PI3K-Akt pathway.257 PD-1 and PD-L1 signaling is implicated to play a

role in negative selection in the thymus.270 In the periphery, T cell tolerance is

maintained by PD-1 by limiting early activation and expansion.270 Furthermore, PD-L1

expression on tolerogenic DCs was also shown to be necessary for the induction of

CD8+ T cell peripheral tolerance.271 Finally, PD-L1 signaling is also important for the

induction of peripheral CD4+FOXP3+ regulatory T cells.272 As expected, PD-1 deficient

mice display signs of lupus-like autoimmune disease.273

Finally anergy can also be induced in response to metabolic cues from the

environment.261 Studies using rapamycin have demonstrated induction of anergy in T

cells even in the presence of signal 1 and signal 2 (TCR and CD28) stimulation due to

the central role of mTOR in cell metabolism.195 This was further demonstrated by

Page 53: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

40

utilizing leucine and glucose antagonists to induce anergy in the presence of both signal

1 and signal 2 of T cell activation.274 This sensing of hypoxia is believed to be the result

of adenosine monophosphate-activate protein kinase (AMPK), which is activated as a

result of low ATP levels in the cells and functions to counteract mTOR signaling.261

Clonal anergy in vitro can be reversed by the exogenous addition of IL-2.275 The

term “adaptive tolerance” has been coined to describe T cell anergy in vivo. This state,

which involves distinct signaling pathways, requires persistent exposure to antigen and

is refractory to exogenous IL-2 addition.276

1.3.4.3 Activation-induced cell death

Activation-induced cell death (AICD), is an important mechanism for T cell

homeostasis and for the prevention of autoimmunity in the peripheral immune

system.277,278 Upon primary antigen exposure, T cells undergo a primary expansion

phase and are resistant to AICD. During the contraction phase and upon re-stimulation,

T cells become sensitive to AICD.278 AICD is mediated in the target cell by the binding

of Fas (CD95) on the target cell to either soluble FasL (suicide) or a neighboring T cell

(fratricide).277 Fas belongs to the TNFR superfamily of transmembrane proteins. This

superfamily also contains other death receptors including TNFR1, DR3 (TNFRSF25),

DR4 (TRAILR1), DR5 (TRAILR2) and DR6 (TNFRSF21).279 Fas-induced apoptosis is

regulated primarily by the death-inducing signaling complex (DISC) comprised of the

adaptor FADD (Fas-associated death domain) protein and the proteases caspase-8 and

caspase-10.278 These activate downstream effector (executioner) caspase-3, caspase-6

Page 54: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

41

and caspase-7 which mediate cell death by cleaving several critical proteins required for

cell function.279 Key cytokines such as IL-2, IL-4 and IFNγ increase sensitivity to

AICD.278 The role of Fas-induced AICD has an important role in prevention of

autoimmunity. In humans, autoimmune lymphoproliferative disorder (ALPS) is an

inherited disease based on mutations in the Fas, and in some cases the FasL and

caspase-10 genes.278,280 In mice, the MRL (Murphy Roths Large) strain develops multi-

organ autoimmunity, serum autoantibodies and lymphadenopathy which are the

hallmarks of SLE in humans.281 MRL/lpr mice were found to have a spontaneous

mutation in the lpr (fas) gene, and MRL/lpr mice have an accelerated autoimmune

pathogenesis compared to MRL mice.281 Finally, FasL is also expressed on astrocytes

and on cells in immunoprivileged sites, such as Sertoli cells, thyroid epithelium and

endothelial corneal cells.278

1.3.5 Suppression by immunomodulatory cells

In addition to cell-intrinsic mechanisms for maintaining peripheral tolerance,

extrinsic immunomodulation by regulatory cells is crucial for preventing excessive

immune activation and autoimmunity. Regulatory or tolerogenic cell subsets from both

the myeloid and lymphoid hematopoietic lineage have been identified and

described.282,283 As the focus of this section is on peripheral T cell tolerance, the cell

types that warrant further discussion are dendritic cells, regulatory B cells (Bregs) and

regulatory T cells (Tregs), all of which can suppress T cell function.282

Page 55: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

42

1.3.5.1 Dendritic cells

As one of the three professional APCs, DCs provide a functional bridge between

innate and adaptive immunity.284 Upon sensing PAMPs and DAMPs through PRRs,

immature DCs become activated, upregulate their expression of co-stimulatory

molecules (CD80/86) and release inflammatory cytokines.285,286 DC subsets are highly

varied in phenotype and function and are found both in lymphoid and non-lymphoid

organs.284,287 DCs are broadly divided into two major subtypes based on their function

and ontogeny. Myeloid-derived classical, or conventional DCs (cDCs), express co-

stimulatory molecules and sample antigen for presentation to and activation of CD4+ T

cells.284 Plasmacytoid DCs (pDCs) arise from lymphoid progenitors and one of their

primary functions is to mediate anti-viral immune responses by production of large

quantities of type I interferons.286,287

Selective depletion of both pDCs and cDCs leads to fatal autoimmunity in mice

indicating a crucial role for DCs in maintaining immune tolerance.288 As discussed

previously, both resident and migratory DCs in the thymus, play an important role in

central T cell tolerance.242 On the other hand, DCs also important for maintenance of

peripheral T cell tolerance in the absence of inflammatory or pathogenic stimuli (i.e.

steady-state). The first evidence for the tolerogenic role of steady-state DCs was

described by Hawiger et al. in 2001.289 By producing a fusion protein comprised of DEC-

205 (a mAb towards a DC-specific endocytic receptor) and hen egg lysozyme (HEL),

HEL antigen was targeted directly to DCs. Despite production of IL-2 and an initial

proliferation of CD4+ T cells in these mice, they failed to produce IFNγ, IL-4 or IL-10.289

Page 56: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

43

After 7 days, most activated T cells either underwent deletion or became anergic to re-

stimulation with HEL.289 Thus in the steady-state, DCs prevent T-helper subset

differentiation and mediate T cell anergy. It was demonstrated further that steady-state

presentation of antigen by DCs also induced CD8+ tolerance through co-inhibitory PD-1

and CTLA-4 signaling.271 Subsequent studies have delineated a wide range of DC

subsets with immunoregulatory roles.290 These include induction of FOXP3+ Tregs,

secretion of suppressive molecules like IL-10, TGF-β and indoleamine 2,3-dioxygenase

(IDO) and induction of apoptosis through FasL expression.285,290 Currently, tolerogenic

DCs (TolDCs) are believed to be immature or maturation-resistant DCs, with low

surface expression of co-stimulatory molecules, that modulate T cell function through

one or more of the aforementioned mechanisms.285,286 TolDCs from both mice and

humans can be generated in vitro using pharmacological treatments such as

dexamethasone, rapamycin, 1α,25-dihydroxyvitamin D3, sanglifehrin A or cytokine

treatment with IL-10, IDO or TGF-β and are being studied for their ability to promote

tolerance in autoimmunity and transplantation.285

1.3.5.2 Regulatory B cells

Since their discovery, B cells have been primarily conceived of as potent

activators of the immune system through antibody production and APC functions.

However, an increasing body of evidence in the literature points to the role of B cells as

negative regulators of the immune system.240,291,292 As early as the 1970s, the

suppressive role of B cells was observed in a guinea-pig model of delayed-type

Page 57: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

44

hypersensitivity.293 In 1996, Janeway’s group generated evidence for a role of B cells in

mediating self-tolerance. B-cell deficient mice displayed an increased severity of and

inability to recover from experimental autoimmune encephalomyelitis (EAE), a murine

model of multiple sclerosis.294

Currently, B regulatory cells (Bregs) in both mice and humans lack a unique

phenotypic marker similar to FOXP3+ in CD4+CD25+ Tregs. Instead, all of these Breg

subsets are identified by their functional capacity to produce large amounts of IL-

10.291,292 In mice, the major mechanisms of Breg immunoregulation consist of inhibition

of TH1 differentiation, inhibition of DC maturation and induction of Tregs.295 Moreover,

Breg activation is believed to occur through CD40 or TLR stimulation.296,297 Two primary

Breg populations have been identified in mice. A transitional B cell population in the

marginal zone (T2-MZP-B cells) were identified by Evans et al. as suppressive cells in a

mouse model of collagen-induced arthritis.298 These cells are

CD19+CD21hiCD23+CD24hi and their suppressive capacity is IL-10 dependent.298 A

rare population of Bregs in the spleen (1-2%), termed B10 cells were identified by

Yanaba et al. in 2008.299 These cells were CD1dhiCD5+ and suppressed contact

hypersensitivity in mice in an IL-10 dependent manner.299 A subsequent study also

demonstrated that these cells could differentiate into Ab-producing plasmablasts after

transiently producing IL-10.300 It was posited that these Ab-production by these cells

could further suppress immune activation by reducing antigen load.

In humans, various Breg subsets have been identified and have been implicated

in protection from autoimmunity.291 The mechanisms of human Breg-mediated

Page 58: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

45

immunosuppression involve TH1 differentiation and proliferation, but require further

characterization.291,295 Blair and colleagues, identified a subset of B cells that are

CD19+CD24hiCD38hi and produce IL-10 in response to CD40 stimulation in vitro.301 It

was demonstrated that this subset of Bregs was dysfunctional in patients suffering from

SLE.301 A reduced Breg frequency is also observed in other autoimmune pathologies

such as rheumatoid arthritis and multiple sclerosis.302,303 The multiple roles of Bregs in

maintaining and promoting immune tolerance have only recently been discovered.

Bregs have been implicated in promoting allograft tolerance in mouse models and a B

cell signature has been observed in operational tolerance in kidney transplantation.304–

306 Thus, Bregs are a potential candidate for adoptive cell therapy in tolerance induction

trials. However, further studies are necessary to characterize the phenotypes, functions

and isolation techniques of putative Breg populations in humans.

1.3.5.3 Regulatory T cells

Peripheral T cell tolerance is in part maintained by thymic selection and T cell-

intrinsic inactivation mechanisms.229 However, regulatory T cells (Tregs) are now

recognized as a major mechanisms of T cell suppression and prevention of

autoimmunity.307,308 The concept of “suppressor T cells” was first posited through the

work of Gershon and Kondo in 1970.309 Subsequent studies identified the IL-2Rα chain

CD25, as a marker for an immunosuppressive T cell population that was derived from

the thymus and was crucial for the prevention of autoimmunity.310,311 Finally, in 2003 the

X-linked transcription factor FOXP3, was identified as a master regulator of Tregs.312,313

Page 59: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

46

Once foxp3 is expressed, it induces the transcription of signature Treg genes including

foxp3 itself.314 These cells are crucial for the maintenance of self-tolerance. FOXP3-

mutated Scurfy mice exhibit fatal autoimmune inflammation with increased TH1, TH2 and

TH17 activation.315 In humans, FOXP3 inactivating mutations result in immune

dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome.316

However, constitutive FOXP3 expression is not a requisite for the function of certain

regulatory T cells such as Type 1 regulatory (Tr1) cells.317 In addition to Tr1 cells other

T cells with immunoregulatory functions include Th3 cells, CD8+ Treg, CD8αα intra-

epithelial lymphocytes, double negative (DN) T cells and γδ T cells.246,308,318 However,

due to their central role in peripheral tolerance and relevance to this dissertation, this

section will focus on CD4+CD25+FOXP3+ cells as the major type of Treg.

1.3.5.3.1 CD4+CD25+FOXP3+ Tregs

In normal naive mice, Tregs comprise 5-10% of all CD4+ T cells.319 As previously

discussed, natural or thymic Tregs (tTregs), arise in the thymus through the

developmental processes of clonal diversion.320 Alternatively, limited numbers of

peripheral Tregs (pTregs) can be generated in the steady-state through the TGF-β

dependent mechanisms.321 This is posited to occur primarily in tolerogenic tissue

environments such as mucosal surfaces.307 However, pTregs can expand significantly

in the context of immune inflammation or due to pharmacological treatments.322

Moreover, in vitro TCR signaling in the presence of TGF-β and IL-2 can also give rise to

induced Tregs (iTregs) that express FOXP3.323 It is important to note that while these

Page 60: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

47

different Treg populations exhibit FOXP3 expression, there are functional and genetic

differences between them. For instance, pTregs generated in vivo show potent in vitro

suppressive capacity whereas TGF-β induced iTregs are not as suppressive and do not

acquire the complete gene signature of pTregs.322 Feuerer et al. conducted an in-depth

microarray analysis of various Treg subtypes in mice and found differently expressed

genes between each subtype.324 Therefore, further study is required to find phenotypic

markers that could be used to isolate and study each subset of Tregs. Recently, Helios,

an Ikaros-family transcription factor, was studied as a candidate to distinguish tTregs

from pTregs.325 However, subsequent studies showed that Helios could be expressed in

activated T cells as well as iTregs.322 Another candidate marker, the cell surface

molecule Neuropilin-1 (Nrp-1) was observed to be expressed on tTreg.326 However,

pTregs can upregulate Nrp-1 during inflammatory settings.322 Thus, neither Helios nor

Nrp-1 are steady markers for natural thymic-derived Tregs in mice while their

expression in human Tregs require further investigation.322 Isolation of pure Tregs in

both mice and humans for functional studies is hindered by the lack of an exclusive cell

surface marker. Tregs express cell surface markers that are integral to their function

but are also expressed on other cell types such as CD25, CTLA-4, lymphocyte

activation gene-3 (LAG-3) and T cell immunoreceptor with Ig and ITIM domains

(TIGIT).327,328 A further complication observed in human T cells, FOXP3 in the absence

of suppressor Treg function.329 In human Tregs, the cell surface molecule CD127 (IL-

7Rα), was found to be inversely correlated to Treg suppressive function.330 Thus,

several studies have begun investigating Tregs that exhibit the

CD4+CD25+FOXP3+CD127low phenotype for human clinical studies.282 The critical role

Page 61: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

48

for Tregs in maintaining peripheral tolerance is evident from studies in patients with

autoimmunity. Several studies in patients with SLE, MS, RA and type 1 diabetes (T1D)

demonstrate perturbations in Treg frequencies compared to healthy controls.308,322

However, the wider role for Tregs in several other contexts is also being recognized

including cancer and persistent viral infections.331,332 Finally, Tregs are an important

candidate for tolerance induction in transplantation.321

1.3.5.3.2 Mechanisms of Treg suppression

Tregs are considered major regulators of the immune system due to their broadly acting

suppressive functions. These functions can be grouped into four major mechanisms of

action; i) inhibition of APC maturation and function, ii) cytolysis of target cells, iii)

metabolic disruption of effector T cells (Teff), iv) secretion of immunosuppressive

factors. These mechanisms have been extensively reviewed in the literature by us and

other groups.318,328,333–335 One salient feature of Tregs that is important to note is that

unlike activated T effector cells, Tregs do not produce IL-2. Instead, they rely on

paracrine IL-2 signaling and genetic ablation of either IL-2 or IL-2R in mice results in a

50% reduction in the proportion of Treg thymocytes.320 The major mechanisms of Treg

immunosuppressive function are outlined in Figure 1-2 below. Moreover, the major cell

surface receptors and immunosuppressive molecules produced by Treg cells and their

effects are presented in Table 1-2.328

Page 62: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

49

Figure 1-2 Treg-mediated mechanisms of immunosuppression

The major mechanisms of Treg function include APC modulation, direct cytolysis of target

cells, metabolic disruption of effector T (Teff) cells and secretion of a number of

immunosuppressive molecules. Copyright© Chruscinski et al. 2015.

Reprinted from Rambam Maimonides Med. J. 6, e0024, Role of Regulatory T Cells (Treg)

and the Treg Effector Molecule Fibrinogen-like Protein 2 in Alloimmunity and Autoimmunity,

Chruscinski, A. Sadozai, H et al., (2015).328

Page 63: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

50

Table 1-2 Treg effector molecules

Cell Type Ligand/

Receptor

Target Cell Mechanism

CTLA-4 Treg B7 molecules

(CD80/CD86)

DC Inhibition of DC activation through the trans-endocytosis

and degradation of CD80 and CD86 molecules by Treg

Sterically hinders the association of naïve T cells with DC

through coordinated activity with LFA-1

Negative regulation of effector T cell survival by signaling

through Foxp3

IL-2 Activated

T cells

High affinity

IL-2 receptor

Treg IL-2 deprivation by Treg in low-affinity TCR and antigen:MHC

interactions induce T cell apoptosis

TIGIT Treg,

T cells

NK cells

CD155 (PVR)

CD112

(PVRL2)

DC Inhibition of IL-12 (p40) production by DC

Binds CD155 (PVR) and CD112 (PVRL2) on APCs

Increases IL-10 expression inducing tolerogenic DC which

suppress T cell proliferation and IFNγ production

LAG-3 Treg MHC-II DC Inhibits DC maturation

Inhibits co-stimulation of naïve T cells by DC

CD39

/CD73

Activated

Treg

Treg Activated

T cells

DC

CD39 converts ATP in the extracellular space into ADP and

AMP decreasing inflammation

CD39 increases suppressive activity of Treg

CD73 converts AMP to adenosine which inhibits DC function

and activated T cells

IL-10 Treg IL-10R T cells

DC

Inhibits T cell proliferation, decreases production of IL-2,

TNF-α and IL-5

Impairs Th1 responses by inhibiting DC activation and

secretion of IL-2

TGF-β Treg TGF-βR T cells Direct suppression of effector T cells

Inhibits cytokine production and cytotoxic function of T cells

IL-35 Treg IL-35R Naïve T cells,

DC

Direct inhibition of T cell proliferation

Induction of naïve T cells to become activated IL-35 Treg

Gzmb Treg Perforin-

independent

entry into

target cell

Activated

T cells

DC

Induction of apoptosis in target cells

FGL2 T cells,

Treg,

activated

Treg

FcγRIIB/ RIII DC Inhibition of DC maturation

Suppression of Th1 and Th17 effector T cell responses

Abbreviations: ADP, adenosine diphosphate; AMP, adenosine monophosphate; APC, antigen presenting cell; ATP,

adenosine triphosphate; CTLA-4, cytotoxic-T-lymphocyte-associated protein 4; DC, dendritic cell; FGL2, fibrinogen-

like protein 2; Foxp3, forkhead box p3; Gzmb, granzyme B; IL, interleukin; LAG-3, lymphocyte activation gene 3; LAT,

linker for activation of T cells; LFA-1, lymphocyte function-associated antigen 1; MHC, major histocompability

complex; PD-1, programmed cell death-1; PVR, poliovirus receptor; PVRL, poliovirus receptor ligand; TCR, T cell

receptor; TGF, Transforming Growth Factor; TIGIT, t cell immunoreceptor with Ig and ITIM domains.

Page 64: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

51

1.4 Strategies to Induce Transplant Tolerance

1.4.1 Overview

The seminal work of Billingham, Brent and Medawar demonstrated that immune

tolerance in the absence of immunosuppression was theoretically possible.227 Their

approach was based on work performed by Ray Owen in the 1940s, who showed that

freemartin (dizygotic) twin cattle had shared placental circulation and chimeric immune

systems.336 Billingham and Medawar among others, demonstrated that these dizygotic

twins could accept skin grafts from each other.336 However, induction of tolerance in

their murine studies was considered a purely theoretical approach not deemed

applicable to clinical immune tolerance. However, research in the past two decades

have greatly furnished our understanding of human immune function, particularly in the

activation of T cells and the immunobiology of suppressor cell types.243,337 Moreover,

our understanding of hematopoiesis and immune system development, has allowed for

approaches based on Medawar’s initial concept of neonatal acquired tolerance.338

Currently, multiple strategies are being employed to induce long-term allograft specific

tolerance in the clinic. These strategies can be divided into three major approaches for

tolerance induction. These are i) co-stimulatory blockade ii) administering or inducing

Treg iii) hematopoietic stem cell transplantation.282,318,337,339,340 This section will discuss

all three major approaches with a primary focus on therapies that have gone or are

currently undergoing clinical trials for tolerance induction in solid organ transplantation.

Page 65: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

52

1.4.2 Co-stimulatory blockade

Due to the central role of co-inhibitory and co-stimulatory receptors in T cell

activation, they are an attractive target for tolerance inducing regimens. As mentioned

previously, these molecules belong to either the Ig family (e.g. CD28, CTLA-4, PD1),

the TIM family (T cell Ig and mucin domain), the TNF-TNFR family (e.g. CD40, OX40) or

are cell surface adhesion molecules (e.g. LFA-1).99,337 The first well-characterized

regulatory axis was that observed between CD28, CTLA-4 and CD80/86.101 As

mentioned previously, Belatacept (CTLA-4Ig) has recently been demonstrated to have

efficacy over CsA but its use alone cannot induce tolerance..215,217 Furthermore, two

important considerations argue against the value of this approach in favour of a CD28

selective blockade. First, CTLA-4 signaling is also important for Treg function and

second, that CD80 was recently found to bind to PD-L1 on T cells resulting in

inhibition.320,341 Thus, the selective CD28 agonist TGN1412 was developed and tested

in a phase I safety trial in healthy volunteers. However, this resulted in a cytokine storm

in all patients marked by severe systemic inflammatory symptoms and all patients

required intensive care till the symptoms subsided.342 Subsequent studies are now

being conducted with engineered anti-CD28 antibodies that preserve CTLA-4 function

while selectively blocking CD28. These studies have shown to promote Tregs and

prolong allograft survival in murine and NHP preclinical models.337

There is also interest in targeting the CD40-CD154 pathway for tolerance

induction in transplantation due to its crucial role in promoting both cellular and humoral

immunity.343 Targeting CD154 was demonstrated to promote long-term graft survival in

Page 66: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

53

both mice and NHP studies.344 Human trials with anti-CD154 were quickly halted as it

was not yet known that platelets express CD154 and that administration of anti-CD154

resulted in thromboembolic complications in these patients.345 Targeting CD40 with

novel non-depleting mAbs has showed efficacy in NHP models of renal, cardiac and

islet transplantation.346 Currently, an anti-CD40 mAb (ASKP1240) is undergoing phase

IIa clinical trials in kidney transplant recipients.346

Cell surface adhesion molecules are crucial for cell trafficking to sites of

inflammation. The adhesion molecule LFA-1 (lymphocyte function associated antigen-

1), is expressed on memory T cells. It is purported to have a role in T cell activation in

the immunological synapse and T cell migration into tissues by binding to ICAM

(intracellular adhesion molecule) on inflamed endothelium.347 Efalizumab (anti-LFA-1

mAb) was first approved for psoriasis and underwent clinicial trials in islet and renal

transplant recipients. Initial results demonstrated encouraging results including insulin-

free survival and an increased Treg signature when used in combination with

sirolimus.337 However, further development for transplantation and autoimmune disease

was halted after the development of progressive multifocal leukoencephalopathy (PML),

a fatal disorder associated with the JC polyomavirus.337 An additional focus has been to

disrupt the pathway between the T cell surface antigen CD2, expressed highly on

effector memory T cells with LFA-3 (lymphocyte functional associated antigen 3).

Alefacept, a LFA-3 fusion protein has been shown to prolong allograft survival in

synergy with anti-CD154 treatment in NHP kidney transplant recipients.348 However,

subsequent studies in NHP models in the absence of anti-CD154 treatment did not

demonstrate the same efficacy.337 Other pathways that have shown promise in

Page 67: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

54

preclinical transplant tolerance studies are those associated with PD-1 and inducible T

cell co-stimulator protein (ICOS).337,346 ICOS is of particular interest as it has been

shown to play a critical role in TH17 differentiation, which is now being recognized as a

major subset involved in the alloresponse.106,349 To date, none of the co-stimulatory

blockade treatments tested clinically have resulted in organ-specific immune

tolerance.337 Further study is required to unravel the complex interplay between each of

these co-stimulatory and co-inhibitory pathways to generate rational and potentially,

combinatorial targeting of these receptors.

1.4.3 Induction or administration of Tregs

Research in the past two decades have unveiled the crucial role of Tregs in

promoting and maintaining allograft tolerance.282,321 The two approaches currently being

studied to utilize Tregs for transplant tolerance are induction of Tregs in vivo through

pharmacological modulation and infusion of ex vivo generated Tregs.350 Treatment with

mTOR inhibitors in humans have been shown to expand CD4+CD25+FOXP3+ Tregs in

diabetes patients and healthy volunteers.199,351 In the context of solid organ

transplantation, Levitsky et al. demonstrated that conversion from tacrolimus to

sirolimus in liver transplant recipients resulted in increased Tregs in peripheral blood

and bone marrow.211 Sirolimus treated patients also displayed an immunoregulatory

gene signature and diminished MLR responses.211 A similar expansion of Tregs was

recently reported in a renal transplant cohort converted from CNIs to everolimus, an

analog of rapamycin.352 Other candidate drugs for the expansion of Tregs include IL-2,

Page 68: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

55

ATG and the aforementioned LFA-3 fusion protein, Afelacept. However, to date, none of

these have been tested for Treg expansion in clinical trials of solid organ

transplantation.350 Furthermore, the major focus of current clinical Treg therapy for

transplantation is on the transfer of ex vivo generated Tregs.282 In 1993, Qin et al. first

demonstrated that the capacity to adoptively transfer tolerance in murine allografts, i.e.

“infectious tolerance” involved a CD4+ T cell population.353 Recent studies have now

shown that transfer of Tregs can treat both acute and chronic allograft rejection, as well

as, prevent GVHD (graft-versus-host-disease) in murine models.318,321 Initial studies in

small human cohorts demonstrated that infusion of Tregs could prevent the induction of

GVHD in patients after allogeneic hematopoietic stem cell transplantation (HSCT).354

These patients received either ex vivo derived natural, or thymic Tregs (tTregs) or

umbilical cord blood purified Tregs. These studies were too preliminary to analyze

efficacy of treatment but they demonstrated the safety of performing Treg infusion in

human patients.355 The ONE study is a multi-center phase I/IIa trial that commenced in

2014, with the aim of examining autologous ex vivo expanded Tregs in renal transplant

patients (www.onestudy.org).356 The study will also test other regulatory cell types such

as Tr1 cells, TolDCs and regulatory macrophages. Participants will be followed for 60

weeks and biopsy-proven rejection has been established as the clinical endpoint for this

trial.356 This study will yield crucial information about the feasibility and efficacy of Treg-

based cell therapy for tolerance induction in transplantation. The primary concerns for

using ex vivo Tregs are the cost-effectiveness of GMP manufacture of the Treg product,

the shelf-life of manufactured products and finally, the stability of the Treg phenotype in

vivo.355

Page 69: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

56

1.4.4 Hematopoietic stem cell transplantation

Both autologous and allogeneic hematopoietic stem cell transplantation (HSCT)

have been implemented clinically for the purposes of tolerance induction (Table 1-

3).338,357–359 Based on the concept of mixed chimerism, combined kidney and bone

marrow/HSCT has been examined as a tolerance inducing regimen in limited numbers

of patients.340,360 Alternatively, based on the preclinical observations of Van Bekkum et

al., autologous HSCT has been utilized for nearly two decades to abrogate various

types of autoimmune disease.361,362 Although some of the putative tolerogenic

mechanisms differ between allogeneic and autologous HSCT, both treatments

represent an attempt to induce tolerance by “re-education” of the recipient’s immune

system.339,340 As the use of autologous and allogeneic HSCT for malignant disease has

been reviewed extensively elsewhere, this section will focus on HSCT for tolerance

induction in human patients.363–365

Page 70: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

57

Table 1-3 Overview of Autologous versus Allogeneic HSCT

1.4.4.1 Hematopoietic stem cells

The bombings of Hiroshima and Nagasaki, and the dawning of the atomic age

revealed the intense myeloablative effects of ionizing radiation.366 It was later observed

that allogeneic bone marrow transplantation could rescue lethally myelosuppressed

mice and humans.366 In a series of seminal studies starting in 1961, Till and McCulloch

demonstrated that the bone marrow consisted of a sub-population of cells with the

ability to give rise to multiple myeloerythroid cell types and to self-renew.367–369 These

AUTOLOGOUS HSCT ALLOGENEIC HSCT

INDICATIONS Myeloma

Non-Hodgkin Lymphoma

Autoimmune disease

Acute myeloid leukemia

Acute lymphoblastic leukemia

Fanconi Anaemia

Organ Transplantation

CONDITIONING

REGIMENS

Chemotherapy+/irradiation Chemotherapy+/irradiation

STEM CELL

SOURCES

Autologous Peripheral Blood

Stem Cells

Allogeneic Peripheral Blood Stem

Cells

Cord Blood Stem cells

COMPLICATIONS Treatment-related mortality and

Toxicity

(NO GVHD)

Disease Relapse

Infectious Disease

Treatment-related mortality and

Toxicitity

(GVHD)

Disease Relapse

Infectious Disease

Page 71: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

58

cells were termed hematopoietic stem cells (HSCs) and are now defined by their

multipotent capacity to generate all of the hematopoietic lineages and to self-replicate

into daughter cells.370 Hematopoiesis is a highly regulated hierarchical process with

HSCs at the apex of that hierarchy as the multipotent progenitors of all hematopoietic

lineages (Figure 1-3).366,371 According to canonical understanding, these multipotent

cells follow a stepwise development into oligopotent and finally unipotent cell types that

give rise to all the lineages.366 However, in 2015, Notta et al., provided evidence against

the existence of the oligopotent common myeloid progenitor (CMP) stage in both mice

and human bone marrow which is believed to give rise to myeloid, erythroid and

megakaryocyte lineages.372 This study re-defined classical views of hematopoietic

development and further study is required to elucidate the potential heterogeneity within

each major progenitor cell type for a more accurate map of hematopoiesis.

Another major focus of research in immunology has been the identification of cell

surface markers for HSCs in humans and mice. Early studies identified a population of

cells with potent HSC capacity in vivo that were termed LSK and constituted about

0.05% of adult mouse bone marrow. These cells lacked expression of lineage-specific

markers (e.g. B220,CD3,Gr-1,Ter119) and displayed stem cell antigen 1 (Sca1) and the

trans-membrane tyrosine kinase c-kit (CD117), the receptor for stem cell factor also

known as steel factor. 373,374 LSK cells nevertheless, are a heterogeneous population of

long-term (LT-), intermediate-term (IT-) and short-term (ST-) HSCs in addition to

multipotent progenitors (MPPs) (Figure 1-3). These represent three distinct populations

classified by their repopulating activity and longevity in mice.371 The SLAM (signaling

lymphocytic activation molecule) family of glycoproteins have been used to identify LT-

Page 72: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

59

HSCs.375,376 LT-HSCs comprise 10% of the LSK population and are

CD150(slamf1)+CD48-CD49blo.371 Whereas murine HSCs lack expression of CD34, in

humans, CD34 expression delineates cells with HSC and MPP activity.371,377 In 2011,

Notta et al. demonstrated that that single cells from the CD34+CD90+CD49f+

phenotypic subset could fully reconstitute humanized NOD-SCID-IL2RyKO (NSG)

recipient mice providing proof of a LT-HSC population in humans.378

In clinical medicine, bone marrow obtained from the iliac crest was the first

source of HSCs.379 However, due to the highly invasive nature of this procedure, there

was a shift towards using peripheral blood mobilized stem cells (PBSCs) in the

1990s.380 This technique involves treatment with chemotherapy along with G-CSF to

promote stem cell entry into peripheral circulation. Studies have shown that treatment

with AMD3100, an inhibitor of the chemokine receptor (CXCR4), which is involved in

HSC homing to the bone marrow, is more effective than G-CSF alone.381,382 PBSCs are

then isolated using magnetic-activated cell sorting (MACS) for CD34+ cells.383 Several

studies have demonstrated the safety and efficacy of this treatment.383 However, bone

marrow transplantation remains common in the pediatric population due to improved

outcomes versus PBSCs.357

Page 73: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

60

Figure 1-3 Schematic of mouse and human hematopoeitic development

A comparative depiction of surface markers of major cell types in the hematopoietic hierarchies

in both mice and humans. Recent evidence generated by Notta et al. will require redefining

classically accepted hierarchies.372 This schematic nevertheless, remains useful for highlighting

the differences between murine and human hematopoietic lineages. CMP – common myeloid

progenitor, ETP – early thymic progenitor, GMP – granulocyte/macrophage progenitor,HSC-

hematopoietic stem cell, LMPP- lymphoid primed multipotent progenitor, MEP –

megakaryocyte/erythrocyte progenitor, MPP – multipotent progenitor, MLP – multi-lymphoid

progenitor. Reprinted from Cell Stem Cell, Volume 10. Doulatov et al., Hematopoiesis: A

Human Perspective, Pg. 124. (2012) with permission from Elsevier.371

Page 74: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

61

1.4.4.2 Allogeneic HSCT and mixed chimerism

Allogeneic bone marrow transplantation (BMT) was first used to treat primary

immune deficiencies and cancer.384 In the past thirty years, a few groups have also

attempted to utilize allogeneic HSCT to induce solid organ transplant tolerance.360 In

1984, Ildstad and Sachs demonstrated acceptance of allografts in mice treated with

total body irradiation and reconstituted with mixed bone marrow from syngeneic and

allogeneic strains of mice.385 GVHD was avoided through use of T-cell depleted bone

marrow. In 1989, Sharabi and Sachs used a non-lethal conditioning regimen using T

cell depleting mAbs along with local thymic irradiation, to induce long-term mixed

chimerism and graft acceptance.386 Studies in mice utilizing the mixed chimerism

approach have shown an indispensable role for intra-thymic deletion of donor-reactive T

cells.340 Further tolerization is posited to occur through the various peripheral self-

education mechanisms.340 Studies in NHP models demonstrated that allograft

tolerance could be established but mixed chimerism was transient potentially due to the

large repertoire of memory T cells in NHPs versus in-bred laboratory mice.387 This

indicates a potential role for regulatory mechanisms in maintaining tolerance in NHP

models after mixed chimerism is lost.360 Studies in NHP models also presented

evidence for the utility of co-stimulatory blockade in enhancing but not establishing long-

term mixed chimerism.388

Currently, three groups have examined the potential of mixed chimerism for

tolerance induction in clinical transplantation utilizing three different protocols. At

Page 75: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

62

Stanford, total-lymphoid irradiation (TLI) and ATG was used to induce mixed chimerism

in HLA-matched kidney transplant recipients. Of the 16 patients receiving this treatment,

15 developed long-term mixed chimerism (>6 months) and 8 patients were successfully

withdrawn from all IS between 6-12 months post-HSCT.389 In the most recent report by

this group, it was shown that out of 22 patients that received the protocol, 18 patients

who established persistent mixed chimerism (>12 months), were successfully withdrawn

from IS and no incidence of GVHD or rejection was observed for up to a 7 year post-

withdrawal observation period.390 The same protocol was tested in HLA-mismatched

kidney transplant recipients but did not result in tolerance rendering this approach

currently unsuitable for non-HLA identical organ transplants.360,391 A second protocol

was established at the Massachusetts General Hospital (MGH) based on a non-

myeloablative conditioning regimen involving cyclophosphamide and thymic

irradiation.392 This trial was performed in 10 HLA-mismatched kidney transplant

recipients with varying underlying indications for transplant. 7 out of 10 patients were

successfully withdrawn from IS after 8-14 months. Mixed chimerism was transient and

non-detectable after 21 days of HSCT with only 4 patients showing longer (<105 days)

lasting mixed chimerism. Nevertheless, treated patients demonstrated reduced donor-

specific MLR and CTL activity in vitro with elevated mRNA levels of FOXP3 in the

graft.392 The long-term follow-up to this patient cohort shows mixed results for tolerance

induction. As of the last report in 2014, only 4 out of 10 patients remained IS-free

without any complications, with one patient IS-free for a period of 10 years.393 While

there was no serious opportunistic infections, 3 patients were required to re-start IS

after 6-8 years as they started to show histological signs of rejection and 3 patients

Page 76: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

63

experienced graft loss due to rejection or thrombotic microangiopathy.393 All patients

suffered from HSCT-associated engraftment syndrome that causes fever, skin rash and

diarrhea.394 A third clinical protocol was developed at Northwestern University based on

a high-intensity conditioning regimen using cyclophosphamide, fludarabine and total

body irradiation (TBI- 200 cGy).395 Of note, this trial involved manipulation of the PBSC

grafts by the company Regenerex LLC, to enrich for HSCs and a population of CD8+

facilitating cells (FCs). The full phenotype of these proprietary FCs are hitherto

undisclosed. As of 2015, this group has reported that 12 patients out of a cohort of 19,

had been withdrawn from IS for between 8 months to 48 months without incidence of

acute rejection.396 The high-intensity nature of this regimen resulted in stable donor

chimerism in the recipients with only one patient developing stable mixed chimerism.

Although only one patient reportedly developed GVHD, this treatment was associated

with severe neutropenia and a high incidence of opportunistic bacterial or fungal

infections.396 The 3 clinical approaches for tolerance induction using mixed chimerism

have demonstrated its utility for prolonging graft survival in limited numbers of patients.

However, the results from all 3 studies are mixed with some patients undergoing

rejection episodes or requiring IS after withdrawal.397 The primary successes of this

technique appear to be associated with the length of the mixed chimeric state. After

mixed chimerism is lost, there is a potential for the activation of Treg-mediated

tolerogenic mechanisms as demonstrated by the elevated FOXP3 expression in the

graft in the MGH cohort.392 Although, there was not significant incidence of GVHD

associated with any of the 3 protocols, stable allogeneic chimerism always poses the

risk for GVHD.397 Finally, finding HLA-matched allogeneic donors is an additional

Page 77: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

64

challenge associated with the mixed chimerism approach. These considerations warrant

that alternative mechanisms for immune re-education be investigated as well.

1.4.4.3 Autologous HSCT

In 1989, Van Bekkum et al. studied autologous BMT as a treatment for adjuvant-

induced arthritis in rats.361 Due to the technical complications of autologous HSCT in

rodent models, bone marrow from syngeneic or congenic (CD45.1 vs. CD45.2) mice is

utilized and deemed to be an autologous HSCT.398 As the authors noted in the rat

model, treatment with TBI and syngeneic bone marrow transfer resulted in significant

reduction in arthritis severity. As a control for TBI, a group of rats was also treated with

CsA but did not cause significant recovery from arthritis versus the autologous BMT

group.361

As a result of work with rodent models, autologous HSCT was quickly adopted as

a treatment for patients with treatment-refractory autoimmune disease (AID). It is

estimated that as of 2011, nearly 3000 patients worldwide had been treated with

autologous HSCT for AID.399 Currently, conditioning regimens for autologous HSCT are

highly varied and are generally classified into the following categories, i) “high intensity”

regimens involving the use of high doses of busulphan or TBI, ii) “low-intensity”

regimens involve fludarabine-based, or cyclophosphamide-alone regimens,

iii)“intermediate-intensity” chemotherapeutic regimens such as BEAM (carmustine,

etoposide, cytarabine and melphalan) or cyclophosphamide in combination with ATG

primarily for rheumatological conditions (e.g.SSc, SLE).400 Clinical trials of autologous

Page 78: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

65

HSCT have been performed in a wide range of autoimmune pathologies including

multiple sclerosis, systemic sclerosis (SSc), SLE, Crohn’s disease, juvenile arthritis and

T1D.400,401 These trials have shown treatment safety and efficacy with an increased

patient quality of life and stabilization of disease.400,402 Recent clinical trials have shown

significant abrogation of autoimmune disease and have shown that autologous HSCT is

superior to current standard of care for those diseases. In 2011 Burt et al., reported the

efficacy of non-myeloablative autologous HSCT in systemic sclerosis.403 Ten out of

nineteen patients treated with HSCT had improved modified Rodnan skin scores

(mRSS) and pulmonary function as measured by forced vital capacity, compared to the

9 cyclophosphamide-alone treated control arm patients. Furthermore, 8 out of 9 control

arm patients had disease progression compared to none in the HSCT-treated arm.403

Another seminal clinical trial was recently performed in MS patients by our collaborator

Dr. Harold Atkins at the Ottawa Hospital Research Institute.404 This phase II, single-arm

trial included 24 patients (aged 18-50) who had progressive disease and poor

prognosis. Patients were treated with busulphan, cyclophosphamide and ATG followed

by autologous HSCT and monitored for a median period of 6.7 years post-HSCT. Prior

to HSCT, the cohort of 24 patients had 167 clinical relapses over 140 patient-years and

188 gadolinium-enhancing lesions over 48 MRI (magnetic resonance imaging) scans.404

Post-HSCT, there were no new T2-weighted or Gd lesions over 188 MRI scans and no

clinical relapses for a follow up period of up to 12 years. The Expanded Disability Status

Scale (EDSS) score was improved in 35% of patients. HSCT-associated mortality was

low with 1 out of 24 patients succumbing to treatment-related toxicity.362 This study was

the first evidence of long-term disease abrogation in MS due to autologous HSCT.

Page 79: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

66

Although this clinical study did not report the immunological changes associated with

HSCT, other groups have studied potential immune potential mechanisms of immune

re-education associated with autologous HSCT. One of the primary mechanisms is the

re-setting of the immune system with a shift from memory T and B cells to newly

emerging naive cells.400,405 Recently, Muraro et al. performed high-throughput

sequencing of the TCRβ chain before and after HSCT in patients being treated for

MS.359 This study showed that while CD4+ repertoires were almost entirely renewed

post-HSCT, CD8+ were not effectively depleted and reconstituted the CD8+

compartment.359 In 2016, Delemarre et al. also provided evidence of a crucial effect of

autologous HSCT on the Treg compartment. By comparing Treg TCR diversity pre- and

post-HSCT, the authors demonstrate that Treg TCR diversity was significantly

increased after HSCT.406 The primary limitations of this study were that it was

performed in a pediatric AID patient population for patients suffering from juvenile

arthritis and dermatomyositis, and the patient cohort was small (n=4). Further studies in

adults and various time points post-HSCT will be required to uncover the role of Treg

diversity in abrogating autoimmunity post-HSCT. Nevertheless, this study provides

additional proof of the potent immune re-education capacity of autologous HSCT.

Another important phenotypic observation in AID patients receiving autologous HSCT is

a significant increase of CD4+CD25+FOXP3+ Tregs in patients compared to pre-

transplant levels.407,408 In 2013, Abrahamsson et al. performed multiparameter

immunophenotyping in MS patients receiving non-myeloablative HSCT.408 In addition to

a transient but significant increase in CD4+FOXP3+ Tregs, the study also unveiled

perturbations in other cell populations that may be important for promoting tolerance.

Page 80: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

67

These involved a significant increase in CD56high NK cells that have putative

immunoregulatory roles and a pronounced depletion of CD8+CD161high.408

CD8+CD161high cells represent a population termed mucosal-associated invariant T cells

that are found to infiltrate the brain in MS patients, and produce high levels of the pro-

inflammatory cytokines IFNγ and IL-17.408,409 These results indicate that tolerance

induction and disease-abrogation post-HSCT in these patients is likely a multifactorial

process that results from a system-wide resetting of the immune system. In 2014, a

clinical report also described that HSCT in SSc patients also resulted in the emergence

of a putative Breg population designated as CD19+CD24highCD38high at 6 and 12 months

post-transplant.410 This was also associated with a significant reduction in B memory

cells (CD19+CD27+IgD-) coinciding with an increase in naive (CD19+CD27+IgD+) B cells

.411 Further studies from this group are yet to be reported. In particular, the

immunological functions of this Breg population will need to be elucidated in the context

of AID. The evidence from clinical studies of autologous HSCT indicate several

phenotypic and functional alterations that can play a role in tolerance induction. These

studies provide a rationale for expanding its use in AID and potentially in organ

transplantation. The primary complications of autologous HSCT are treatment-related

mortality, which can be potentially as high as 15%, as well as treatment-related

morbidity including opportunistic infections, infertility and risk of malignant diseaase.358

The major advantages of this approach however are that there is no risk of GVHD and

that autologous PBSCs are readily available and easily stored.

At the Multi-Organ Transplant Program of the University Health Network, we

observed a clinical case of a patient who underwent autologous HSCT for underlying

Page 81: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

68

malignant disease and subsequently maintained stable graft function after IS withdrawal

(Levy, unpublished data). This resulted in the establishment the ASCOTT study, a

phase I clinical trial in liver transplant patients who will undergo autologous HSCT for

tolerance induction (NCT02549586). In parallel to the clinical study, there is a need for a

preclinical murine model to provide mechanistic proof-of-concept of the utility of this

technique in SOT. The establishment and investigation of the mouse model is the focus

of this dissertation.

Page 82: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

69

Hypotheses and Aims

The overall aim of this thesis project was to establish a murine model of autologous

HSCT and examine its potential for tolerance induction in a murine allograft model.

Previously, in our lab we established the heterotopic cardiac allograft model first

described by Corry et al.130,206 The specific hypothesis that we tested in this thesis was

as follows; Tolerance will develop to MHC-mismatched cardiac allografts in mice

following autologous HSCT.

The aims of this study were to:

1. Establish a mouse model of autologous HSCT which reconstitutes all hematopoietic

compartments in the recipient.

2. Examine the potential of HSCT to induce tolerance in a murine model of heterotopic

cardiac transplantation.

3. Elucidate immunological mechanisms post-HSCT in allograft recipient mice.

Page 83: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

70

Materials and Methods

3.1 Mice

C57BL/6J (H-2b), B6.CD45.1 (Ptprca, H-2b), BALB/cJ (H-2d) and C3H/HeJ (H-2k) were

purchased from the Jackson Laboratory (Bar Harbor, ME) and were kept in specific-

pathogen free housing at the University Health Network Animal Resource Center.

Animals were treated in accordance with guidelines set by the Canadian Council for

Animal Care and the University Health Network. All mice used were female and

between 6 to 12 weeks of age. CD45.1 and CD45.2 (BL/6) congenic mice were used as

HSC donor and recipient respectively so that post-HSCT hematopoietic lineages could

be tracked and to determine degree of chimerism . This allows for analysis of pre- and

post-HSCT hematopoietic populations.

3.2 Heterotopic cardiac transplantation

Intra-abdominal heterotopic cardiac transplantation was performed as previously

described by Corry et al.130 All surgeries were performed by microsurgeon Dr. William

(Wei) He from the Levy lab. Briefly, six week old donor BALB/cJ mice were injected i.p.

with pentobarbital and placed under an operating microscope (25x). 1ml of 300U

heparin was then injected into the inferior vena cava. Donor hearts were harvested by

ligating the inferior vena cava (IVC), the superior vena cava and the azygous vein with

6-0 silk sutures and dividing them superior to the ligatures. The ascending aorta and

pulmonary artery were then separated and the pulmonary artery was transected at the

Page 84: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

71

point of bifurcation. The heart was then excised and placed in a 4°C cold saline solution

(Baxter Healthcare, IL).

Recipient 12-week-old recipient C57BL/6J mice were anesthetized with an i.p. injection

of pentobarbital and placed under the operating microscope. A long midline abdominal

incision was made and the contents of the abdomen were moved aside and covered

with gauze, exposing the abdominal aorta and IVC. The abdominal aorta and IVC were

mobilized and clamped off at the point of bifurcations of the renal and iliac vessels. The

lumbar vessels were ligated with 10-0 sutures. The donor’s aorta was anastomosed

with the recipient’s abdominal aorta. In similar fashion, the donor’s pulmonary artery

was sutured to the recipient’s IVC. Afterwards the clamps on the recipient’s abdominal

aorta and IVC were removed. This allows for retrograde blood flow from the recipient

aorta to the donor coronary arteries draining into the donor right atrium where it is

pumped into the right ventricle and eventually via the donor pulmonary artery to the

recipient IVC.

Graft function was assessed daily via manual palpation and scored for beating rate and

strength on a scale from 0-3 with 0 being a total cessation of graft function. Grafts that

continued to beat ≥ 70 days post-transplant were considered to be accepted. After 70

days, or at cessation of graft beating, mice were sacrificed and graft function was

verified by direct visual examination. Grafts were subsequently harvested for

histological studies. Prior to sacrifice, saphenous vein blood was collected in EDTA

coated capillary tubes and submitted for complete blood counts using the Hemavet

950FS (Drew Scientific, Miami Lakes, FL).

Page 85: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

72

3.3 Flow cytometry

Flow Reagents. The following anti-mouse mAbs were utilized for FACS analyses of

reconstitution studies. All mAbs were purchased from Biolegend (San Diego, CA):

Brilliant Violet™ (BV) 650 anti-CD45.1 (clone-A20), BV785 anti-CD45.2 (clone-104),

BV510 anti-CD3ε (clone-145-2C11), BV605 anti-CD19 (clone-6D5), PerCP/Cy5.5 anti-

CD4 (clone-GK1.5), APC/Cy7 anti-mouse CD8α (clone-53-6.7). For Treg staining, the

following anti-mouse mAbs were used: FITC anti-CD4 (clone-GK1.5), APC anti-CD25

(clone-PC61), PE anti-Foxp3 (clone-150D). In all flow cytometric studies, Fc blocking

was performed using a mAb to mouse CD16/32 (TruStain FcX™ clone 93) from

Biolegend. For all studies viability was assessed by staining with fixable viability dye

eFluor®450 (eBioscience, San Diego, CA) also referred to as pacific blue.

Cell Suspensions. Single cell suspensions of spleen and bone marrow cells were

prepared as follows. Spleens were mechanically disassociated on a 40 µm nylon filter.

Bone marrow was obtained from crushed femurs and tibias. Subsequently, red blood

cell lysis was performed using RBC lysis buffer (eBioscience) followed by a final

filtration step through a 40 µm filter. For surface staining, 1x106 spleen or bone marrow

cells were re-suspended in 100 µL of FACS buffer (1XPBS supplemented with 1%FBS

and 5 mM EDTA) and incubated with flow mAbs at 4°C for 30 minutes (min). After

surface staining, cells were washed twice and incubated with pacific blue viability dye

for 30 min at 4°C. Finally, cells were fixed using 2% paraformaldehyde (PFA), washed 2

times and re-suspended in 400 µl of FACS buffer for analysis. For staining of peripheral

blood, 100 µl of saphenous vein blood was treated initially with RBC lysis buffer

Page 86: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

73

(eBioscience). After stopping the reaction, cells were surface stained and followed by

staining with pacific blue viability dye. Finally, the cells were placed in 1X Fix/Lyse buffer

(eBioscience) to further lyse red blood cells and fix cells for further analysis. Cells were

washed 2 times and re-suspended in 400 µl of FACS buffer for analysis.

Analysis. Single cell suspensions were analyzed on a BD LSRII flow cytometer (BD

Bioscience, Franklin Lakes, NJ). Data were analyzed using the program FlowJo version

9.6 (Treestar Inc. Ashland, OR). For all analyses, cells were gated on singlets, live cells.

Lymphocytes were gated on using forward and side scatter.

3.4 Purification of LSK cells

LSK isolation protocols were adapted from earlier studies.412,413 Femurs and tibias were

harvested from four 12 week old donor CD45.1 female mice. Bone marrow was

obtained by crushing femurs in 10cm tissue culture dishes (Sarstedt AG&Co,

Numbrecht, Germany) containing 10ml PBS using a glass mortar. Crushed femurs were

washed by centrifuging 1200RPM for 5 min in α-modified Eagle’s Medium (Invitrogen,

Carlsbad, CA) with 10% fetal bovine serum (ThermoFisher, Waltham, MA) and 0.5 µM

2-mercaptoethanol (Sigma-Aldrich, St. Louis, MO). The pellet was filtered through a 40-

µm nylon filter and centrifuged as above. Subsequently the cell pellet was re-suspended

in 4 mL of RBC Lysis Buffer (eBioscience, San Diego, CA) and incubated at room

temperature (RT). After 5 min, the reaction was stopped and cells were counted prior to

lineage depletion.

Page 87: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

74

Lineage negative (Lin-) cells were enriched by labelling bone marrow cells with the

mouse lineage cell depletion kit (Miltenyi Biotec, Bergisch Gladbach, Germany) followed

by magnetic separation using the autoMACS® Pro Separator (Miltenyi Biotec). Lin- cells

were counted and stained with APC anti-mouse CD117 (c-kit) antibody (clone 2B8-

Biolegend, San Diego, CA) and PE anti-mouse Ly-6A/E (Sca1) antibody (clone D7-

Biolegend). As the lineage cell depletion kit contains biotinylated mAbs, FITC

Streptavidin was utilized to assess enrichment pre- and post-lineage depletion. A 10-

fold enrichment in Lin- cells was observed post-lineage depletion (Fig 4-1). After 30 min

incubation at 4°C, cells were washed twice and re-suspended in 200 μL of FACS buffer.

5 µl of the viability dye Propidium Iodide (PI) was added to the cells, and the sample

was submitted the Sickkids-UHN Flow Cytometry Facility for LSK sorting using a BD

FACSAriaII cell sorter (BD Biosciences, San Jose, CA).

3.5 Hematopoietic stem cell transplantation

Sorted LSK cells were washed 2 times with PBS and counted using a hemocytometer.

CD45.2 B6 recipient mice were lethally irradiated at a dose of 13 Grays (Gy) using a

GAMMACELL® 40 cesium-137 irradiator (Best Theratronics, Ottawa, ON). The dose

was fractionated into two doses of 6.5 Gy. Within 2h of the final irradiation recipient

mice received intravenous (i.v.) injections of 200 µl of PBS containing 5000 viable

CD45.1 LSK cells. HSCT-treated mice were weighed weekly and were monitored daily

for morbidity and mortality.

Page 88: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

75

3.6 Treatment groups

The HSCT treatment protocol was adapted from earlier studies for use in our murine

heterotopic heart transplant model.414 Recipient CD45.2 BL/6 female mice (12 weeks of

age) were allografted as described above. To prevent acute allograft rejection, allograft

recipients were treated with rapamycin (Pfizer, New York City, NY) at a dosage of 2

mg/kg per day for a total of 14 days starting on day -1 to day 13 post-heart

transplantation. Six days post-transplant, the mice were treated with HSCT as described

above. This group, designated as TxHSCT, and all control groups used in this study are

tabulated in Table 3-1. The control groups included were as follows: TxRej-an

allogeneic transplant group that did not receive any treatment (BALB/c > C57BL/6);

TxRapa-an allogeneic transplant group treated with 14 days of rapamycin (BALB/c >

C57BL/6); TxSyn-a syngeneic transplant group that received no treatment and an

untreated non-transplanted C57BL/6 (CD45.2) control group (Non-Tx).

Table 3-1 Heterotopic heart transplantation treatment groups

Treatment Group Graft Rapamycin (1mg/kg) HSCT

1. TxRej ALLOGRAFT NO NO

2. TxRapa ALLOGRAFT YES- 14 days NO

3. TxHSCT ALLOGRAFT YES-14 days YES – POD7

4.TxSyn SYNGENEIC NO NO

5. Non-Tx Controls C57BL/6

NO NO NO

Page 89: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

76

Figure 3-1 Schematic of HSCT treatment for tolerance induction in cardiac

allotransplant model

HSCT treatment were examined in a previously established murine model of heterotopic

heart transplantation. A short course of rapamycin treatment was used to prevent acute

rejection and expand Tregs. On post-operative day 7 (POD7), mice underwent TBI

(13Gy) and were reconstituted with 5000 LSKs from congenic CD45.1 donor mice.

Rapamycin will be withdrawn at Day 13. Allograft survival was monitored via manual

palpation and grafts surviving > 70 days was deemed accepted. On POD45, mice from

each treatment group (Table 3-1) were sacrificed for immunological studies.

Page 90: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

77

3.7 Histology and immunohistochemistry

Transplanted and native BALB/c hearts were extracted and dissected along the

transverse (horizontal) axis into 2 nearly equivalent sections. The apex of the graft

proximal to the anastomoses with the recipient’s vessels was discarded and not utilized.

The two sections from each heart were placed into 10% buffered formalin and

embedded in OCT (Sakura Fintek, Torrence, CA) respectively.

The tissue fixed in 10% buffered formalin was submitted for sectioning and staining to

the Pathology Research Program Core Facility (PRP) at the Toronto General Hospital.

The tissue was subsequently embedded in paraffin, cut into 5 µm thick sections and

stained with hematoxylin and eosin (H&E) and Masson’s Trichrome (MT) dyes.

Immunoperoxidase staining was performed using anti-rat/mouse Foxp3 (clone FJK-16s,

eBioscience, San Diego, CA), anti-mouse CD3 (clone 17A2, eBioscience) and anti-

mouse B220 (BD Pharmingen-RA3-6B2) antibodies. Representative images of the

histology were taken by a trained pathologist and selected slides were scanned for

morphometric analysis at the STTARR research facility at the University Health Network

using an Aperio ScanScope XT whole slide scanner (Leica Biosystems, Wetzlar,

Germany). Positively stained cells were quantitated using TissueStudio® (Definiens,

Carlsbad, CA).

3.8 Mixed lymphocyte reaction

Mixed lymphocyte reactions were performed as per the Current Protocols in

Immunology.415 Spleens were extracted from donor (BALB/c), recipient (C57BL/6) and

Page 91: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

78

third-party (C3H/HeJ) mice, mechanically disassociated and RBCs were lysed. The

splenocytes were then layered onto Lympholyte-M (Cedarlane, Burlington, ON) and

spun at 1250xg at RT for 20 min. The interphase was collected and washed twice with

complete αMEM (prepared as described above). Allogeneic donor (BALB/c), third-party

(C3H) and syngeneic (C57BL/6) splenocytes were irradiated with 20 Gy using a

GAMMACELL® 40 cesium-137 irradiator (Best Theratronics, Ottawa, ON). In a U-

bottom 96-well suspension cell plate (Sarstedt AG&Co, Numbrecht, Germany), 2x105

responder splenocytes from C57BL/6 mice were co-cultured in triplicate with irradiated

4x105 syngeneic (C57BL/6), donor (BALB/c) or third-party (C3H) stimulator cells. Mixed

lymphocyte co-cultures were incubated at 37°C for 48 hours. After 48 hours, 1 µCi of

3H-thymidine (PerkinElmer, Boston, MA) was added to each well and cells were

incubated for an additional 18 hours. Plates were harvested using a UNIFILTER-96

Filtermate Harvester (PerkinElmer) and counts per minute (CPM) were recorded on a

TopCount Microplate scintillation counter (PerkinElmer). The proliferation data are

presented as fold change by calculating a “Stimulation Index” (SI) as per the Current

Protocols in Immunology .415 This is calculated by dividing the arithmetic mean of CPM

from triplicate experimental cultures by CPM from triplicate control (syngeneic) cultures.

3.9 Flow cytometry for donor-specific antibodies (DSA)

A flow cytometry-based cross match assay was adapted from earlier studies.206,416 Sera

was obtained from saphenous vein peripheral blood of C57BL/6 graft recipients and

non-transplanted controls. 1x106 donor BALB/c splenocytes were treated with Fc block

and incubated in duplicate with 2.5 µl of serum from recipient or non-transplanted mice

Page 92: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

79

for 30 min at 4°C. Afterwards, cells were washed and incubated with polyclonal FITC-

conjugated anti-mouse IgG (Immunology Consultants Laboratory, Portland, OR)

followed by APC anti-mouse CD3ε (Biolegend-clone:145-2C11). Cells were finally

stained with pacific blue viability dye and analyzed using a BD LSRII flow cytometer (BD

Bioscience, Franklin Lakes, NJ). Data were analyzed using the program FlowJo version

9.6 (Treestar Inc. Ashland, OR). Levels of anti-donor MHC Class I antibodies were

determined by gating on live CD3+ cells and reported as median fluorescence

intensities (MFI).

3.10 Statistics

Data are shown as mean ± SEM unless otherwise stated. Statistical significance was

determined using Students t-tests or one-way ANOVA followed by Tukey’s post-hoc test

on Prism version 5 (Graphpad Software, La Jolla, CA). Survival data was plotted on

Kaplan-Meier curves using log-rank tests to assess for statistical significance. Statistical

results with P≤0.05 were considered significant.

Page 93: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

80

Results

4.1 Purification of LSK cells and dose selection

To establish our autologous HSCT model, we aimed to isolate and purify the Lin-

Sca1+c-kit+ (LSK) population of murine HSCs from donor CD45.1 mice.413 As described

in the methods (section 3.4), we implemented a MACS-based lineage depletion protocol

followed by FACS for the stem cell markers Sca1 and c-kit (Figure 4-1). LSK were gated

on propidium iodide (PI)-negative populations to exclude dead cells. LSK cells were re-

suspended in PBS and injected intravenously to recipient CD45.2 mice. Subsequently,

we performed a dose-escalation study to investigate which dose resulted in complete

CD45.1 chimerism in the long-term. 1500, 3000 and 5000 LSKs were injected into mice

conditioned with 13Gy of TBI. At Day 100 post-HSCT, mice were sacrificed and level of

CD45.1 chimerism was determined using flow cytometry (Figure 4-2). All treatment

groups achieved > 97% CD45.1 chimerism in peripheral blood mononuclear cells with

no statistically significant differences between treatment groups. However, best results

were observed in the 5000 LSK cell group which tended to display higher (>99%)

CD45.1 chimerism.

Page 94: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

81

Figure 4-1 Isolation of LSK cells

This figure shows gating strategy for LSK cells pre- and post- lineage depletion to outline the

LSK purification process. Bone marrow was isolated by crushing femurs from CD45.1 donor

mice. RBC lysis was performed and cells were stained with the murine lineage depletion kit

antibody cocktail (Miltenyi Biotec). Lineage depletion was performed using autoMACS robotic

separation. Lin- cells were enriched 10-fold after magnetic sorting. Cells were stained with

mAbs to c-kit (APC) and Sca1 (PE) and submitted for LSK sorting on the Sca1 and c-kit

double positive population. As the lineage depletion cocktail was biotinylated, FITC-

streptavidin was used to distinguish between lineage positive and negative cells. Propidium

iodide was used as a viability dye for the exclusion of dead cells. Data are representative of 3

independent isolations using pooled bone marrow from n=4 mice at each time point.

Page 95: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

82

4.2 HSCT with LSK cells results in full hematopoietic reconstitution

and is not impaired by rapamycin treatment

We selected 5000 LSK cells as our optimal stem cell dosage. Subsequently, we

examined the ability of LSK cells to fully reconstitute all the hematopoietic lineages post-

HSCT. We were also concerned about performing LSK engraftment under the cover of

rapamycin, a potent immunosuppressant. As per Figure 3-1, our experimental plan for

allograft tolerance induction involves using a short course of rapamycin treatment pre-

Figure 4-2 LSK cell dose selection

Flow cytometry plots of CD45.1 versus CD45.2 populations in peripheral blood

mononuclear cells from mice treated with 5000, 3000 and 1500 LSK cells

respectively. There were no statistically significant differences in long-term CD45.1

chimerism in any of the treatment groups as determined by ANOVA. Gating was

performed on pacific blue negative (viable) cells. Data are representative of n=3

mice per group.

Page 96: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

83

and post-HSCT to prevent acute rejection and expand Tregs. Hence, we investigated

the effects of rapamycin treatment on long-term hematopoietic reconstitution after LSK

cell transfer. In the absence of an allograft, hematopoietic reconstitution was assessed

in a treatment group of mice receiving LSK cells alone compared to a treatment group

receiving LSK cells plus rapamycin as per our dosing regimen outlined in Figure 3-1.

At Day 100, mice from both the LSK and LSK+Rapa groups were sacrificed and

peripheral blood, spleen and bone marrow cells were stained for CD45.1, CD45.2, CD4,

CD8 and CD19 (Figure 4-3). There were no statistically significant differences between

the two groups in terms of CD45.1 (donor) chimerism in PBMC (Figure 4-3A). Both

groups had over 99% donor chimerism indicating successful engraftment of LSK cells in

the absence and presence of rapamycin treatment. Furthermore, flow cytometric

analyses demonstrated that frequencies of CD4+, CD8+ and CD19+ lymphocytes were

not significant different between groups in PBMC (Figure 4-3B), splenic lymphocytes

(Figure 4-3C) and bone marrow lymphocytes (Figure 4-3D). We also performed

complete blood count analysis (CBC) to examine full hematopoietic reconstitution in

both treatment groups (Table 4-1). Both treatment groups were within the normal range

for white blood cells, neutrophils (PMNs), platelets and hematocrit indicating

multilineage long-term reconstitution. At Day 100 post-LSK cell transfer, there were no

significant differences between the two groups for any of the aforementioned

parameters.

Page 97: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

84

Figure 4-3 Rapamycin does not impair long-term hematopoietic reconstitution

after HSCT with LSK cells

To study the effects of rapamycin on long-term LSK cell reconstitution, mice were

irradiated and treated with either 5000 LSK cells or 5000 LSK cells and rapamycin.

Hematopoietic reconstitution was assessed by flow cytometry. There were no

statistically significant differences between groups in A) CD45.1 chimerism in PBMC or

the frequencies of CD4+, CD8+ and CD19+ lymphocytes in B) peripheral blood C) spleen

or D) bone marrow. Differences between groups means were analyzed using ANOVA

followed by Tukey’s post-hoc test. Data are represented as mean ± SEM (n=4 mice per

group).

Page 98: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

85

Table 4-1 Complete blood counts at Day 100 post-HSCT (data are shown as mean

±SEM)

Treatment

Group

WBCs

(1x103/uL)

PMNs

(1x103/uL)

Lymphocytes

(1x103/uL)

Hct

(%)

Platelets

(1x103/uL)

Normal Ranges 1.8-10.7 0.1-2.4 0.9-9.3 35.1-45.4 592-3000

LSK

(n=4)

6.5±1.4 1.2±0.2 4.2±0.8 38.7

±0.5

798.8

±110.7

LSK + Rapa

(n=4)

6.4±1.3 1.1±0.1 4.5±0.9 36.3

±0.6

783.7

±55.5

WBC- White blood cells PMN – polymorphonuclear cells (neutrophils) Hct - Hematocrit

Page 99: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

86

4.3 HSCT promotes long-term cardiac allograft survival

HSCT was observed to significantly prolong allograft survival compared to

untreated and rapamycin-alone treated allograft recipients (Figure 4-4). TxHSCT mice

had a median survival time (MST) of 55 days with one recipient surviving long term till

Day 70. As expected, the untreated recipients (TxRej) suffered from acute rejection

culminating in 100% graft loss by Day 10 with a MST of 8 days. TxRapa recipients had

a MST of 31 days indicating that rapamycin alone cannot induce long-term graft survival

in our MHC-mismatch model. As expected, isograft recipients (TxSyn) had 100% long

term survival and were visually examined to be beating at the Day 70 endpoint (Figure

3-1). A small group (n=3) of isograft recipients were also treated with 5000 LSKs and

13Gy of TBI to examine whether radiation-induced damage could trigger graft loss in

the absence of immunological rejection. At Day 70, 100% of TxSynHSCT allografts

were surviving despite a reduced cardiac function score as assessed by manual

palpation (data not shown). These results indicate that while TBI is not sufficient to

induce graft loss in the context of an isograft it may result in graft damage.

Histological studies were performed on grafts sacrificed at peak of rejection for

each group. Peak of rejection for each group was based on median graft survival time

and reduction graft function as defined by manual palpation score. Thus, peak of

rejection was deemed to be POD 5 for the non-treated rejecting (TxRej), POD 20 for the

TxRapa group and POD 30 for the TxHSCT. Syngeneic grafts from the TxSyn group

were sacrificed at Day 30 and used as histological controls. Upon visual examination of

H&E histology at 100X magnification (Figure 4-5A), it was observed that both the TxRej

and TxRapa groups exhibited hallmarks of acute cellular rejection marked by dense

Page 100: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

87

mononuclear infiltrates (depicted by asterisks on image) and myocyte necrosis. In

contrast, allografts from the TxHSCT group had markedly reduced cellular infiltration

and tissue morphology comparable to that of syngeneic isografts which survive

indefinitely. A higher magnification (400X) array of H&E stains is also presented for

each group (Figure 4-5B). Visual inspection of these representative images revealed

extensive infiltration of mononuclear cells in the endothelium (vasculitis- depicted by

black arrows on image) and in cardiac tissue in the non-treated rejecting (TxRej) and

the TxRapa (rapamycin-only) allografts. In contrast, blood vessels from TxHSCT

allografts have very limited vasculitis and markedly reduced cellular infiltration. Blood

vessels from the TxSyn group have near normal appearance and are patent.

Representative images of immunoperoxidase staining for CD3+ cells are depicted for

cardiac grafts harvested from each treatment group at the same time point as the H&E

stained grafts (Figure 4-5C). Visual inspection of these images reveals that non-treated

rejecting (TxRej) and TxRapa allografts have significantly high numbers of graft-

infiltrating CD3+ cells. In contrast, TxHSCT allografts exhibit moderate to low numbers

of intra-graft CD3+ cells while syngeneic grafts (TxSyn) exhibit very low CD3+ cell

infiltration.

Immunostaining was also performed to investigate graft-infiltrating B, Tconv and

Treg cells in each treatment group. A single time point was selected for comparative

morphometric analyses. On POD 45, grafts were harvested and stained for infiltrating

CD3, B220 and FOXP3 positive cells (Figure 4-6). Grafts from the TxRej group which

reject entirely by POD 11, were not included for analysis due to extensive necrotic

damage and poor morphology at POD 45. Visual analyses of these representative

Page 101: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

88

images reveal that TxHSCT allografts have significantly fewer CD3+ cells (Figure 4-6A),

markedly higher FOXP3+ infiltrates and considerably lower B220+ cells (Figure 4-6C)

than TxRapa allografts. Syngeneic grafts from the TxSyn group exhibit low levels of all

infiltrating cell types (B cell, Tconv and Treg).

These aforementioned images were analyzed for morphometry of the

immunoperoxidase staining using TissueStudio® (Definiens, Carlsbad,CA).

Morphometry readouts for each marker (CD3, FOXP3 and B220) are depicted in Figure

4-7. Allografts from the TxRapa group had significantly higher infiltration of CD3+ cells

per mm2 (3344 ± 636) versus TxHSCT (1217 ± 266) and TxSyn (1141 ± 273) treatment

groups (Figure 4-7A). Mice treated with HSCT and rapamycin also displayed drastically

diminished levels of infiltrating B cells (B220+) versus TxRapa (Figure 4-7B). The

numbers of B220+ cells per mm2 in the TxRapa grafts (228 ± 7.3) were over 10-fold

higher than the numbers in the TxHSCT (17 ± 4.1) group, which had nearly the same

numbers of B cells as the TxSyn control group (20 ± 4.0). We also wished to examine

graft-infiltrating Tregs. Treatment with HSCT and rapamycin resulted in statistically

higher numbers of FOXP3+ infiltrating cells per unit area, compared to rapamycin alone

or in syngeneic control grafts (Figure 4-7C). FOXP3+ cells per mm2 in the TxHSCT

allografts (350 ± 25) were over 3-fold higher than in TxRapa (98 ± 18) and over 10-fold

higher than in control TxSyn (28 ± 14) grafts. Treg to T cell ratios were examined in the

grafts by expressing the immunostaining data as a ratio of FOXP3+ to CD3+ cells

normalized for cells per unit area (Figure 4-7D). Graft prolongation in the TxHSCT group

correlated with elevated Treg to T cell ratios. TxHSCT (20.8 ± 5.6%) grafts were

observed to have statistically higher Treg to Tconv ratios versus the TxRapa (5.1 ±

Page 102: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

89

0.9%) and the control TxSyn (2.7 ± 1.3%) grafts. Collectively, these data suggest that

graft prolongation in the TxHSCT group are associated with significantly reduced Tconv

and B lymphocytes and increased levels of Tregs. TxHSCT allografts closely resembled

sygeneic grafts in morphology and lack of cellular infiltration, except for graft-infiltrating

FOXP3+ Tregs. These results indicate that HSCT and rapamycin treatment protects

against cell-mediated rejection versus rapamycin alone. Thus, graft loss in the TxHSCT

group may be due to the toxicity associated with the TBI conditioning regimen.

Page 103: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

90

Figure 4-4 HSCT prolongs cardiac allograft survival

Heterotopic heart transplant survival in HSCT-treated and control groups of mice. Upon

cessation of beating, mice were sacrificed and graft loss was confirmed by visual

examination. Allografts in HSCT-treated recipients survived significantly longer (▲:

median survival time = 55 days) than untreated recipients (●: median survival time = 8

days) or rapamycin-alone treated recipients (■: median survival time = 31 days) with 1

graft surviving indefinitely. Control syngeneic graft recipients survived indefinitely as

expected (▼: survival > 70 days). Syngeneic graft recipients were treated with

autologous HSCT to assess the effects of the conditioning regimen (TBI) on graft

survival. Syngeneic recipients treated with HSCT also had indefinite survival (♦: survival

> 70 days). *** P<0.001 versus rapamycin only and untreated controls as determined by

log-rank test.

Page 104: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

91

Figure 4-5 HSCT treatment preserves cardiac allograft morphology

Cardiac grafts were sectioned and stained with H&E and anti-CD3 at various timepoints

coinciding with peak of rejection. Syngeneic grafts which survive indefinitely are used as

histological controls. A) Grafts from all treatment groups stained with H&E at 100X magnification.

TxRej and TxRapa grafts exhibit myocyte damage, dense mononuclear infiltrates and vasculitis.

TxHSCT and syngeneic (TxSyn) grafts have preserved morphology with the absence of cellular

infiltrates. Asterisks(*) indicate foci of dense mononuclear cellular infiltration. B) Grafts from all

treatment groups stained with H&E at 400X magnification. High magnification reveals that TxRej

and TxRapa grafts have vasculitis. TxHSCT and TxSyn have patent vessels with normal

appearance and absence of vasculitis. Arrows point to vessels. C) Representative CD3+

immunoperoxidase staining of all treatment groups. TxRapa and TxRej grafts exhibit foci of high

CD3+ cell infiltration. In contrast, visual examination reveals significantly reduced CD3+ cells in

HSCT treated grafts (TxHSCT) and sygeneic grafts. Representative graft histology (n=3-4 mice

per group) are presented.

Page 105: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

92

Figure 4-6 Representative immunoperoxidase staining of graft infiltrating cells

Cardiac grafts from POD 45 were harvested, sectioned and stained for anti-CD3,

FOXP3 and B220 cells. Untreated rejecting (TxRej) grafts were not included due to

extensive necrosis and poor tissue morphology. Representative images from each

group are presented at 200X magnification (n=3/4 mice per group). A) Representative

staining for graft infiltrating CD3+ cells. TxRapa grafts exhibit extensive CD3+ cell

infiltration. In contrast TxHSCT allografts and TxSyn isografts display markedly reduced

CD3+ cell infiltration. B) Representative staining for graft infiltrating FOXP3+ cells.

Examination of TxHSCT allografts reveals high numbers of intra-graft FOXP3+ cells in

contrast to TxRapa allografts or syngeneic (TxSyn) grafts. C) Representative

immunostaining for B220+ cells. Upon visual examination, TxRapa allografts exhibit

markedly increased B220+ (B cell) infiltration in the graft compared to HSCT treated

(TxHSCT) and isografts (TxSyn).

Page 106: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

93

Figure 4-7 Morphometric analyses of immunoperoxidase staining

Graphs of morphometric analysis performed on immunohistochemistry stains performed

on post-operative day 45 (POD45). Representative images are provided in Figure 4-6.

Data are presented as means ± SEM for each group A) Absolute number of CD3+ cells

per unit area, B) absolute number of B220+ cells per unit area, C) absolute number of

FOXP3+ cells per unit area, D) ratio of FOXP3+ to CD3+ cells in the grafts. Differences in

group means were analyzed by ANOVA followed by Tukey’s post-hoc test

****P<0.0001, ***P<0.001,**P<0.01,*P<0.05.

Page 107: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

94

4.4 HSCT-treated mice maintain primary immune response in

vitro

At POD 45 we performed a one-way MLR to assess T cell proliferation in

response to donor (BALB/C) and third-party (C3H) stimulators (Figure 4-5A and 4-5B

respectively). There were statistically significant differences in MLR responses to

BALB/C between TxRej mice and all other treatment groups. As expected, untreated

allograft recipients (TxRej) that had rejected their grafts by Day 10 demonstrated a

secondary immune response in vitro (SI =15.0 ± 1.3). In contrast, allografted mice

receiving HSCT and rapamycin (TxHSCT) maintained a primary immune response to

donor (SI = 9.8 ± 0.9). Despite rejecting their grafts by Day 32, allograft recipients given

rapamycin alone (TxRapa) also maintained a primary MLR response to BALB/C

stimulators (SI = 7.6 ± 1.2). Both syngeneic graft recipients (TxSyn) and non-

transplanted controls (Non-Tx) which had not been previously exposed to BALB/C

antigen demonstrated a primary immune response as expected (SI = 5.8 ± 0.4 and SI =

6.1 ± 1.0 respectively). There were no statistically significant differences in SI between

the TxHSCT, TxRapa, TxSyn or Non-Tx control groups. When co-cultured with third-

party (C3H) stimulators, all treatment groups had a primary MLR response (SI < 5 for all

groups).

Page 108: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

95

Figure 4-8 Lymphocytes from HSCT treated mice maintain primary immune

response in vitro

A mixed lymphocyte reaction (MLR) was performed using responder splenocytes from

each treatment group. MLR proliferation was assessed by incorporation of 3H-thymidine

and expressed as stimulation indices (SI) as per the Current Protocols in Immunology.

Background counts were assessed by co-culturing with syngeneic stimulator

splenocytes and were <500 CPM. Experimental counts were assessed by co-culturing

with allogeneic stimulator splenocytes from A) donor (BALB/C) or B) third-party (C3H)

mice and were > 1500 CPM. Data are presented as mean ± SEM. Differences between

group means were analyzed by ANOVA followed by Tukey’s post-hoc

test.***P<0.001,**P<0.01,*P<0.05.

Page 109: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

96

4.5 HSCT treatment markedly diminishes DSA and expands

Tregs

At POD 45, we also performed quantitation of Tregs in the spleens of mice from all

treatment groups and donor-specific antibodies in recipient mice sera (Figure 4-6). We

investigated the frequencies of CD25+FOXP3+ cells in the CD4+ compartment of splenic

lymphocytes. As a proportion of CD4+ cells, Tregs were significantly higher in the

TxHSCT group (7.2 ± 0.7%) compared to all other treatment groups (Figure 4-6A).

Although Tregs tended to be higher in the TxRapa group (4.2 ± 0.7%) compared to

TxSyn (2.9 ± 0.3%), TxRej (3.7 ± 0.5%) and Non-Tx (3.0 ± 0.6%) control mice, these

differences were not statistically significant. On POD 45, sera were also isolated from

mice in all treatment groups and non-transplanted controls. As described in the methods

section (section 3.9), we performed a flow cross-match assay to assess levels of DSA.

As expected, in isografted TxSyn and non-transplanted control mice which had not

previously been exposed to BALB/C antigen, DSA was deemed non-detectable (Figure

4-6B). Even though these samples had background fluorescence (MFI =128.5 ± 13.3

and 90.5 ± 13.1 for Non-Tx and TxSyn respectively), it was not significantly different to

control wells in which no serum was added (80.2 ± 11.5%). Thus, these background

levels were deemed to be below the limit of detection. We observed that the TxHSCT

group had markedly diminished levels of DSA (MFI = 2854 ± 143). These levels were

15-fold lower than the DSA levels (MFI = 45049 ± 3498) in untreated recipients (TxRej) .

We also noted that despite treatment with a potent immunosuppressant, mice given

Page 110: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

97

rapamycin alone (MFI = 21476 ± 1799) continued to exhibit high levels of DSA (8-fold

greater than TxHSCT) albeit lower than DSA in the TxRej group.

Figure 4-9 Splenic Treg and DSA quantitation

HSCT treatment markedly increases proportions of Tregs and reduces DSA levels. A)

Quantification of flow cytometric profiles. CD25+FOXP3+ Treg are shown as a proportion

of CD4+ cells in the spleen. B) Sera from mice in each transplanted group and non-

transplanted controls were analyzed for DSA using a flow cross-match assay.

Alloantibody levels are depicted by median fluorescence intensity (MFI). The

segmented axis is used to compare results that show several fold-differences.

Syngeneic graft recipients and non-transplanted controls had MFI levels comparable to

non-serum controls and were deemed below limit of detection. Data in both A and B are

expressed as mean ± SEM. Differences between group means were analyzed by

ANOVA followed by Tukey’s post-hoc test.****P<0.0001,**P<0.01,*P<0.05.

Page 111: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

98

Discussion

Although solid organ transplantation is a highly successful therapy for patients

with end stage organ failure, the need for long term immunosuppression limits long term

survival and quality of life. One solution to this problem is a therapy that can induce

organ-specific immune tolerance in the clinic.

A number of approaches are being studied both pre clinically and clinically

including allogeneic stem cell transplantation to produce mixed chimerism, infusion of

Treg cells and the identification of biomarkers that will identify patients who have

developed tolerance.354,360,417 Each of these treatments has distinct advantages and

disadvantages that warrant further discussion. The success of the mixed chimerism

approach is predicated on the successful establishment of multi-lineage hematopoietic

chimerism. Studies performed in pre-clinical models demonstrated the capacity of multi-

lineage chimerism to induce tolerance by both central (deletional) and peripheral

mechanisms of tolerance.360 The trafficking of donor APC to the host thymus in these

murine models results in intra-thymic chimerism and deletion (negative selection) of

alloreactive clones.418,419 Evidence for clonal deletion in human patients was provided

by high-throughput sequencing of the TCRβ chain in three tolerant patients who had

been treated with the MGH combined kidney and bone marrow (CKBM) transplant

protocol.420 In contrast, non-tolerant recipients of the MGH protocol or conventional

kidney transplant recipients did not display similar reductions of donor-reactive

clones.420 Allogeneic HSCT resulting in mixed or complete chimerism also involves

mechanisms of peripheral tolerance.397 An early increase of Treg proportions has been

Page 112: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

99

reported in post-allogeneic HSCT in patients from all three clinical cohorts (MGH, SU

and NW).395,421,422 Patients treated with the Stanford protocol, which involves total

lymphoid irradiation and ATG, also displayed increased ratios of NKT to T cells. These

observations are functionally validated by the donor-specific hyporesponsiveness

observed in vitro using lymphocytes from patients who were successfully weaned off IS

using the allogeneic HSCT approach.395,422 Despite these promising results, it is now

clear that the allogeneic HSCT approach has severe limitations. The use of allogeneic

bone marrow poses a risk for GVHD. At MGH, the HLA-matched CKBM has also been

utilized in patients with multiple myeloma and end-stage renal disease. In this cohort, 2

out of 7 patients developed chronic GVHD while one patient suffered from acute

GVHD.423 However, GVHD was largely absent in all of the three recent clinical cohorts

of patients treated with allogeneic HSCT.397,424 GVHD was reportedly avoided by using

TLI and ATG in the SU group while the NW group posited that this avoidance of GVHD

was a result of using proprietary facilitating cells (FCx).397 However, long-term follow up

has revealed that GVHD does occur in a subset of patients over time. In a recent clinical

abstract, Leventhal et al. reported that two patients from the NW cohort developed

GVHD.425 Finally, it is important to note that the success rate of tolerance induction in

these trials is low. In the MGH trial of CKBM transplant in HLA-mismatched patients,

only 7 out of 10 treated patients were successfully weaned off IS. Moreover, three out of

these 7 patients were subsequently returned to systemic IS after experiencing rejection

episodes within 5 years of IS withdrawal.393,426 Only a single patient has achieved long-

term (~11 years) IS-free graft survival. Similarly, in the SU cohort, long-term follow up

data is only available for HLA-matched patients. In 22 HLA-matched patients, only 16

Page 113: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

100

could be successfully withdrawn from IS and currently the longest time off IS for a

patient is under 6 years.390,427 A total of 6 HLA-mismatched patients have also been

treated with the SU protocol recently but tolerance induction results from this cohort are

not yet available.390 Finally, in the NW cohort of patients, only 12 HLA-mismatched

patients are currently off IS for a maximum period of 6 years. Furthermore, this cohort

has also experienced a high rate of opportunistic infections with 11 out of 19 treated

patients developing bacterial or fungal infections that required treatment.396 In their most

recent report, Leventhal et al. also describe the loss of two renal transplants due to

infection.425 Collectively, these results indicate that the allogeneic HSCT approach is not

the most effective therapy for tolerance induction in clinical transplant recipients. A

crucial observation in all three clinical cohorts was that a large subset of patients had

only transient (<100 days) mixed chimerism. Patients weaned off IS in the MGH cohort

had transient chimerism persisting for only 2-3 weeks.424 Patients treated with the SU

protocol developed a stable mixed chimerism (2-4 years) but long-term data on the

persistence of chimerism in these patients are not yet available.397,424 Finally, 11 out of

12 IS-free patients in the NW cohort developed full donor chimerism, which significantly

increases the risk for GVHD.396 A further consideration is that out of the 41 patients from

all three cohorts that are currently reported to be off IS, only 16 are HLA-mismatched.397

In the absence of the tolerizing effects of the mixed chimerism state, the risk for GVHD

and/or graft rejection for HLA-mismatched patients will significantly increase in the long-

term. The mixed chimerism approach is hypothesized to induce lifelong tolerance on the

indispensable condition of lifelong multilineage hematopoietic chimerism. Whereas mice

achieve lifelong mixed chimerism, studies have shown that mixed chimerism was only

Page 114: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

101

transient in NHP models of renal transplantation.360,388 Studies demonstrated that while

tolerance was induced in these NHP models even with transient mixed chimerism, the

transplants could only be performed while peripheral mixed chimerism lasted.360 Thus, a

loss of mixed chimerism or the development of full donor chimerism, the risk of GVHD

and development of opportunisitic infections is a serious limitation to the use of this

approach to induce tolerance in these patients.

The approach of using ex vivo expanded autologous Tregs for the purposes of

tolerance induction in transplantation has also recently attracted interest.428,429 The

concept of infusing autologous Tregs is promising due to their central role in maintaining

peripheral tolerance. Studies in murine models have shown that adoptive transfers of

Tregs can induce allograft tolerance.430 However, no such study has been reported to

date in human patients. As discussed previously, the ONE study is a multi-center phase

I/IIa trial that will examine the potential of ex vivo expanded Tregs to induce tolerance in

renal transplantation (www.onestudy.org).356 Thus far, Treg infusions in humans have

been used successfully to prevent GVHD in limited numbers of allogeneic bone marrow

recipients.431 Two studies have also recently shown safety of Treg infusion in Type 1

diabetes patients but long-term follow up of these patients is required to determine if

self-tolerance can be established.431 Despite the encouraging safety profiles, there are

several concerns regarding Treg infusions as a therapy for organ transplantation. First,

current trials have utilized polyclonal freshly isolated and expanded Tregs. However,

studies have shown that alloantigen-specific Tregs can be generated in vitro and have

superior suppressive capacity compared to polyclonal Tregs.432,433 The type of Treg

used and the methods for ex vivo generation of these cells will necessitate further

Page 115: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

102

optimization in the context of organ transplantation. Furthermore, it will be important to

ensure that infused Tregs stably continue to express FOXP3. The demethylation of key

regions of the FOXP3 gene are important for maintaining stability.434 Thus, the

demethylating agent azacytidine has been tested in a phase I trial to promote Treg

stability and prevent GVHD in patients receiving allo-HSCT for acute myeloid leukemia.

It was demonstrated that azacytidine increased post-transplant numbers of Tregs and

prevented GVHD while promoting a favourable CD8+ graft versus leukemia effect.435

Thus, attempts to induce long-term FOXP3 stability might also activate graft-specific

CD8+ T cells. The longevity of these infused Tregs is also a key concern. It has been

posited that continued treatment with IL-2 may allow for long-term survival of infused

Tregs but as IL-2 is also required for effector T cells and is used regularly for cancer

treatment, this approach may counteract the tolerogenic effects of the Treg transfer.431

A final consideration is that Treg transfer is a highly personalized therapy requiring ex

vivo manipulation of cells in a GMP grade facility. The cost of a single injection of Tregs

in a UK-based GMP facility was reported to be £20,000 (CAD 35,000) and multiple

injections may be required to successfully induce tolerance thereby significantly

increasing the cost per patient for this treatment.431 As a whole, these considerations

raise concerns about the practicality of Treg infusions as a treatment and may be

ultimately non-viable for the larger population of organ transplant recipients.

In the quest to achieve immunosuppression-free graft survival in organ

transplantation, several groups have also focused on the discovery of biomarkers that

can identify patients that are “operationally tolerant” and can be safely weaned off

immunosuppression.20,306,436 As discussed previously, operational tolerance is defined

Page 116: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

103

as a state of rejection-free graft survival in the absence of immunosuppression for at

least one year.21 The identification of reliable biomarkers would improve the field of

organ transplantation in several ways.437 First, it would significantly ameliorate long-term

health and quality of life for patients who are successfully weaned off

immunosuppression. Second, it would provide a technique to gauge the establishment

of tolerance and define end-points for the various clinical trials attempting to induce

clinical transplant tolerance. Finally, identification of genomic or proteomic biomarkers

for tolerance would provide mechanistic insight into tolerance induction in humans,

which is currently not well understood.437 Currently, reliable biomarkers for operational

tolerance have only been identified in cohorts of liver and renal transplant patients.20 It

has been observed that due to the immunoprivileged status of the liver, nearly 20% of

liver transplant recipients are operationally tolerant to their graft.437 The number of

operationally tolerant kidney recipients is far less with only 100 cases of reliable

operational tolerance described since the 1970s.438 A further complication is that the

biomarkers identified are not unique to all organ types. There are also differences

between intra-graft and peripheral expression profiles in operationally tolerant

patients.437 In operationally tolerant liver transplant patients, studies have identified

transcripts associated with NK and γδ cells as potential biomarkers in peripheral

blood.439 Conversely, in kidney transplant recipients, operational tolerance appears to

be related to B cell associated gene expression signatures.305,306 Thus, there is no

universal biomarker for tolerance that can reliably predict or assess operational

tolerance in organ transplantation. While the search for biomarkers of tolerance is an

Page 117: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

104

important endeavour within the field of transplantation immunology, there is an unmet

need for therapies that can actively induce tolerance in the clinic.

Autologous HSCT involves the re-setting of the immune system by reconstituting

a myeloablated host with previously isolated autologous bone marrow or peripheral

blood stem cells.440 Recently, patients treated with autologous HSCT for AID have been

shown to halt progression of disease and ameliorate quality of life. Based on these

reports, we have established a murine model of autologous HSCT following SOT. In this

thesis we have established HSCT as a potential mechanism of re-educating the

immune response in the setting of allotransplantation. A comparison of allogeneic

versus autologous HSCT is provided in Table 1-3.

For the first aim of this thesis, we established a murine model of autologous

HSCT that was capable of long-term multilineage hematopoietic reconstitution.

Currently, the majority of HSCT in mouse models is performed with total or T cell-

depleted bone marrow.441 However, the clinical relevance of using bone marrow can be

called into question given that currently, autologous HSCT in humans predominantly

utilizes mobilized and magnetically sorted CD34+ PBSCs.380 Another concern is that

bone marrow functions as a reservoir for memory T and B cells, as well as, long-lived

plasma cells.442–444 Thus, using highly purified LSK cells can prevent carryover of

mature, potentially alloreactive memory cells. Due to the indispensable use of

rapamycin in our murine allograft model, we also sought to test whether rapamycin

would interfere with LSK cell engraftment and long-term hematopoietic reconstitution.

Our studies demonstrated that performing LSK cell transfers under rapamycin treatment

Page 118: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

105

does not impair long-term hematopoietic reconstitution. Furthermore, we utilized a

peripheral blood CBC analyzer to demonstrate full reconstitution of WBC, platelets,

PMNs and RBCs (hematocrit). Thus, we propose that LSK cells are an ideal population

of readily purified murine HSCs for future studies of autologous HSCT in mice.

We also demonstrated as part of aim 2 of this thesis, that autologous HSCT

significantly prolonged cardiac allograft survival and in some cases produced long term

survival in recipients of fully MHC-mismatched heart allografts. As expected, at the

histological peak of rejection, grafts from both the TxRej and TxRapa groups

demonstrated classical histological signs of acute graft rejection consisting of dense

mononuclear infiltrates, myocyte necrosis and vasculitis. In contrast, grafts from the

TxHSCT group did not show strong evidence of cell mediated rejection but ultimately,

most grafts were most. We also performed immunostaining for CD3 on grafts harvested

from the treatment groups at earlier time points. Our immunostaining revealed a far

higher intensity of immunoperoxidase CD3 staining in the TxRej and TxRapa groups

versus the TxHSCT and the TxSyn groups. Morphometric analyses revealed that grafts

from TxHSCT mice had significantly reduced numbers of CD3+ infiltrating cells. In fact,

these numbers were not significantly higher than those found in syngeneic grafts which

display inflammation associated immune infiltration but do not undergo cell-mediated

rejection. Allografts treated with HSCT also demonstrated a higher absolute number of

FOXP3+ infiltrating cells and a significantly higher ratio of FOXP3+ to CD3+ cells, versus

rapamycin-only (TxRapa) or syngeneic grafts. These results are in accordance with

previous studies from our lab, performed in murine models of cardiac

allotransplantation.206,445 In both studies, long-term allograft survival was associated

Page 119: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

106

with an increased ratio of FOXP3+ (Tregs) to CD3+ (T cells). Using a rapamycin-induced

model of cardiac tolerance with BALB/C (H2d) donors and C3H/HeJ (H2k) recipients, we

showed that tolerance was dependent on graft-infiltrating Treg cells.206 Furthermore,

depletion of Tregs using anti-CD25 mAbs resulted in abrogation of tolerance. These

observations have also been reported in other studies. Lee et al. in 2005, demonstrated

an indispensable role for FOXP3+ cells in a murine model of cardiac allograft tolerance

using CD154 co-stimulation blockade and donor-specific transfusion (DST).446

Thymectomy or anti-CD25 mAbs prevented establishment of tolerance. Moreover, this

study showed that trafficking of Tregs to the graft was dependent on the expression of

the chemokine receptor CCR4 as CCR4 KO mice were also incapable of developing

tolerance using CD154 and DST.446 Studies in clinical transplantation have also showed

an important role for FOXP3 infiltration as a marker for tolerance. Bestard et al. have

shown that the FOXP3 to CD3 ratio is an accurate marker for predicting rejection in

kidney transplant patients with rejection occurring in patient who have low FOXP3/CD3

ratios.447 In the cohort of patients developing tolerance post allo-HSCT at MGH, Kawai

et al. reported an increased intragraft gene expression of FOXP3.392 As previously

described, Levitsky et al. reported markedly improved immunoregulatory profiles in liver

transplant patients converted from tacrolimus (CNI) to sirolimus (rapamycin).211 In

particular, this study noted that increased numbers of Tregs in the peripheral blood were

strongly correlated with increased FOXP3 to CD3 ratios in the graft of sirolimus-

converted patients.211 These observations are also in accordance with the data

generated in this thesis demonstrating that increased splenic Treg proportions in the

TxHSCT group are correlated with increased intra-graft FOXP3+ to CD3+ ratios. It is

Page 120: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

107

also important to note that FOXP3+ cells may infiltrate the graft prior to rejection. In a

recent study, Boer et al. assessed infiltration of natural (thymic) Tregs in

endomyocardial biopsies of heart transplant recipients who had suffered from acute

rejection or were rejection-free.448 Whereas, natural Tregs were increased in patients

undergoing acute rejection, total Treg numbers were higher in non-rejecting patient

biopsies, or biopsies taken from rejecting patients prior to onset of acute rejection.448

These results implicated an important role for Tregs in preventing rejection. In our

study, the preferential recruitment of FOXP3+ cells to the HSCT-treated grafts versus

rapamycin-alone treated grafts, suggests that HSCT treatment expands Tregs with far

greater efficacy than rapamycin alone. Our immunostaining data for graft-infiltrating

B220+ (B cells) showed that B cells were also significantly lower (>10 fold) in TxHSCT

grafts versus TxRapa grafts. Flow cytometric analyses of the splenic compartment in

the TxRej, TxRapa, TxHSCT and TxSyn groups revealed no significant differences in

frequencies of B cells (data not shown). These observations indicate either the

depletion of alloreactive B cell clones or a B cell targeted suppressive mechanism.

Thus, further studies are required to uncover the exact mechanism of HSCT on B cells

re-education in our model. Recent studies have also uncovered the importance of the

antigen presenting role of graft-infiltrating B cells. In 2014, Zeng et al. showed that

chronic rejection of murine cardiac allografts was predicated on the antigen-presenting,

as opposed to the antibody producing roles of B cells.136 These studies require further

validation in human patients to examine if B cells play analogous roles in chronic

rejection of human cardiac transplants. Nevertheless, these observations warrant

further study as autologous HSCT potentially offers a mechanism for reducing the risk

Page 121: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

108

for chronic allograft rejection, the most common cause of late-term graft loss in human

heart transplant patients.449

A potential cause of graft loss in the HSCT-treated group may be attributed to the

effects of TBI on cardiac tissue. Radiation-induced heart disease (RIHD) in patients

undergoing radiotherapy is now a well described clinical occurrence.450,451 RIHD

includes a wide spectrum of cardiac pathologies including cardiomyopathy, myocardial

fibrosis, pericarditis, valvular disease, coronary artery disease and arrhythmias.451

Cardiovascular disease is a leading cause of death in Hodgkin’s lymphoma patients

who often receive radiotherapy.452 Similarly, meta-analysis studies have shown a 62%

increase in cardiac deaths in breast cancer patients undergoing clinical trials involving

radiotherapy.453 The mechanisms of radiation-induced cardiac damage are not fully

understood but are known to involve endothelial damage and recruitment of

inflammatory cells all of which culminate in tissue fibrosis.451 In the acute phase post

exposure to ionizing radiation, damaged endothelial cells express adhesion molecules

and recruit inflammatory cells that express TNF, IL-1, IL-6 and IL-8.451 Classical pro-

fibrotic cytokines such as platelet-derived growth factor (PDGF), basic fibroblast growth

factor (bFGF) and TGF-β, are also released in the acute phase. This sets the stage for

a chronic fibrosis phase resulting in collagen deposition that affects the function of

myocytes, vascular endothelium and the pericardium.451 It has also been proposed that

upregulated NF-κB and chronic oxidative stress can mediate vascular damage post

radiation exposure.450 These widespread effects of radiation-induced cardiac damage

may explain the graft loss in the TxHSCT group even in the absence of high numbers of

mononuclear cell infiltration and vasculitis. This is also supported by Masson’s trichrome

Page 122: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

109

staining of TxHSCT grafts harvested at POD 45, which display extensive staining for

collagen deposition (data not shown). Therefore, we propose that future studies using

our model could adopt a lower dose of TBI or switch to chemotherapy-based

myeloablative regimens.

The immunological studies in our model provide evidence for a “re-education” of

the immune system in the setting of allotransplantation. Data generated here strongly

suggests the role for active immune regulation mechanisms as opposed to clonal

deletion. This assertion is supported by the observation of significantly higher

proportions of splenic and intra-graft Tregs in the TxHSCT group compared to all other

groups on POD 45. This observation is in accordance with studies in human patients

undergoing autologous HSCT for AID.408 Several mechanisms may be involved in the

elevated proportions of splenic Treg in TxHSCT mice. First, rapamycin treatment in the

presence of a newly emerging T cell repertoire could significantly skew the T cell

populations towards a high ratio of Tregs to effector T cells. Battaglia et al. showed that

rapamycin can cause preferential expansion and increase of Treg proportions in vitro.199

In 2007, Noris et al. also observed Treg expansion in patients who received sirolimus

after cytoreductive therapy.454 Thus, rapamycin treatment should have also resulted in

preferential expansion of Tregs in the TxRapa group but splenic Treg proportions in

TxRapa were not significantly higher than Non-Tx control, TxRej and TxSyn mice on

POD 45. Thus, alternative mechanisms of Treg expansion may also be contributing to

the high proportions of splenic Treg observed in the TxHSCT group. Delemarre et al.

recently showed that autologous HSCT in human patients, results in a renewal and

diversification of the Treg TCR repertoire.406 The authors also performed murine

Page 123: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

110

congenic (CD90.1/90.2) bone marrow transplants using 7.5Gy of TBI mice to analyze

Treg repopulation. It was observed that Treg populations from donor and host are

maintained in host mice at 3 weeks post-BMT while at 7 weeks, the entire Treg

compartment is donor derived.406 These thymic derived congenic donor Treg also

displayed proliferative capacity and an increased IL-10 mRNA expression compared to

host Treg. This increased suppressive function was purported to be a result of the

increased Treg TCR diversity that was observed in human patients post-HSCT.406 As

discussed above, HSCT-treatment in our model may also have significantly expanded

Tregs that persisted both intra-graft and in the periphery at higher levels compared to all

other treatment groups. Further work is warranted in the setting of both murine and

human autologous HSCT in organ transplantation to examine the phenotype and

functionality of pre- and post-HSCT Tregs.

Another line of evidence for a peripheral tolerogenic mechanism in our model, is

the data from in vitro MLR assays. Mice in the TxHSCT group maintained a primary

immune response to BALB/C stimulators indicating a peripheral suppressive versus a

central (deletional) mechanism of tolerance to donor antigen in this setting. An elevated

proportion of Tregs in these may be the major contributors towards suppressing donor

specific T cell responses in vitro and in vivo. As demonstrated by Muraro et al., HSCT in

human patients results in a nearly complete renewal of the CD4+ but not the CD8+ T cell

repertoire. Based on our results so far, we cannot rule out the contribution of CD8+ T

cells as well as innate immune cells such as NK cells, towards graft loss post-HSCT in

our model. Of note, we also observed that while all rapamycin only (TxRapa) treated

mice rejected by POD 32 and did not have increased Treg levels on POD 45, these

Page 124: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

111

mice displayed a primary MLR response in vitro. These results can be explained by a

number of reasons. First, rapamycin is known to significantly reduce DC maturation and

function.455 In 2007, Turnquist et al. demonstrated that ex-vivo rapamycin-conditioned

murine DCs were potent suppressors of allogeneic MLRs and could promote indefinite

cardiac allograft survival.456 Thus, rapamycin conditioning in these mice may have led to

anergy induction and deletion of potentially alloreactive CD4+ T cell clones as a result of

binding to immature DCs. Furthermore, rapamycin has been observed to play a

contradictory role in promoting both CD8+ memory as well as CD4+ Tregs due to similar

metabolic demands.457 Araki et al. showed that rapamycin treatment significantly

improved LCMV-specific CD8+ memory cells signaling a potential immune activating

role for rapamycin.197 However, in 2010, Ferrer et al. used a transgenic mouse model to

show rapamycin could improve CD8+ cell responses towards antigen when presented in

the context of a bacterial pathogen but not towards antigen presented in the context of

an allograft.458 Furthermore, there is also some evidence for the role of mTOR inhibition

in promoting apoptosis of memory CD4+ T cells.459 These data show that rapamycin can

significantly diminish alloreactive memory T cell responses thereby maintaining a

primary immune response to alloantigen in vitro. Therefore, the rejection of allografts in

TxRapa mice may be attributed to the role of B cells, NK cells and other innate cell

types. In fact, it was shown that rapamycin-only treated mice had high levels of DSA.

Our study also showed a significant re-education of the B cell repertoire post-

HSCT. As discussed above, there was a marked reduction in graft-infiltrating B cells in

TxHSCT mice. Our data also showed an important role for re-education of B cells in the

periphery. DSA levels in TxHSCT mice were 16-fold and 8-fold lower than in untreated

Page 125: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

112

rejecting and rapamycin only treated mice respectively. Studies performed in human

AID patients treated with autologous HSCT have demonstrated a potent re-education of

the B cell repertoire.411,460 In 2009, Alexander et al. reported the effects of autologous

HSCT in a small cohort of 7 SLE patients.460 HSCT in these patients re-set the B cell

repertoire towards a predominantly naive B cell repertoire and significant lowered (9-

fold) the frequencies of CD19+CD27+IgD- memory B cells in peripheral blood.

Furthermore, autoantibodies were also significantly abolished in these patients. In 6 out

of 7 patients, anti-dsDNA antibodies dropped to below limit of detection, while in 4

patients, antinuclear antibodies (ANA) fell to below clinically significant values.460 The

mechanisms underlying this abrogation of autoantibody production are not well

understood. However, it is known that long-lived plasma cells (LLPCs) in the bone

marrow a major source of antibody production.461 Thus, autologous HSCT may alter this

repertoire through newly emerging B cells outcompeting these LLPCs or directly

inhibiting their survival through an unknown suppressor mechanism. Either of these

mechanisms may be underlying the significant reduction of DSA observed post-HSCT in

our mouse model and warrant further study.

Page 126: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

113

Conclusions

In conclusion, this thesis has established HSCT as a potential means to re-

educate the immune response in the setting of allotransplantation The re-education

affected both T and B cell responses. These results are consistent with the data

generated by Atkins et al showing that autologous HSCT can abrogate MS progression

in human patients.404 Thus, our data provide a rationale for the clinical study to examine

the use of autologous HSCT in the setting of liver transplantation to induce tolerance

(ASCOTT- NCT02549586).

Page 127: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

114

Future Directions

The studies performed and the results generated by this dissertation provide

many avenues for further research. The two major approaches for future research in

this area are to further elucidate the mechanisms associated with autologous HSCT in

murine models and to perform research using clinical samples from the ASCOTT trial

for investigating re-education in the human SOT recipients. The autologous HSCT and

cardiac allograft model established in this thesis can be further studied to gain

mechanistic insights into the tolerogenic potential of autologous HSCT in SOT.

However, this approach can also be expanded to other murine allograft models such as

kidney and lung.226 Our studies demonstrated that Tregs were significantly increased in

the TxHSCT group implicating a role of Tregs in prolonging cardiac allograft survival in

this model. Thus, future experiments could administer anti-CD25 mAbs to HSCT-treated

mice and monitor for accelerated rejection as performed in a previous study by our

group.206 As discussed in the previous section, Delemarre et al. also demonstrated that

donor derived Tregs post-BMT from congenic donors, had high proliferative capacity

and increased IL-10 expression.406 Thus, future functional studies in our mouse model

should examine the mRNA levels of key suppressive cytokines (TGF-β, IL-10 and IL-34)

in Tregs from TxHSCT, TxRapa and TxRej groups. Another key observation was the

marked reduction of DSA in the mice treated with HSCT. Future studies can be

performed to uncover the mechanisms through which this reduction may occur. LLPCs,

as described earlier, are known reside in the bone marrow and function as long-term

producers of alloantibodies.88 Hence, multiparameter flow cytometric profiling of B cell

Page 128: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

115

subsets including LLPCs, should yield evidence for perturbations in the B cell repertoire

as observed in human patients undergoing autologous HSCT for AID.411 Multiplex

ELISA can also be carried out using available bead-arrays such as Legendplex™ from

Biolegend (San Diego, CA) for a wide range of pro- and anti-inflammatory cytokines

such as IL-10, TGF-β, IL-4, IFNγ and IL-1. We deem radiation-induced cardiac damage

to be a major cause of graft loss in our studies. Therefore, future studies could

incorporate our LSK-based HSCT model with chemotherapy-based myeloablative

regimens. In 2014, Gorczynski et al. showed that T cell-depleted BMT using busulphan

and cyclophosphamide as a conditioning regimen resulted in significantly prolonged

skin allograft survival and reduced in vitro responsiveness to donor antigen.462 Thus,

LSK studies can be performed in this setting to evade the potential damage associated

with TBI. Finally, future work can also focus on examining intra-graft mRNA expression.

In our previous work, we demonstrated that tolerant murine cardiac allografts

demonstrated a 6-gene profile associated with tolerance.206 Future experiments could

involve studying the expression of these 6 genes in HSCT-treated allografts in our

model.

In parallel to the murine work, the establishment of the ASCOTT trial

(NCT02549586), offers an exciting opportunity to study the mechanisms of HSCT-

mediated immune re-education in SOT patients. As demonstrated by the TCR

sequencing studies of Muraro et al. and Delemarre et al. an in-depth view of renewal

the Tconv/Treg repertoire can be generated using this approach.359,406 Similar studies in

SOT patients can be performed to examine alloreactive T cell/Treg repertoires. Due to

the large proportion of T cells that can respond to alloantigen as a result of heterologous

Page 129: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

116

immunity (5-10%), it will be essential to examine the ability of autologous HSCT to reset

this alloreactive repertoire.56 For HSCT-treated patients who have a living-donor

transplant, future studies will have the advantage of opportunity to perform in vitro MLR

and CTL assays to assess donor-specific responsiveness. Finally, as the

immunobiology of graft rejection is far more complex in NHPs and humans compared to

mice, future studies will require detailed immunophenotyping studies in SOT patients

who undergo HSCT. These studies can follow a three-pronged approach. First, as the

studies in AID patients demonstrate, several cell types are perturbed post-HSCT.408

Thus immunophenotyping of PBMCs in these patients will require a high-throughput

approach to examine several cell subsets. Currently, flow cytometry is limited at 16-17

markers due to issues of spectral overlap. However, with the advent of mass cytometry

(CyTOF), currently around 45-50 cell markers can be examined in a single sample.463

Thus future studies in ASCOTT patients can utilize CyTOF as a high-throughput

phenotyping platform in conjunction with multiplex ELISA for proteomic and multiplex

qPCR for genomic assessments in these patients. This three-pronged approach has the

potential to not only characterize the tolerant patients in this trial but also discover

potentially novel mechanisms of tolerance induction in SOT.

Page 130: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

117

References

1. Sayegh, M. H. & Carpenter, C. B. Transplantation 50 years later--progress,

challenges, and promises. N Engl J Med 351, 2761–2766 (2004).

2. Merrill, J. P., Murray, J. E., Harrison, J. H. & Guild, W. R. Successful

homotransplantation of the human kidney between identical twins. J Am Med

Assoc 160, 277–282 (1956).

3. STARZL, T. E. et al. IMMUNOSUPPRESSION AFTER EXPERIMENTAL AND

CLINICAL HOMOTRANSPLANTATION OF THE LIVER. Ann. Surg. 160, 411–39

(1964).

4. Barnard, C. N. Human cardiac transplantation. An evaluation of the first two

operations performed at the Groote Schuur Hospital, Cape Town. Am. J. Cardiol.

22, 584–596 (1968).

5. Starzl, T. E. et al. Orthotopic homotransplantation of the human liver. Ann. Surg.

168, 392–415 (1968).

6. Calne, R. Y. et al. CYCLOSPORIN A IN PATIENTS RECEIVING RENAL

ALLOGRAFTS FROM CADAVER DONORS. Lancet 312, 1323–1327 (1978).

7. Kahan, B. D. Drug therapy: cyclosporine. New England Journal of Medicine 321,

1725–1738 (1989).

8. Salvadori, M. & Bertoni, E. What’s new in clinical solid organ transplantation by

2013. World J. Transplant. 4, 243–66 (2014).

9. Meier, D., Rumbo, M. & Gondolesi, G. E. Current status of allograft tolerance in

intestinal transplantation. Int. Rev. Immunol. 33, 245–260 (2014).

10. Shapiro, A. M. et al. Islet transplantation in seven patients with type 1 diabetes

mellitus using a glucocorticoid-free immunosuppressive regimen. N. Engl. J. Med.

Page 131: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

118

343, 230–8 (2000).

11. Rana, A. et al. Survival benefit of solid-organ transplant in the United States.

JAMA Surg. 150, 252–259 (2015).

12. Garcia, G. G., Harden, P., Chapman, J. & Garcia, G. G. The Global Role of

Kidney Transplantation Full Text Introduction Outcomes of Kidney Transplantation

The Place of Kidney Transplantation in Treatment for ESRD Global Disparities in

Access to Kidney Transplantation Improving Access to Transplantation. 278–281

(2012).

13. Canadian Institute for Health Information. Canadian organ replacement register

annual report: Treatment of end-stage organ failure in Canada, 2004 to 2013. 1–

129 (2014).

doi:https://secure.cihi.ca/free_products/2011_CORR_Annua_Report_EN.pdf

14. Lechler, R. I., Sykes, M., Thomson, A. W. & Turka, L. A. Organ transplantation--

how much of the promise has been realized? Nat. Med. 11, 605–13 (2005).

15. Lodhi, S. A., Lamb, K. E. & Meier-Kriesche, H. U. Solid organ allograft survival

improvement in the United States: The long-term does not mirror the dramatic

short-term success. Am. J. Transplant. 11, 1226–1235 (2011).

16. Ojo, A., Held, P., Port, F., Wolfe, R., Leichtman, A., Young, E., Arndorfer, J., et

al. Chronic renal failure after transplantation of a nonrenal organ. N Engl J Med

349, 931–940 (2003).

17. Roussel, J. C. et al. Outcome of Heart Transplants 15 to 20 Years Ago: Graft

Survival, Post-transplant Morbidity, and Risk Factors for Mortality. J. Hear. Lung

Transplant. 27, 486–493 (2008).

18. Legendre, C., Canaud, G. & Martinez, F. Factors influencing long-term outcome

after kidney transplantation. Transplant International 27, 19–27 (2014).

19. Lamb, K. E., Lodhi, S. & Meier-Kriesche, H.-U. Long-term renal allograft survival

Page 132: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

119

in the United States: a critical reappraisal. Am. J. Transplant. 11, 450–62 (2011).

20. Heidt, S. & Wood, K. J. BIOMARKERS OF OPERATIONAL TOLERANCE IN

SOLID ORGAN TRANSPLANTATION. Expert Opin. Med. Diagn. 6, 281–293

(2012).

21. Chandrasekharan, D., Issa, F. & Wood, K. J. Achieving operational tolerance in

transplantation: How can lessons from the clinic inform research directions?

Transplant International 26, 576–589 (2013).

22. Huston, D. P. The biology of the immune system. Jama 278, 1804–1814 (1997).

23. Warrington, R., Watson, W., Kim, H. L. & Antonetti, F. R. An introduction to

immunology and immunopathology. Allergy, Asthma Clin. Immunol. 7, 1–8 (2011).

24. Murphy, K. Janeway’s Immunobiology. (Garland Science, Taylor and Francis

Group, LLC, 2012). doi:10.1007/s13398-014-0173-7.2

25. Owen, J. A., Punt, J., Stranford, S. A., Jones, P. P. & Kuby, J. Kuby immunology.

(W.H. Freeman, 2013).

26. Parkin, J. & Cohen, B. An overview of the immune system. Lancet 357, 1777–

1789 (2001).

27. Hemmi, H. et al. A Toll-like receptor recognizes bacterial DNA. Nature 408, 740–

745 (2000).

28. Schulz, O. et al. Toll-like receptor 3 promotes cross-priming to virus-infected cells.

Nature 433, 887–892 (2005).

29. Brown, G. D. et al. Dectin-1 is a major beta-glucan receptor on macrophages. J

Exp Med 196, 407–412 (2002).

30. Schenten, D. & Medzhitov, R. The control of adaptive immune responses by the

innate immune system. Adv Immunol 109, 87–124 (2011).

Page 133: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

120

31. Iwasaki, A. & Medzhitov, R. Control of adaptive immunity by the innate immune

system. Nat Immunol 16, 343–353 (2015).

32. Market, E. & Papavasiliou, F. N. V(D)J recombination and the evolution of the

adaptive immune system. PLoS Biol 1, E16 (2003).

33. Raff, M. C. T and B Lymphocytes and Immune Responses. Nature 242, 19–23

(1973).

34. Thomas, L. R., Cobb, R. M. & Oltz, E. M. Dynamic regulation of antigen receptor

gene assembly. Adv Exp Med Biol 650, 103–115 (2009).

35. Chong, A. S. & Alegre, M.-L. The impact of infection and tissue damage in solid-

organ transplantation. Nat. Rev. Immunol. 12, 459–471 (2012).

36. Wood, K. J. & Goto, R. Mechanisms of Rejection: Current Perspectives.

Transplantation 93, 1–10 (2012).

37. Castellano, G. et al. Therapeutic targeting of classical and lectin pathways of

complement protects from ischemia-reperfusion-induced renal damage. Am. J.

Pathol. 176, 1648–1659 (2010).

38. Milland, J. & Sandrin, M. S. ABO blood group and related antigens, natural

antibodies and transplantation. Tissue Antigens 68, 459–466 (2006).

39. Gould, D. S. & Auchincloss, H. Direct and indirect recognition: The role of MHC

antigens in graft rejection. Immunology Today 20, 77–82 (1999).

40. Peugh, W. N., Superina, R. A., Wood, K. J. & Morris, P. J. The role of H-2 and

non-H-2 antigens and genes in the rejection of murine cardiac allografts.

Immunogenetics 23, 30–37 (1986).

41. Vyas, J. M., Van der Veen, A. G. & Ploegh, H. L. The known unknowns of antigen

processing and presentation. Nat. Rev. Immunol. 8, 607–18 (2008).

42. Neefjes, J., Jongsma, M. L., Paul, P. & Bakke, O. Towards a systems

Page 134: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

121

understanding of MHC class I and MHC class II antigen presentation. Nat Rev

Immunol 11, 823–36 (2011).

43. Jensen, P. E. Recent advances in antigen processing and presentation. Nat.

Immunol. 8, 1041–8 (2007).

44. Cresswell, P., Ackerman, A. L., Giodini, A., Peaper, D. R. & Wearsch, P. A.

Mechanisms of MHC class I-restricted antigen processing and cross-presentation.

Immunological Reviews 207, 145–157 (2005).

45. Roche, P. a & Furuta, K. The ins and outs of MHC class II-mediated antigen

processing and presentation. Nat. Rev. Immunol. 15, 203–216 (2015).

46. Steimle, V., Siegrist, C. A., Mottet, A., Lisowska-Grospierre, B. & Mach, B.

Regulation of MHC class II expression by interferon-gamma mediated by the

transactivator gene CIITA. Science 265, 106–109 (1994).

47. Gorer, P. A., Lyman, S. & Snell, G. D. Studies on the Genetic and Antigenic Basis

of Tumour Transplantation. Linkage between a Histocompatibility Gene and

‘Fused’ in Mice. Proc. R. Soc. B Biol. Sci. 135, 499–505 (1948).

48. Stuart, P. M. in eLS (John Wiley & Sons, Ltd, 2001).

doi:10.1002/9780470015902.a0000921.pub4

49. Janeway, C. A., Travers, P., Walport, M. & Shlomchik, M. Immunobiology: The

Immune System In Health And Disease. Immuno Biol. 5 892 (2001).

doi:10.1111/j.1467-2494.1995.tb00120.x

50. Ayala García, M. A., González Yebra, B., López Flores, A. L. & Guaní Guerra, E.

The Major Histocompatibility Complex in Transplantation. J. Transplant. 2012, 1–

7 (2012).

51. Deakin, J. E. et al. Evolution and comparative analysis of the MHC Class III

inflammatory region. BMC Genomics 7, 281 (2006).

Page 135: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

122

52. Chinen, J. & Buckley, R. H. Transplantation immunology: Solid organ and bone

marrow. J. Allergy Clin. Immunol. 125, (2010).

53. Spierings, E. Minor histocompatibility antigens: Past, present, and future. Tissue

Antigens 84, 374–360 (2014).

54. Dierselhuis, M. & Goulmy, E. The relevance of minor histocompatibility antigens in

solid organ transplantation. Curr. Opin. Organ Transplant. 14, 419–425 (2009).

55. Sherman, L. A. & Chattopadhyay, S. The molecular basis of allorecognition.

Annu. Rev. Immunol. 11, 385–402 (1993).

56. Lakkis, F. G. & Lechler, R. I. Origin and biology of the allogeneic response. Cold

Spring Harb. Perspect. Med. 3, (2013).

57. Moreau, A., Varey, E., Anegon, I. & Cuturi, M. C. Effector mechanisms of

rejection. Cold Spring Harbor Perspectives in Medicine 3, (2013).

58. MacEdo, C. et al. Contribution of naive and memory t-cell populations to the

human alloimmune response. Am. J. Transplant. 9, 2057–2066 (2009).

59. Suchin, E. J. et al. Quantifying the frequency of alloreactive T cells in vivo: new

answers to an old question. J. Immunol. 166, 973–981 (2001).

60. Pietra, B. A., Wiseman, A., Bolwerk, A., Rizeq, M. & Gill, R. G. CD4 T cell-

mediated cardiac allograft rejection requires donor but not host MHC class II. J.

Clin. Invest. 106, 1003–10 (2000).

61. Ochando, J. C., Krieger, N. R. & Bromberg, J. S. Direct versus indirect

allorecognition: Visualization of dendritic cell distribution and interactions during

rejection and tolerization. Am. J. Transplant. 6, 2488–2496 (2006).

62. Ali, J. M., Bolton, E. M., Bradley, J. A. & Pettigrew, G. J. Allorecognition pathways

in transplant rejection and tolerance. Transplantation 96, 681–8 (2013).

63. Garrod, K. R. et al. NK cell patrolling and elimination of donor-derived dendritic

Page 136: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

123

cells favor indirect alloreactivity. J. Immunol. (Baltimore, Md 1950) 184, 2329–

2336 (2010).

64. Benham, A. M., Sawyer, G. J. & Fabre, J. W. Indirect T cell allorecognition of

donor antigens contributes to the rejection of vascularized kidney allografts.

Transplantation 59, 1028–1032 (1995).

65. Auchincloss, H. et al. The role of ‘indirect’ recognition in initiating rejection of skin

grafts from major histocompatibility complex class II-deficient mice. Proc. Natl.

Acad. Sci. U. S. A. 90, 3373–7 (1993).

66. Ali, J. M. et al. Diversity of the CD4 T Cell Alloresponse: The Short and the Long

of It. Cell Rep. 14, 1232–1245 (2016).

67. Wise, M. P., Bemelman, F., Cobbold, S. P. & Waldmann, H. Linked suppression

of skin graft rejection can operate through indirect recognition. J. Immunol. 161,

5813–5816 (1998).

68. Yamada, A. et al. Recipient MHC class II expression is required to achieve long-

term survival of murine cardiac allografts after costimulatory blockade. J Immunol

167, 5522–5526 (2001).

69. Herrera, O. B. et al. A Novel Pathway of Alloantigen Presentation by Dendritic

Cells. J. Immunol. 173, 4828–4837 (2004).

70. Valujskikh, A., Lantz, O., Celli, S., Matzinger, P. & Heeger, P. S. Cross-primed

CD8(+) T cells mediate graft rejection via a distinct effector pathway. Nat.

Immunol. 3, 844–51 (2002).

71. Harper, S. J. F. et al. CD8 T-cell recognition of acquired alloantigen promotes

acute allograft rejection. Proc. Natl. Acad. Sci. U. S. A. 112, 12788–12793 (2015).

72. Nankivell, B. J. & Alexander, S. I. Rejection of the kidney allograft. N. Engl. J.

Med. 364, 485–486 (2011).

Page 137: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

124

73. Shah, M. M., Presser, N. & Fung, J. J. in Abdominal Solid Organ Transplantation

(eds. Pinna, A. D. & Ercolani, G.) 3–32 (Springer International Publishing, 2015).

74. Sarwal, M. M. & Sarwal, R. D. in eLS (John Wiley & Sons, Ltd, 2001).

doi:10.1002/9780470015902.a0001232.pub2

75. Colvin, R. B. & Smith, R. N. Antibody-mediated organ-allograft rejection. Nat. Rev.

Immunol. 5, 807–817 (2005).

76. Pruitt, S. K., Baldwin, W. M. 3rd & Sanfilippo, F. The role of C3a and C5a in

hyperacute rejection of guinea pig-to-rat cardiac xenografts. Transplant. Proc. 28,

596 (1996).

77. Lee, C.-Y. et al. The involvement of FcR mechanisms in antibody-mediated

rejection. Transplantation 84, 1324–1334 (2007).

78. Puttarajappa, C., Shapiro, R. & Tan, H. P. Antibody-mediated rejection in kidney

transplantation: a review. J. Transplant. 2012, 193724 (2012).

79. Gelfand, E. W. Intravenous immune globulin in autoimmune and inflammatory

diseases. N. Engl. J. Med. 367, 2015–25 (2012).

80. Faguer, S. et al. Rituximab therapy for acute humoral rejection after kidney

transplantation. Transplantation 83, 1277–80 (2007).

81. Paterno, F. et al. Bortezomib for acute antibody-mediated rejection in liver

transplantation. Am. J. Transplant. 12, 2526–2531 (2012).

82. Shimizu, A. & Yamada, K. Histopathology of xenografts in pig to non-human

primate discordant xenotransplantation. Clinical Transplantation 24, 11–15

(2010).

83. Platt, J. L. et al. Immunopathology of hyperacute xenograft rejection in a swine-to-

primate model. Transplantation 52, 214–220 (1991).

84. Klymiuk, N., Aigner, B., Brem, G. & Wolf, E. Genetic modification of pigs as organ

Page 138: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

125

donors for xenotransplantation. Molecular Reproduction and Development 77,

209–221 (2010).

85. Christie, J. D. et al. The registry of the international society for heart and lung

transplantation: 29th adult lung and heart-lung transplant report - 2012. J. Hear.

Lung Transplant. 31, 1073–1086 (2012).

86. Cornell, L. D., Smith, R. N. & Colvin, R. B. Kidney Transplantation: Mechanisms

of Rejection and Acceptance. Annu. Rev. Pathol. Mech. Dis 3, 189–220 (2008).

87. Win, T. S. & Pettigrew, G. J. Humoral autoimmunity and transplant vasculopathy:

when allo is not enough. Transplantation 90, 113–120 (2010).

88. Clatworthy, M. R. B cell modulation in transplantation. Clin. Exp. Immunol. 178,

61–63 (2014).

89. Asgari, E., Zhou, W. & Sacks, S. Complement in organ transplantation. Curr.

Opin. Organ Transplant. 15, 486–91 (2010).

90. Elgueta, R. et al. Molecular mechanism and function of CD40/CD40L engagement

in the immune system. Immunological Reviews 229, 152–172 (2009).

91. Garside, P. et al. Visualization of specific B and T lymphocyte interactions in the

lymph node. Science (80-. ). 281, 96–99 (1998).

92. Crotty, S. Follicular helper CD4 T cells (TFH). Annu. Rev. Immunol. 29, 621–663

(2011).

93. Patel, J. K., Kittleson, M. & Kobashigawa, J. A. Cardiac allograft rejection.

Surgeon 9, 160–7 (2011).

94. Crespo, M. et al. Acute humoral rejection in renal allograft recipients: I. Incidence,

serology and clinical characteristics. Transplantation 71, 652–658 (2001).

95. Colvin, R. B. Antibody-mediated renal allograft rejection: diagnosis and

pathogenesis. J. Am. Soc. Nephrol. 18, 1046–1056 (2007).

Page 139: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

126

96. Kaplan, B. et al. Successful rescue of refractory, severe antibody mediated

rejection with splenectomy. Transplantation 83, 99–100 (2007).

97. Lafferty, K. & Cunningham, A. a New Analysis of Allogeneic Interactions. Aust. J.

Exp. Biol. Med. Sci. 53, 27–42 (1975).

98. Wekerle, T., Kurtz, J., Bigenzahn, S., Takeuchi, Y. & Sykes, M. Mechanisms of

transplant tolerance induction using costimulatory blockade. Curr. Opin. Immunol.

14, 592–600 (2002).

99. Pilat, N., Sayegh, M. H. & Wekerle, T. Costimulatory pathways in transplantation.

Seminars in Immunology 23, 293–303 (2011).

100. Paterson, A. M., Vanguri, V. K. & Sharpe, A. H. SnapShot: B7/CD28

Costimulation. Cell 137, (2009).

101. Krummel, M. F. & Allison, J. P. CD28 and CTLA-4 have opposing effects on the

response of T cells to stimulation. J Exp Med 182, 459–465 (1995).

102. Walunas, T. L. et al. CTLA-4 can function as a negative regulator of T cell

activation. Immunity 1, 405–413 (1994).

103. Curtsinger, J. M. et al. Inflammatory cytokines provide a third signal for activation

of naive CD4+ and CD8+ T cells. J. Immunol. 162, 3256–62 (1999).

104. Boyman, O. & Sprent, J. The role of interleukin-2 during homeostasis and

activation of the immune system. Nat. Rev. Immunol. 12, 180–90 (2012).

105. Ingulli, E. Mechanism of cellular rejection in transplantation. Pediatric Nephrology

25, 61–74 (2010).

106. Liu, Z., Fan, H. & Jiang, S. CD4+ T-cell subsets in transplantation. Immunol. Rev.

252, 183–191 (2013).

107. D’Elios, M. M. et al. Predominant Th1 cell infiltration in acute rejection episodes of

human kidney grafts. Kidney Int. 51, 1876–1884 (1997).

Page 140: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

127

108. Sawitzki, B. et al. IFN-gamma production by alloantigen-reactive regulatory T cells

is important for their regulatory function in vivo. J. Exp. Med. 201, 1925–35

(2005).

109. Surquin, M. et al. IL-4 deficiency prevents eosinophilic rejection and uncovers a

role for neutrophils in the rejection of MHC class II disparate skin grafts.

Transplantation 80, 1485–1492 (2005).

110. Barbara, J. A. et al. Islet allograft rejection can be mediated by CD4+, alloantigen

experienced, direct pathway T cells of TH1 and TH2 cytokine phenotype.

Transplantation 70, 1641–9 (2000).

111. Laan, M. et al. Neutrophil recruitment by human IL-17 via C-X-C chemokine

release in the airways. J. Immunol. 162, 2347–52 (1999).

112. Badell, I. R. & Ford, M. L. T follicular helper cells in the generation of alloantibody

and graft rejection. Curr. Opin. Organ Transplant. 21, 1–6 (2016).

113. Ridge, J. P., Di Rosa, F. & Matzinger, P. A conditioned dendritic cell can be a

temporal bridge between a CD4+ T-helper and a T-killer cell. Nature 393, 474–

478 (1998).

114. Schoenberger, S. P., Toes, R. E., van der Voort, E. I., Offringa, R. & Melief, C. J.

T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions.

Nature 393, 480–483 (1998).

115. Barry, M. & Bleackley, R. C. Cytotoxic T lymphocytes: all roads lead to death. Nat.

Rev. Immunol. 2, 401–9 (2002).

116. Oh, S. I. et al. Correlation of Fas and Fas ligand expression with rejection status

of transplanted heart in human. Transplantation 71, 906–909 (2001).

117. Kim, J. et al. The activating immunoreceptor NKG2D and its ligands are involved

in allograft transplant rejection. J. Immunol. 179, 6416–6420 (2007).

Page 141: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

128

118. Glocker, E.-O. et al. Inflammatory bowel disease and mutations affecting the

interleukin-10 receptor. N. Engl. J. Med. 361, 2033–45 (2009).

119. Meier-Kriesche, H. U., Schold, J. D. & Kaplan, B. Long-term renal allograft

survival: Have we made significant progress or is it time to rethink our analytic

and therapeutic strategies? Am. J. Transplant. 4, 1289–1295 (2004).

120. Stehlik, J. et al. The registry of the international society for heart and lung

transplantation: 29th official adult heart transplant report - 2012. J. Hear. Lung

Transplant. 31, 1052–1064 (2012).

121. Trulock, E. P. et al. Registry of the International Society for Heart and Lung

Transplantation: Twenty-fourth Official Adult Lung and Heart-Lung

Transplantation Report-2007. J. Hear. Lung Transplant. 26, 782–795 (2007).

122. Joosten, S. A., Sijpkens, Y. W. J., Van Kooten, C. & Paul, L. C. Chronic renal

allograft rejection: Pathophysiologic considerations. Kidney International 68, 1–13

(2005).

123. Rocha, P. N., Plumb, T. J., Crowley, S. D. & Coffman, T. M. Effector mechanisms

in transplant rejection. Immunol. Rev. 196, 51–64 (2003).

124. Costello, J. P., Mohanakumar, T. & Nath, D. S. Mechanisms of chronic cardiac

allograft rejection. Texas Hear. Inst. Journalart Inst. J. 40, 395–399 (2013).

125. Kfoury, A. G. et al. Cardiovascular Mortality Among Heart Transplant Recipients

With Asymptomatic Antibody-Mediated or Stable Mixed Cellular and Antibody-

Mediated Rejection. J. Hear. Lung Transplant. 28, 781–784 (2009).

126. McKenna, R. M., Takemoto, S. K. & Terasaki, P. I. Anti-HLA antibodies after solid

organ transplantation. Transplantation 69, 319–326 (2000).

127. Petrossian, G. A. et al. Relation between survival and development of coronary

artery disease and anti-HLA antibodies after cardiac transplantation. Circulation

80, III122–5 (1989).

Page 142: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

129

128. Nath, D. S., Basha, H. I. & Mohanakumar, T. Antihuman leukocyte antigen

antibody-induced autoimmunity: role in chronic rejection. Curr. Opin. Organ

Transplant. 15, 16–20 (2010).

129. Leong, H. S. et al. Vimentin autoantibodies induce platelet activation and

formation of platelet-leukocyte conjugates via platelet-activating factor. J. Leukoc.

Biol. 83, 263–271 (2008).

130. Corry, R. J., Winn, H. J. & Russell, P. S. Primarily vascularized allografts of hearts

in mice. The role of H-2D, H-2K, and non-H-2 antigens in rejection.

Transplantation 16, 343–350 (1973).

131. Russell, P. S., Chase, C. M., Winn, H. J. & Colvin, R. B. Coronary atherosclerosis

in transplanted mouse hearts. II. Importance of humoral immunity. J Immunol 152,

5135–5141 (1994).

132. Uehara, S. et al. Chronic cardiac transplant arteriopathy in mice: Relationship of

alloantibody, C4d deposition and neointimal fibrosis. Am. J. Transplant. 7, 57–65

(2007).

133. Hirohashi, T. et al. A novel pathway of chronic allograft rejection mediated by NK

cells and alloantibody. Am. J. Transplant. 12, 313–321 (2012).

134. Graham, J. A. et al. Viral infection induces de novo lesions of coronary allograft

vasculopathy through a natural killer cell-dependent pathway. Am. J. Transplant.

9, 2479–2484 (2009).

135. Fateh-Moghadam, S. et al. Cytomegalovirus infection status predicts progression

of heart-transplant vasculopathy. Transplantation 76, 1470–1474 (2003).

136. Zeng, Q. et al. B cells mediate chronic allograft rejection independently of

antibody production. J. Clin. Invest. 124, 1052–6 (2014).

137. Sis, B. et al. Endothelial gene expression in kidney transplants with alloantibody

indicates Antibody-mediated damage despite lack of C4d staining. Am. J.

Page 143: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

130

Transplant. 9, 2312–2323 (2009).

138. Rosenblum, M. D., Gratz, I. K., Paw, J. S. & Abbas, A. K. Treating human

autoimmunity: current practice and future prospects. Sci Transl Med 4, 125sr1

(2012).

139. Webber, A., Hirose, R. & Vincenti, F. Novel strategies in immunosuppression:

issues in perspective. Transplantation 91, 1057–1064 (2011).

140. Aliabadi, A., Cochrane, A. B. & Zuckermann, A. O. Current strategies and future

trends in immunosuppression after heart transplantation. Curr Opin Organ

Transpl. 17, 540–545 (2012).

141. Adams, D. H., Sanchez-Fueyo, A. & Samuel, D. From immunosuppression to

tolerance. J Hepatol 62, S170–s185 (2015).

142. Heilman, R. L., Mazur, M. J. & Reddy, K. S. Immunosuppression in simultaneous

pancreas-kidney transplantation: progress to date. Drugs 70, 793–804 (2010).

143. Du Pasquier, R. A., Pinschewer, D. D. & Merkler, D. Immunological mechanism of

action and clinical profile of disease-modifying treatments in multiple sclerosis.

CNS Drugs 28, 535–558 (2014).

144. Hartono, C., Muthukumar, T. & Suthanthiran, M. Immunosuppressive drug

therapy. Cold Spring Harb Perspect Med 3, a015487 (2013).

145. Brennan, D. C. et al. A randomized, double-blinded comparison of Thymoglobulin

versus Atgam for induction immunosuppressive therapy in adult renal transplant

recipients. Transplantation 67, 1011–1018 (1999).

146. Mohty, M. Mechanisms of action of antithymocyte globulin: T-cell depletion and

beyond. Leukemia 21, 1387–1394 (2007).

147. Wilde, M. I. & Goa, K. L. Muromonab CD3: a reappraisal of its pharmacology and

use as prophylaxis of solid organ transplant rejection. Drugs 51, 865–894 (1996).

Page 144: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

131

148. Webster, A. C., Playford, E. G., Higgins, G., Chapman, J. R. & Craig, J. C.

Interleukin 2 receptor antagonists for renal transplant recipients: a meta-analysis

of randomized trials. Transplantation 77, 166–176 (2004).

149. Ruck, T., Bittner, S., Wiendl, H. & Meuth, S. G. Alemtuzumab in multiple sclerosis:

Mechanism of action and beyond. International Journal of Molecular Sciences 16,

16414–16439 (2015).

150. Hanaway, M. J. et al. Alemtuzumab induction in renal transplantation. N. Engl. J.

Med. 364, 1909–1919 (2011).

151. Jordan, S. C., Choi, J. & Vo, A. Kidney transplantation in highly sensitized

patients. Br. Med. Bull. 114, 113–125 (2015).

152. Halloran, P. F. Immunosuppressive Drugs for Kidney Transplantation. N. Engl. J.

Med. 351, 2715–2729 (2004).

153. Jolles, S., Sewell, W. a C. & Misbah, S. a. Clinical uses of intravenous

immunoglobulin. Clin. Exp. Immunol. 142, 1–11 (2005).

154. Marfo, K., Lu, A., Ling, M. & Akalin, E. Desensitization protocols and their

outcome. Clin. J. Am. Soc. Nephrol. 6, 922–936 (2011).

155. Karam, S. & Wali, R. K. Current State of Immunosuppression: Past, Present, and

Future. Crit. Rev. Eukaryot. Gene Expr. 25, 113–34 (2015).

156. Kohei, N., Hirai, T., Omoto, K., Ishida, H. & Tanabe, K. Chronic antibody-

mediated rejection is reduced by targeting B-cell immunity during an introductory

period. Am. J. Transplant. 12, 469–476 (2012).

157. Vo, A. a et al. Benefits of rituximab combined with intravenous immunoglobulin for

desensitization in kidney transplant recipients. Transplantation 98, 312–9 (2014).

158. Ejaz, N. S. et al. Review of bortezomib treatment of antibody-mediated rejection in

renal transplantation. Antioxid. Redox Signal. 21, 2401–18 (2014).

Page 145: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

132

159. Patel, J. et al. Reduction of alloantibodies via proteasome inhibition in cardiac

transplantation. J. Heart Lung Transplant. 30, 1320–6 (2011).

160. Maltzman, J. S. & Koretzky, G. A. Azathioprine: old drug, new actions. J. Clin.

Invest. 111, 1122–4 (2003).

161. Tiede, I. et al. CD28-dependent Rac1 activation is the molecular target of

azathioprine in primary human CD4+ T lymphocytes. J Clin Invest 111, 1133–

1145 (2003).

162. Elion, G. B. The George Hitchings and Gertrude Elion Lecture. The pharmacology

of azathioprine. Ann N Y Acad Sci 685, 400–407 (1993).

163. Murray, J. E., Tilney, N. L. & Wilson, R. E. Renal transplantation: a twenty-five

year experience. Ann Surg 184, 565–573 (1976).

164. Starzl, T. E., Demetris, A. J. & Van Thiel, D. Liver transplantation (1). N Engl J

Med 321, 1014–1022 (1989).

165. Gisbert, J. P. & Gomollon, F. Thiopurine-induced myelotoxicity in patients with

inflammatory bowel disease: a review. Am J Gastroenterol 103, 1783–1800

(2008).

166. van Sandwijk, M. S., Bemelman, F. J. & Ten Berge, I. J. Immunosuppressive

drugs after solid organ transplantation. Neth J Med 71, 281–289 (2013).

167. Maripuri, S. & Kasiske, B. L. The role of mycophenolate mofetil in kidney

transplantation revisited. Transpl. Rev 28, 26–31 (2014).

168. Placebo-controlled study of mycophenolate mofetil combined with cyclosporin and

corticosteroids for prevention of acute rejection. European Mycophenolate Mofetil

Cooperative Study Group. Lancet 345, 1321–1325 (1995).

169. Allison, A. C. & Eugui, E. M. Mechanisms of action of mycophenolate mofetil in

preventing acute and chronic allograft rejection. Transplantation 80, S181–90

Page 146: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

133

(2005).

170. Eskin-Schwartz, M. & Mimouni, D. in Blistering Diseases: Clinical Features,

Pathogenesis, Treatment 483–491 (2015). doi:10.1007/978-3-662-45698-9_48

171. Barker, C. F. & Markmann, J. F. Historical overview of transplantation. Cold

Spring Harb. Perspect. Med. 3, (2013).

172. E.Chapman, A. E. C. The anti-inflammatory and immunosuppressive effects of

glucocorticoids, recent developments and mechanistic insights. Mol. Cell.

Endocrinol. 335, 2–13 (2011).

173. Luther, C. et al. Prednisolone treatment induces tolerogenic dendritic cells and a

regulatory milieu in myasthenia gravis patients. J.Immunol. 183, 841–848 (2009).

174. Rea, D. et al. Glucocorticoids transform CD40-triggering of dendritic cells into an

alternative activation pathway resulting in antigen-presenting cells that secrete IL-

10. Blood 95, 3162–7 (2000).

175. Liu, J. et al. Calcineurin is a common target of cyclophilin-cyclosporin A and

FKBP-FK506 complexes. Cell 66, 807–815 (1991).

176. Peng, S. L., Gerth, a J., Ranger, a M. & Glimcher, L. H. NFATc1 and NFATc2

together control both T and B cell activation and differentiation. Immunity 14, 13–

20 (2001).

177. Akool, E.-S. et al. Molecular mechanisms of TGF beta receptor-triggered signaling

cascades rapidly induced by the calcineurin inhibitors cyclosporin A and FK506. J.

Immunol. 181, 2831–2845 (2008).

178. Azzi, J. R., Sayegh, M. H. & Mallat, S. G. Calcineurin inhibitors: 40 years later,

can’t live without ... J. Immunol. 191, 5785–91 (2013).

179. Pirsch, J. D., Miller, J., Deierhoi, M. H., Vincenti, F. & Filo, R. S. A comparison of

tacrolimus (FK506) and cyclosporine for immunosuppression after cadaveric renal

Page 147: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

134

transplantation. FK506 Kidney Transplant Study Group. Transplantation 63,

(1997).

180. Macdonald, A. S. Use of mTOR inhibitors in human organ transplantation. Expert

Rev. Clin. Immunol. 3, 423–36 (2007).

181. Touzot, M., Soulillou, J. P. & Dantal, J. Mechanistic target of rapamycin inhibitors

in solid organ transplantation: from benchside to clinical use. Curr. Opin. Organ

Transplant. 17, 626–633 (2012).

182. Chi, H. Regulation and function of mTOR signalling in T cell fate decisions. Nat.

Rev. Immunol. 12, 325–38 (2012).

183. Powell, J. D., Pollizzi, K. N., Heikamp, E. B. & Horton, M. R. Regulation of

immune responses by mTOR. Annu. Rev. Immunol. 30, 39–68 (2012).

184. Weichhart, T., Hengstschlager, M. & Linke, M. Regulation of innate immune cell

function by mTOR. Nat. Rev. Immunol. 15, 599–614 (2015).

185. Pollizzi, K. N. & Powell, J. D. Regulation of T cells by mTOR: The known knowns

and the known unknowns. Trends in Immunology 36, 13–20 (2015).

186. Limon, J. J. & Fruman, D. A. Akt and mTOR in B cell activation and differentiation.

Front. Immunol. 3, (2012).

187. Sancak, Y. et al. The Rag GTPases bind raptor and mediate amino acid signaling

to mTORC1. Science 320, 1496–501 (2008).

188. Jacinto, E. et al. Mammalian TOR complex 2 controls the actin cytoskeleton and

is rapamycin insensitive. Nat. Cell Biol. 6, 1122–1128 (2004).

189. Schreiber, K. H. et al. Rapamycin-mediated mTORC2 inhibition is determined by

the relative expression of FK506-binding proteins. Aging Cell 14, 265–273 (2015).

190. Delgoffe, G. M. et al. The kinase mTOR regulates the differentiation of helper T

cells through the selective activation of signaling by mTORC1 and mTORC2. Nat.

Page 148: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

135

Immunol. 12, 295–303 (2011).

191. Oh, W. J. & Jacinto, E. mTOR complex 2 signaling and functions. Cell Cycle 10,

2305–2316 (2011).

192. Lehman, J. A., Calvo, V. & Gomez-Cambronero, J. Mechanism of ribosomal

p70s6 kinase activation by granulocyte macrophage colony-stimulating factor in

neutrophils. Cooperation of a MEK-related, Thr421/Ser424 kinase and a

rapamycin-sensitive, mTOR-related Thr389 kinase. J. Biol. Chem. 278, 28130–

28138 (2003).

193. Benjamin, D., Colombi, M., Moroni, C. & Hall, M. N. Rapamycin passes the torch:

a new generation of mTOR inhibitors. Nat. Rev. Drug Discov. 10, 868–880 (2011).

194. Sehgal, S. N. Sirolimus: Its discovery, biological properties, and mechanism of

action. in Transplantation Proceedings 35, (2003).

195. Powell, J. D., Lerner, C. G. & Schwartz, R. H. Inhibition of cell cycle progression

by rapamycin induces T cell clonal anergy even in the presence of costimulation.

J. Immunol. 162, 2775–2784 (1999).

196. Thomson, A. W., Turnquist, H. R. & Raimondi, G. Immunoregulatory functions of

mTOR inhibition. Nat. Rev. Immunol. 9, 324–37 (2009).

197. Araki, K. et al. mTOR regulates memory CD8 T-cell differentiation. Nature 460,

108–12 (2009).

198. Delgoffe, G. M. et al. The mTOR Kinase Differentially Regulates Effector and

Regulatory T Cell Lineage Commitment. Immunity 30, 832–844 (2009).

199. Battaglia, M. et al. Rapamycin promotes expansion of functional

CD4+CD25+FOXP3+ regulatory T cells of both healthy subjects and type 1

diabetic patients. J. Immunol. 177, 8338–47 (2006).

200. Kang, J., Huddleston, S. J., Fraser, J. M. & Khoruts, A. De novo induction of

Page 149: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

136

antigen-specific CD4+CD25+Foxp3+ regulatory T cells in vivo following systemic

antigen administration accompanied by blockade of mTOR. J. Leukoc. Biol. 83,

1230–1239 (2008).

201. Zeiser, R. et al. Differential impact of mammalian target of rapamycin inhibition on

CD4+CD25+Foxp3+ regulatory T cells compared with conventional CD4+ T cells.

Blood 111, 453–462 (2008).

202. Zhou, L. et al. {TGF}-β-induced Foxp3 inhibits {TH}17 cell differentiation by

antagonizing {RORγt} function. Nature 453, 236–240 (2008).

203. Sauer, S. et al. T cell receptor signaling controls Foxp3 expression via PI3K, Akt,

and mTOR. Proc. Natl. Acad. Sci. U. S. A. 105, 7797–7802 (2008).

204. Basu, S., Golovina, T., Mikheeva, T., June, C. H. & Riley, J. L. Cutting edge:

Foxp3-mediated induction of pim 2 allows human T regulatory cells to

preferentially expand in rapamycin. J. Immunol. 180, 5794–8 (2008).

205. Nakamura, T., Nakao, T., Yoshimura, N. & Ashihara, E. Rapamycin Prolongs

Cardiac Allograft Survival in a Mouse Model by Inducing Myeloid-Derived

Suppressor Cells. Am. J. Transplant 15, 2364–77 (2015).

206. Urbanellis, P. et al. The regulatory T cell effector molecule fibrinogen-like protein

2 is necessary for the development of rapamycin-induced tolerance to fully MHC-

mismatched murine cardiac allografts. Immunology 144, 91–106 (2015).

207. Horibe, E. K. et al. Rapamycin-conditioned, alloantigen-pulsed dendritic cells

promote indefinite survival of vascularized skin allografts in association with T

regulatory cell expansion. Transpl. Immunol. 18, 307–18 (2008).

208. Cai, L. et al. Galectin-9 in combination with rapamycin induces cardiac allograft

tolerance in mice. Transplantation 96, 379–86 (2013).

209. Peddi, V. R., Wiseman, A., Chavin, K. & Slakey, D. Review of combination

therapy with mTOR inhibitors and tacrolimus minimization after transplantation.

Page 150: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

137

Transplantation Reviews 27, 97–107 (2013).

210. Sterneck, M. et al. Everolimus and early calcineurin inhibitor withdrawal: 3-year

results from a randomized trial in liver transplantation. Am. J. Transplant. 14, 701–

710 (2014).

211. Levitsky, J. et al. Systemic immunoregulatory and proteogenomic effects of

tacrolimus to sirolimus conversion in liver transplant recipients. Hepatology 57,

239–248 (2013).

212. Eisen, H. J. et al. Everolimus versus mycophenolate mofetil in heart

transplantation: A randomized, multicenter trial. Am. J. Transplant. 13, 1203–1216

(2013).

213. Topilsky, Y. et al. Sirolimus as primary immunosuppression attenuates allograft

vasculopathy with improved late survival and decreased cardiac events after

cardiac transplantation. Circulation 125, 708–720 (2012).

214. Kobashigawa, J. a et al. Cardiac allograft vasculopathy by intravascular

ultrasound in heart transplant patients: substudy from the Everolimus versus

mycophenolate mofetil randomized, multicenter trial. JACC. Heart Fail. 1, 389–99

(2013).

215. Larsen, C. P. et al. Rational development of LEA29Y (belatacept), a high-affinity

variant of CTLA4-Ig with potent immunosuppressive properties. Am. J.

Transplant. 5, 443–453 (2005).

216. Vanrenterghem, Y. et al. Belatacept-based regimens are associated with

improved cardiovascular and metabolic risk factors compared with cyclosporine in

kidney transplant recipients (BENEFIT and BENEFIT-EXT studies).

Transplantation 91, 976–983 (2011).

217. Vincenti, F. et al. Belatacept and Long-Term Outcomes in Kidney Transplantation.

N. Engl. J. Med. 374, 333–43 (2016).

Page 151: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

138

218. Vincenti, F. et al. Randomized phase 2b trial of tofacitinib (CP-690,550) in de

novo kidney transplant patients: Efficacy, renal function and safety at 1 year. Am.

J. Transplant. 12, 2446–2456 (2012).

219. Vincenti, F. et al. Evaluation of the effect of tofacitinib exposure on outcomes in

kidney transplant patients. Am. J. Transplant. 15, 1644–1653 (2015).

220. Evenou, J.-P. et al. The potent protein kinase C-selective inhibitor AEB071

(sotrastaurin) represents a new class of immunosuppressive agents affecting

early T-cell activation. J. Pharmacol. Exp. Ther. 330, 792–801 (2009).

221. Pascher, a et al. Protein Kinase C Inhibitor Sotrastaurin in De Novo Liver

Transplant Recipients: A Randomized Phase II Trial. Am. J. Transplant 15, 1283–

92 (2015).

222. Russ, G. R. et al. Efficacy of sotrastaurin plus tacrolimus after de novo kidney

transplantation: Randomized, phase II trial results. Am. J. Transplant. 13, 1746–

1756 (2013).

223. Sykes, M. Immune tolerance in recipients of combined haploidentical bone

marrow and kidney transplantation. Bone Marrow Transplant. 50, S82–S86

(2015).

224. Sebastiaan Heidt, K. J. W. BIOMARKERS OF OPERATIONAL TOLERANCE IN

SOLID ORGAN TRANSPLANTATION. 6, 281–293 (2013).

225. Kingsley, C. I., Nadig, S. N. & Wood, K. J. Transplantation tolerance: Lessons

from experimental rodent models. Transplant International 20, 828–841 (2007).

226. Chong, A. S., Alegre, M. L., Miller, M. L. & Fairchild, R. L. Lessons and limits of

mouse models. Cold Spring Harb. Perspect. Med. 3, (2013).

227. Billingham, R. E., Brent, L. & Medawar, P. B. Actively Acquired Tolerance of

Foreign Cells. Nature 172, 603–606 (1953).

Page 152: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

139

228. Hogquist, K. a, Baldwin, T. a & Jameson, S. C. Central tolerance: learning self-

control in the thymus. Nat. Rev. Immunol. 5, 772–782 (2005).

229. Mueller, D. L. Mechanisms maintaining peripheral tolerance. Nat. Immunol. 11,

21–27 (2010).

230. Melchers, F. Checkpoints that control B cell development. J. Clin. Invest. 125,

2203–10 (2015).

231. Tobón, G. J., Izquierdo, J. H. & Cañas, C. A. B lymphocytes: development,

tolerance, and their role in autoimmunity-focus on systemic lupus erythematosus.

Autoimmune Dis. 2013, 827254 (2013).

232. Pelanda, R. & Torres, R. M. Central B-cell tolerance: where selection begins. Cold

Spring Harb. Perspect. Biol. 4, (2012).

233. Hardy, R. R. & Hayakawa, K. B cell development pathways. Annu. Rev. Immunol.

19, 595–621 (2001).

234. Grandien, A., Fucs, R., Nobrega, A., Andersson, J. & Coutinho, A. Negative

selection of multireactive B cell clones in normal adult mice. Eur J Immunol 24,

1345–1352 (1994).

235. Wardemann, H. et al. Predominant autoantibody production by early human B cell

precursors. Science 301, 1374–7 (2003).

236. Gay, D., Saunders, T., Camper, S. & Weigert, M. Receptor editing: an approach

by autoreactive B cells to escape tolerance. J. Exp. Med. 177, 999–1008 (1993).

237. Tiegs, S. L., Russell, D. M. & Nemazee, D. Receptor editing in self-reactive bone

marrow B cells. J. Exp. Med. 177, 1009–20 (1993).

238. Wardemann, H. & Nussenzweig, M. C. B-Cell Self-Tolerance in Humans.

Advances in Immunology 95, 83–110 (2007).

239. Brink, R. Regulation of B cell self-tolerance by BAFF. Seminars in Immunology

Page 153: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

140

18, 276–283 (2006).

240. Mauri, C. & Menon, M. The expanding family of regulatory B cells. International

Immunology 27, 479–486 (2015).

241. Griesemer, A. D., Sorenson, E. C. & Hardy, M. a. The role of the thymus in

tolerance. Transplantation 90, 465–74 (2010).

242. Klein, L., Kyewski, B., Allen, P. M. & Hogquist, K. a. Positive and negative

selection of the T cell repertoire: what thymocytes see (and don’t see). Nat. Rev.

Immunol. 14, 377–91 (2014).

243. Alpdogan, O. & Van Den Brink, M. R. M. Immune tolerance and transplantation.

Seminars in Oncology 39, 629–642 (2012).

244. Bhandoola, A., von Boehmer, H., Petrie, H. T. & Z????iga-Pfl??cker, J. C.

Commitment and Developmental Potential of Extrathymic and Intrathymic T Cell

Precursors: Plenty to Choose from. Immunity 26, 678–689 (2007).

245. Michie, A. M. & Zuniga-Pflucker, J. C. Regulation of thymocyte differentiation:

Pre-TCR signals and beta-selection. Seminars in Immunology 14, 311–323

(2002).

246. Ferreira, L. M. R. Gammadelta T cells: innately adaptive immune cells? Int. Rev.

Immunol. 32, 223–48 (2013).

247. Wang, L. & Bosselut, R. CD4-CD8 lineage differentiation: Thpok-ing into the

nucleus. J. Immunol. 183, 2903–2910 (2009).

248. Mathis, D. & Benoist, C. Aire. Annu. Rev. Immunol. 27, 287–312 (2009).

249. Villaseñor, J., Benoist, C. & Mathis, D. AIRE and APECED: Molecular insights into

an autoimmune disease. Immunol. Rev. 204, 156–164 (2005).

250. Takaba, H. et al. Fezf2 Orchestrates a Thymic Program of Self-Antigen

Expression for Immune Tolerance. Cell 163, 975–987 (2015).

Page 154: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

141

251. Wu, L. & Shortman, K. Heterogeneity of thymic dendritic cells. Seminars in

Immunology 17, 304–312 (2005).

252. Yamano, T. et al. Thymic B Cells Are Licensed to Present Self Antigens for

Central T Cell Tolerance Induction. Immunity 42, 1048–1061 (2015).

253. Buhlmann, J. E., Elkin, S. K. & Sharpe, A. H. A role for the B7-1/B7-

2:CD28/CTLA-4 pathway during negative selection. J. Immunol. 170, 5421–5428

(2003).

254. von Boehmer, H. & Melchers, F. Checkpoints in lymphocyte development and

autoimmune disease. Nat. Immunol. 11, 14–20 (2010).

255. Hsieh, C.-S., Lee, H.-M. & Lio, C.-W. J. Selection of regulatory T cells in the

thymus. Nat. Rev. Immunol. 12, 157–167 (2012).

256. Enouz, S. et al. Autoreactive T cells bypass negative selection and respond to

self-antigen stimulation during infection. J Exp Med 209, 1769–1779 (2012).

257. Xing, Y. & Hogquist, K. A. T-Cell tolerance: Central and peripheral. Cold Spring

Harb. Perspect. Biol. 4, 1–15 (2012).

258. Parish, I. a & Heath, W. R. Too dangerous to ignore: self-tolerance and the

control of ignorant autoreactive T cells. Immunol. Cell Biol. 86, 146–152 (2008).

259. Ohashi, P. S. et al. Ablation of ‘tolerance’ and induction of diabetes by virus infec-

tion in viral antigen transgenic mice. Cell 65, 305–317 (1991).

260. Bianchi, T. et al. Maintenance of peripheral tolerance through controlled tissue

homing of antigen-specific T cells in K14-mOVA mice. J Immunol 182, 4665–4674

(2009).

261. Chappert, P. & Schwartz, R. H. Induction of T cell anergy: Integration of

environmental cues and infectious tolerance. Curr. Opin. Immunol. 22, 552–559

(2010).

Page 155: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

142

262. Wells AD. New insights into the molecular basis of T cell anergy: anergy factors,

avoidance sensors, and epigenetic imprint. J. Immunol. 182, 7331–7341 (2009).

263. Jenkins, M. K. & Schwartz, R. H. Antigen presentation by chemically modified

splenocytes induces antigen-specific T cell unresponsiveness in vitro and in vivo.

J. Exp. Med. 165, 302–319 (1987).

264. Harding, F. a, McArthur, J. G., Gross, J. a, Raulet, D. H. & Allison, J. P. CD28-

mediated signalling co-stimulates murine T cells and prevents induction of anergy

in T-cell clones. Nature 356, 607–609 (1992).

265. Valdor, R. & Macian, F. Induction and stability of the anergic phenotype in T cells.

Seminars in Immunology 25, 313–320 (2013).

266. Walunas, T. L. & Bluestone, J. A. CTLA-4 regulates tolerance induction and T cell

differentiation in vivo. J. Immunol. 160, 3855–3860 (1998).

267. Waterhouse P, Penninger JM, Timms E, Wakeham A, Sha- hinian A, Lee KP,

Thompson CB, Griesser H, M. T. Lymphoproliferative disorders with early lethality

in mice deficient in Ctla-4. Science (80-. ). 270, 985–988 (1995).

268. Perez, V. L. et al. Induction of peripheral T cell tolerance in vivo requires CTLA-4

engagement. Immunity 6, 411–417 (1997).

269. Keir ME, ButteMJ, Freeman GJ, S. PD-1 and its ligands in tolerance and

immunity. Annu. Rev. Immunol. 26, 677–704 (2008).

270. Francisco, L. M., Sage, P. T. & Sharpe, A. H. The PD-1 pathway in tolerance and

autoimmunity. Immunological Reviews 236, 219–242 (2010).

271. Probst, H. C., McCoy, K., Okazaki, T., Honjo, T. & van den Broek, M. Resting

dendritic cells induce peripheral CD8+ T cell tolerance through PD-1 and CTLA-4.

Nat. Immunol. 6, 280–286 (2005).

272. Francisco, L. M. et al. PD-L1 regulates the development, maintenance, and

Page 156: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

143

function of induced regulatory T cells. J. Exp. Med. 206, 3015–29 (2009).

273. Nishimura H, Nose M, Hiai H, Minato N, H. T. Development of lupus-like

autoimmune diseases by dis- ruption of the PD-1 gene encoding an ITIM motif-

car- rying immunoreceptor. Immunity 11, 141–151 (1999).

274. Zheng Y, Delgoffe GM, Meyer CF, Chan W, P. J. Anergic T cells are metabolically

anergic. J. Immunol. 183, 6095–6101 (2009).

275. Sykes, M. Immune tolerance: Mechanisms and application in clinical

transplantation. Journal of Internal Medicine 262, 288–310 (2007).

276. Chiodetti, L., Choi, S., Barber, D. L. & Schwartz, R. H. Adaptive tolerance and

clonal anergy are distinct biochemical states. J. Immunol. 176, 2279–2291 (2006).

277. Krammer, P. H., Arnold, R. & Lavrik, I. N. Life and death in peripheral T cells. Nat.

Rev. Immunol. 7, 532–542 (2007).

278. Arakaki, R., Yamada, A., Kudo, Y., Hayashi, Y. & Ishimaru, N. Mechanism of

activation-induced cell death of T cells and regulation of FasL expression. Crit.

Rev. Immunol. 34, 301–14 (2014).

279. Ashkenazi, A. & Salvesen, G. Regulated cell death: signaling and mechanisms.

Annu. Rev. Cell Dev. Biol. 30, 337–356 (2014).

280. Turbyville, J. C. & Rao, V. K. The autoimmune lymphoproliferative syndrome: A

rare disorder providing clues about normal tolerance. Autoimmunity Reviews 9,

488–493 (2010).

281. Liu, J. et al. Genomic view of systemic autoimmunity in MRLlpr mice. Genes

Immun. 7, 156–68 (2006).

282. Ferrer, I. R., Hester, J., Bushell, A. & Wood, K. J. Induction of transplantation

tolerance through regulatory cells: From mice to men. Immunol. Rev. 258, 102–

116 (2014).

Page 157: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

144

283. Scalea, J. R., Tomita, Y., Lindholm, C. R. & Burlingham, W. Transplantation

tolerance induction: Cell therapies and their mechanisms. Frontiers in

Immunology 7, (2016).

284. Merad, M., Sathe, P., Helft, J., Miller, J. & Mortha, A. The dendritic cell lineage:

ontogeny and function of dendritic cells and their subsets in the steady state and

the inflamed setting. Annu. Rev. Immunol. 31, 563–604 (2013).

285. Morelli, A. E. & Thomson, A. W. Tolerogenic dendritic cells and the quest for

transplant tolerance. Nat. Rev. Immunol. 7, 610–621 (2007).

286. Liu, J. & Cao, X. Regulatory dendritic cells in autoimmunity: A comprehensive

review. Journal of Autoimmunity 63, 1–12 (2015).

287. Reizis, B., Bunin, A., Ghosh, H. S., Lewis, K. L. & Sisirak, V. Plasmacytoid

dendritic cells: recent progress and open questions. Annu. Rev. Immunol. 29,

163–83 (2011).

288. Ohnmacht, C. et al. Constitutive ablation of dendritic cells breaks self-tolerance of

CD4 T cells and results in spontaneous fatal autoimmunity. J. Exp. Med. 206,

549–59 (2009).

289. Hawiger, D. et al. Dendritic cells induce peripheral T cell unresponsiveness under

steady state conditions in vivo. J. Exp. Med. 194, 769–779 (2001).

290. Manicassamy, S. & Pulendran, B. Dendritic cell control of tolerogenic responses.

Immunological Reviews 241, 206–227 (2011).

291. Miyagaki, T., Fujimoto, M. & Sato, S. Regulatory B cells in human inflammatory

and autoimmune diseases: From mouse models to clinical research. Int. Immunol.

27, 495–504 (2015).

292. Yang, M., Rui, K., Wang, S. & Lu, L. Regulatory B cells in autoimmune diseases.

Cell. Mol. Immunol. 10, 122–32 (2013).

Page 158: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

145

293. KATZ, S. I., PARKER, D. & TURK, J. L. B-cell suppression of delayed

hypersensitivity reactions. Nature 251, 550–551 (1974).

294. Wolf, S. D., Dittel, B. N., Hardardottir, F. & Janeway, C. A. Experimental

autoimmune encephalomyelitis induction in genetically B cell-deficient mice. J.

Exp. Med. 184, 2271–8 (1996).

295. Chesneau, M., Michel, L., Degauque, N. & Brouard, S. Regulatory B cells and

tolerance in transplantation: From animal models to human. Frontiers in

Immunology 4, (2013).

296. Fillatreau, S., Sweenie, C. H., McGeachy, M. J., Gray, D. & Anderton, S. M. B

cells regulate autoimmunity by provision of IL-10. Nat. Immunol. 3, 944–50

(2002).

297. Lampropoulou, V. et al. TLR-activated B cells suppress T cell-mediated

autoimmunity. J. Immunol. 180, 4763–4773 (2008).

298. Evans, J. G. et al. Novel Suppressive Function of Transitional 2 B Cells in

Experimental Arthritis. J. Immunol. 178, 7868–7878 (2007).

299. Yanaba, K. et al. A Regulatory B Cell Subset with a Unique CD1dhiCD5+

Phenotype Controls T Cell-Dependent Inflammatory Responses. Immunity 28,

639–650 (2008).

300. Maseda, D. et al. Regulatory B10 cells differentiate into antibody-secreting cells

after transient IL-10 production in vivo. J. Immunol. 188, 1036–48 (2012).

301. Blair, P. A. et al. CD19+CD24hiCD38hi B Cells Exhibit Regulatory Capacity in

Healthy Individuals but Are Functionally Impaired in Systemic Lupus

Erythematosus Patients. Immunity 32, 129–140 (2010).

302. Flores-Borja, F. et al. CD19+CD24hiCD38hi B cells maintain regulatory T cells

while limiting TH1 and TH17 differentiation. Sci. Transl. Med. 5, 173ra23 (2013).

Page 159: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

146

303. Knippenberg, S. et al. Reduction in IL-10 producing B cells (Breg) in multiple

sclerosis is accompanied by a reduced naïve/memory Breg ratio during a relapse

but not in remission. J. Neuroimmunol. 239, 80–86 (2011).

304. Deng, S. et al. Cutting edge: transplant tolerance induced by anti-CD45RB

requires B lymphocytes. J. Immunol. 178, 6028–6032 (2007).

305. Brouard, S. et al. Identification of a peripheral blood transcriptional biomarker

panel associated with operational renal allograft tolerance. Proc. Natl. Acad. Sci.

U. S. A. 104, 15448–15453 (2007).

306. M., C. et al. Tolerant kidney transplant patients produce B cells with regulatory

properties. J. Am. Soc. Nephrol. 26, 2588–2598 (2015).

307. Bluestone, J. a, Bour-Jordan, H., Cheng, M. & Anderson, M. T cells in the control

of organ-specific autoimmunity. J. Clin. Invest. 125, 2250–60 (2015).

308. Grant, C. R., Liberal, R., Mieli-Vergani, G., Vergani, D. & Longhi, M. S. Regulatory

T-cells in autoimmune diseases: Challenges, controversies and-yet-unanswered

questions. Autoimmunity Reviews 14, 105–116 (2015).

309. Gershon, R. & Kondo, K. Cell interactions in the induction of tolerance: the role of

thymic lymphocytes. Immunology 18, 723–737 (1970).

310. Sakaguchi, S., Sakaguchi, N., Asano, M., Itoh, M. & Toda, M. Immunologic self-

tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains

(CD25). Breakdown of a single mechanism of self-tolerance causes various

autoimmune diseases. J Immunol 155, 1151–1164 (1995).

311. Asano, M., Toda, M., Sakaguchi, N. & Sakaguchi, S. Autoimmune disease as a

consequence of develop- mental abnormality of a T cell subpopulation. J. Exp.

Med. 184, 387–396 (1996).

312. Fontenot, J. D., Gavin, M. A. & Rudensky, A. Y. Foxp3 programs the development

and function of CD4+CD25+ regulatory T cells. Nat. Immunol. 4, 330–336 (2003).

Page 160: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

147

313. Hori, S., Nomura, T. & Sakaguchi, S. Control of regulatory T cell development by

the transcription factor Foxp3. Science (80-. ). 299, 1057–1061 (2003).

314. Williams, L. M. & Rudensky, A. Y. Maintenance of the Foxp3-dependent

developmental program in mature regulatory T cells requires continued

expression of Foxp3. Nat. Immunol. 8, 277–284 (2007).

315. Khattri, R., Cox, T., Yasayko, S.-A. & Ramsdell, F. An essential role for Scurfin in

CD4+CD25+ T regulatory cells. Nat. Immunol. 4, 337–342 (2003).

316. Bennett, C. L. et al. The immune dysregulation, polyendocrinopathy, enteropathy,

X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat. Genet. 27, 20–

1 (2001).

317. Pot, C., Apetoh, L. & Kuchroo, V. K. Type 1 regulatory T cells (Tr1) in

autoimmunity. Seminars in Immunology 23, 202–208 (2011).

318. Wood, K. J., Bushell, A. & Hester, J. Regulatory immune cells in transplantation.

Nat. Rev. Immunol. 12, 417–30 (2012).

319. Feh??rvari, Z. & Sakaguchi, S. CD4+ Tregs and immune control. Journal of

Clinical Investigation 114, 1209–1217 (2004).

320. Josefowicz, S. Z., Lu, L.-F. & Rudensky, A. Y. Regulatory T cells: mechanisms of

differentiation and function. Annu. Rev. Immunol. 30, 531–564 (2012).

321. Waldmann, H. & Hilbrands, R. Harnessing FOXP3+ regulatory T cells for

transplantation tolerance. J. Clin. … 124, (2014).

322. Yadav, M., Stephan, S. & Bluestone, J. A. Peripherally induced Tregs-role in

immune homeostasis and autoimmunity. Front. Immunol. 4, (2013).

323. Bilate, A. . & Lafaille, J. . Induced CD4+Foxp3+ regulatory T cells in immune

tolerance. Annu. Rev. Immunol. 30, 733–758 (2012).

324. Feuerer, M. et al. Genomic definition of multiple ex vivo regulatory T cell

Page 161: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

148

subphenotypes. Proc Natl Acad Sci U S A 107, 5919–5924 (2010).

325. Thornton, A. M. et al. Expression of Helios, an Ikaros transcription factor family

member, differentiates thymic-derived from peripherally induced Foxp3+ T

regulatory cells. J. Immunol. 184, 3433–3441 (2010).

326. Yadav, M. et al. Neuropilin-1 distinguishes natural and inducible regulatory T cells

among regulatory T cell subsets in vivo. J. Exp. Med. 209, 1713–22, S1–19

(2012).

327. Joller, N. et al. Treg cells expressing the coinhibitory molecule TIGIT selectively

inhibit proinflammatory Th1 and Th17 cell responses. Immunity 40, 569–581

(2014).

328. Chruscinski, A. et al. Role of Regulatory T Cells (Treg) and the Treg Effector

Molecule Fibrinogen-like Protein 2 in Alloimmunity and Autoimmunity. Rambam

Maimonides Med. J. 6, e0024 (2015).

329. Kmieciak, M. et al. Human T cells express CD25 and Foxp3 upon activation and

exhibit effector/memory phenotypes without any regulatory/suppressor function. J.

Transl. Med. 7, 89 (2009).

330. Liu, W. et al. CD127 expression inversely correlates with FoxP3 and suppressive

function of human CD4+ T reg cells. J. Exp. Med. 7, 1701–1711 (2006).

331. Li, S., Gowans, E. J., Chougnet, C., Plebanski, M. & Dittmer, U. Natural regulatory

T cells and persistent viral infection. J. Virol. 82, 21–30 (2008).

332. Beyer, M. & Schultze, J. L. Regulatory T cells in cancer. Blood 108, 804–811

(2006).

333. Caridade, M., Graca, L. & Ribeiro, R. M. Mechanisms underlying CD4+ Treg

immune regulation in the adult: From experiments to models. Frontiers in

Immunology 4, (2013).

Page 162: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

149

334. Schmidt, A., Oberle, N. & Krammer, P. H. Molecular mechanisms of treg-

mediated T cell suppression. Front. Immunol. 3, 51 (2012).

335. Shevach, E. M. Mechanisms of Foxp3+ T Regulatory Cell-Mediated Suppression.

Immunity 30, 636–645 (2009).

336. Brent, L. Transplantation tolerance - a historical introduction. Immunology 1958,

n/a–n/a (2015).

337. Ford, M. L., Adams, A. B. & Pearson, T. C. Targeting co-stimulatory pathways:

transplantation and autoimmunity. Nat. Rev. Nephrol. 10, 14–24 (2014).

338. Sykes, M. Hematopoietic cell transplantation for tolerance induction: animal

models to clinical trials. Transplantation 87, 309–16 (2009).

339. Abrahamsson, S. & Muraro, P. A. Immune re-education following autologous

hematopoietic stem cell transplantation. Autoimmunity (2009).

340. Pilat, N. & Wekerle, T. Transplantation tolerance through mixed chimerism. Nat.

Rev. Nephrol. 6, 594–605 (2010).

341. Butte, M. J., Keir, M. E., Phamduy, T. B., Sharpe, A. H. & Freeman, G. J.

Programmed Death-1 Ligand 1 Interacts Specifically with the B7-1 Costimulatory

Molecule to Inhibit T Cell Responses. Immunity 27, 111–122 (2007).

342. Suntharalingam, G. et al. Cytokine Storm in a Phase 1 Trial of the Anti-CD28

Monoclonal Antibody TGN1412. N. Engl. J. Med. 335, 1018–1028 (2006).

343. Yamada And, A. & Sayegh, M. H. The CD154-CD40 costimulatory pathway in

transplantation. Transplantation 73, S36–9 (2002).

344. Kinnear, G., Jones, N. D. & Wood, K. J. Costimulation blockade: current

perspectives and implications for therapy. Transplantation 95, 527–35 (2013).

345. Kawai, T., Andrews, D., Colvin, R. B., Sachs, D. H. & Cosimi, A. B.

Thromboembolic complications after treatment with monoclonal antibody against

Page 163: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

150

CD40 ligand. Nat Med 6, 114 (2000).

346. Ford, M. L. T Cell Cosignaling Molecules in Transplantation. Immunity 44, 1020–

1033 (2016).

347. Springer, T. A. & Dustin, M. L. Integrin inside-out signaling and the immunological

synapse. Current Opinion in Cell Biology 24, 107–115 (2012).

348. Weaver, T. a et al. Alefacept promotes co-stimulation blockade based allograft

survival in nonhuman primates. Nat. Med. 15, 746–749 (2009).

349. Paulos, C. M. et al. The inducible costimulator (ICOS) is critical for the

development of human T(H)17 cells. Sci. Transl. Med. 2, 55ra78 (2010).

350. Perdigoto, A. L., Chatenoud, L., Bluestone, J. A. & Herold, K. C. Inducing and

administering tregs to treat human disease. Frontiers in Immunology 6, (2016).

351. Monti, P. et al. Rapamycin monotherapy in patients with type 1 diabetes modifies

CD4+CD25+FOXP3+ regulatory T-cells. Diabetes 57, 2341–7 (2008).

352. Sabbatini, M. et al. Oscillatory mTOR inhibition and Treg increase in kidney

transplantation. Clin. Exp. Immunol. 182, 230–240 (2015).

353. Qin, S. et al. ‘Infectious’ transplantation tolerance. Science 259, 974–7 (1993).

354. van der Net, J. B., Bushell, A., Wood, K. J. & Harden, P. N. Regulatory T cells:

first steps of clinical application in solid organ transplantation. Transpl. Int. n/a–n/a

(2015). doi:10.1111/tri.12608

355. van der Net, J. B., Bushell, A., Wood, K. J. & Harden, P. N. Regulatory T cells:

first steps of clinical application in solid organ transplantation. Transpl. Int. n/a–n/a

(2015). doi:10.1111/tri.12608

356. Geissler, E. K. The ONE Study compares cell therapy products in organ

transplantation: introduction to a review series on suppressive monocyte-derived

cells. Transplant. Res. 1, 1–3 (2012).

Page 164: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

151

357. Jenq, R. R. & van den Brink, M. R. M. Allogeneic haematopoietic stem cell

transplantation: individualized stem cell and immune therapy of cancer. Nat. Rev.

Cancer 10, 213–221 (2010).

358. Daikeler, T., Tichelli, A. & Passweg, J. Complications of autologous hematopoietic

stem cell transplantation for patients with autoimmune diseases. Pediatr. Res. 71,

439–444 (2012).

359. Muraro, P. A. et al. T cell repertoire following autologous stem cell transplantation

for multiple sclerosis. J. Clin. Invest. 124, 1168–72 (2014).

360. Sachs, D. H., Kawai, T. & Sykes, M. Induction of tolerance through mixed

chimerism. Cold Spring Harb. Perspect. Med. 4, (2014).

361. van Bekkum, D. W., Bohre, E. P., Houben, P. F. & Knaan-Shanzer, S. Regression

of adjuvant-induced arthritis in rats following bone marrow transplantation. Proc.

Natl. Acad. Sci. U. S. A. 86, 10090–10094 (1989).

362. Atkins, H. L., Muraro, P. A., van Laar, J. M. & Pavletic, S. Z. Autologous

hematopoietic stem cell transplantation for autoimmune disease--is it now ready

for prime time? Biol. Blood Marrow Transplant. 18, S177–83 (2012).

363. Hamadani, M. Autologous hematopoietic cell transplantation: an update for

clinicians. Ann. Med. 46, 619–632 (2014).

364. Ljungman, P. et al. Allogeneic and autologous transplantation for haematological

diseases, solid tumours and immune disorders: current practice in Europe 2009.

Bone Marrow Transpl. 45, 219–234 (2010).

365. Baron, F. & Storb, R. Allogeneic hematopoietic cell transplantation as treatment

for hematological malignancies: A review. Springer Seminars in Immunopathology

26, 71–94 (2004).

366. Weissman, I. L. & Shizuru, J. A. The origins of the identification and isolation of

hematopoietic stem cells, and their capability to induce donor-specific

Page 165: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

152

transplantation tolerance and treat autoimmune diseases. Blood 112, 3543–3553

(2008).

367. Till, J. E. & McCULLOCH, E. a. A direct measurement of the radiation sensitivity

of normal mouse bone marrow cells. Radiat. Res. 14, 213–222 (1961).

368. Becker, a J., McCULLOCH, E. a & Till, J. E. Cytological demonstration of the

clonal nature of spleen colonies derived from transplanted mouse marrow cells.

Nature 197, 452–454 (1963).

369. Siminovitch, L., Mcculloch, E. a & Till, J. E. the Distribution of Colony-Forming

Cells Among Spleen Colonies. J. Cell. Physiol. 62, 327–336 (1963).

370. Seita, J. & Weissman, I. L. Hematopoietic stem cell: Self-renewal versus

differentiation. Wiley Interdisciplinary Reviews: Systems Biology and Medicine 2,

640–653 (2010).

371. Doulatov, S., Notta, F., Laurenti, E. & Dick, J. E. Hematopoiesis: A human

perspective. Cell Stem Cell 10, 120–136 (2012).

372. Notta, F. et al. Distinct routes of lineage development reshape the human blood

hierarchy across ontogeny. Science 351, 1–16 (2015).

373. Spangrude, G. J., Heimfeld, S. & Weissman, I. L. Purification and characterization

of mouse hematopoietic stem cells. Sci. (New York, NY) 241, 58–62 (1988).

374. Ikuta, K. & Weissman, I. L. Evidence that hematopoietic stem cells express

mouse c-kit but do not depend on steel factor for their generation. Proc. Natl.

Acad. Sci. U. S. A. 89, 1502–6 (1992).

375. Kiel, M. J. et al. SLAM family receptors distinguish hematopoietic stem and

progenitor cells and reveal endothelial niches for stem cells. Cell 121, 1109–1121

(2005).

376. Challen, G. A., Boles, N., Lin, K. K.-Y. & Goodell, M. A. Mouse hematopoietic

Page 166: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

153

stem cell identification and analysis. Cytometry. A 75, 14–24 (2009).

377. Civin, C. I. et al. Antigenic analysis of hematopoiesis. III. A hematopoietic

progenitor cell surface antigen defined by a monoclonal antibody raised against

KG-1a cells. J. Immunol. 133, 157–165 (1984).

378. Notta, F. et al. Isolation of single human hematopoietic stem cells capable of long-

term multilineage engraftment. Science 333, 218–221 (2011).

379. Copelan, E. A. Hematopoietic stem-cell transplantation. N. Engl. J. Med. 354,

1813–1826 (2006).

380. Gallardo, D. et al. Is mobilized peripheral blood comparable with bone marrow as

a source of hematopoietic stem cells for allogeneic transplantation from HLA-

identical sibling donors? A case-control study. Haematologica 94, 1282–1288

(2009).

381. Flomenberg, N. et al. The use of AMD3100 plus G-CSF for autologous

hematopoietic progenitor cell mobilization is superior to G-CSF alone. Blood 106,

1867–1874 (2005).

382. Pelus, L. M. Peripheral blood stem cell mobilization: new regimens, new cells,

where do we stand. Curr. Opin. Hematol. 15, 285–92 (2008).

383. Pelus, L. M. Peripheral blood stem cell mobilization: new regimens, new cells,

where do we stand. Curr. Opin. Hematol. 15, 285–92 (2008).

384. Jenq, R. R. & van den Brink, M. R. M. Allogeneic haematopoietic stem cell

transplantation: individualized stem cell and immune therapy of cancer. Nat. Rev.

Cancer 10, 213–221 (2010).

385. Ildstad, S. T. & Sachs, D. H. Reconstitution with syngeneic plus allogeneic or

xenogeneic bone marrow leads to specific acceptance of allografts or xenografts.

Nature 307, 168–170 (1984).

Page 167: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

154

386. Sharabi, Y. & Sachs, D. H. Mixed chimerism and permanent specific

transplantation tolerance induced by a nonlethal preparative regimen. J. Exp.

Med. 169, 493–502 (1989).

387. Kawai, T., Cosimi, a B. & Sachs, D. H. Preclinical and clinical studies on the

induction of renal allograft tolerance through transient mixed chimerism. Curr.

Opin. Organ Transplant. 16, 366–71 (2011).

388. Kawai, T. et al. CD154 blockade for induction of mixed chimerism and prolonged

renal allograft survival in nonhuman primates. Am. J. Transplant. 4, 1391–1398

(2004).

389. Scandling, J. D. et al. Tolerance and chimerism after renal and hematopoietic-cell

transplantation. N. Engl. J. Med. 358, 362–368 (2008).

390. Scandling, J. D. et al. Chimerism, graft survival, and withdrawal of

immunosuppressive drugs in HLA matched and mismatched patients after living

donor kidney and hematopoietic cell transplantation. Am. J. Transplant 15, 695–

704 (2015).

391. Millan, M. T. et al. Mixed chimerism and immunosuppressive drug withdrawal

after HLA-mismatched kidney and hematopoietic progenitor transplantation.

Transplantation 73, 1386–91 (2002).

392. Kawai, T. et al. HLA-mismatched renal transplantation without maintenance

immunosuppression. N. Engl. J. Med. 358, 353–361 (2008).

393. Kawai, T. et al. Long-term results in recipients of combined HLA-mismatched

kidney and bone marrow transplantation without maintenance

immunosuppression. Am. J. Transplant. 14, 1599–1611 (2014).

394. Maiolino, a et al. Engraftment syndrome following autologous hematopoietic stem

cell transplantation: definition of diagnostic criteria. Bone Marrow Transplant. 31,

393–397 (2003).

Page 168: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

155

395. Leventhal, J. et al. Chimerism and tolerance without GVHD or engraftment

syndrome in HLA-mismatched combined kidney and hematopoietic stem cell

transplantation. Sci. Transl. Med. 4, 124ra28 (2012).

396. Leventhal, J. R. et al. Immune reconstitution/immunocompetence in recipients of

kidney plus hematopoietic stem/facilitating cell transplants. Transplantation 99,

288–298 (2015).

397. Strober, S. Use of hematopoietic cell transplants to achieve tolerance in patients

with solid organ transplants. Blood 127, 1539–1543 (2016).

398. Duran-Struuck, R. & Dysko, R. C. Principles of Bone Marrow Transplantation

(BMT): Providing Optimal Veterinary and Husbandry Care to Irradiated Mice in

BMT Studies. J. Am. Assoc. Lab. Anim. Sci. 48, 11–22 (2009).

399. Snowden, J. a et al. Haematopoietic SCT in severe autoimmune diseases:

updated guidelines of the European Group for Blood and Marrow Transplantation.

Bone Marrow Transplant. 1–21 (2011). doi:10.1038/bmt.2011.185

400. Snowden, J. a et al. Haematopoietic SCT in severe autoimmune diseases:

updated guidelines of the European Group for Blood and Marrow Transplantation.

Bone Marrow Transplant. 1–21 (2011). doi:10.1038/bmt.2011.185

401. Arruda, L. C. M. et al. Resetting the immune response after autologous

hematopoietic stem cell transplantation for autoimmune diseases. Curr. Res.

Transl. Med. 64, 107–113 (2016).

402. Arruda, L. C. M. et al. Resetting the immune response after autologous

hematopoietic stem cell transplantation for autoimmune diseases. Curr. Res.

Transl. Med. 64, 107–113 (2016).

403. Burt, R. K. et al. Autologous non-myeloablative haemopoietic stem-cell

transplantation compared with pulse cyclophosphamide once per month for

systemic sclerosis (ASSIST): An open-label, randomised phase 2 trial. Lancet

Page 169: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

156

378, 498–506 (2011).

404. Atkins, H. L. et al. Immunoablation and autologous haemopoietic stem-cell

transplantation for aggressive multiple sclerosis: a multicentre single-group phase

2 trial. Lancet (London, England) (2016). doi:10.1016/S0140-6736(16)30169-6

405. Farge, D. et al. Analysis of immune reconstitution after autologous bone marrow

transplantation in systemic sclerosis. Arthritis Rheum. 52, 1555–1563 (2005).

406. Delemarre, E. M. et al. Autologous stem cell transplantation AIDS autoimmune

patients by functional renewal and TCR diversification of regulatory T cells. Blood

127, 91–101 (2016).

407. de Kleer, I. et al. Autologous stem cell transplantation for autoimmunity induces

immunologic self-tolerance by reprogramming autoreactive T cells and restoring

the CD4+CD25+ immune regulatory network. Blood 107, 1696–702 (2006).

408. Abrahamsson, S. V. et al. Non-myeloablative autologous haematopoietic stem

cell transplantation expands regulatory cells and depletes IL-17 producing

mucosal-associated invariant T cells in multiple sclerosis. Brain 136, 2888–2903

(2013).

409. Dusseaux, M. et al. Human MAIT cells are xenobiotic-resistant, tissue-targeted,

CD161 hi IL-17-secreting T cells. Blood 117, 1250–1259 (2011).

410. Arruda, L. C. M. et al. THU0501 Hematopoietic Stem Cell Transplantation

Increases Naive and Regulatory B Cells While Decreasing Memory B Cells in

Systemic Sclerosis Patients. Ann. Rheum. Dis. 73 , 356 (2014).

411. Arruda, L. C. M. et al. THU0501 Hematopoietic Stem Cell Transplantation

Increases Naive and Regulatory B Cells While Decreasing Memory B Cells in

Systemic Sclerosis Patients. Ann. Rheum. Dis. 73 , 356 (2014).

412. Adolfsson, J. et al. Upregulation of Flt3 expression within the bone marrow Lin-

Sca1+c-kit+ stem cell compartment is accompanied by loss of self-renewal

Page 170: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

157

capacity. Immunity 15, 659–669 (2001).

413. Grassinger, J., Haylock, D. N., Williams, B., Olsen, G. H. & Nilsson, S. K.

Phenotypically identical hemopoietic stem cells isolated from different regions of

bone marrow have different biologic potential. Blood 116, 3185–3196 (2010).

414. Chen, B. J., Cui, X., Sempowski, G. D., Domen, J. & Chao, N. J. Hematopoietic

stem cell dose correlates with the speed of immune reconstitution after stem cell

transplantation. Blood 103, 4344–4352 (2004).

415. Kruisbeek, A. M., Shevach, E. & Thornton, A. M. Proliferative assays for T cell

function. Curr. Protoc. Immunol. Chapter 3, Unit 3.12 (2004).

416. Xu, H. et al. Costimulatory blockade of CD154-CD40 in combination with T-cell

lymphodepletion results in prevention of allogeneic sensitization. Blood 111,

3266–3275 (2008).

417. Mastoridis, S., Martínez-Llordella, M. & Sanchez-Fueyo, A. Biomarkers and

immunopathology of tolerance. Curr. Opin. Organ Transplant. (2015).

doi:10.1097/MOT.0000000000000269

418. Tomita, Y., Khan, A. & Sykes, M. Role of intrathymic clonal deletion and

peripheral anergy in transplantation tolerance induced by bone marrow

transplantation in mice conditioned with a nonmyeloablative regimen. J Immunol

153, 1087–1098 (1994).

419. Khan, A., Tomita, Y. & Sykes, M. Thymic dependence of loss of tolerance in

mixed allogeneic bone marrow chimeras after depletion of donor antigen.

Peripheral mechanisms do not contribute to maintenance of tolerance.

Transplantation 62, 380–387 (1996).

420. Morris, H. et al. Tracking donor-reactive T cells: Evidence for clonal deletion in

tolerant kidney transplant patients. Sci. Transl. Med. 7, 272ra10 (2015).

421. Andreola, G. et al. Mechanisms of donor-specific tolerance in recipients of

Page 171: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

158

haploidentical combined bone marrow/kidney transplantation. Am. J. Transplant

11, 1236–1247 (2011).

422. Scandling, J. D. et al. Tolerance and Withdrawal of Immunosuppressive Drugs in

Patients Given Kidney and Hematopoietic Cell Transplants. Am. J. Transplant. 12,

1133–1145 (2012).

423. Spitzer, T. R. et al. Long-Term Follow-Up of Recipients of Combined Human

Leukocyte Antigen-Matched Bone Marrow and Kidney Transplantation for Multiple

Myeloma With End-Stage Renal Disease. Transplantation 91, 672–676 (2011).

424. Hock, K., Mahr, B., Schwarz, C. & Wekerle, T. Deletional and regulatory

mechanisms coalesce to drive transplantation tolerance through mixed

chimerism. European Journal of Immunology 45, 2470–2479 (2015).

425. Leventhal J, Galvin J, Stare D, Gallon L, Miller J, Mathew J, Abecassis M, I. S.

Seven Year Follow-Up of a Phase 2 Clinical Trial to Induce Tolerance in Living

Donor Renal Transplant Recipients. Am J Transpl. 16,

http://www.atcmeetingabstracts.com/abstract/seven– (2016).

426. Chen, Y.-B., Kawai, T. & Spitzer, T. R. Combined Bone Marrow and Kidney

Transplantation for the Induction of Specific Tolerance. Adv. Hematol. 2016,

6471901 (2016).

427. Scandling, J. D., Busque, S., Shizuru, J. A., Engleman, E. G. & Strober, S.

Induced immune tolerance for kidney transplantation. The New England journal of

medicine 365, 1359–1360 (2011).

428. Brunstein, C. G. et al. Infusion of ex vivo expanded T regulatory cells in adults

transplanted with umbilical cord blood: safety profile and detection kinetics. Blood

117, 1061–70 (2011).

429. Di Ianni, M. et al. Tregs prevent GVHD and promote immune reconstitution in

HLA-haploidentical transplantation. Blood 117, 3921–8 (2011).

Page 172: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

159

430. Wood, K. J. & Sakaguchi, S. Regulatory T cells in transplantation tolerance. Nat.

Rev. Immunol. 3, 199–210 (2003).

431. Safinia, N., Scotta, C., Vaikunthanathan, T., Lechler, R. I. & Lombardi, G.

Regulatory T cells: Serious contenders in the promise for immunological tolerance

in transplantation. Frontiers in Immunology 6, (2015).

432. Sagoo, P. et al. Human regulatory T cells with alloantigen specificity are more

potent inhibitors of alloimmune skin graft damage than polyclonal regulatory T

cells. Sci. Transl. Med. 3, 83ra42 (2011).

433. Putnam, A. L. et al. Clinical grade manufacturing of human alloantigen-reactive

regulatory T cells for use in transplantation. Am. J. Transplant. 13, 3010–3020

(2013).

434. Floess, S. et al. Epigenetic control of the foxp3 locus in regulatory T cells. PLoS

Biol. 5, 0169–0178 (2007).

435. Goodyear, O. C. et al. Azacitidine augments expansion of regulatory T cells after

allogeneic stem cell transplantation in patients with acute myeloid leukemia

(AML). Blood 119, 3361–3369 (2012).

436. Bohne, F. et al. Intra-graft expression of genes involved in iron homeostasis

predicts the development of operational tolerance in human liver transplantation. J

Clin.Invest 122, 368–382 (2012).

437. Turka, L. A. & Lechler, R. I. Towards the identification of biomarkers of

transplantation tolerance. Nat. Rev. Immunol. 9, 521–526 (2009).

438. Dugast, E., Chesneau, M., Soulillou, J. P. & Brouard, S. Biomarkers and possible

mechanisms of operational tolerance in kidney transplant patients. Immunological

Reviews 258, 208–217 (2014).

439. Mart??nez-Llordella, M. et al. Using transcriptional profiling to develop a

diagnostic test of operational tolerance in liver transplant recipients. J. Clin.

Page 173: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

160

Invest. 118, 2845–2857 (2008).

440. Roord, S. T. A. et al. Autologous bone marrow transplantation in autoimmune

arthritis restores immune homeostasis through CD4+CD25+Foxp3+ regulatory T

cells. Blood 111, 5233–41 (2008).

441. Duran-Struuck, R. & Dysko, R. C. Principles of Bone Marrow Transplantation

(BMT): Providing Optimal Veterinary and Husbandry Care to Irradiated Mice in

BMT Studies. J. Am. Assoc. Lab. Anim. Sci. 48, 11–22 (2009).

442. Mazo, I. B. et al. Bone Marrow Is a Major Reservoir and Site of Recruitment for

Central Memory CD8+ T Cells. Immunity 22, 259–270 (2016).

443. Paramithiotis, E. & Cooper, M. D. Memory B lymphocytes migrate to bone marrow

in humans. Proc. Natl. Acad. Sci. U. S. A. 94, 208–12 (1997).

444. Chu, V. T. et al. Eosinophils are required for the maintenance of plasma cells in

the bone marrow. Nat. Immunol. 12, 151–159 (2011).

445. Bartczak, A. et al. Overexpression of Fibrinogen-Like Protein 2 Promotes

Tolerance in a Fully Mismatched Murine Model of Heart Transplantation. Am. J.

Transplant (2015). doi:10.1111/ajt.13696

446. Lee, I. et al. Recruitment of Foxp3+ T regulatory cells mediating allograft

tolerance depends on the CCR4 chemokine receptor. J. Exp. Med. 201, 1037–44

(2005).

447. Bestard, O. et al. Presence of FoxP3+ regulatory T Cells predicts outcome of

subclinical rejection of renal allografts. J. Am. Soc. Nephrol. 19, 2020–2026

(2008).

448. Boer, K. et al. Thymus-Derived Regulatory T Cells Infiltrate the Cardiac Allograft

Before Rejection. Transplantation 99, 1839–46 (2015).

449. Costello, J. P., Mohanakumar, T. & Nath, D. S. Mechanisms of chronic cardiac

Page 174: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

161

allograft rejection. Texas Hear. Inst. Journalart Inst. J. 40, 395–399 (2013).

450. Yusuf, S. W., Sami, S. & Daher, I. N. Radiation-induced heart disease: a clinical

update. Cardiol. Res. Pract. 2011, 317659 (2011).

451. Taunk, N. K., Haffty, B. G., Kostis, J. B. & Goyal, S. Radiation-induced heart

disease: pathologic abnormalities and putative mechanisms. Front. Oncol. 5, 39

(2015).

452. Aleman, B. M. P. et al. Long-term cause-specific mortality of patients treated for

Hodgkin’s disease. J. Clin. Oncol. 21, 3431–3439 (2003).

453. Cuzick, J. et al. Cause-specific mortality in long-term survivors of breast cancer

who participated in trials of radiotherapy. J. Clin. Oncol. 12, 447–453 (1994).

454. Noris, M. et al. Regulatory T cells and T cell depletion: role of immunosuppressive

drugs. J. Am. Soc. Nephrol. 18, 1007–1018 (2007).

455. Thomson, A. W., Turnquist, H. R. & Raimondi, G. Immunoregulatory functions of

mTOR inhibition. Nat. Rev. Immunol. 9, 324–37 (2009).

456. Turnquist, H. R. et al. Rapamycin-conditioned dendritic cells are poor stimulators

of allogeneic CD4+ T cells, but enrich for antigen-specific Foxp3+ T regulatory

cells and promote organ transplant tolerance. J. Immunol. 178, 7018–7031

(2007).

457. Waickman, A. T. & Powell, J. D. mTOR, metabolism, and the regulation of T-cell

differentiation and function. Immunol. Rev. 249, 43–58 (2012).

458. Ferrer, I. R. et al. Cutting Edge: Rapamycin Augments Pathogen-Specific but Not

Graft-Reactive CD8+ T Cell Responses. J. Immunol. 185, 2004–2008 (2010).

459. Chen, S. et al. Effect of inhibiting the signal of mammalian target of rapamycin on

memory T cells. Transplant. Proc. 46, 1642–1648 (2014).

460. Alexander, T. et al. Depletion of autoreactive immunologic memory followed by

Page 175: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

162

autologous hematopoietic stem cell transplantation in patients with refractory SLE

induces long-term remission through de novo generation of a juvenile and tolerant

immune system. Blood 113, 214–23 (2009).

461. Nutt, S. L., Hodgkin, P. D., Tarlinton, D. M. & Corcoran, L. M. The generation of

antibody-secreting plasma cells. Nat. Rev. Immunol. 15, 160–171 (2015).

462. Gorczynski, R., Chen, Z., Khatri, I. & Yu, K. Long-Term Tolerance and Skin

Allograft Survival in CD200tg Mice After Autologous Marrow Transplantation.

Transplantation (2014). doi:10.1097/TP.0000000000000456

463. Cheung, R. K. & Utz, P. J. Screening: CyTOF-the next generation of cell

detection. Nat. Rev. Rheumatol. 7, 502–503 (2011).

Page 176: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

163

Copyright Acknowledgements

This thesis contains one figure (Figure 1-2) that was reproduced under the Creative

Commons License from an open-access publication for which I am a co-author. A

second figure (Figure 1-3) from Doulatov et al.371 was reproduced with permission from

Elsevier and the license is provided on the next page.

ELSEVIER LICENSE

Page 177: Autologous Hematopoietic Stem Cell …...ii Autologous Hematopoietic Stem Cell Transplantation for Tolerance Induction in a Mouse Model of Solid Organ Transplantation Hassan Sadozai

164

TERMS AND CONDITIONS

Jul 27, 2016

This Agreement between Hassan A Sadozai ("You") and

Elsevier ("Elsevier") consists of your license details and the

terms and conditions provided by Elsevier and Copyright

Clearance Center.

License Number 3904400629746

License date Jul 08, 2016

Licensed Content Publisher Elsevier

Licensed Content Publication Cell Stem Cell

Licensed Content Title Hematopoiesis: A Human Perspective

Licensed Content Author Sergei Doulatov,Faiyaz Notta,Elisa

Laurenti,John E. Dick

Licensed Content Date 3 February 2012

Licensed Content Volume Number 10

Licensed Content Issue Number 2

Licensed Content Pages 17

Start Page 120

End Page 136

Type of Use reuse in a thesis/dissertation

Portion figures/tables/illustrations

Number of figures/tables/illustrations 1

Format print

Are you the author of this Elsevier article? No

Will you be translating? No

Order reference number

Original figure numbers Figure 2

Title of your thesis/dissertation Autologous Hematopoietic Stem Cell

Transplantation for Tolerance Induction in

Solid Organ Transplantation

Expected completion date Sep 2016

Elsevier VAT number GB 494 6272 12

Requestor Location Hassan A Sadozai

Unit 311

218 Queens Quay West

Toronto, ON M5J2Y6

Canada

Attn: