immunotherapy of leukemic stem cells using natural killer ... · immunotherapy of leukemic stem...

273
Immunotherapy of Leukemic Stem Cells using Natural Killer Cell lines by Brent A. Williams A thesis submitted in conformity with the requirements for the degree of Doctor of Medical Sciences Institute of Medical Sciences University of Toronto © Copyright by Brent A. Williams 2015

Upload: others

Post on 27-May-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

Immunotherapy of Leukemic Stem Cells using

Natural Killer Cell lines

by

Brent A. Williams

A thesis submitted in conformity with the requirements

for the degree of Doctor of Medical Sciences

Institute of Medical Sciences

University of Toronto

© Copyright by Brent A. Williams 2015

Page 2: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

ii

Immunotherapy of Leukemic Stem Cell using

Natural Killer Cell lines

Brent A. Williams

Doctor of Philosphy

Institute of Medical Sciences

University of Toronto

2015

Abstract

The field of cancer immunobiology has focused on the interaction of bulk tumour cells with the

immune system. The discovery of stem cells in a number of cancers including leukemia has

raised new questions about the how the immune system interacts with these cancer stem cells and

has implications for immune based therapies. Here the natural killer cell lines NK-92 and

KHYG-1 are tested against primary acute myeloid leukemia and cell lines utilizing techniques to

address the impact on bulk and leukemic stem cells. NK-92 and KHYG-1 are both shown to

preferentially target leukemic stem cells in primary AML and cell lines using a novel clonogenic

cytotoxicity assay. Further, both cell lines can prolong survival in AML xenograft models using

primary AML and cell lines. NK-92 gene modified to express CD16 can mediate ADCC against

leukemic stem cells in vitro and in vivo. KHYG-1 can have significant enhancement against

leukemia when pretreated with anti-NKp30 and NKp44 monoclonal antibodies via reverse

ADCC, overcoming resistance of leukemic cell lines and primary AML samples. These data

support the notion that cell therapy agents can target leukemic stem cells, be enhanced by

antibodies, and lead to enhanced survival, providing a rationale for clinical trials testing this

approach in AML patients with minimal residual disease lacking a suitable allogeneic transplant

donor.

Page 3: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

iii

Dedication

In loving memory of my father, Bernard K. Williams, my first mentor. While not a scientist by

trade, he taught me the structure of the atom, the theory of evolution and the power of rationale

thought.

Page 4: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

iv

Acknowledgments

I would like to acknowledge my supervisor Dr. Armand Keating for taking me into the lab group

to pursue the question of how the immune system recognizes cancer stem cells and guiding the

process of discovery into uncharted regions. Further, I appreciate his decades of translational

research experience which I have been able to tap in the formulation of laboratory experiments

and models with the primary objective of designing novel therapeutic strategies for the treatment

of leukemia. Finally, I wish to acknowledge his career mentorship in pursuing my goal of

becoming an independent clinician scientist.

I would like to thank my committee members Dr. Mark Minden and Dr. Pam Ohashi for their

scientific critique of experimental design and results. They have been patient with delays and

experimental dead-ends and provided insight in surmounting difficult scientific challenges.

Their expectation of scientific rigor has elevated the quality of my work and enhanced my

approach to experimental design and translational research. Dr. Minden provided critical input

in the selection of AML cell lines and primary AML samples and their use in developing

xenogeneic leukemia models.

I must give special thanks to Dr. Xing-Hua Wang our lab manager who is a pillar of the lab

group and has provided continual support of my work. I learned most of the lab techniques

utilized in the thesis from him. Further, he generously provided his expertise in animal models

and experimentation to make some of the key discoveries presented here.

Next I would like to acknowledge the members of the Keating lab, past and present who have

provided insights and colleagial support over my PhD studies. Thanks to Sonia Montanari,

Roula Antoon, Iran Rashedi for sharing the ‘pain’ of the PhD experience with me and providing

critical insights into my work during lab meetings and other casual conversations. I would like

to acknowledge the work of Dr. Yoko Kosaka, a former post-doc, who started the NSG animal

colony for our lab and started us using a cutting edge immunodeficient animal model to develop

xenogeneic cancer models.

I have also had pleasure of having several highly motivated summer students work with me who

assisted in conducting pilot experiments. Sonam Maghera assisted in clonogenic cytotoxicity

assays that yielded puzzling results at the time, but led to much of the work done in Chapter 4 on

Page 5: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

v

reverse ADCC. Richard Cheng also assisted in evolving the clonogenic assay to explore NK cell

cytotoxicity using antibodies.

I also must acknowledge a fruitful collaboration with Dr. Jeff Leyton formerly Dr. Raymond

Reilly’s postdoctoral fellow and currently Assistant Professor at Sherbrooke University. We

both had set out to develop a therapeutic strategy to target leukemic stem cells; he using

radioconjugated anti-CD123 antibodies and myself with ADCC capable NK cell lines and anti-

CD123 antibodies. The overlap in approaches was clear and we began to discuss the optimal in

vivo readouts for novel therapeutics targeting leukemic stem cells often debating the superiority

of primary and secondary engraftment versus survival outcomes. In the end we settled on using

all methods which is reflected in this thesis and in our collaborative work using radioconjugated

antibody therapy to target leukemic stem cells which has recently been published. I must thank

Jeff for criticially reviewing early versions of chapter 3 and paying attention to the small details.

While material from this collaborative work does not appear in the thesis it allowed for me to

engage in a productive collaboration while dealing with delays in key reagents.

I would like to thank Dr. Neal DenHollander, director of the UHN HLA bank, for stimulating

immunological discussions and facilitating the link of the Princess Margaret Leukemia Bank

with the HLA bank which allowed us to address key questions of NK cell line recognition. I

would like to acknowledge Dr. Hans Hitzler, my clinical mentor in pediatric

hematology/oncology for allowing me flexibility to attend leukemia clinics in the context of a

busy research program.

I would like to thank my former research supervisor and mentor, Dr. David Hoskin, who taught

me basic immunology as an undergraduate and medical student and instilled a rigorous approach

to the scientific method and experimental design. His continual support of all my recent research

applications is greatly appreciated.

I would like to acknowledge the financial support of the National Cancer Institute Terry Fox

Foundation and Canadian Institute of Health research which provided my primary salary support

over the PhD research period. Also, additional support came from the Cell Therapy Program at

Princess Margaret Hospital, the Hospital for Sick Children, University of Toronto, Ontario

Cancer Institute, Canadian Hematology Society who have provided financial support for both

salary and operating costs.

Page 6: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

vi

I would like to thank my mother and father, Bonnie and Bernie, for their enduring support of me

and my career aspirations. I would like to acknowledge my children Hayden, Ethan and Kate

who keep me balanced and engaged in the important things in life. Special thanks to Hayden and

Ethan for building a ‘lab’ under the stairs in the basement as a backup place to do experimental

work if my scientific career doesn’t work out. Finally I would like to acknowledge the love and

support of my wife, Jeannine, to accept the long years of training I have chosen. It has been

challenging and without her support this work would not have been possible.

Page 7: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

vii

Quotation

Dans les champs de l'observation, le hasard ne favorise que les esprits prepares.

In the fields of observation, chance favors only the prepared mind.

Louis Pasteur, 1822-1895

Page 8: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

viii

Table of Contents

Dedication ..................................................................................................................................... iii

Acknowledgments ........................................................................................................................ iv

Quotation ..................................................................................................................................... vii

Table of Contents ....................................................................................................................... viii

List of Tables .............................................................................................................................. xiii

List of Figures ............................................................................................................................. xiv

1 Chapter 1: Literature review ..................................................................................................... 1

1.1 Overview ............................................................................................................................. 1

1.2 Acute myeloid leukemia (AML) ......................................................................................... 1

1.2.1 Epidemiology .......................................................................................................... 1

1.2.2 Pathology ................................................................................................................ 2

1.2.3 Immunophenotype of AML .................................................................................... 5

1.2.4 Minimal residual disease (MRD) ............................................................................ 6

1.2.5 Chemotherapy for AML ......................................................................................... 8

1.2.6 Leukemia cell lines ................................................................................................. 9

1.2.7 Animal models of leukemia .................................................................................. 10

1.2.8 Cancer stem cell hypothesis .................................................................................. 14

1.2.9 Clinical relevance of LSCs ................................................................................... 17

1.3 Cytotoxicity assays ........................................................................................................... 20

1.3.1 Bulk cytotoxicity assays ....................................................................................... 20

1.3.2 Flow cytometric cytotoxicity assays ..................................................................... 21

1.3.3 Clonogenic cytotoxicity assays ............................................................................. 21

1.4 Cancer immunobiology ..................................................................................................... 24

1.4.1 Lymphocytes ......................................................................................................... 24

Page 9: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

ix

1.4.2 NK cells ................................................................................................................ 24

1.4.3 NK cell lines ......................................................................................................... 41

1.5 Antibody therapy for cancer ............................................................................................. 43

1.6 Cell therapy for cancer ...................................................................................................... 46

1.6.1 Allogeneic hematopoietic stem cell transplantation ............................................. 46

1.6.2 Adoptive immunotherapy ..................................................................................... 47

2 Chapter 2: Hypotheses and experimental approach ................................................................. 57

2.1 Thesis aims ........................................................................................................................ 57

2.2 Hypotheses ........................................................................................................................ 57

2.2.1 Leukemic stem cells are present in cell line KG1 and are sensitive to NK-92

mediated cytotoxicity ............................................................................................ 57

2.2.2 Primary AML leukemic stem cells have greater sensitivity to NK92 than bulk

leukemia and can be targeted by CD16+NK-92 and anti-CD123 mAb

mediated ADCC in vivo ........................................................................................ 58

2.2.3 KHYG-1 has less cytotoxicity than NK-92 against leukemic targets which can

be modulated by antibody pretreatment of targets and effectors .......................... 58

3 Chapter 3: Clonogenic assays measure leukemia stem cell killing not detectable by

chromium release and flow cytometric cytotoxicity assays ..................................................... 60

3.1 Abstract ............................................................................................................................. 61

3.2 Introduction ....................................................................................................................... 62

3.3 Materials and methods ...................................................................................................... 64

3.3.1 Cell lines ............................................................................................................... 64

3.3.2 Antibodies and reagents ........................................................................................ 64

3.3.3 Chromium release assay ....................................................................................... 64

3.3.4 Flow cytometry and cell sorting ........................................................................... 65

3.3.5 Flow cytometric cytotoxicity assay ...................................................................... 65

3.3.6 Methylcellulose and liquid reculturing cytotoxicity assays .................................. 66

3.3.7 Animals ................................................................................................................. 67

Page 10: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

x

3.4 Results ............................................................................................................................... 68

3.4.1 Clonogenic capacity of KG1 in vitro and in vivo ................................................. 68

3.4.2 Immunophenotyping and fractionation studies of KG1 ........................................ 71

3.4.3 Chromium release assay (CRA) and flow cytometric cytotoxicity assay of

NK-92 and chemotherapy drugs versus KG1 ....................................................... 74

3.4.4 Clonogenic and proliferation assays of NK-92 and chemotherapy drugs versus

KG1 ....................................................................................................................... 77

3.5 Discussion ......................................................................................................................... 80

4 Chapter 4: Irradiated CD16+NK-92 prolongs survival in an AML xenograft model in

combination with anti-CD123 monoclonal antibody therapy by targeting leukemic stem

cells through antibody dependent cell mediated cytotoxicity (ADCC) ................................... 84

4.1 Abstract ............................................................................................................................. 85

4.2 Introduction ....................................................................................................................... 86

4.3 Methods ............................................................................................................................. 88

4.3.1 Cell lines and primary samples ............................................................................. 88

4.3.2 Chromium release assay ....................................................................................... 88

4.3.3 ADCC chromium release assay ............................................................................ 89

4.3.4 Flow cytometry ..................................................................................................... 89

4.3.5 Cell sorting ............................................................................................................ 89

4.3.6 Methylcellulose cytotoxicity assay ....................................................................... 90

4.3.7 Animals ................................................................................................................. 90

4.3.8 Statistics ................................................................................................................ 90

4.4 Results ............................................................................................................................... 91

4.4.1 NK-92 is cytotoxic against primary AML by granule exocytosis ........................ 91

4.4.2 Chromium release assay versus methylcellulose cytotoxicity assay .................... 93

4.4.3 NK-92 preferentially kills leukemic stem cells relative to bulk leukemia ............ 94

4.4.4 Primary AML xenograft model ............................................................................ 97

Page 11: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

xi

4.4.5 In vitro treatment of primary AML cells by irradiated NK-92 reduce

engraftment potential ............................................................................................ 99

4.4.6 Irradiated NK-92 reduce leukemic stem cell fraction in secondary

transplantation assay ........................................................................................... 101

4.4.7 NK-92 prolongs survival in a primary AML xenograft model ........................... 103

4.4.8 iCD16+NK-92 can mediate ADCC against bulk and stem cell antigens in vitro 106

4.4.9 CD16+NK-92 improves survival in an AML xenograft model with

enhancement by anti-CD123 mAb therapy ......................................................... 109

4.5 Discussion ....................................................................................................................... 113

5 Chapter 5: NK cell line killing of leukemia cells is enhanced by reverse antibody

dependent cell mediated cytotoxicity (R-ADCC) via NKp30 and NKp44 and target cell

Fcγ receptor II (CD32) ........................................................................................................... 118

5.1 Abstract ........................................................................................................................... 119

5.2 Introduction ..................................................................................................................... 120

5.3 Methods ........................................................................................................................... 122

5.3.1 Cell lines and primary samples ........................................................................... 122

5.3.2 Chromium release assay ..................................................................................... 122

5.3.3 Antibody pretreatment of NK cell effectors ....................................................... 122

5.3.4 Flow cytometry ................................................................................................... 123

5.3.5 High throughput sampling flow cytometry ......................................................... 123

5.3.6 Animals ............................................................................................................... 124

5.3.7 Statistics .............................................................................................................. 124

5.4 Results ............................................................................................................................. 125

5.4.1 NK-92 and KHYG-1 cytotoxicity against leukemia cell lines ........................... 125

5.4.2 High throughput screening flow cytometry of NK-92 and KHYG-1 surface

receptors .............................................................................................................. 127

5.4.3 Anti-Class I HLA blockade of AML targets ....................................................... 129

5.4.4 Effect of pretreating NK-92 and KHYG-1 with activating receptor specific

antibodies ............................................................................................................ 131

Page 12: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

xii

5.4.5 Relationship of Fcγ receptor expression and enhancement of cytotoxicity ........ 142

5.4.6 Effect of anti-NKp30 pretreatment on NK cell line cytotoxicity against

clonogenic OCI/AML5 ....................................................................................... 147

5.4.7 In vitro effect of anti-NKp30 pretreated iKHYG-1 against OCI/AML5

capacity for leukemic progression in an NSG xenograft model ......................... 149

5.4.8 Effect of anti-NKp30 pretreatment of iKHYG-1 on therapeutic efficacy for

OCI/AML5 or primary AML xenografted mice ................................................. 152

5.5 Discussion ....................................................................................................................... 155

6 Chapter 6: General Discussion ............................................................................................... 163

6.1 Overview ......................................................................................................................... 163

6.2 Methodologic approaches to measuring the impact of immune effectors on leukemic

stem cell and bulk leukemia ............................................................................................ 164

6.3 Natural cytotoxicity of NK cell lines with and without ADCC enhancement against

leukemic stem cells ......................................................................................................... 166

6.4 Natural cytotoxicity of NK cell lines with and without reverse-ADCC enhancement

against leukemic stem cells ............................................................................................. 169

6.5 Translational relevance ................................................................................................... 172

7 Chapter 7: Conclusions and future directions ........................................................................ 174

8 Chapter 8: References ............................................................................................................ 176

Copyright Acknowledgements (pending) ................................................................................... 249

Page 13: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

xiii

List of Tables

Table 1.1: WHO histopathological classification of AML ............................................................. 4

Table 1.2: NK cell inhibiting receptors ......................................................................................... 32

Table 1.3: NK cell activating receptors ........................................................................................ 35

Table 1.4: FDA approved therapeutic monoclonal antibodies for cancer* .................................. 45

Table 1.5: Comparison of autologous cell therapy and cell line therapy ...................................... 55

Table 3.1: Frequency of KG1 stem cell frequency using liquid culture repopulation .................. 68

Table 3.2: Frequency of KG1 stem cell frequency using two fold serial dilutions in 96 well

confluence assay (5000 to 0.3 per well) ........................................................................................ 68

Table 3.3: Frequency of KG1 stem cell frequency using cell sorting and 96 well confluence assay

(1000 to 1 per well) ....................................................................................................................... 69

Table 5.1: Differential expression of cell surface activating, inhibiting and apoptosis inducing

molecules on NK-92 and KHYG-1 ............................................................................................. 128

Table 5.2: HLA type of primary AML panel and sensitivity to NK-92 and KHYG-1 +/- HLA

blockade ...................................................................................................................................... 130

Table 5.3: Comparison of antibody pretreatment effects on KHYG-1 cytotoxicity and synergy

assessment at 0.1 µg/ml .............................................................................................................. 137

Table 5.4: Comparison of antibody pretreatment effects on KHYG-1 cytotoxicity and synergy

assessment at 0.01 µg/ml ............................................................................................................ 138

Page 14: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

xiv

List of Figures

Figure 1.1: NK cell degranulation by stimulation with activating ligands only ........................... 26

Figure 1.2: NK cell inhibition of degranulation by stimulation with activating and inhibitory

ligands ........................................................................................................................................... 26

Figure 1.3: NK cell degranulation by stimulation with activating and inhibitory ligands ........... 26

Figure 1.4: NK cell antibody dependent cell mediated cytotoxicity ............................................ 39

Figure 1.5: Schematic of adoptive immunotherapy using LAK cells ........................................... 48

Figure 1.6: Schematic of adoptive immunotherapy using TILs ................................................... 49

Figure 1.7: Design of CD19 CAR utilized in a clinical trial for CLL .......................................... 52

Figure 1.8: Schematic of CD19+ CAR T-cell recognition of CD19+ CLL cells ......................... 53

Figure 1.9: Schematic of adoptive immunotherapy using cell lines ............................................. 54

Figure 3.1: NOD/SCID leukemia initiating frequency of KG1 .................................................... 70

Figure 3.2: Immunophenotype of KG1 ......................................................................................... 71

Figure 3.3: Reconstitution of CD38 distribution following cell sorting KG1 .............................. 73

Figure 3.4: Chromium release assay of NK-92 against KG1 and K562 ....................................... 74

Figure 3.5: Chromium release assay versus flow cytometric cytotoxicity assay ......................... 75

Figure 3.6: Chromium release assay versus flow cytometric drug assay ..................................... 76

Figure 3.7: Liquid reculturing cytotoxicity assay ......................................................................... 77

Figure 3.8: Methylcellulose cytotoxicity assay ............................................................................ 79

Figure 4.1: Chromium release assay of NK-92 a against a primary AML sample at a range of

Effector:Target ratios with and without calcium chelation ........................................................... 91

Page 15: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

xv

Figure 4.2: Chromium release assay of NK-92 against a panel of primary AML patient samples

at a range of Effector:Target ratios ............................................................................................... 92

Figure 4.3: Clonogenic cytotoxicity assay of NK-92 against OCI/AML2 and OCI/AML3 ........ 93

Figure 4.4: NK-92 cytotoxicity against sorted leukemic stem cells (CD34+CD38-) and bulk

leukemia (CD34+CD38+) ............................................................................................................. 94

Figure 4.5: Schematic of methylcellulose cytotoxicity assay ....................................................... 95

Figure 4.6: NK-92 against primary AML blasts using the methylcellulose cytotoxicity assay

compared to the chromium release assay ...................................................................................... 96

Figure 4.7: Primary AML immunophenotype .............................................................................. 97

Figure 4.8: Survival of NSG mice with primary AML versus 1st passage AML derived from BM

....................................................................................................................................................... 98

Figure 4.9: Schematic of in vitro cytotoxicity assay with in vivo engraftment readout ............... 99

Figure 4.10: In vitro cytotoxicity assay with in vivo engraftment readout ................................. 100

Figure 4.11: In vivo cytotoxic impact of iNK-92 on secondary BM engraftment of AML cells and

LSCs ............................................................................................................................................ 102

Figure 4.12: Schematic of NK-92 therapy for primary AML xenografted NSG mice ............... 103

Figure 4.13: NK-92 therapy of primary AML xenografted NSG mice ...................................... 104

Figure 4.14: iNK-92 therapy of primary AML xenografted NSG mice ..................................... 105

Figure 4.15: Immunophenotyping of CD16+NK-92, NK-92 and OCI/AML 2, 3 and 5 ............ 106

Figure 4.16: CD16+NK-92 in vitro ADCC assay against primary AML ................................... 107

Figure 4.17: CD16+NK-92 in vitro ADCC assay against OCI/AML5 ...................................... 108

Figure 4.18: Schematic of iCD16+NK-92 +/- a single anti-CD123 mAb dose for primary AML

xenografted NSG mice ................................................................................................................ 109

Page 16: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

xvi

Figure 4.19: iCD16+NK-92 +/- a single anti-CD123 mAb dose for primary AML xenografted

NSG mice .................................................................................................................................... 110

Figure 4.20: Schematic of CD16+NK-92 +/- five doses of anti-CD123 mAb therapy for primary

AML xenografted NSG mice ...................................................................................................... 111

Figure 4.21: iCD16+NK-92 with and without 7G3 or isotype control treatment for primary AML

xenograft model .......................................................................................................................... 112

Figure 5.1: NK-92 and KHYG-1 cytotoxicity against a panel of leukemia cell lines ................ 125

Figure 5.2: NK-92 and KHYG-1 cytotoxicity against K562 with and without calcium chelation

..................................................................................................................................................... 126

Figure 5.3: KHYG-1 cytotoxicity against 4 primary AML samples +/- class I HLA blockade . 130

Figure 5.4: Effect of antibody pre-treatment of activating receptors on NK-92 and KHYG-1

cytotoxicity against leukemia cell lines ...................................................................................... 132

Figure 5.5: Effect of antibody pre-treatment of activating receptors on NK-92 and KHYG-1

cytotoxicity against primary AML samples ................................................................................ 133

Figure 5.6: Effect of antibody pre-treatment with isotype control on NK-92 and KHYG-1

cytotoxicity against leukemia cell lines ...................................................................................... 135

Figure 5.7: Effect of antibody pre-treatment with anti-NKp30 and anti-NKp44 on NK-92 and

KHYG-1 cytotoxicity against leukemia cell lines ...................................................................... 136

Figure 5.8: Effect of antibody pre-treatment with anti-NKp30 and anti-NKp44 on NK-92 and

KHYG-1 cytotoxicity against leukemia cell lines and primary AML ........................................ 139

Figure 5.9: Impact of antibody pre-treatment with anti-NKp30 and anti-NKp44 on NK-92 and

KHYG-1 cytotoxicity against esophageal cancer cell lines ........................................................ 141

Figure 5.10: Fc gamma receptor expression on leukemia cell lines (K562, KG1, KG1a,

OCI/AML3, OCI/AML5) ........................................................................................................... 143

Page 17: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

xvii

Figure 5.11: Fc gamma receptor expression on esophageal cancer cell lines (OE-33, FLO-1,

KYAE-1, SKGT-4) ..................................................................................................................... 144

Figure 5.12: Regression analysis of CD32 expression and delta cytotoxicity of NKp30 and

NKp44 pretreated NK-92 and KHYG-1 ..................................................................................... 146

Figure 5.13: Methylcellulose cytotoxicity assay of NK-92 and KHYG-1 +/- pretreatment with

antibodies against OCI/AML5 .................................................................................................... 148

Figure 5.14: OCI/AML5 induced malignant ascites .................................................................. 150

Figure 5.15: In vitro incubation of OCI/AML5 with iKHYG-1 +/- anti-NKp30 and in vivo

proliferation in NSG mice ........................................................................................................... 151

Figure 5.16: Bone marrow engraftment of OCI/AML5 injected iv into NSG mice ................... 152

Figure 5.17: Treatment of OCI/AML5 leukemia in NSG mice with iKHYG-1 +/- NKp30

pretreatment ................................................................................................................................ 153

Figure 5.18: Treatment of primary AML xenografted NSG mice with iKHYG-1 +/- NKp30

pretreatment ................................................................................................................................ 154

Figure 6.1: Antibody-dependent cell-mediated cytotoxicity (ADCC) ....................................... 168

Figure 6.2: Reverse antibody-dependent cell-mediated cytotoxicity (R-ADCC) ....................... 170

Page 18: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

xviii

List of Appendices

Appendix I: NK-92 HTS flow cytometry percent positivity and MFI of antigens ..................... 212

Appendix II: KHYG-1 HTS flow cytometry percent positivity and MFI of antigens ................ 221

Appendix III: OCI/AML3 HTS flow cytometry percent positivity and MFI of antigens .......... 231

Appendix IV: OCI/AML5 HTS flow cytometry percent positivity and MFI of antigens .......... 240

Page 19: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

xix

List of Abbreviations

ADCC Antibody dependent cell mediated cytotoxicity

ALDH Aldehyde dehydrogenase

ALL Acute lymphoblastic leukemia

AML Acute myeloid leukemia

APC Allophycocyanin

ATTC American Type Culture Collection

BM Bone marrow

CAR Chimeric antigen receptor

CD Cluster of differentiation

CML Chronic myeloid leukemia

CRACC CD2-like leceptor activating cytotoxic cells

CSC Cancer stem cell

CTL Cytotoxic T lymphocyte

DAP12 DNAX activation protein of 12 kDa

DNAM-1 DNAX accessory molecule 1

EC50% Effective concentration to achieve 50% of maximal effect

EGIL European group for the immunological classification of leukaemias

EFS Event free survival

ERK2 Extracellular regulated kinase 2

FADD Fas associated death domain

FITC Fluorescein isothyocyanate

FLT3 fms-related tyrosine kinase 3

GFP Green fluorescent protein

GMCSF Granulocyte monocyte colony stimulating factor

GVL Graft versus leukemia effect

HSCT Hematopoietic stem cell transplantation

HLA Human leukocyte antigen

HSC Hematopoietic stem cell

HSCT Hematopoeitic stem cell transplant

HSP Heat shock protein

ICAM Intercellular adhesion molecule

IFN-γ Interferon gamma

Ig Immunoglobulin

IS Immunologic synapse

KIR Killer immunoglobulin like receptor

ITAM Immunoreceptor tyrosine-based activating motifs

ITD Internal tandem repeats

ITIM Immunoreceptor tyrosine-based inhibitory motifs

ITSM Immunoreceptor tyrosine-based switch motifs

LAK Lymphokine activated killer

LFA Lymphocyte function antigen

LIR Leukocyte immunoglobulin-like inhibitory receptors

LSC Leukemic stem cell

MIL Marrow infiltrating lymphocyte

mAb Monoclonal antibody

mRNA Messenger RNA

Page 20: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

xx

MCA Methylcellulose cytotoxicity assay

MDS Myelodysplastic syndrome

MPS Myeloproliferative syndrome

MRD Minimal residual disease

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

NCR Natural cytotoxicity receptor

NK Natural killer

NMP1 Nuclear Matrix Protein 1

DNMT3A DNA (cytosine-5)-methyltransferase 3A

NKG2D Natural killer group 2 D

NRG NOD/RAG gamma null

NOD Non-obese diabetic

NSB NOD/SCID/β2microglobulin null

NSG NOD-SCID gamma null

OCI Ontario cancer institute

OS Overall survival

PBL Peripheral blood lymphocyte

PE Phycoerythrin

PCR Polymerase chain reaction

Prkdc Protein kinase, DNA-activated, catalytic polypeptide

PML-RARα Promyelocytic myeloid leukemia-retinoic acid α receptor

PNET Primitive neuroectodermal tumour

RANK1 Receptor Activator of Nuclear Factor κ B

SCID Severe combined immunodeficiency

SP Side population

ScFv Single-chain variable fragment

Th1 T helper 1

TIL Tumour infiltrating lymphocyte

TNF Tumour necrosis factor

Treg T regulatory cell

TRAIL TNF-related apoptosis-inducing ligand

TWEAK Tumor necrosis factor receptor superfamily member 12A

WHO World Health Organization

Page 21: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

1

1 Chapter 1: Literature review

1.1 Overview

The field of cancer immunology has focused primarily on the interaction of cells of the immune

system with bulk tumour cells. The discovery of cancer stem cells has brought into question

whether prior studies of immune effector cell interaction with cancer cells can be extrapolated to

these rare subset of cells. This poses many new biological questions regarding how the immune

system interacts, recognizes and responds to cancer stem cells. Further, there are significant

therapeutic implications to the cancer stem cell hypothesis if only a rare subpopulation of cells

has the capability to initiate, progress and recapitulate cancer after therapy. In the case of acute

myeloid leukemia (AML), the stem cell compartment comprises a rare subpopulation initially

identified as CD34+CD38- AML cells. (Lapidot, Sirard et al. 1994; Bonnet and Dick 1997)

While chemotherapy can cure a minority of AML patients, many relapse due to the presence of

minimal residual disease. Hematopoietic stem cell transplantation (HSCT) transplantation is one

curative approach for high risk or relapse AML, demonstrating the potential for cellular based

therapies to treat AML. There is a paucity of studies focused on the interaction of the cellular

immune system with cancer stem cells, leaving many avenues for novel investigation and the

potential to the develop future therapies designed to treat leukemic stem cells.

1.2 Acute myeloid leukemia (AML)

1.2.1 Epidemiology

The age-adjusted incidence rate of AML in the U.S. (1975–2011) was 3.51/100,000/year with

males having slightly higher incidence (4.36) than females (2.92).(Howlader N) These rates are

comparable with other North American and western European countries.(Jemal, Thomas et al.

2002; Deschler and Lubbert 2006) This leads to approximately 13,000 new cases of AML

annually in the United States(Ries LAG 2008) and 1,300 in Canada. The first peak in incidence

occurs in the first year of life and decreases up to age 4, and remains at this level through

childhood and early adulthood,(Gurney, Davis et al. 1996) increasing again progressively in late

adulthood(Howlader N). There is a ten-fold increase in the age-specific incidence of AML in the

population older than 65, leading to a high prevalence of cases (~40%) in this age group, with

the incidence increasing to 24/100,000/year in the over 80 age group.(Deschler and Lubbert

Page 22: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

2

2006) AML accounts for approximately 90% of all acute leukemias in adults and 15% of cases in

children.(Lowenberg, Downing et al. 1999) The incidence of leukemia is highest in the United

States, Australia and Western Europe.(Jemal, Thomas et al. 2002) Projections of an aging world

population mapped onto the age-specific incidence rates for AML lead to the conclusion of a

increase in incidence of AML that will strain current resources available in most countries. Short

term, these demographic shifts in Western countries have led to predictions for a need to increase

the number of practicing oncologists treating all cancers by 40% from 2014 to 2020.(Yang,

Williams et al. 2014) This will be a greater issue for AML because of the sharp increase in

incidence above 65 of both de novo AML cases, as well as myelodysplastic syndrome, which

predisposes to AML. This highlights the need for targeted, minimally toxic and affordable

therapeutic options for AML patients.

1.2.2 Pathology

AML can be categorized by the both French American British (FAB) system(Bennett, Catovsky

et al. 1976) and the World Health Organiziation (WHO) systems (Vardiman, Thiele et al. 2009).

FAB categories include M0 (minimially differentiated), M1 (without maturation), M2

(granulocytic), M3 (acute promyelocytic), M4 (myelomonocytic), M4Eo (myelomonocytic with

eosinophilic differentiation) M5 (monocytic), M6 (erthyroleukemia), M7 (megakaryoblastic).

Acute promyelocytic leukemia is defined by a characteristic translocation (PML-RARα) and is

treated with a unique treatment regimen distinguishing it from other subclass of AML.

World Health Organization (WHO) classification has expanded to identify numerous subtypes of

AML (Table 1), mostly falling into the following categories: a) AML with recurrent genetic

abnormalities, b) AML with myelodysplasia-related changes, c) therapy-related myeloid

neoplasms and d) AML not otherwise categorized. AML with recurrent genetic abnormalities

may lead to better [e.g. t(8:21), inv16, t(15;17)] or worse survival outcomes (e.g. MLL

rearrangement, complex cytogenetics). Topoisomeriase II therapy (e.g. etoposide) can lead to

second malignancies that are particularly resistant to therapy. Patients with therapy related

AML or progressed from myelodysplatic syndrome (MDS) or myeloproliferative syndrome

(MPS), fare poorly. M3, or acute promyelocytic and M6, erythroleukemia and M7,

megakaryoblastic leukemia are considered distinct disease entities each requiring specific

Page 23: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

3

therapy. The remaining categories are treated using similar protocols consisting of a backbone

of daunorubicin and cytarabine.

Additional molecular abnormalities can contribute to poor prognosis. A study of 854 patients in

United Kingdom Medical Research Council (MRC) AML trials demonstrated the presence of

FLT3 internal tandem duplicates (ITD) correlated with lower complete remission rate, higher

induction death rate, increased relapse risk, worse event-free survival (EFS) and overall survival

(OS) (p <0.001 for all).(Kottaridis, Gale et al. 2001) In another study, presence of FLT3 ITD

was the most predictive negative prognostic factor. (Meshinchi, Woods et al. 2001) FLT3 ITD

was evaluated in 91 pediatric patients with AML from CCG 2891 and those with and without

this abnormality had an eight year EFS of 7% and 44%.(Meshinchi, Woods et al. 2001). Another

important genetic abnormality with prognostic significance is mutation of neucleophosmin

(NPM1), which correlates with a chemosensitive disease and has a favourable prognosis in the

absence of other negative prognostic factors as reviewed by Falini et al.(Falini, Martelli et al.

2011). A meta-analysis conducted on ten studies with a total of 6,219 patients revealed that

CEBPA mutation conferred a favourable prognosis.(Li, Deng et al. 2014) Thus, the revised

WHO criterion has included both mutations of NPM1 and CEPBA as provisional diagnostic

categories for AML.

Page 24: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

4

Table 1.1: WHO histopathological classification of AML

General diagnostic category Examples

Acute myeloid leukemia

with recurrent genetic

abnormalities

AML with t(8;21)(q22;q22); RUNX1-RUNX1T1

AML with inv(16)(p13.1q22) or t(16;16)(p13.1;q22); CBFB-

MYH11

APL with t(15;17)(q22;q12); PML-RARA

AML with t(9;11)(p22;q23); MLLT3-MLL

AML with t(6;9)(p23;q34); DEK-NUP214

AML with inv(3)(q21q26.2) or t(3;3)(q21;q26.2); RPN1-EVI1

AML (megakaryoblastic) with t(1;22)(p13;q13); RBM15-MKL1

Provisional entity: AML with mutated NPM1

Provisional entity: AML with mutated CEBPA

Acute myeloid leukemia

with myelodysplasia-

related changes

AML with a prior history of MDS or have specific MDS related

cytogenetic abnormalities (e.g. -5 or -7) or exhibit dysplasia in

50% or more of the cells in 2 or more myeloid lineages

Therapy-related myeloid

neoplasms

AML secondary to etoposide therapy

Acute myeloid leukemia,

not otherwise specified

AML with minimal differentiation

AML without maturation

AML with maturation

Acute myelomonocytic leukemia

Acute monoblastic/monocytic leukemia

Acute erythroid leukemia

Acute megakaryoblastic leukemia

Acute basophilic leukemia

Acute panmyelosis with myelofibrosis

*Adapted from Vardiman 2009 et al. pg 939, table 2.(Vardiman, Thiele et al. 2009)

Page 25: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

5

1.2.3 Immunophenotype of AML

AML cells typically express antigens found on normal myeloid progenitor and differentiated

cells, such as macrophages and monocytes, with aberrant expression of other lineage markers.

AML expresses the pan-leukocyte marker CD45 and other myeloid markers such as CD11b,

CD13 and CD33. A review of 106 adult AML cases was conducted to assess immunophenotypic

variation based on FAB classification using a 22 antibody panel.(Khalidi, Medeiros et al. 1998)

The most commonly expressed antigens were CD45 (97.2%), CD33 (95.3%), and CD13

(94.3%). Lymphoid-associated antigens were expressed in approximately half of cases with the

frequency in descending order being: CD20 (17%), CD7 (16%), CD19 (9.8%), CD2 (7.5%),

CD3 (6.7%), CD5 (4.8%), and CD10 (2.9%). CD56, typically found on NK cells can also be

found on AML cells, but not on normal myeloid cells. CD56 expression in (t8;21) AML was

associated with a higher rate of relapse.(Iriyama, Hatta et al. 2013)

The European Group for the Immunological Classification of Leukaemias (EGIL) had proposed

in the past that AML be defined immunologically by the expression of 2 or more of the

following myeloid markers: myeloperoxidase, CD13, CD33, CDw65, and CD117, but only three

of FAB subtypes could be easily defined by immunological markers alone: M0, M6, M7.(Bene,

Castoldi et al. 1995) The prognostic significance of 21 antigens was evaluated in 177 adult AML

patients and no single antigen expressed on blasts predicted patient survival. However, patients

with blasts expressing all 5 myeloid markers (myeloperoxidase, CD13, CD33, CDw65, CD117)

had a higher complete remission rate (p< 0.0001), improved disease-free survival (p =0.02) and

overall survival (p =0.008), than patients whose cells expressed fewer than 5 of these markers.

(Legrand, Perrot et al. 2000) This finding suggests that more differentiated leukemia responds

better to the therapy than an undifferentiated leukemia.

Page 26: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

6

1.2.4 Minimal residual disease (MRD)

Morphological assessment of bone marrow samples using light microscopy can only determine

when leukemic blasts are abundant and greater than the normal frequency of 5% of cells in the

bone marrow. AML patients are routinely assessed with bone marrow aspiration and molecular

testing, which may include PCR for common translocations and fusion transcripts or flow

cytometry to detect cells with leukemia associated phenotypes.(Baer 2002) Most patients can be

put into a complete morphological remission with current protocols, but many relapse within two

years due to minimal residual disease that has not been irradicated.(Burnett, Wetzler et al. 2011)

Minimal residual disease detection for AML is not part of standard of care at most institutions,

but there is considerable evidence to support measuring this parameter for prognostication and

risk stratification for dose intensification.

1.2.4.1 Polymerase chain reaction based MRD

Polymerase chain reaction (PCR) involves the amplification of a sequence of DNA using DNA

polymerase in combination with sequence specific primers and in a controlled thermocycling

program. Using reverse transcriptase a PCR reaction can also copy mRNA sequences into DNA

and subsequently amplify abnormal mRNA sequences. Both techniques can be used for initial

diagnostic purposes and MRD detection. Variations on methodology include semi-quantitative

and real-time quantitative PCR approaches. A limiting factor with this approach is that many

patients do not have a gene translocation that facilitates amplification.

T(8;21), inv(16), FLT3 internal tandem duplication (ITD) (20-30%) represent the most common

molecular abnormalities in AML amenable to detection by PCR based MRD methods, with

significant implications for relapse and overall survival.(Kottaridis, Gale et al. 2001; Meshinchi,

Woods et al. 2001; Stirewalt, Willman et al. 2001) MRD for FLT3 using PCR had a high degree

of sensitivity (0.01-0.001%).(Stirewalt, Willman et al. 2001).

AML patients with recurring reciprocal translocations can have MRD detected using reverse

transcriptase PCR (RT-PCR) to detect aberrant mRNA transcripts.(Strehl, Konig et al. 2001;

Olesen, Clausen et al. 2004) RT-PCR based MRD for acute promyelocytic leukemia (APL) with

t15;17 (PML-RARα fusion transcript) has been essential to the assessment of therapeutic

response, and sensitive approaches using ‘nested’ or ‘semi-nested’ RT-PCR have been

Page 27: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

7

utilized.(Miller, Levine et al. 1993; Gau, Young et al. 2000; Chendamarai, Balasubramanian et

al. 2012) Sensitivity for this approach ranges from 1 in 10,000 in some instances to 1 in

1,000,000 in others.

1.2.4.2 Flow cytometry based MRD

Flow cytometry approaches to MRD detection have the advantage of being applied to a larger

proportion of AML patients using leukemia-associated phenotypes to track reduction of disease

at end of induction or consolidation. Flow cytometry techniques allow for a sensitivity in

detecting leukemic cells of approximately 1 in 10,000 (0.01%). Presence of MRD post therapy

has been shown to be predictive of survival in ALL(Dworzak 2002; Borowitz, Devidas et al.

2008; Bassan, Spinelli et al. 2009) and AML(San Miguel, Vidriales et al. 2001; Buccisano,

Maurillo et al. 2010), but MRD detection is more routinely used in risk stratification for ALL

patients in the clinical trial setting or in standard of care management. San Miguel et al. assessed

BM samples of AML patients in first morphologic remission in 126 patients with aberrant

phenotypes at diagnosis, which demonstrated low risk of relapse with MRD <1 in 10,000 bone

marrow cells with progressively higher relapse rates with each log increase in MRD.(San

Miguel, Vidriales et al. 2001) In a large multicenter study of pediatric AML, an MRD marker

was identified in 90% of 232 AML patients and those with MRD higher than 0.1% at the end of

a second induction cycle predicted relapse.(Rubnitz, Inaba et al. 2010) MRD positivity prior to

allogeneic stem cell transplantation is also highly predictive of relapse in both the myeloablative

and non-myeloablative settings.(Walter, Gyurkocza et al. 2014) Application of MRD techniques

to the detection of leukemic stem cells is a potential extension of this approach currently under

investigation.

1.2.4.3 Leukemic stem cell MRD

While MRD is clinically predictive of relapse free and overall survival, most studies do not

attempt to detect residual leukemic stem cells. However, MRD positive patients do not always

relapse, suggesting that in some cases the assay is not detecting leukemic stem cells, but rather

more differentiated progenitors or perhaps pre-leukemic stem cells(Shlush, Zandi et al. 2014). It

stands to reason that in MRD positive patients who relapse, there remains a proportion of these

detected cells which are bona fida leukemic stem cells. Several attempts have been made to

Page 28: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

8

measure leukemic stem cell burden pre and post therapy which is discussed further in the section

on leukemic stem cells (Clinical relevance of LSCs).

1.2.5 Chemotherapy for AML

Overall survival outcomes for patients with acute myeloid leukemia (AML) have been slower to

improve over the last 20 years relative to acute lymphoblastic leukemia (ALL)(Lowenberg,

Downing et al. 1999; Burnett, Wetzler et al. 2011) indicating a need for novel therapies for this

disease. The mainstay of therapy for AML is based on a core regimen of daunorubicin and

cytarabine, and 70-85% of AML patients treated with current chemotherapy protocols are able to

achieve a morphologic remission (Hurwitz, Mounce et al. 1995) (Ribeiro, Razzouk et al. 2005),

defined as less than 5% myeloblasts in a cellular marrow with trilineage hematopoiesis.

However, despite meeting these criteria many patients relapse because of recurrence from MRD

leading to a five year survival of approximately 40% in adults(Lowenberg, Downing et al. 1999)

and 60% in children(Rubnitz 2012) with high risk groups faring much worse (<10%)

(Grimwade, Hills et al. 2010). Survival outcomes remain particularly poor for patients over the

age of 60 (Laubach and Rao 2008). In these relapsing patients, eradication of leukemic stem

cells has not occurred.

Dose intensification of induction with standard-dose cytarabine and daunorubicin at 45 or 90

mg/m2 for 3 days was assessed in a large prospective trial showing that adverse events were

similar in both arms and a significantly higher complete remission rate was achieved with higher

dosing of daunorubicin (67.6 vs 57.2%), however, OS was only significantly improved in

patients with favourable or intermediate risk cytogenetics.(Fernandez, Sun et al. 2009) The

number of cycles of consolidation post remission varies between institutions with no clear

evidence to determine the optimal number.(Rowe and Tallman 2010) One other important drug

in AML therapy is the topoisomerase II inhibitor, etoposide, which has single agent activity

against AML and can be incorporated into induction or consolidation protocols depending on the

risk category, age and cardiac status of the patient.(Ho, Brado et al. 1991)

Page 29: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

9

1.2.6 Leukemia cell lines

Leukemia cell lines have been derived from primary human AML and stored in cell banks such

as the American Type Culture Collection (ATTC). The development of cancer cell lines has

accelerated basic and translational discoveries in oncology by providing a consistent and readily

available source of tumour cells to assay. There are numerous leukemia derived cell lines well

characterized in the literature.(Drexler 2010) Cell lines are required to have been in continuous

culture from the primary tissue beyond a year and demonstrate a degree of homogeneity. The

list below includes cell lines used in this work.

1.2.6.1 K562

K562 is a cell line derived from the pleural fluid of a CML patient in blast crisis.(Lozzio and

Lozzio 1975) It was subsequently characterized as having a near triploid karyotype with a

Philadelphia chromosome, Fc gamma receptor positivity, with a tentative assignment as

granulocytic lineage (Klein, Ben-Bassat et al. 1976) Further studies of this cell line

demonstrated expression of glycophorin, a red cell sailoglycoprotein, suggesting erythroid

lineage.(Andersson, Nilsson et al. 1979) More recent immunophenotyping of K562

demonstrated the presence of myelomonocytic (CD13, CD15, CD33, CD65) and

erythrocytic/megakaryocytic (CD9, CD41, CD61 and CD235a) lineage markers(Toba, Kishi et

al. 1996) prompting its classification as an erythrocytic/megakaryocytic cell line(Drexler 2010).

It has downregulated class I HLA expression, which makes it susceptible to NK cell-mediated

lysis, in turn making it a standard target in the assessment of NK cell cytolytic function.

However, it is not a true AML cell line, because it is derived from a CML patient in blast crisis

with mixed lineage markers.

1.2.6.2 KG1 and KG1a

KG1 is a commonly used leukemia cell line derived from a patient with erythroleukemia (M7) in

myeloblastic relapse (Koeffler and Golde 1978) reported as having an immunophenotype of

CD34+CD38+. It has demonstrated the capacity to cause leukemia in SCID mice with injection

of 107 cells, leading to bone marrow infiltration, peripheral blood leukemia and end stage disease

between 6-8 weeks.(Sawyers, Gishizky et al. 1992) KG1a is a subclone derived from KG1 with

a more primitive immunophenotype, being predominantly CD34+CD38-.(Koeffler, Billing et al.

1980) Both cell lines have fast cycling time and are are known to engraft in immunodeficient

Page 30: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

10

mice. FcγII receptors (CD32) are expressed on KG1 and KG1a (25 and 17%) respectively.(Wu,

Markovic et al. 1996). While both KG1 and KG1a express CD34, KG1a is CD33 negative(Silla,

Chen et al. 1995), which is consistent with its more primitive CD34+CD38- immunophenotype.

1.2.6.3 OCI/AML2, OCI/AML3 and OCI/AML5

Cell lines derived from primary AML samples at the Ontario Cancer Institute have been

designated with the prefix OCI/AML and a number based on the order of development. These

are novel AML lines not available from the ATCC. OCI/AML2 was derived from a 65 year old

male with AML M4 at diagnosis. Its immunophenotype is negative for CD3 and CD14 and

positive for CD13, CD15, CD19 and CD33 (Wang, Curtis et al. 1989) It has hyperdiploid

complex cytogenetic translocations. OCI/AML3 was developed from a 57 year old male with

AML M4. The cells carry an NPM1 gene mutation (Type A) and NDMT3A R8826C

mutation(Wang, Curtis et al. 1989). It also has a hyperdiploid karyotype with complex

cytogenetics and hemizygous for RB1. OCI/AML5 was derived from a 77 year-old male with

AML M4 (relapse), with complex hyperdiploid karyotype and a doubling time between 30-50

hours.(Wang, Koistinen et al. 1991) Immunophenotypically it is negative for CD3, CD7, CD19,

CD14 and positive for CD34, CD68, TdT, HLA-DR and CD8. OCI/AML2 and OCI/AML3

grow in standard medium with 10% FBS supplementation and do not require additional cytokine

supplementation, but OCI/AML5 also requires 5637 bladder carcinoma conditioned medium

(10%) for optimal growth.

1.2.7 Animal models of leukemia

While early cancer drug discovery focused on in vitro assays, animal models have progressively

become more prevalent in the preclinical assessment of cancer therapeutics and an expectation in

most cases prior to testing in humans. However, in vitro toxicity assays remain a useful

screening tool to identify agents which may be of therapeutic benefit. These assays include

measures of mitochondrial viability (e.g. MTT assay), membrane permeability (e.g chromium

release assay) and flow cytometric approaches (e.g. forward and side scatter properties,

propidium iodide staining and annexin V staining). However, in vitro efficacy of an agent often

does not translate to efficacy in animal models of disease, due to issues of biological complexity.

Unlike in vitro assays, animal models allow for an assessment of the pharmacokinetic clearance

Page 31: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

11

of an agent, organ toxicity and interactions with the immune system and three dimensional

aspects of tumour biology.

Syngeneic murine cancer models involve use of cancer cell lines to induce cancer in the same

strain of mouse as the original tumour. This allows for murine tumour cell lines to be

transplanted into mice that are typically unable to reject the tumour and facilitate study of

therapeutic agents in a fully immunocompetent animal model. Testing of proven clinically

active oncology drugs in these models demonstrate efficacy providing some support for their use

in cancer drug discovery.(Darro, Decaestecker et al. 2005) However, the biology of murine

cancers differ from human cancer, drawing into question their use as preclinical screening tools.

Voskoglou-Nomikos et al. conducted a review of 31 anti-cancer drugs and compared their

performance in human cell line, human xenograft, and syngeneic murine models, relative to their

efficacy at the phase II clinical trial stage in breast, non-small cell lung, ovary, and colon

cancers. Syngeneic murine models of cancer were not predictive of performance in human

clinical trials. However, human cell line and human xenograft models were predictive for non-

small cell lung cancer and ovarian cancer, and cell line models were predictive for breast

cancer.(Voskoglou-Nomikos, Pater et al. 2003) This lack of translation has been an issue,

whereby agents appeared effective in murine disease models, but did not work in humans. To

address this issue, advances in murine xenograft models were developed using genetically

immunodeficient mice permissive for human tumour growth, allowing for biological

characterization of human tumours in vivo and preclinical assessment of novel agents.

The development of murine human xenograft models dates back to the 1970s, but there have

been three main advances that have had the greatest impact(Shultz, Ishikawa et al. 2007). The

first was the discovery and characterization of protein kinase DNA-activated catalytic

polypeptide (Prkdc) mutated CB17 mice(Bosma, Custer et al. 1983). The second was back

crossing the Prkdc mutation onto a non-obese diabetic (NOD) background(Shultz, Schweitzer et

al. 1995), yielding NOD/SCID mice. The third was a null or truncated mutation in the common

interleukin-2 receptor gamma chain to yield a NOD/SCID gamma null mouse(Ito, Hiramatsu et

al. 2002; Shultz, Banuelos et al. 2003; Traggiai, Chicha et al. 2004; Ishikawa, Yasukawa et al.

2005).

Page 32: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

12

1.2.7.1 Early xenograft models

The first attempt to xenograft human AML utilized thymectomized, irradiated mice rescued with

syngeneic bone marrow infusions that were infused sc with AML cells that formed transient

local tumours(Franks, Bishop et al. 1977) Subsequently, neonatal thymectomized mice were

irradiated and conditioned with cytarabine to facilitate growth of primary AML cells injected

subcutaneously leading to localized tumours (Palu, Selby et al. 1979). Following this, athymic

nude mice(Flanagan 1966; Segre, Nemhauser et al. 1995) which lack functional T and B cells

were utilized to study AML cell lines (MO7E and TF-1) in vivo, but required gene modification

to produce essential cytokines to allow for engraftment (Thacker and Hogge 1994). Human

AML cell lines such as HL-60(Yamada, Mori et al. 1983; Potter, Shen et al. 1984) and KG-1

expanded in these models better(Machado, Gerard et al. 1984), but tended to generate localized

myelosarcomas with minimal bone marrow engraftment(Nilsson, Giovanella et al. 1977) making

them poor leukemia models. The most significant limiting factor with these early xenograft

models was that primary leukemia samples did not engraft well.(Nara and Miyamoto 1982;

Caretto, Forni et al. 1989)

1.2.7.2 SCID mice

Severe combined immunodeficient (SCID) mice were first described in 1983 by Bosma et

al.(Bosma, Custer et al. 1983) due to autosomal recessive mutation of chromosome 16, resulting

in disruption of the protein kinase DNA-activated catalytic polypeptide (Prkdc) gene.(Blunt,

Finnie et al. 1995; Kirchgessner, Patil et al. 1995; Miller, Hogg et al. 1995; Blunt, Gell et al.

1996) Abnormal Prkdc genes lead to deactivation of a DNA recombinase enzyme, resulting in

inability to rearrange T- and B-cell antigen receptors, due to failure of coding joint

formation(Lieber, Hesse et al. 1988) in the final step of VDJ recombination(Malynn, Blackwell

et al. 1988). This causes a generalized deficiency in humoral and cellular immunity, with

incomplete penetrance, allowing for the production of only a small number of functional T- and

B-cells, resulting in severe immunodeficiency.(Bosma and Carroll 1991) The ‘leakiness’ of the

SCID mutation is strain dependent, and mice can have high levels of immunoglobulin (Ig) with

production increasing with age and some functional T- and B-cell rearrangements.(Nonoyama,

Smith et al. 1993) The SCID mouse resulted in superior engraftment of human hematopoietic

tissue(McCune, Namikawa et al. 1988), as demonstrated by the generation of long-term

production of human T-cells after injection of peripheral blood leukocytes

Page 33: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

13

intraperitoneally(Mosier, Gulizia et al. 1988). The earliest leukemia xenograft models used

SCID mice to engraft human pre-B acute lymphoblastic leukemia primary samples(Kamel-Reid,

Letarte et al. 1989). This model was then used in an attempt to get primary AML samples to

engraft. However, SCID mice have high levels of NK cell activity, limiting xenograft potential.

1.2.7.3 NOD/SCID mice

In addition to T- and B- cell deficits, NOD/SCID mice have reduced numbers and function of

NK cells, but some activity does remain.(Shultz, Schweitzer et al. 1995) This is evident in the

fact that only 70% of all AML samples exhibit detectible engraftment in NOD/SCID mice

(Ailles, Gerhard et al. 1999) and many of these samples engraft at less than 10%, measured at 6-

12 weeks. Limitations of the NOD/SCID mouse model includes development of thymic

lymphomas by 8.5 months, radiosensitivity and residual NK cell activity, which limit

engraftment of xenogeneic tissue.(Shultz, Schweitzer et al. 1995)

1.2.7.4 NOD/SCID Interleukin-2 receptor gamma null mice

The IL-R gamma chain(IL-2Rγc) is common to the cytokine receptors IL-2, IL-4, IL-7, IL-9, IL-

15 and IL-21 and is essential to multiple immune functions (reviewed by Sugamura)(Sugamura,

Asao et al. 1996). Deficiencies in IL-2Rγc alone lead to severe T- and B-cell deficiencies and a

blockade of NK cell development.(Cao, Shores et al. 1995; DiSanto, Muller et al. 1995; Ohbo,

Suda et al. 1996). The development of IL-2Rγc null mice back-crossed onto the NOD/SCID

background(Ito, Hiramatsu et al. 2002; Ishikawa, Yasukawa et al. 2005; Shultz, Lyons et al.

2005), led to a significant improvement in the engraftment of human hematopoietic tissue

(benign and malignant), as they are fully deficient in T-, B- and NK cells. However, these mice

retain some elements of innate immunity in the myeloid cell compartment (ie neutrophils,

monocytes and macrophage). A study of leukemic cell engraftment was undertaken in 3 strains

of immunodeficient mice: NOD/SCID, NOD/SCID/β2microglobulin null (NSB) and

NOD/SCID/IL-2Rγc null (NSG).(Agliano, Martin-Padura et al. 2008) Engraftment of five

primary AML samples in the three murine strains revealed that only 2/5 samples engrafted in

NOD/SCID or NSB mice, whicle 5/5 engrafted in NSG mice.

Another similar study demonstrated a 6-fold increase in bone marrow engraftment of human

HSC in NSG mice relative to NOD/SCID mice.(Shultz, Lyons et al. 2005) Also, NOD/SCID

Page 34: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

14

mice develop thymic lymphomas(Christianson, Greiner et al. 1997), and NSB mice(Shultz,

Ishikawa et al. 2007) have accelerated lymphomagenesis, leading to significantly shorter

lifespans than NSG mice, which creates confounding issues when assaying human malignant

cells.

1.2.8 Cancer stem cell hypothesis

John Bennett was the first to clearly describe a patient with leukemia and implicating it as a

disease of the blood, which he termed “leucocythaemia”.(Bennett 1845) Rudolph Virchow soon

after described another patient with leukemia, who had a very high white blood cell count, and

termed this condition, “Leukämie”, derived from two Greek words; leukos (white) and

haima(blood). (Virchow 1856) In addition, to this important diagnostic clinical discovery,

Virchow also made the observation of tumour pleimorphism, postulated that, ‘cells came from

cells’, and that cancer cells were derived from normal cells. Studies by Furth et al. using inbred

mouse strains and syngeneic leukemic cell lines demonstrated that the tumour initiating

frequency was between 1 and 100.(Furth 1935) More remarkable, was their subsequent finding

that a leukemia could be transmitted with a single cell to a murine host.(Furth, Kahn et al. 1937)

However, the underlying basis for the heterogeneity of leukemia, with apparently rare tumour

initiating cells, was not considered.

The demonstration of normal hematopoietic stem cells by Till and McCulloch,(McCulloch and

Till 1960) led them to explore the underlying process that determined cell fate decision to self-

renew or differentiate. They proposed a stochastic model based on the frequencies of HSCs as

detected by the colony forming unit-spleen (CFU-S) assay, given that the number colonies

derived from serially transplanted CFU-S approximated a gamma distribution.(Till, McCulloch

et al. 1964). They acknowledged that their study had been demonstrated in only one

hematopoietic stem cell model and remained open to the possibility of non-stochastic

mechanisms in stem cell fate decision-making. These concepts were extended to tumour models,

with a debate forming between a stochastic and stem cell model of tumour progression. The

concept of a cancer stem cell in leukemia was proposed by Bruce et al. (Bruce and Ash 1963)

and provided an alternative to the stochastic model to explain rare tumour initiating cells. One

additional feature of this theory was that cancer cells could differentiate along a hierarchy

Page 35: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

15

somewhat analogous to normal hematopoietic stem cells. Early cell cycle kinetic studies by

Clarkson et al. in cell line and murine models of acute leukemia demonstrated that the majority

of leukemic blasts were postmitotic. Further, two additional populations were identified; a fast

and slow cycling group representing the minority of cells.(Clarkson 1969) The authors proposed

that the slow cycling fraction represented a stem cell population analogous to a hematopoietic

stem cell. LSCs in AML were the first identified cancer stem cell population, which were shown

to be enriched in the CD34+ CD38- fraction of whole blasts, as measured by the ability to

engraft in the bone marrow of SCID mice. Further, these cells possessed the capacity to

differentiate into more mature blasts (CD34+CD38+ AML cells).(Lapidot, Sirard et al. 1994;

Bonnet and Dick 1997)

A stem cell hierarchy for AML was demonstrated and cancer stem cells have subsequently been

demonstrated in a variety of cancers, including brain tumors(Singh, Clarke et al. 2003; Singh,

Hawkins et al. 2004), breast cancer(Al-Hajj, Wicha et al. 2003), multiple myeloma(Matsui, Huff

et al. 2004) and colon cancer(O'Brien, Pollett et al. 2007; Ricci-Vitiani, Lombardi et al. 2007).

Some cell lines have demonstrated cancer stem cells within cell C6 glioma cell lines and

multiple myeloma cell lines.(Kondo, Setoguchi et al. 2004; Matsui, Huff et al. 2004)

A recent report of dual LSC populations in a large number of primary AML samples further

refine the CD34+CD38- compartment as follows: Lin-CD34+CD38-CD90-CD45RA+ and Lin-

CD34+CD38-CD90-CD45RA-.(Goardon, Marchi et al. 2011) There are other non-

immunophenotypically based functional definitions which have enriched AML LSCs, including

side population (SP) cells(Wulf, Wang et al. 2001) and aldehyde dehydrogenase (ALDH)

expressing cells(Cheung, Wan et al. 2007). Colon cancer-initiating cells have been

identified(O'Brien, Kreso et al. 2009), and recent evidence indicates that they are sensitive to NK

cell-mediated killing due to low HLA expression, and increased expression of NKp30 and

NKp44 ligands, while differentiated colon cancer cells are less so.(Tallerico, Todaro et al. 2013)

1.2.8.1 Leukemia stem cell markers

While LSCs were postulated following the discovery of normal hematopoietic stem cells by Till

and McCulloch(McCulloch and Till 1960), until cell surface markers and cell sorting techniques

developed to distinguish and separate putative populations, it could not be confirmed

Page 36: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

16

experimentally. While there have been many reported LSC markers a few have been of

particular relevance and will be described here.

1.2.8.1.1 CD123

CD123 is present on ~99% of leukemic stem cells(Jordan, Upchurch et al. 2000), but is not

expressed on normal hematopoietic stem cells(Huang, Chen et al. 1999), making it an attractive

therapeutic target. CD123 is the interleukin 3 receptor alpha chain, or low affinity receptor. It is

a type I transmembrane glycoprotein. The alpha chain is shared by the IL-5 and granulocyte

monocyte-colony stimulating factor (GM-CSF) receptors. However, when coupled to the

interleukin 3 receptor beta chain (CDw131), the binding infinity increases dramatically,

facilitating signal transduction from low concentrations of IL-3. IL-3 is important in driving

myeloid differentiation and can activate STAT5. IL3R knockout mice do not have major

hematologic impairment, but have reduced granulcotye-monocyte colony forming capacity

(CFU-GM). CD123 is expressed on committed hematopoietic progenitors and mediates

differentiation and proliferation. Variable levels of CD123 have been reported on CD34+ HSCs,

but it is low on HSCs. CD123 is expressed on cells of the hematopoietic system (monocytes,

neutrophils, basophils, eosinophils, megakaryocytes and erythroid precursors, mast cells,

macrophages, some B lymphocytes) and non-hematopoietic tissue (Leyding cells of the testis,

placenta, and brain).(Moretti, Lanza et al. 2001)

1.2.8.1.2 CD34

CD34 is a cell surface glycoprotein that as been widely used as a marker to identify and isolate

hematopoietic stem cells (Berenson, Andrews et al. 1988; Andrews, Singer et al. 1989; Ema,

Suda et al. 1990) and progenitors. It also marks vascular endothelial cells(Baumhueter, Dybdal

et al. 1994) other tissue-specific stem cells, including muscle satellite cells and epidermal

precursors, as well as mast cells and eosinophils.(Nielsen and McNagny 2008) The definitive

function of CD34 has not been determined, but has been proposed to promote the proliferation

while blocking differentiation of progenitor cells.(Krause, Fackler et al. 1996) Further, CD34

may have roles in chemotaxis and in asymmetric cell division. CD34-knockout mice have a

delay in both erythroid and myeloid differentiation, with reduced colony-forming activity of

hematopoietic progenitors derived from bone marrow, and inability to be cultured ex vivo with

standard cytokines support.(Cheng, Baumhueter et al. 1996) However, CD34-knockout mice

Page 37: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

17

develop normally and have normal hematopoiesis in the adult mouse. CD34 selection has been

shown to enrich LSCs in primary AML samples.(Lapidot, Sirard et al. 1994)

1.2.8.1.3 CD38

While not a stem cell marker per se, the absence of CD38 expression has been used to further

enrich CD34+ hematopoietic or leukemic stem cell populations, and it is therefore an important

differentiation marker that can distinguish stem cells from progenitors.(Lapidot, Sirard et al.

1994) Human CD38 is made up of a single chain of 300 amino acids with a molecular weight of

45 kDa and is expressed by hematopoietic and non-hematopoietic cells, including NK cells and

monocytes. Other CD38+ cells include smooth and striated muscle cells, renal tubules, retinal

gangliar cells and cornea (reviewed in Malavasi)(Malavasi, Deaglio et al. 2008). CD38 is

involved in signal transduction, cell adhesion and calcium signaling. The binding to the ligand

CD31, initiates a signaling cascade that includes phosphorylation of sequential intracellular

targets and increases cytoplasmic Ca2+

levels, mediating different biological events, depending

on the cell type (e.g., activation, proliferation, apoptosis, cytokine secretion and homing). While

absence of CD38 was used to establish classic LSC definitions, subsequent work demonstrated

the LSCs can exist in the CD34+CD38+ compartment of a significant number of primary AML

samples.(Taussig, Miraki-Moud et al. 2008)

1.2.9 Clinical relevance of LSCs

Most clinical studies of leukemic stem cells (LSCs) have focused on the CD34+CD38- definition

with or without addition of CD123 or CLL-1(van Rhenen, Feller et al. 2005; van Rhenen,

Moshaver et al. 2007; Witte, Ahlers et al. 2011). The latter two antigens were discovered

through use of flow cytometric analysis of CD34+CD38- AML cells using panels of CD markers

rather than from biological studies in immunodeficient mice. While some studies have

demonstrated that higher NOD/SCID engraftment capacity correlates with worse survival

outcomes(Pearce, Taussig et al. 2006), this does not provide evidence to exclusively support the

cancer stem cell hypothesis because a stochastic model of tumour initiation and maintenance

could also explain these findings. A more recent study demonstrated that patient’s whose whole

AML samples had a gene expression profile similar to LSCs or HSCs had worse survival

outcomes, and this could be used to better risk stratify patients independent of known prognostic

factors.(Eppert, Takenaka et al. 2011) While this provides some evidence that leukemic stem

Page 38: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

18

cell signatures influence clinical outcome, it does not definitively show that treatment of LSCs is

required for cure. This is consistent with earlier studies showing that less differentiated

leukemia, as determined by immunophenotype, has a worse prognosis.(Legrand, Perrot et al.

2000)

Only two clinical studies to date have studied the clinical relevance of functionally validated

immunophenotypic LSC definitions established using immunodeficient murine models. The first

was a study of 92 AML patients that showed a high burden of CD34+CD38- AML cells at

diagnosis correlated with poor survival and conventional MRD positivity after the third cycle of

chemotherapy(van Rhenen, Feller et al. 2005). A similar retrospective study in 17 pediatric

AML patients from the BFM clinical trial group also demonstrated a high CD34+CD38- cell

burden led to increased relapse and worse survival outcomes.(Witte, Ahlers et al. 2011) While

this provides an additional level of evidence to support the leukemia stem cell hypothesis, it is

not definitive. CD34+CD38- LSCs are chemo-resistant due to prolonged G0 status(Guan,

Gerhard et al. 2003) and drug efflux potential(Costello, Mallet et al. 2000; Wulf, Wang et al.

2001), and are presumed to be responsible for recapitulating leukemic disease.

The demonstration that a high burden of CD34+CD38- AML cells at diagnosis correlates with

worse survival outcome does not directly confirm that these cells are the subset that leads to

relapse post therapy. To directly demonstrate this requires assessing the response of these cells

to chemotherapy and demonstrating that the residual CD34+CD38- cell burden post-therapy is

correlated with relapse and survival. This approach has been utilized for prognosticating in pre-

B ALL, where MRD post induction is predictive of survival(Dworzak 2002; Borowitz, Devidas

et al. 2008; Bassan, Spinelli et al. 2009), and is currently used in risk stratification in numerous

major clinical trial groups worldwide (e.g. Children’s Oncology Group and BFM Group). This

ability to risk stratify and intensify therapy rationally has contributed to improved outcomes in

the treatment of ALL. Studies designed to assess the prognostic value of MRD have taken bone

marrow and peripheral blood samples for MRD at day 8, 14 and 29 (e.g. chemotherapy clinical

trial COG AALL 0331), while other studies have utilized later time points including at the end of

therapy. There is no published study that has risk stratified patients using either CD34+CD38-

burden at diagnosis or post-therapy, and dose intensified the higher risk group either with

additional chemotherapy or allogeneic bone marrow transplantation.

Page 39: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

19

1.2.9.1 Strategies to detect LSCs in patients

In AML both PCR and flow cytometry can be used to assess for MRD at various time points

during and post therapy. While PCR testing may be more sensitive (~1 in 105) than flow

cytometry (~1 in 104)(Buccisano, Maurillo et al. 2009), it is an assessment of bulk residual

disease and cannot be used to differentiate leukemic stem cells from blasts. Flow cytometry on

the other hand, assesses cells on a single cell basis, allowing for examination of residual LSCs in

peripheral blood and bone marrow. LSCs can be detected easily in peripheral blood using CD34

and CD38 staining, but in bone marrow detection is confounded by normal HSCs, which also are

immunophenotypically CD34+CD38-.

The two clinical studies evaluating LSC burden at diagnosis used different gating strategies to

identify LSCs(van Rhenen, Feller et al. 2005; Witte, Ahlers et al. 2011). In a follow-up study by

van Rhenen et al., LSCs could be detected in 55 patients with either AML or high risk

myelodysplastic syndrome in remission using several 4 and 5 color staining panels. This study

established that LSCs can be distinguished from HSCs in normal bone marrow by lineage marker

expression (e.g. CD2, CD5, CD7, CD19, CD11b, CD22 and CD56), lineage marker

asynchronous antigen expression (e.g. CD13-CD33+), lineage marker overexpression (e.g.

CD33++) and underexpression (e.g. HLA-DR low). Further, the LSC specific markers

CD123(Jordan, Upchurch et al. 2000) and CLL-1(van Rhenen, van Dongen et al. 2007) were

also demonstrated to effectively differentiate LSCs from HSCs. CD123 may be present at low

levels on normal HSCs and can upregulate post chemotherapy, confounding its use as a LSC

marker immediately after therapy for some patients (i.e. post induction). CLL-1 does not

upregulate post chemotherapy, but is not as frequently or highly expressed on LSCs as

CD123(van Rhenen, Moshaver et al. 2007). CLL-1 is restricted to the hematopoietic lineage, in

particular to myeloid cells present in peripheral blood and bone marrow, is absent on HSCs, but

highly expressed on LSCs. Recently, it was reported that Tim3 provides another marker that can

differentiate HSCs from LSCs (Jan, Chao et al. 2011). Tim3 is a Th1-specific cell surface

protein that regulates macrophage activation, and is expressed on LSCs. CD96 has also been

identified as a LSC marker.(Hosen, Park et al. 2007)

Page 40: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

20

1.3 Cytotoxicity assays

The concept that rare cancer stem cells initiate and propagate tumours have profound

implications for the interpretation of cytotoxicity assays where the assumption of tumour

homogeneity has been made. Typical cytotoxicity assays test a snap shot assessment of bulk

tumour cells to determine whether a tumour has signs of apoptosis (e.g. annexin V positivity),

mitochondrial damage (e.g. MTT assessment of mitochondrial activity), cell permeability (e.g.

51CrO4 release assay and PI staining in flow cytometry), or some other readout deemed indicative

of viability or cell death. Leukemic stem cells have been demonstrated to have resistance to

chemotherapeutic agents relative to bulk leukemia (Costello, Mallet et al. 2000), emphasizing the

importance of measure cytotoxicity against the leukemic stem cell.

Where a cancer stem cell has been identified and validated in vivo for a particular cancer using

specific cell surface markers, cell sorting can be used in conjunction with one of the previously

mentioned approaches to address toxicity against the cancer stem cell population. However,

most primary cancers and cell lines do not have an identifiable cancer stem cell population that

can be generalized, making this approach problematic. Even where cancer stem cell definitions

have been validated and one can use conventional cytotoxicity assays, functional assessments

against their proliferative and clonogenic capacity are important parameters to measure when

evaluating novel cancer therapeutics.

1.3.1 Bulk cytotoxicity assays

Most cytotoxicity assays have focused on the impact on bulk tumour populations. The classic

MTT assay measures the mitochondrial enzyme activity as a measure of viability and is used

commonly in quantitation of drug sensitivity of tumour cells. However, the assay is confounded

when testing mixtures of immune effectors and targets that cannot easily be separated,

confounding the viability readouts. This is one reason that the chromium release assay has been

commonly used to evaluate the impact of immune effectors on tumour targets. The basic

principle involves labelling tumour targets with 51

CrO4 prior to co-incubation with immune

effectors and measuring the released chromium relative to spontaneous and maximum release

values.(Brunner, Mauel et al. 1968) While newer types of assays have been developed, the

majority are designed to measure cytotoxicity against bulk tumour. The calcein AM assay is one

such approach that uses a non-radiolabelled dye to facilitate identification of viable and dead

Page 41: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

21

cells.(Neri, Mariani et al. 2001) The colorimetric change can be detected using a

spectrophotometer. However, the assay is more labor intensive and prone to signal-noise issues.

1.3.2 Flow cytometric cytotoxicity assays

Flow cytometry based cytotoxicity assays have focused on the use of propidium iodide which

can enter dead cells with porous membranes and is easily detectable.(Jones and Senft 1985)

Additional viability or dead cell stains can be used individually or in combination to allow for

more fine discrimination of viability at the single cell level. Annexin V detection is another

means, which allows for the detection of early apoptosis which leads to flipping of membrane

phospholipids enabling Annexin V to bind. Annexin V can be conjugated to standard

fluorochromes such as FITC to facilitate use in flow cytometric assays to detect early apoptotic

cells.(Anthony, McKelvie et al. 1998) The advantage of this approach is to identify a greater

proportion of dead cells than with PI. Combining PI and Annexin V can also be used in time

course studies to show progression of apoptosis, which begins with Annexin V positivity

followed by PI positivity. Necrosis and apoptosis can be identified using this approach by

tracking the position of cells in a two-by-two plot. Necrosis tends to proceed directly to PI

positivity, without conversion to Annexin V positivity. However, flow cytometric assays have

been typically used to measure cytotoxicity at one time point, either percent or cumulative

cytotoxicity. Percent cytotoxicity is a snap shot at one time of the proportion of cells assayed.

While this is typically extrapolated to the entire cell population, this neglects the possibility of

cellular breakdown after death, leading to an underestimate of cytotoxicity. This can be

controlled for using counting beads in all tubes, enabling a more accurate measure of

cytotoxicity. The longer a cytotoxicity assay runs, the more relevant cellular disintegration

becomes and the greater the underestimate of cytotoxicity. The percent cytotoxicity from flow

cytometric assays has yielded equivalent cytotoxic readouts as the chromium release

assay.(Ozdemir, Ravindranath et al. 2003)

1.3.3 Clonogenic cytotoxicity assays

The utility of the aforementioned standard methodologies to assess cytotoxic agents against

tumor targets have been questioned by a growing body of literature supporting rare cancer stem

cells in a number of cancers.(Lapidot, Sirard et al. 1994; Bonnet and Dick 1997; Al-Hajj, Wicha

et al. 2003; Singh, Hawkins et al. 2004; O'Brien, Pollett et al. 2007; Ricci-Vitiani, Lombardi et

Page 42: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

22

al. 2007) If only a small subfraction of tumour cells have true unlimited proliferative potential

then extrapolating the results of bulk tumour assays to the cancer stem cell fraction may not be

reflective of toxicity against the cancer stem cell fraction. Therefore, attempting to utilize

methods to address the impact of therapeutic agents against CSCs is highly relevant to the

development of curative strategies. It also has profound implications for studies of cytotoxicity

against bulk tumour samples, which have focused on bulk tumour assays and have more

confounding issues due to mixed effector and target populations. Although clonogenic readouts

have been used in the past to assess chemotherapy drugs(Curtis, Minden et al. 1995; Jacobs and

Wood 2005) they have rarely applied to evaluation of immune effectors.

The clonogenic assay was developed by Puck and Marcus in 1956, initially as a system to grow

HeLa cells(Puck, Marcus et al. 1956) and then to grow epithelial cells from normal human tissue

(liver, conjunctiva, kidney and appendix)(Cieciura, Marcus et al. 1956). This involved the use of

semi-solid medium supplemented with growth factors and nutrients to grow colonies derived

from single cells. This predated the discovery of hematopoietic stem cells in 1961 by Till and

McCulloch using the in vivo spleen colony forming assay.(Till and McCulloch 1961) An

effective means to generate murine bone marrow derived colonies was done in 1966 using agar

plates with mouse kidney or embryonic feeder layers.(Bradley and Metcalf 1966) The first

attempt to grow primary tumours in semi-solid medium was by McCulloch’s research group with

murine myeloma cells, which grew quantifiable colonies.(Park, Bergsagel et al. 1971)

Subsequently, they applied this approach to study sensitivity of murine and human hematopoietic

stem cells to chemotherapy drugs.(Ogawa, Bergsagel et al. 1973) Further applications of an

agar based colony forming assay for primary acute myeloid leukemia samples was demonstrated

to predict clinical response and resistance.(Park, Amare et al. 1980)

Currently, clonogenic assays typically involve growing the cells either in methylcellulose or agar

supplemented with various growth factors and cytokines. Proliferation of a subset of cells results

in the appearance of colonies that can be enumerated typically at 10-14 days. The value of

clonogenic assays is that it allows assessment of single cells with high proliferative capacity.

Tumour stem and progenitor cells have the ability to form colonies in a semisolid state matrix

consisted of either agar or methylcellulose providing a readout for testing cytotoxic agents.

However, limitations of clonogenic assays include low frequency of clonogenic cells, two log

range for cytotoxicity evaluation, and clump artifacts for some types of cells.(Hoffman 1991)

Page 43: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

23

Clonogenic chemotherapy assays are good at predicting patients likely be resistant to a particular

chemotherapy agent, but not as accurate at determining those who will be sensitive. Clonogenic

assays have the potential of individualizing patient treatment as they can predict clinical

responses, but have not been incorporated into standard of care for any given cancer, possibly

because of the inability to grow a high proportion of primary tumours using this approach.

Moreover, the sensitivity of clonogenic leukemic cells to chemotherapy predicts the clinical

response in AML.(Park, Amare et al. 1980) These data suggest that clonogenicity is a clinically

relevant parameter. While clonogenic cells are not cancer stem cells per se, they include a subset

of malignant progenitors and stem cells with significant proliferative capacity and the ability to

contribute to disease progression. Inhibition of colony formation, therefore, is an important

measure of cytotoxicity that assays single cells and is a reasonable surrogate for toxicity against

cancer stem cells.

Few studies have examined the in vitro sensitivity of cancer stem cells to immune effector

killing. In one study, lymphokine activated killer (LAK) cells and allogeneic lymphocytes were

shown to exert a modest cytotoxic effect on AML cancer stem cells (CD34+ CD38-) that were

intrinsically resistant to the chemotherapeutic agent, daunorubicin.(Costello, Mallet et al. 2000)

This was done by sorting the cells and performing a chromium release assay. A limitation of that

study was that the LAK cells were a mixed population of IL-2 activated T and NK cells, making

it difficult to discern the contribution of either cell type.

Another study using the clonogenic assay to examine the role of KIR mismatched NK cells

against primary AML showed a reduction in colonies.(Langenkamp, Siegler et al. 2009) This

study also utilized the chromium release assay and reported secondary replating of clonogenic

cells. Similarly, autologous activated marrow-infiltrating T-lymphocytes (MILs) have

demonstrated superior in vitro cytotoxicity against clonogenic MM cells compared to activated

peripheral blood lymphocytes (PBLs) isolated from the same patient. Normal hematopoietic

progenitors were not affected by MILs or PBLs, providing pre-clinical evidence for the safety of

this therapy.(Noonan, Matsui et al. 2005) However, a direct comparison between bulk tumor and

clonogenic cell killing was not considered in this work. Further, neither study utilized

appropriate controls to differentiate killing during the 4 hour co-incubation versus over the 2

week low density incubation in methylcellulose, making a comparison of methods difficult to

interpret.

Page 44: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

24

1.4 Cancer immunobiology

1.4.1 Lymphocytes

The cellular immune system comprises lymphocytes and myeloid cells, which evolved primarily

to protect organisms from microbial infection. However, the immune system has also

demonstrated the capacity to prevent cancer from developing, as evidenced by the increased rates

of leukemia and lymphoma in patients and animals with severe combined immunodeficiency.

These findings support the notion of immune tumour surveillance, and biological studies confirm

the anti-tumour activities of NK cells and T-cells. This capacity may have initially evolved in

the context of virally driven lymphoproliferative disorders and cancers. Both adaptive and innate

cellular immune systems can play a role in tumour immune surveillance and/or anti-tumour

responses. Primarily, these responses are mediated by lymphocytes. The lymphocyte subset

includes B-, T- and NK cells of which the latter two play important roles in tumour immune

surveillance.(Swann and Smyth 2007) T- and NK cells both contain cytotoxic granules

containing perforin and granzymes that can be exocytosed onto target cells, leading to apoptosis.

In addition, they can kill via death ligands on their surface such as Fas Ligand or TRAIL.

1.4.2 NK cells

Natural killer (NK) cells were discovered in the mouse by Herberman et al. and Kiessling et al.

as a population of spleen-derived non-T, non-B lymphocytes with cytolytic activity.(Herberman,

Nunn et al. 1975; Herberman, Nunn et al. 1975; Kiessling, Klein et al. 1975; Kiessling, Klein et

al. 1975) Subsequently, an analogous population was discovered in humans also termed NK

cells(Jondal and Pross 1975; Pross and Jondal 1975). Deficiency of NK cells in humans is rare

and can predispose to fatal infection.(Orange 2006) While murine NK cells are similar to

human, there are unique species-specific receptors. In humans, NK cells are classically defined

as CD3-CD19-CD56+ lymphocytes with variable expression of CD16. Cytolytic NK cells with

high perforin content express low levels of CD56, while cytokine secreting or ‘helper’ type NK

cells express high degrees of CD56 and often low levels of CD16. One proposed NK maturation

process involves differentiation from a CD56bright

CD16neg

cell predominantly in the BM, to a

CD56+CD16+ cell present in BM and SPL and finally a CD56dim

CD16+ cell predominantly in

the spleen.(Armas 2009)

Page 45: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

25

NK cells have had have been postulated to play a role in immune tumour surveillance(Trinchieri

1989) and play a complementary role to T-cells as many virally infected or tumour cells

downregulate class I HLA as means to evade detection and destruction by the immune system.

NK cells are able to recognize and destroy cells that have downregulated HLA class I molecules

on their surface. This enables NK cells to destroy virally-infected cells or tumor cells

unrecognized by T cells, which rely on antigen presentation in the context of HLA class I.

Therefore, when cells downregulate HLA class I, they are susceptible to NK cell recognition and

cytolysis, as described by the “missing self” hypothesis.(Karre, Ljunggren et al. 1986) This

function was later attributed to the Ly49, which when engaged by H-2Dd in C57BL/6 mice

inhibited NK cell function in an MHC class I specific manner.(Karlhofer, Ribaudo et al. 1992)

Further, Karlhofer et al. noted that absence of MHC I ligand on non-malignant tissue did not

result in NK cell cytotoxicity, implicating other NK recognition mechanisms. Subsequently,

analogous inhibitory receptors were discovered in the human(Moretta, Bottino et al. 1990;

Moretta, Tambussi et al. 1990) with identification of their ligands as class I HLA being

determined later.(Dohring and Colonna 1996) These human HLA specific receptors were

collectively termed inhibitory killer immunoglobulin-like receptors (KIRs).

A balance of inhibitory and activating signals determines whether an NK cell kills its

target(Sutlu and Alici 2009). This process is depicted in simplified form with only one

activating and inhibiting receptor (Figure 1.1, Figure 1.2, Figure 1.3), but NK cell signaling is an

integrative process involving many activating and inhibiting receptors working in concert to

provide specificity to its recognition capability. While the ‘missing self’ hypothesis explains

how NK cell autoreactivity can prevented by inhibitory receptors that respond to self MHC

(mouse) or HLA (human), it does not explain how activation occurs in the context of this

regulatory mechanism. The concept of ‘induced self’ involves the upregulation of stress related

antigens such as heat shock proteins, which can occur following infection or malignant

transformation of a cell. In the case of NKG2D ligands, when upregulated, these can override

Ly49 mediated inhibition as reviewed by Malarkannan et al.(Malarkannan 2006) This supports

the notion of NK regulation as an integration of signaling through different activating and

inhibiting receptors which detect ‘induced’ and/or ‘missing’ self, allowing for recognition of

‘altered self’. The balance of signaling depends on the nature and density of NK receptors and

target cell ligands.

Page 46: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

26

Figure 1.1: NK cell degranulation by stimulation with activating ligands only NK cells can be stimulated through a number of different activating receptors that may be

present on targets. In the presence an activating ligand (L) only granule exocytosis would be

triggered resulting in cytolysis.

Figure 1.2: NK cell inhibition of degranulation by stimulation with activating and

inhibitory ligands

In the presence of both an inhibitory and activating ligand (L) on a target cell, granule exocytosis

would be inhibited resulting in no cytolysis, provided that the effects of the inhibitory receptor

predominate.

Figure 1.3: NK cell degranulation by stimulation with activating and inhibitory ligands

In the presence of high concentration of activating ligand (L) on the target cell relative to

inhibiting ligand, granule exocytosis could be triggered resulting in cytolysis, despite

engagement of the inhibiting receptor.

Page 47: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

27

1.4.2.1 Granule exocytosis

The major mechanism of NK cell cytotoxicity is via exocytosis of granules containing perforin

and granzymes, first characterized by Podack et al., whereby they isolated T-cell granules,

identified protein components and demonstrated cytolysis(Podack and Konigsberg 1984)

(reviewed by Henkart et al.)(Henkart 1985). Following an encounter with another cell, an NK

cell will form an immunological synapse (IS).(McCann, Vanherberghen et al. 2003) If there is

sufficient activation signals on the potential target cell and lack of inhibitory signals, this will

trigger a cytolytic response. This requires cytoskeletal rearrangement and re-orientated of the

granules to the IS, followed by fusion into the synapse, and subsequent contact of granule

contents with the target cell membrane.

Granules contain perforin, first described by Dennart and Podack et al., which can create pores in

membranes of target cells following cytolytic effector degranulation.(Dennert and Podack 1983;

Podack and Dennert 1983), and have a structure similar to complement component

C9(Lichtenheld, Olsen et al. 1988). Serine proteases termed granzymes are also contained

within granules, with Granzyme A being the first characterized member of this family in T cell

granules.(Masson, Zamai et al. 1986; Masson and Tschopp 1987) Granzymes are facilitated

entry by perforin, where they are able to initiate apotosis by both caspase-dependent and

independent pathways. Perforin is a 70 Kda protein that requires free calcium and neutral pH to

optimally integrate into the target membrane.

Perforin knockout mice have been developed that have normal numbers of CD8+ T cells and NK

cells, but lack cytolytic function against allogeneic, virally infected, or tumour targets.(Kagi,

Ledermann et al. 1994) Further, perforin knockout mice have impaired control of tumour

progression in three different models of cancer (syngeneic tumour cell line, carcinogen exposure

and oncogenic virus), implicating T and NK cells in tumour immune surveillance.(van den

Broek, Kagi et al. 1996) In this study, unprimed wild type mice were able to mediate anti-

tumour effects, suggesting a role for NK cells, which lack the requirement for priming to

effectively mediate cytolysis. Finally, antibody-targeted depletion of NK cells in mouse studies

have supported the results from perforin knockout mice, showing NK cell-deficient mice are

more susceptible to 3-methylcholanthrene-induced sarcomas than wild-type mice(Smyth, Swann

et al. 2005) thus supporting NK cells as relevant in tumour immune surveillance.

Page 48: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

28

Perforin deficiency caused by either a homozygous nonsense or missense mutation leads to

familial hemophagocytic lymphohistiocytosis (type 2) in humans, accounting for 60% of

cases.(Stepp, Dufourcq-Lagelouse et al. 1999) Granule exocytosis is dependent in part on

Munc13-4, a granule trafficking protein. Mutation in Munc13-4, has been shown to present in at

least 20% of familial hemophagocytic lymphohistiocytosis patients designated as type

3.(Feldmann, Callebaut et al. 2003)

Six granzymes have been identified in humans; A, B, H, K, M and tryptase-

2/granzyme3.(Hameed, Lowrey et al. 1988) Granzymes have one of four substrate specificities

cleaving after particular amino acids: tryptase (Arg or Lys), Asp-ase (Asp), Met-ase (Met or

Leu), and chymase (Phe, Tyr, or Trp).(Kam, Hudig et al. 2000) Granzyme B tends to cleave

proteins following aspartic acid residues in the substrate P1 position. Granzyme A, is a tryptase,

which cleaves substrates with basic residues in the P1 position (e.g arginine). The other human

granzymes have other enzymatic functions: H (chymase), K (tryptase) and M (metase).(Kam,

Hudig et al. 2000).

Granzyme B induces DNA damage through activation of caspase activity and has been shown to

partially process procaspase 3, which requires release of other proapoptotic factors from the

mitochondria to complete apoptosis.(Sutton, Wowk et al. 2003) By contrast, granzyme A is

unable to activate caspases, but instead targets nuclear proteins directly to induce DNA single-

stranded DNA breaks and fragmentation by a caspase-independent pathway.(Beresford, Xia et al.

1999) These nuclear proteins include histones, lamins, and DNA damage repair proteins, Ku70

and PARP-1.(Lieberman 2010) Granzyme A has also been shown to target mitochondria and

cause non-apoptotic death by cleaving the complex I protein NDUFS3, and ultimately generating

superoxide anions(Martinvalet, Dykxhoorn et al. 2008). Granzyme knockout mice do not have

significant immunodeficiency or decreased cytotoxicity of T- and NK cells in vitro, indicating

redundancy, and supporting a primary role of perforin in cytolysis of lymphocyte effectors.

Page 49: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

29

1.4.2.2 Tumour necrosis factor family mediated cytotoxicity

The tumour necrosis factor family in humans has been well studied, with 27 members as of

2013(Aggarwal 2003; Bremer 2013). All are capable of binding ligands present on target cells to

induce apoptosis (extrinsic pathway) and are expressed primarily by NK and CTL. TNF-α and

TNF-β were the first discovered members of this family. Most TNF ligands are type II

transmembrane proteins containing an extracellular domain that can be cleaved by

metalloproteinases to generate soluble cytokines, which can mediate cytotoxic effects. The most

relevant effectors of cytotoxicity are Fas ligand and TNF-related apoptosis inducing ligand

(TRAIL). Major roles for FasL and TRAIL have been shown for tumour immunity. Fas ligand

is a type II transmembrane protein, which also has a secreted soluble form (Tanaka et al., 1996)

expressed on both cytotoxic CD8 T-cells and NK cells. Binding by Fas ligand of target Fas

allows for calcium-independent cytotoxicity against a range of tumour cells. This is typically

demonstrated by using EGTA, a calcium chelator, when conducting cytotoxicity assays.

Residual killing in calcium-free conditions is attributed to ligand-mediated killing.

While granule exocytosis models of cell death have focused primarily on perforin and

granzymes, FasL has been found to be localized to the outer membrane of cytoplasmic granules,

implicating it in granule mediated cytotoxicity.(Kojima, Kawasaki-Koyanagi et al. 2002) This

finding might also provide a mechanism of control over ligand mediated killing which must be

regulated, otherwise normal cells expressing Fas could be killed by casual contact with immune

effectors bearing FasL. However, conventional approaches to inhibit granule exocytosis have

demonstrated that ligand mediated killing occurs in the absence of granule exocytosis,

questioning the proposed model of Kojima et al.

Fas (CD95), the binding partner for Fas Ligand, is present on a wide range of normal tissues

such as liver as well as on malignant cells. Cytotoxic T-lymphocytes (CTL), in the absence of

perforin, granule exocytosis and calcium, were shown to be cytotoxic against L1210 cells

transfected with Fas, providing early evidence for a ligand mediated killing pathway.(Rouvier,

Luciani et al. 1993) Fas Ligand binding of Fas leads to trimerization of Fas, which causes

aggregation of death domains in the cytoplasmic region of the receptor, and subsequent

Page 50: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

30

signalling via the adaptor molecule FADD (Fas associated death domain), ultimately leading to

apoptosis.

TRAIL is also expressed by NK and CTL, and depends on FADD-dependent signalling(Kuang,

Diehl et al. 2000). In addition to the apoptosis-signalling TRAIL receptors, DR4 and DR5, there

are receptors that lack a functional death domain, thought to serve as a ‘decoy’ mechanism to

regulate TRAIL-mediated apoptosis. TRAIL is also upregulated on NK cells following exposure

to IL-2, IL-15 and IFNs.(Smyth, Cretney et al. 2005; Smyth, Swann et al. 2005)

The physiological function of apoptosis by death ligands and receptors primarily functions as

mechanism of homeostatic regulation of lymphocyte and other hematopoietic cell populations.

The generation of FasL (gld) and Fas (lpr) double-knockout mice has demonstrated the

important role of death-receptor mediated pathways of cytotoxicity (Watanabe-Fukunaga,

Brannan et al. 1992; Takahashi, Tanaka et al. 1994). FasL -/- (gld) and Fas -/- (lpr) mice

develop autoimmune nephritis and other features analogous to human systemic lupus

erythematosus (SLE) (Matiba et al., 1997). The lymphoproliferation and autoantibody

production displayed in the lpr and gld mice confirms their role in the control and depletion of

‘self’ reactive lymphocytes. Fas receptor mutations occur in humans as well, leading to an

autosomal lymphoproliferative disorder(Fisher, Rosenberg et al. 1995; Rieux-Laucat, Le Deist et

al. 1995).

TRAIL-deficient mice were generated and had a more rapid onset of fibrosarcoma formation in

response to exposure to MCA, a carcinogen. Further, infusion of TRAIL-blocking antibodies

into wildtype mice revealed an anti-metastatic role for this receptor in the Renca tumour

metastases.(Cretney, Takeda et al. 2002) Also, TRAIL deficient mice have less ability to

mediate graft-versus-tumour effects in experimental murine models of transplantation.(Schmaltz,

Alpdogan et al. 2002)

TRAIL is constitutively expressed on murine NK cells in the liver and can suppress tumor

metastasis. Administration of anti-TRAIL blocking antibody increased experimental liver

metastases of TRAIL-sensitive tumor cell lines. This anti-metastatic effect of TRAIL was not

observed in NK cell-depleted or IFN-γ-deficient mice, which lack TRAIL on NK cells derived

Page 51: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

31

from the liver.(Takeda, Hayakawa et al. 2001) NK cell control of Renca carcinoma hepatic

metastases in the liver was partially TRAIL-dependent as evidenced by administration of IL-12

which upregulated TRAIL expression on liver, spleen, and lung NK cells.(Smyth, Cretney et al.

2001)

1.4.2.3 NK cell recognition receptors

1.4.2.3.1 NK adhesion molecules

Prior to engagement of formal recognition receptors, NK cells must adhere to cells that they are

probing for abnormal cell surface antigens. This is accomplished by several receptors shared

with other lymphocytes including CD2, CD11a, CD18, CD54 and CD58. Resting human NK

cells express, lymphocyte function antigen-1 (LFA-1) (CD11a/CD18), LFA-3 (CD58) and

intercellular adhesion molecule-1 (ICAM-1)(CD54) which increase after incubation with IL-2.

Increases in NK cell adhesion molecule expression was associated with enhanced formation of

E:T cell conjugates and cytotoxicity, which could be partially inhibited by blocking CD2,

CD11a, or CD54 with specific antibodies.(Robertson, Caligiuri et al. 1990)

1.4.2.3.2 NK cell inhibitory receptors

NK cells express a variety of inhibitory receptors that regulate their cytotoxicity against normal

tissue. There are two major families of MHC-specific inhibitory receptors in humans. The first

is the Ig superfamily, which includes the killer immunoglobulin Ig-like receptors (KIRs) and

leukocyte Ig-like inhibitory receptors (LIRs). The second is the C-type lectin-like receptor

superfamily (Carretero et al., 1998; Colonna and Samaridis, 1995; Raulet et al., 2001). Each NK

cell expresses multiple receptors in various combinations, which leads to subpopulations of NK

cells able to detect the loss of MHC class I proteins (Gazit et al., 2004; Moretta et al., 1996). The

major HLA class I-specific NK inhibitory receptors in humans are listed in Table 1.2.

Page 52: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

32

Table 1.2: NK cell inhibiting receptors

Name(s) CD marker Ligands

KIR

KIR2DL1 CD158a HLA-C2

KIR2DL2 CD158b HLA-C1

KIR2DL3 CD158b2 HLA-C1

KIR2DL4 CD158d HLA-G

KIR3DL1 CD158e2 HLA-Bw4

KIR3DL2 CD158k HLA-A3, HLA-A11

KIR2DL5 CD158f Unknown

LIR

LIR-1, ILT-2 CD85j HLA-A, -B, -C,-G, hCMV UL18

LIR-2, ILT-4 CD85d HLA-G

C-Type lectin-like

NKG2A/CD94 CD159a/CD94 HLA-E

NKR-P1A CD161 LLT1 (CLEC2D)

1.4.2.3.2.1 Inhibitory killer immunoglobulin like receptors

The KIRs are type I transmembrane glycoproteins that are classified into two groups based on

whether there are two (KIR2D) or three (KIR3D) Ig-like domains in the extracellular region of

the protein(Moretta, Bottino et al. 1996), and possess either inhibitory or activating functions.

Inhibitory KIRs have a long cytoplasmic tail with an immunoreceptor tyrosine-based inhibitory

motif (ITIM) that delivers an inhibitory signal. The consensus sequence for an ITIM is:

V/L/IxYxxL (where V= valine, Y= tyrosine, L= leucine, I= isoleucine, and x= any amino acid).

Following KIR binding to its cognate ligand tyrosine residues in the ITIM become

phosphorylated and recruit Src homology region 2-containing protein tyrosine phosphatase

(SHP)-1and SHP-2(Burshtyn, Scharenberg et al. 1996; Olcese, Lang et al. 1996). Further work

on KIR2DL1 signalling demonstrated that β-Arrestin 2(Yu, Su et al. 2008; Bari, Bell et al. 2009)

first binds to the phosphorylated ITIM and subsequently recruits SHP-1 and SHP-2. These

phosphatases prevent or interfere with activating signals. KIR2DL1 recognizes class I HLA-C2

group and KIR2DL2 and 3 recognized class I HLA-C2 group. KIR3DL1 recognizes HLA-Bw

group and KIR3DL2 recognizes HLA A3 and A11.(Lanier 2005) The specificity of KIR2D

receptors is dependent on the presence of either a K or N residue at position 80 of the HLA-C

molecule.(Mandelboim, Reyburn et al. 1996)

Page 53: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

33

1.4.2.3.3 Leukocyte immunoglobulin-like inhibitory receptors

The leukocyte Ig-like inhibitory receptors (LIRs) have 13 family members with homology to

LIR1, an inhibitory receptor expressed on NK cells. LIR1 receptor contains four cytoplasmic

ITIMs that delivers inhibitory signals upon binding to a conserved region in MHC class I

proteins and the human cytomegalovirus class I homolog (UL18). The crystal structure of LIR-1

reveals two immunoglobulin-like domains resembling the basic structure of KIRs, but with a

different structural binding domain. (Chapman, Heikema et al. 2000). LIR-1 is able to recognize

a large range of MHC class I proteins including HLA-A, HLA-B and HLA-C with most efficient

binding to HLA-G(Colonna, Navarro et al. 1997) (Colonna, Nakajima et al. 1999).

1.4.2.3.4 C-type lectin-like receptors

CD94 and NKG2 family proteins (NKG2A, NKG2C, and NKG2E) are all type II integral

membrane glycoproteins containing C-type, carbohydrate recognition domains which covalently

assemble together to form heterodimers and recognize HLA class I allotypes(Lazetic, Chang et

al. 1996). These receptors are expressed on NK cells and a subset of CTLs (Carretero et al.,

1998). CD94 can associate with NKG2A (which has an ITIM), creating an inhibitory receptor

that binds to HLA-E (Lee, Llano et al. 1998) NKR-P1A (CD161) is a C-type lectin-like receptor

initially identified on rat NK cells and then later discovered to be expressed on human NK cells

(Lanier, Chang et al. 1994). This initial report was unable to confirm the definitive function of

NKR-P1A. A subsequent study of NKR-P1A using a rat NK cell line demonstrated activating

function against tumour targets.(Ryan, Niemi et al. 1995) However, a study of human NKR-P1A

demonstrated that lectin-like transcript-1(LLT1) was a natural ligand and LLT1 expressing

targets inhibited NK cell cytotoxicity via NKP-P1A supporting that it was an inhibitory receptor

in humans.(Aldemir, Prod'homme et al. 2005; Rosen, Bettadapura et al. 2005; Rosen, Cao et al.

2008)

1.4.2.4 NK cell activating receptors

NK cells also have a number of activating receptors that recognize ligands on target cells that can

lead to signal transduction, degranulation and target cell death. Direct evidence for the existence

of NK activating receptors was first provided by the development of monoclonal antibodies that

Page 54: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

34

could blocked the NK cell-mediated cytotoxicity.(Sivori, Vitale et al. 1997; Pessino, Sivori et al.

1998; Vitale, Bottino et al. 1998; Pende, Parolini et al. 1999)

In humans, the major NK activating receptors include NKG2D(Houchins, Yabe et al. 1991;

Pende, Cantoni et al. 2001), CD16(Mandelboim, Malik et al. 1999) and the natural cytotoxic

receptors, which include NKp46(Sivori, Vitale et al. 1997), NKp44(Vitale, Bottino et al. 1998)

and NKp30(Pende, Parolini et al. 1999). Molecular cloning of the NCRs reveals that they share

no homology with each other or other human proteins.(Moretta et al., 2002). Resting human

NK cell activation has been studied using insect cells transfected with various individual and

combinations of ligands, demonstrating that different receptors are required for adhesion,

polarization, degranulation with some being activating, co-activating or co-

stimulating.(Bryceson, March et al. 2006) Activating receptors signal via an adaptor protein

containing an immunoreceptor tyrosine based activation motif (ITAM), such as CD3ζ, FcεRIγ,

DAP10 and DAP12.(Lanier 2003) The consensus sequence for an ITAM is: YxxL/I(x)6-

8YxxL/I (where Y= tyrosine, L= leucine, I= isoleucine and x= any amino acid) The major NK

activating receptors in humans and their ligands are summarized (Table 1.3).

Page 55: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

35

Table 1.3: NK cell activating receptors

Name CD marker Ligand(s)

Activating

NKp30 CD337 heparan sulfate proteoglycans, BAT3 (BAG-6), B7-H6

NKp44 CD336 Influenza hemagluttinin

NKp46 CD335 Influenza hemagluttinin, heparan sulfate proteoglycans

DNAM-1 CD226 CD112, CD155

FcγRIII CD16 IgG (Fc portion)

NKG2C CD159c HLA-E

NKG2D CD314 MICA, MICB, ULB-1, -2, -3, -4, -5, -6, Raet-1

KIR2DS1 CD158h HLA-C2

KIR2DS4 CD158i HLA-Cw4

LEU-9 CD7 SECTM1, Galectin

Hyaluronate receptor CD44 Hyaluronan

BY55 CD160 HLA-C

Lag3 CD223 HLA Class II

2B4 CD244 CD48

CRACC CD319 CRACC

NTB-A CD352 CD352, SH2D1A, SAPPTN6, PTN1

Adhesion and

activating

Protectin CD59 C8, C9

Mac-1 CD11b ICAM-1, Fibrinogen

LFA-1 CD11a ICAM-1,-2,-3,-4,-5

LFA-2 CD2 CD58 (LFA-3)

CLEC2C CD69 Unknown

TACTILE CD96 CD155

1.4.2.4.1 NKG2D

NKG2D is a type II transmembrane glycoprotein expressed on the surface of most human and

murine NK cells with some expression on CD8+ and γδ T-cells (Eagle and Trowsdale 2007).

NKG2D has little homology to the other NKG2 gene family members (ie. A and C) and does not

form a heterodimer with CD94. Initially, NKG2D was shown to recognize the stress inducible

protein MICA, facilitating cytotoxicity by NK and T-cells.(Bauer, Groh et al. 1999)

Engagement of NKG2D activates NK cells independent of other activating stimuli (Jamieson et

al., 2002; Pende et al., 2001), leading to signalling via the adaptor molecules DAP10 (human and

murine) or DAP12 (murine), and subsequent cytotoxic response(Rosen, Araki et al. 2004). One

report has shown some capacity for human NK cells to signal via DAP12.(Karimi, Cao et al.

2005) NKG2D has several ligands identified including MICA, MICB and ULBP1–6.(Eagle and

Trowsdale 2007; Eagle, Traherne et al. 2009) These proteins have structural homology to MHC

Page 56: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

36

class I proteins and can be upregulated on cells in stress conditions. NKG2D ligands are also

induced in response to DNA damage, which can arrest cell cycle and activate DNA repair,

including in cancer cells with unstable genomes.(Gasser, Orsulic et al. 2005) NKG2D ligands

have been demonstrated on some tumours(Groh, Rhinehart et al. 1999; Diefenbach, Jamieson et

al. 2000). In particular, NKG2D has been implicated in the recognition of leukemic

blasts.(Diermayr, Himmelreich et al. 2008)

1.4.2.4.2 NKG2C

NKG2C is an activating receptor of the NKG2 family (Lazetic, Chang et al. 1996), which

associates with CD94 to form a functional resceptor, and utilizes DAP12 for signalling.(Lanier,

Corliss et al. 1998) HLA-E is the natural ligand of NKG2C(Braud, Allan et al. 1998). The

contribution of NKG2C to tumour recognition has not been clearly defined.

1.4.2.4.3 Natural cytotoxicity receptors

The natural cytotoxicity receptors (NCRs) are members of the Ig superfamily that associate with

ITAM-bearing adaptor molecules. The NCRs are involved in recognition and killing of tumour

cells and when blocked by monoclonal antibodies, reduce cytotoxicity of NK cells.(Sivori, Vitale

et al. 1997; Pessino, Sivori et al. 1998; Vitale, Bottino et al. 1998; Pende, Parolini et al. 1999)

NKp46 and NKp30 are expressed exclusively on activated and resting NK cells, while NKp44 is

upregulated after activation(Fuchs, Cella et al. 2005).

NCRs have also been linked to clinical outcome in AML, where it has been demonstrated that

many AML patients have an NCRdull

NK cell immunophenotype with lower levels of NKp30,

and NKp46, which can be reversed following successful therapy.(Fauriat, Just-Landi et al. 2007)

Further, this group demonstrated that AML blasts can directly interact with NK cells to reduce

NCR expression, demonstrating a form of immune evasion. Analysis of their cohort of 71

patients showed that patients with NK cell immunophenotypes predicted survival. Patients with

NKp30 dull vs bright NK cell populations had a 33% versus 60% overall survival, while for

NKp46 dull versus bright subgroups it was 28% versus 72%.

Page 57: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

37

1.4.2.4.3.1 NKp30

NKp30 was identified by Pende et al. in 1999 through the generation of novel murine anti-

human anti-NK cell antibodies and screening them in a reverse ADCC assay using the FcγRII

expressing P815 cell line.(Pende, Parolini et al. 1999) The gene is located in the class III major

histocompatibility complex.(Gruen and Weissman 2001) NKp30 is a 30 kDa glycoprotein that

contains one V-type Ig-like extracellular domain capable of recognition and cytoxicity of targets

that are relatively resistant to NKp46/44-mediated killing.(Pende, Parolini et al. 1999) NKp30

and NKp46 are typically co-expressed on NK cells. NKp30 has 13% identity and 15% similarity

to NKp46.(Pende, Parolini et al. 1999)

NKp30 is selectively expressed on all human NK cells (Pende et al., 1999) except those present

in the lymph nodes and in the endometrium during the menstrual cycle(Manaster, Mizrahi et al.

2008). The transmembrane portion of NKp30 contains an arginine residue, which is probably

involved in the association with CD3ζ chains for the transduction of the downstream activating

signals (Pende et al., 1999). NKp30 recognizes heparan sulfate proteoglycans (HSP) similar to

NKp46.(Bloushtain, Qimron et al. 2004) Subsequently, The nuclear factor HLA-B-associated

transcript 3 (BAT3) was demonstrated to be a ligand of NKp30 required for tumor rejection in a

multiple myeloma model.(Pogge von Strandmann, Simhadri et al. 2007) A fragment of BAT3

(amino acids 686-936) was subsequently shown to be constitute a subdomain that was essential

and sufficient to inhibit NKp30-mediated NK cell cytotoxicity.(Binici, Hartmann et al. 2013)

Another ligand for NKp30 was subsequently identified as a B7-H6, a B7 family

member.(Brandt, Baratin et al. 2009) Shedding of B7-H6 has been shown to serve as mechanism

of immune tumour evasion from NK cell-mediated killing.(Schlecker, Fiegler et al. 2014)

1.4.2.4.3.2 NKp44

NKp44 is the second NCR identified on human NK cells encoding a 44Dka surface glycoprotein

involved in cytotoxicity against MHC class I-deficient targets.(Vitale, Bottino et al. 1998;

Cantoni, Bottino et al. 1999) The simultaneous blocking of both NKp44 and NKp46 led to a

significantly increased inhibition of NK cell cytotoxicity.(Vitale, Bottino et al. 1998)

Engagement of NCRs with mAbs demonstrated NKp46 and NKp30 signalling via CD3ζ and

NKp44 signalling via DAP12(Vitale, Bottino et al. 1998; Cantoni, Bottino et al. 1999) converged

on a common signalling pathway that was distinct from that of CD16 or KIR2DS4, and was

Page 58: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

38

inhibited by engagement of CD94/NKG2A.(Augugliaro, Parolini et al. 2003) NKp44 is not

expressed on resting NK cells, but requires activation for its expression.(Vitale, Bottino et al.

1998) NKp44 recognizes viral hemagglutinnins(Arnon, Lev et al. 2001; Mandelboim, Lieberman

et al. 2001; Mandelboim and Porgador 2001) such as influenza hemagglutinin which directly

binds and activates NK cell cytotoxicity(Arnon, Lev et al. 2001; Arnon, Achdout et al. 2004)

1.4.2.4.3.3 NKp46

NKp46 is a 46 kDa glycoprotein with two C2 immunoglobulin-like domains expressed on

resting and activated human NK cells only, and when crosslinked leads to calcium mobilization,

cytotoxicity and cytokine release.(Sivori, Vitale et al. 1997; Pessino, Sivori et al. 1998) The

crystal structure of NKp46 reveals similarity to the LIR1 and KIR2D receptors.(Foster, Colonna

et al. 2003) Signalling of NKp46 is mediated via the association with the ITAM containing

adaptor molecules CD3ζ and FcεRγ.(Lanier 2003) NKp46 recognizes hemagglutinin molecules

of different influenza strains(Arnon, Lev et al. 2001; Mandelboim, Lieberman et al. 2001; Arnon,

Achdout et al. 2004). NKp46 has further been shown to recognize heparan sulfate proteoglycans

(HSP).(Bloushtain, Qimron et al. 2004)

1.4.2.4.4 NKp80

NKp80 is an 80 kDa activating homodimeric C-type lectin-like type II transmembrane protein

(similar to NKG2D) shown to stimulate NK cell cytotoxicity and induces calcium influx in

human NK cells after triggering by specific antibodies (Vitale, Falco et al. 2001). NKp80 signals

via a hemi-ITAM-like sequence with an essential tyrosine at position 7, resulting in Syk

phosphorylation required for cytotoxic responses.(Dennehy, Klimosch et al. 2011) Activation-

induced C-type lectin (AICL) was identified as a ligand of NKp80 and is expressed by

monocytes, macrophages and granulocytes.(Hamann, Montgomery et al. 1997) NKp80 mediates

NK cell cytolysis of malignant myeloid cells expressing AICL.

1.4.2.4.5 CD16

CD16 (FcγRIIIa) is a type I transmembrane receptor containing two extracellular Ig-like

domains. Most human NK cells express CD16(Trinchieri and Valiante 1993), which can

facilitate antibody-dependent cellular cytotoxicity (ADCC) by NK cells. CD16 signals via

association with the adaptor molecules CD3ζ and FcεRIγ, which contain activation ITAM

Page 59: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

39

motifs(Vivier, Rochet et al. 1991). The primary function of CD16 is to bind to the Fc portion of

IgG molecules that have opsonised cellular targets (Figure 1.4). NK cell cytotoxicity can be

specifically activated upon binding to Fcγ of IgG. This molecule therefore serves to bridge both

humoral and innate immune systems, allowing NK cells to co-ordinate with an antibody response

to a foreign antigen. CD16 was reported to be the most potent activating receptor on freshly iso-

lated human NK cells, able to elicit strong cytotoxicity and cytokine production signalling via

phospholipase-C-gamma and phosphatidylinositol-3-kinase, leading to degranulation.(Bryceson,

March et al. 2006) CD16 has also been shown to exert cytotoxicity against virally infected cells

and tumor cells, independent of its antibody binding capacity. In addition to IgG Fc specificity,

a cell surface CD16-specific ligand has been demonstrated through use of a CD16-Ig fusion

protein assay system, but this molecule is yet to be isolated and identified.(Mandelboim, Malik et

al. 1999)

Figure 1.4: NK cell antibody dependent cell mediated cytotoxicity

Engagement of the Fc portion of a target bound antibody by Fcγ receptor IIIA (CD16) on NK

cells leads to signal transduction and granule exocytosis.

Page 60: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

40

1.4.2.4.6 Activating killer immunoglobulin like receptors (KIRs)

There are six genes encoding for the KIRs clustered on chromosome 19(Lanier 2005) that lead to

an activating signal when bound to class I HLA(Moretta, Sivori et al. 1995) Activating KIRs

are type I transmembrane glycoproteins that consist of either two (KIR2D) or three (KIR3D)

extracellular C2-type Ig-like domains. However, they possess a charged amino acid in their

transmembrane domain and contain a short cytoplasmic tail, without any known signalling motif.

(Biassoni, Cantoni et al. 1996) Instead, they are associated (via the charged amino acid) with the

ITAM-containing signalling protein, DAP12. KIR2DS1 recognizes HLA-C2 while KIR2DS2

recognizes HLA-C1. KIR2DS4 recognizes HLA-C and a non-MHC class I protein expressed on

melanomas, (Katz, Gazit et al. 2004) resulting in enhanced NK killing. KIR3DS1 recognizes

HLA-B Bw4 while KIR2DS3 and KIR2DS5 have unknown ligands.

1.4.2.4.7 2B4

2B4 is an activating cell surface glycoprotein related to CD2 found on human NK and T-

cells(Lanier 2005), whose ligand was shown to be CD48(Brown, Boles et al. 1998). It contains

two immunoglobulin-like external domains and its cytoplasmic tail contains four

immunoreceptor tyrosine-based switch motifs (ITSM). The ITSM consensus sequence is:

TxYxxV/I (where T= threonine Y= tyrosine, V= valine, I= isoleucine and x= any amino acid).

The ITSM motif defines a family of receptors sharing a common signalling pathway and

includes the NTB-A and CRACC receptors that are also expressed by NK cells and are also able

to activate their killing.(Veillette 2006)

Upon phosphorylation of the tyrosine in the ITSM motif, 2B4 binds to the SAP or EAT-2(Perez-

Quintero, Roncagalli et al. 2014), intracellular adaptor proteins, or to the Src homology 2

domain-containing protein tyrosine phosphatase (SHP)-1 and SHP-2 tyrosine phosphatases. 2B4

is capable of triggering effector cell functions when bound by antibody or its natural ligand

CD48.(Nakajima, Cella et al. 1999; Sivori, Parolini et al. 2000) Further, transfection of CD48

into NK-resistant target cells can render them sensitive to NK cell-mediated cytotoxicity and

lead to secretion of IFN-γ following effector and target cell contact. (Tangye, Cherwinski et al.

2000) SAP mutations in humans leads to dysfunctional 2B4 mediated activation of NK cells and

Page 61: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

41

switching the receptor to an inhibitory function(Nakajima, Cella et al. 2000; Parolini, Bottino et

al. 2000), leading to the X-linked lymphoproliferative disorder.(Nakajima and Colonna 2000)

1.4.2.4.8 DNAM-1

DNAM-1 (CD226) is a member of the Ig superfamily, is expressed by human NK cells, T cells, a

subset of B cells, monocytes and platelets,(Lanier 2005) and recognizes CD155 (polio virus

receptor) and CD112 (nectin-2).(Bottino, Castriconi et al. 2003) Interactions between DNAM-1

on NK cells and its ligands on tumour targets facilitate NK cell–mediated cytotoxicity and

cytokine production.(Bottino, Castriconi et al. 2003; Tahara-Hanaoka, Shibuya et al. 2004)

Moreover, it was shown that the interaction of DNAM-1 with CD112 and CD155 contributes to

the NK-mediated lysis of dendritic cells (Pende et al., 2006).

1.4.3 NK cell lines

Eleven true NK cell lines have been established(Drexler 2010): HANK-1 (Nasal-type NK/T-cell

lymphoma)(Kagami, Nakamura et al. 1998), IMC-1, KHYG-1 (Aggressive NK-cell leukemia)

(Yagita, Huang et al. 2000), MEC04, NK-92 (NHL with LGL cells)(Gong, Maki et al. 1994),

NK-Y (Nasal NK-cell lymphoma)(Tsuchiyama, Yoshino et al. 1998) NKL (NK-LGL

leukemia)(Robertson, Cochran et al. 1996), SNK-1, SNK-6 (Nasal NK/T-cell

lymphoma)(Nagata, Konno et al. 2001), SRIK-NKL and YT (ALL + thymoma) 1983(Yodoi,

Teshigawara et al. 1985). Six of these NK cell lines were further characterized by Drexler et al.

in 2000 (HANK1, KHYG-1, NK-92, NKL, NK-YS and YT) (Drexler and Matsuo 2000)

showing that five of these cell lines were IL-2-dependent with a common immunophenotype:

CD1-, CD2+, CD3-, CD4-, CD5-, CD7+, CD8-, CD16-, CD56+, CD57-, T-cell receptor (TCR)-.

It is notable that transformation led to a loss of CD16 expression in all these cell lines which is a

hallmark of endogenously derived NK cells that allows for antibody-dependent cell-mediated

cytotoxicity (ADCC). SNK-6 was later characterized by Matsuo et al. as another bona fida NK

cell line.(Matsuo 2003) Using a calcein cytotoxicity assay, only NK-92, KHYG-1 and SNT-8

showed significant cytotoxicity against K562 targets.(Matsuo 2003) Thirty-five cell surface

markers were assessed on these seven NK cell lines to establish a common profile and variations

between NK cell lines.(Matsuo 2003) KHYG-1 was the only NK cell line notably positive for

CD8 and strongly positive for CD158a (KIR2DL1) and CD158b (KIR2DL2).

Page 62: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

42

1.4.3.1 NK-92

NK-92 is an IL-2 dependent cell line derived from a patient with a NK-cell non-Hodgkin

lymphoma with a CD56+CD3-CD16- immunophenotype, which retained cytotoxic antitumour

activity, consistent with activated NK cells(Gong, Maki et al. 1994). Potential use of NK-92 as a

purging agent in preparing autologous transplant products had been conducted using a neomycin-

resistant K562 line to spike normal PBMCs with 10% K562 and then incubate with NK-92 and

measure colony growth in methylcellulose.(Klingemann, Wong et al. 1996). NK-92 was highly

cytotoxic against K562 CFU. It was further shown that irradiation of NK-92 with 1000 cGy

inhibited proliferation, but no significant impact on cytotoxicity.(Klingemann, Wong et al. 1996)

Also, cytotoxicity of these irradiated cells persisted for 2 days after IL-2 deprivation, but then

rapidly decreased. Finally, NK-92 had no impact on CFU-GEMM, BFU-E or CFU-C from

normal hematopoietic cells.

NK-92 has been demonstrated to have enhanced cytotoxicity over endogenously derived NK

cells against a variety of human leukemia cell lines and primary leukemic blasts in vitro (Yan Y

et al. 1998). Further, non-irradiated NK-92 can impact survival in a primary AML xenograft

SCID model, which can be enhanced by the administration of IL-2 to mice. Non-irradiated NK-

92 cells (20x106 x 5 doses) were not able to engraft in SCID mice or cause disease.

1.4.3.2 Gene-modified variants of NK-92

Several gene modified variants of NK-92 have been generated to enhance its function as a

therapeutic agent or for basic studies of NK cell function. The approach to introduction of new

genes has been either from electroporation or lentiviral transduction. These include low and high

affinity CD16+, IL-2, KIRs and chimeric antigen receptors.

1.4.3.3 CD16+NK-92

NK cells typically express CD16 and are able to engage in antibody-dependent cell-mediated

cytotoxicity (ADCC) against antibody-coated targets ,allowing for a bridge between the adaptive

and innate immune responses. NK-92 lost CD16 expression during malignant transformation,

making it incapable of ADCC. Two different major CD16 alleles occur in humans, a low (176F)

and high (176V) affinity, each with a particular amino acid at position 176. The distribution of

Page 63: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

43

CD16 alleles in the populations is is 0.56 (176F) and 0.44 (176V) yielding a genotypic

distribution of 61% V/F, 26% F/F and 13% V/V.(Wu, Edberg et al. 1997)

However, gene modified variants of NK-92 were generated using a retroviral system (pBMN-

IRES-EGFP vector), whereby both the low and high affinity Fc gamma receptor (CD16) were

transduced into parent line NK-92 with and without green fluorescence protein (GFP).

Individual clones have different integration sites of the retroviral expression vector. These

CD16+NK-92 cells expressing either low (NK-92.176F and NK-92.176F.GFP) or high (NK-

92.176V and NK-92.176V.GFP) affinity CD16 have demonstrated ADCC in vitro against

CD20+ targets treated with Rituximab.(Binyamin, Alpaugh et al. 2008) However, no evidence

of in vivo efficacy has been demonstrated.

1.4.3.4 KHYG-1

Another NK cell line with therapeutic potential is KHYG-1 which was originally derived from a

patient with an NK cell leukemia with a p53 mutation.(Yagita, Huang et al. 2000) Furthermore,

KHYG-1 has a high degree of cytotoxicity against tumour targets over a range of culture

conditions, mediated in part by constitutively phosphorylated ERK2, granzyme M release(Suck,

Branch et al. 2005), and constitutively polarized granules.(Suck, Branch et al. 2006) Like NK-

92, irradiation of KHYG-1 prevents proliferation, but preserves cytotoxicity(Suck 2006), making

it a potential therapeutic agent for the treatment of cancer. A CD16+ gene modified KHYG-1

has been generated which can mediate ADCC against CD20+ target in combination with

Rituximab as low as 0.1 µg/ml).(Kobayashi, Motoi et al. 2014)

1.5 Antibody therapy for cancer

The discovery of a means to generate murine monoclonal antibodies by George Köhler and

César Milstein garnered the 1984 Nobel Prize in Medicine and paved the way for a new class of

therapeutics.(Kohler and Milstein 1975) Monoclonal antibody therapy has transformed therapy

for numerous diseases, including cancer. Initial antibodies were murine (suffix -omab) and of

limited therapeutic value, because they were rejected as foreign antigens. Chimeric

murine/human antibodies reduced the immunogenicity (suffix -ximab), but still had issues with

rejection due to residual murine elements in the antibody variable regions. Humanized

antibodies (suffix -zumab) further modified the antibodies to have a greater percentage of human

sequences, excepting the complementary determining regions. Fully human antibodies (suffix -

Page 64: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

44

umab) of a given specificity were subsequently developed using transgenic mice expressing

human immunoglobulin genes and phage display technology. Rituximab (anti-CD20 chimeric

antibody) was the first monoclonal antibody approved for use in cancer and tested

experimentally in a clinical trial for lymphoma in 1998.(McLaughlin, Grillo-Lopez et al. 1998)

NK cells play a major role in the therapeutic activity of humanized monoclonal antibodies, such

as rituximab (anti-CD20) or alemtuzumab (anti-CD52), because ADCC is the principal

mechanism of action of these mAbs (Bowles, Wang et al. 2006) There are now 30 FDA

approved therapeutic monoclonal antibodies, twelve specific to cancer therapy and four with

ADCC as a major mechanisms of action (Table 1.1).(Scott, Allison et al. 2012)

Page 65: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

45

Table 1.4: FDA approved therapeutic monoclonal antibodies for cancer*

Antibody name Target Disease Mechanism

Generic Trade

Trastuzumab Herceptin ERBB2 Breast cancer Inhibition of ERBB2 signalling and

ADCC

Bevacizumab Avastin VEGF Colon cancer Inhibition of VEGF

Cetuximab Erbitux EGFR Squamous cell

carcinoma

Inhibition of EGFR signalling and

ADCC

Panitumumab Vectibix EGFR Colon cancer Inhibition of EGFR signalling

Ipilimumab Yervoy CTLA4 Inhibition of CTLA4 signalling

Rituximab Mabthera CD20 Lymphoma ADCC, direct induction of apoptosis

and complement dependent

cytotoxicity

Alemtuzumab Campath CD52 B-cell ALL Direct induction of apoptosis and

complement dependent cytotoxicity

Ofatumumab Arzerra CD20 CLL ADCC and complement dependent

cytotoxicity

Gemtuzamab

ozogamicin

Mylotarg CD33 AML Toxic conjugate calicheamicin

Brentuximab

vedotin

Adcetris CD30 Hodgkin’s

lymphoma

Toxic conjugate auristatin

90Y-labelled

ibritumomab

Zevalin CD20 Follicular B-cell

lymphoma

Radioconjugate 90

Y

131I-labelled

tositumomab

Bexxar CD20 Follicular

lymphoma/NHL

Radioconjugate 131

I

*Adapted from Table 3, Scott et al. Nat Rev Cancer 12:284 (Scott, Allison et al. 2012)

Other non-FDA approved chimeric, humanized or fully human antibodies are at various stages of

preclinical or clinical development. The murine anti-human CD123 monoclonal antibody 7G3

has been modified into two humanized versions CSL360 and CSL362. CSL360 has the variable

region of 7G3. CSL362 has been Fc optimized to bind CD16A on NK cells with better affinity,

as well as affinity matured to better bind to CD123 by its variable region. CSL360 was tested in

a phase I clinical trial with 40 relapsed and refractory AML patients, with no major toxicities and

minimal therapeutic benefit in most patients, except two responses, one being a durable

remission (He, Busfield et al. 2014). CSL362 testing is ongoing in another phase I clinical.

Other antibodies in the development pipeline include Daratumomab (fully human anti-CD38

antibody) for multiple myeloma, Galiximab (chimeric anti-CD80 antibody) for B-cell lymphoma

and Lucatumumab (human anti-CD40 antibody) for treatment of multiple myeloma, non-

Page 66: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

46

Hodgkin’s lymphoma and Hodgkin’s lymphoma. Given that monoclonal antibody therapy is

dependent on the innate cellular immune system in many cases, it is logical to consider means to

augment the efficacy of therapeutic monoclonal antibodies in combination with an ADCC-

capable cell therapy treatment.

1.6 Cell therapy for cancer

1.6.1 Allogeneic hematopoietic stem cell transplantation

Bone marrow transplantation or allogeneic hematopoietic stem cell transplantation (HSCT) was

pioneered by E. Donnell Thomas, garnering him the 1990 Nobel prize in medicine. In 1956, he

performed the first successful HSCT for leukemia from a twin donor. Given the syngeneic

nature of the transplant, there were no issues of graft rejection.(Thomas, Lochte et al. 1957)

Subsequently, six patients were treated with unmatched HSCT, after receiving radiation and

chemotherapy. These patients ultimately died, as there was no knowledge of the issue posed by

HLA incompatibility of randomly selected donors. Further work was done on allografting bone

marrow using dogs as a model system.(Ferrebee, Lochte et al. 1958) When HLA typing became

a possibility, the first treatments of leukemia using matched sibling donors was

conducted(Buckner, Epstein et al. 1970), followed by the first matched unrelated HSCT(Clift,

Hansen et al. 1979). This work paved the way for a novel immunotherapeutic approach that

remains today the definitive therapy for many advanced and refractory leukemias.

The benefit of allogeneic hematopoietic stem cell transplantation (HSCT) is greatest for chronic

myeloid leukemia(Arora, Weisdorf et al. 2009), with some benefit for AML(Reiffers, Gaspard et

al. 1989; Woods 2001; Litzow, Tarima et al. 2010) and slight benefit in ALL(Burke, Cao et al.

2009). However, HSCT remains the only effective immunotherapy for AML that can improve

survival and cure some relapsed and refractory AML patients. Further, patients who are deemed

high risk and some intermediate risk patients are typically treated with a HSCT after completion

of chemotherapy, provided that there is a suitable matched related (optimal) or unrelated donor

(less optimal). However, Only 40% of patients will have a matched sibling donor, limiting

consistent application of HSCT (Ruggeri et al 2005). Therefore, a significant number of high

risk AML patients requiring a bone marrow transplant will not have a suitable donor. These high

Page 67: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

47

risk patients over 60 years of age without a matched donor for transplantation have a five-year

survival of approximately 5% (Frohling S et al., 2006).

The primary means by which an allogeneic HSCT treats leukemia is by application of high dose

chemotherapy and subsequent immunotherapeutic effect mediated by T-cells. This graft-versus-

leukemia effect (GVL) facilitates elimination of MRD due to alloreactivity of donor T-cells. One

other type of HSCT transplant involves a half-matched relative donor (haplotype transplant).

Without T-cell reduction, such a half-matched transplant would lead to fatal graft versus host

disease. However, by T-cell depleting and stem cell enriching the haplotype transplant product,

GVL can be minimized at the cost of increased infections post-transplant and less optimal

engraftment compared to a conventional HSCT. In this context, NK cells are able to mediate a

powerful GVL, even in the absence of T-cells. While GVL was initially thought to be

exclusively mediated by T cells recognizing tumour cells via major and minor histocompatibility

elements(Pierce, Field et al. 2001), there is growing evidence supporting an important role of NK

cells in mediating GVL(Ruggeri, Mancusi et al. 2005).

1.6.2 Adoptive immunotherapy

Adoptive immunotherapy involves the removal of cells from a patient, typically ex vivo

expansion in a sterile culture system and reinfusion into the patient. Typically, lymphocytes

derived from leukopheresis have been used and then cultured with various cytokines to enhance

the cytotoxicity of T and/or NK cells in the peripheral blood white cell populations. More

advanced protocols may enrich cell populations with magnetic cell sorting or gene modify the

cells. Several representative approaches to adoptive immunotherapy are described below.

1.6.2.1 Lymphokine activated killer (LAK) cells

The field of adoptive immunotherapy began with attempts to infuse peripheral blood derived

lymphocytes termed lymphokine activated killer (LAK) cells by Steven Rosenberg in

1985.(Rosenberg, Lotze et al. 1985) This involved development of ex vivo lymphocyte

expansion protocols using IL-2 containing growth medium to expand T and NK cell mixed

populations into a population of MHC-unrestricted LAK cells. The initial patients treated with

this approach had metastatic melanoma and some responses were seen, demonstrating proof-of-

principle. A larger randomized study of IL-2 with or without LAK cells showed a trend toward

Page 68: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

48

increased survival in the IL-2 + LAK therapy group in metastatic melanoma patients

only.(Rosenberg, Lotze et al. 1993) LAK cells were tested in patients with AML post

autologous transplant, with or without IL-2, and extended remissions were obtained in small

numbers of patients.(Fefer, Benyunes et al. 1993) To improve on this approach, tumour

infiltrating lymphocytes (TILs) were harvested and expanded in a similar fashion ex vivo with

IL-2.

Figure 1.5: Schematic of adoptive immunotherapy using LAK cells

1.6.2.2 Tumour infiltrating lymphocytes (TIL)

Following the failure of LAK cell therapy to successfully treat metastatic melanoma patients,

TILs were explored as a potentially therapeutic cell population, initially in animal models of

cancer by Rosenberg et al.(Rosenberg, Spiess et al. 1986) This required the development of

techniques to homogenize solid tissue and extract lymphocyte populations and expand them ex

vivo with IL-2 in culture. The harvest approach was refined using patient-derived renal tumours,

and the subsequently generated TILs exhibited promising cytotoxicity.(Belldegrun, Muul et al.

1988) In a subsequent trial of TIL therapy in six patients metastatic melanoma, indium-111

labelled TILs were infused and demonstrated to home back to sites of tumour using serial whole

body gamma camera imaging (Fisher, Packard et al. 1989) Use of IL-2 alone and in combination

with LAK and TIL was evaluated, with some evidence of improvement in responses with IL-2

Page 69: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

49

and TIL therapy but not with with LAK cell therapy.(Rosenberg, Lotze et al. 1989) More

recently, patients with refractory metastatic melanoma treated with a cytoreductive regimen

followed by TIL therapy had objective response rates 50-70% of cases. (Dudley, Yang et al.

2008) While not curative, this approach demonstrated the potential for cellular immunotherapy

to a impact metastatic solid malignancy.

Figure 1.6: Schematic of adoptive immunotherapy using TILs

1.6.2.3 Autologous NK cells

Adoptive immunotherapy with autologous NK cells derived from the patient has been dependent

on expansion protocols and purification techniques enabling immunotherapy with a relatively

pure NK cell population, distinguishing this approach from LAK cell therapy, where the

population originates from both NK and T cell populations in the peripheral blood. The

expansion process is similar to that for LAK cells (Figure 1.5) requiring IL-2 or IL-15, except

there is a T-cell depletion step. Autologous NK cell therapy was tested in a phase I trial with 11

metastatic colorectal cancer patients treated with 0.001-0.3x109 cells/dose iv for 1-4 doses per

cycle and up to 6 cycles with no toxicities noted.(Krause, Gastpar et al. 2004) In another trial,

seven metastatic melanoma and one metastatic renal cell carcinoma patients treated with NK cell

therapy consisting of 4.7x1010

cells in combination with chemotherapy had no toxicities noted,

Page 70: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

50

but also no therapeutic effect.(Parkhurst, Riley et al. 2011) Three other trials of autologous NK

cell therapy for a variety of malignancies are in process [reviewed in(Cheng, Chen et al. 2013)].

1.6.2.4 Allogeneic NK cells

NK cells are able to recognize and destroy cancer cells, which have downregulated HLA class I

molecules on their surface, via lack of signaling from inhibitory KIRs, provided at least one

activating receptor is engaged by the target. Ruggeri et al. were the first to demonstrate that, in

the context of a haplotype HSCT, when there is a KIR ligand mismatch between donor NK cells

and recipient HLA, a potent GVL can occur leading to improved survival for AML

patients.(Ruggeri, Capanni et al. 2002) This prompted attempts to use haploidentical allogeneic

NK cells from related donors. The first trial of haploidentical NK cells was conducted by Jeffrey

Miller’s group at the University of Minnesota in 2005 for patients with AML.(Miller, Soignier et

al. 2005) They utilized two low-dose immunosuppressive regimens to facilitate in vivo

expansion of the transferred NK cells in the outpatient setting: low dose cyclophosphamide with

methylprednisolone and fludarabine. Poor prognosis AML patients received more intense

cyclophosphamide and fludarabine regimens prior to NK cell transfer. They were able to detect

expansion of NK cells, a rise in IL-15 levels, and induction of a complete response in 5/19 poor

prognosis patients. A phase I trial of haploidentical relative donor NK cell therapy for advanced

non-small cell carcinoma (16 patients) of the lung revealed no toxicity, two partial responses and

stabilization of disease in five patients.(Iliopoulou, Kountourakis et al. 2010) A trial at St. Jude

in pediatric patients with favourable or intermediate risk AML in remission was conducted using

lower doses of cyclophosphamide and fludarabine than the Minnesota group prior to

administration of haploidentical NK cells and lower doses of IL-2 to facilitate in vivo

expansion.(Rubnitz, Inaba et al. 2010) All patients were in remission at two years post therapy.

A phase II study of haploidentical NK cells for intermediate risk AML was initiated at St. Jude

Children’s research hospital and is ongoing. Another phase I trial of haploidentical relative

donor NK cell therapy for 14 ovarian and 6 breast cancer patients was undertaken, achieving

transient donor chimerism, but limited efficacy, possibly due to Treg stimulation by IL-2

administration.(Geller, Cooley et al. 2011)

Page 71: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

51

1.6.2.5 Chimeric antigen receptor T-Cells

An evolutionary step in autologous adoptive immunotherapy has been the advent of chimeric

antigen receptor (CAR) expressing T-cells. CARs are antigen-specific receptors constructed

from the single chain variable fragment (scFv) of antibodies fused to the signalling domains of

the T-cell receptor ζ chain or FcRγ (termed first generation). The first functional CAR was

developed in 1989 (Gross, Gorochov et al. 1989) Subsequent CAR designs have included an

additional one (second generation), or two (third generation), co-stimulatory signalling domains

derived from CD28, 4-1BB or other similar molecule. CARs are then transduced into a patient’s

T-cells, typically by gamma retroviral(Scholler, Brady et al. 2012) or lentiviral(Biffi, Bartolomae

et al. 2011) transduction. Carl June’s research group developed a CD19 second generation CAR

with CD3ζ and 41BB signalling domains (Figure 1.7) used to generated CD19 CAR T-cells to

treat a CLL patient with refractory disease.(Porter, Levine et al. 2011) The patient went into

complete remission and developed a delayed tumour lysis syndrome and had a persistent B-cell

aplasia after the treatment. A follow-up study including this patient and two others confirmed

the efficacy of this approach with two achieving a complete remission with establishment of

memory CAR T-cells.(Kalos, Levine et al. 2011) A subsequent trial of CD19 CAR T-cell

therapy in five adults with refractory, MRD+ or MRD- ALL led to maintenance or conversion to

MRD- in all cases.(Brentjens, Davila et al. 2013) Four patients were subsequently treated with a

HSCT and one relapsed at 90 days. Another trial of CD19 CAR T-cells for eight patients with

follicular lymphoma, CLL and splenic marginal zone lymphoma demonstrated six partial

remissions, one complete remission and one patient with stable disease.(Kochenderfer, Dudley et

al. 2013) Four of these patients also developed B-cell aplasia.

Page 72: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

52

Figure 1.7: Design of CD19 CAR utilized in a clinical trial for CLL

Plasmid map of the CD19 CAR construct incorporating a single-chain variable fragement (ScFv)

comprised of a variable heavy (VH) and light chain (VL) from an anti-CD19 mAb, with signaling

domains (4-1BB and CD3ζ) and hinge (human CD8α) region. The plasmid is transduced into

patients T-cells using a self-inactivating lentiviral vector creating antigen specific T-cells

targeting CD19 expressing cells. (Porter, Levine et al. 2011)

Page 73: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

53

T-cell

Zeta chain + 41BB

CD 19

VL

VH

Linker

CLL cell

Figure 1.8: Schematic of CD19+ CAR T-cell recognition of CD19+ CLL cells

T-cells transduced with a CD19 specific chimeric antigen receptor can recognize and target

CD19+ targets such as CLL cells and normal B-cells.

Once a CAR T-cell product has been infused the cells can multiply to an unknown degree

limiting the significance of the starting dose and preventing modulation of cell proliferation and

activity once in circulation. Complications from CAR T-cells include a cytokine release

syndrome which can be fatal as reported for ERb2 second-generation CAR T-cell

therapy(Morgan, Yang et al. 2010) However, the cytokine release syndrome can be successfully

treated with anti-IL-6 antibody therapy, which can dampen the excess release of cytokines by

CAR T-cells. In addition, CD19 CAR T-cell therapy is not specific to leukemia cells and leads

to a permanent B-cell aplasia and state of hypogammaglobulinemia. This side effect can be

managed with life-long IV immunoglobulin treatments, but is a significant and undesired side

effect of targeting a tumour-associated antigen present on normal B-cells.

Page 74: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

54

1.6.2.6 Cell lines

The ability to create cell lines from primary tumour samples has been critical to facilitate

experimental work leading to our current understanding of tumour biology and in the

development of effective cancer therapies. Malignant transformation can occur in the cells of the

immune system such as T or NK cells, with occasional retention of the ability to recognize and

lyse other tumour cells, making these rare cell lines potential therapeutic agents. There have

been two cell lines tested in clinical trials; T-ALL104(Visonneau, Cesano et al. 2000) and NK-

92(Arai, Meagher et al. 2008; Tonn, Schwabe et al. 2013). The current approach to use of cell

line therapy requires the maintenance of a master frozen cell bank, from which doses can be

expanded in a GMP facility and infused into the patient (Figure 1.9).

Figure 1.9: Schematic of adoptive immunotherapy using cell lines

Page 75: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

55

The advantages of using a cell line versus other form of cellular immunotherapy is that they are

more standardized, expand easily with predictable kinetics and any gene modifications need only

be performed once. This is particularly important given that autologous cells from patients have

intrinsic variability and often come from patients who are immunosuppressed from

chemotherapy. Inter-patient variability in cell number, cytotoxicity and growth potential limit

current cellular immunotherapy strategies. Disadvantages include the potential for rejection of

allogeneic tissue, although there have been only occasional immune responses detectable against

NK-92. The potential for engraftment and inducing a malignancy is also present, but can be

abrogated by irradiated of cells. In the future, suicide gene systems can be incorporated into cell

lines to allow for in vivo expansion of the cells and subsequent induction of cytotoxicity. The

advantages and disadvantages of cell line therapy are listed in Table 1.5.

Table 1.5: Comparison of autologous cell therapy and cell line therapy

Issue Cell therapy approach

Autologous Cell line

Cell acquisition Variable, requires leukopheresis Expanded from frozen stock

Cell expansion Variable, expansion within ~3-6

weeks

Expansion within 2-3 weeks

Cytotoxicity Variable Less variable

Gene modification Labour intensive- done for each

patient

Done once

Rejection Low potential High potential, but not observed

Malignancy risk Low potential. Transduction of

CAR T-cells has low risk of

transformation

Some potential- obviated by

irradiation in current protocols and in

the future potentially by suicide gene

insertion

In vivo expansion Yes, limited by homeostatic

regulation of lymphocytes, but

can be overcome by

preconditioning with

chemotherapy.

No, current protocols irradiated cells

preventing in vivo expansion. Could

be done with cells that are either

cytokine dependent or transduced

with suicide genes.

Memory Yes, CAR T-cell therapy can

lead to immunologic memory.

Unclear with other forms of

autologous cell therapy such as

LAK and TIL or NK.

No, cell lines cannot form memory

cells.

Page 76: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

56

1.6.2.6.1 T-ALL104

The first attempt to utilize cell lines as therapeutic agents was with cell line, TALL-104 derived

from a patient with T-cell acute lymphoblastic leukemia which had the ability to kill a wide

range of tumour targets in an MHC unrestricted manner(Cesano and Santoli 1992). TALL-104

was tested in a phase I study of fifteen patients with metastatic breast cancer (infiltrating ductal,

lobular or medullary carcinoma) (Visonneau, Cesano et al. 2000). In this study, lethally

irradiated cells were utilized at doses ranging from 106 to 10

8 cells/kg daily for five days

(induction). Nine patients had progression and were taken off study, while the remainder with

stable disease received monthly two day doses (maintenance) for up to six months. Most

toxicities were limited to grade I/II, with one patient experiencing grade IV liver toxicity from

hepatic tumor necrosis (108 cells/kg dose level), which occurred three weeks after induction.

Responses in some patients were noted, including improvement in liver metastases and reduction

of bony pain. Anti-HLA antibodies were detectable in only one patient.

1.6.2.6.2 NK-92

NK-92 has been evaluated in four completed or ongoing phase I clinical trials. The first

published trial of NK-92 therapy was a phase I study for renal cell carcinoma and melanoma

with 12 patients. Minimal toxicities were noted including one grade III fever and grade IV

hypoglycemia.(Arai, Meagher et al. 2008) The second published trial of NK-92 enrolled thirteen

patients (age, 9-71 years) with advanced solid tumors/sarcomas, and two with

leukemia/lymphoma, who received two infusions of NK-92 cells, given 48 h apart.(Tonn,

Schwabe et al. 2013) Dose escalation was done with 1x109, 3x10

9 and 1x10

10 cells/m

2 with

minimal side effects including one patient with fever and another with back pain that responded

to morphine. A third phase I is ongoing at Princess Margaret Hospital for relapsed and refractory

hematologic malignancies. Eight patients with lymphoma who relapsed after autologous

transplantation and three patients with refractory multiple myeloma have been treated in this

trial, none of whom developed serious side effects. Two of the lymphoma patients survived long

term, indicating a potential cure for patients who were otherwise incurable by intensive standard

of care therapy. A fourth trial of NK-92 for relapsed refractory AML patients has been opened at

the University of Pittsburg and is recruiting patients.

Page 77: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

57

2 Chapter 2: Hypotheses and experimental approach

2.1 Thesis aims

AML is a cancer that has been shown to be hierarchically structured with rare leukemic stem

cells at the base. These LSCs are resistant to chemotherapy and lead to relapse after

conventional therapy. Hematopoietic stem cell transplantation is an effective therapy for AML

demonstrating that a cellular immunotherapy can be effective at curing patients and thus

eliminating LSCs. Further, NK cells have been implicated in improved survival in haplotype

stem cell transplants with KIR receptor ligand mismatches. However, there is a paucity of

studies on the interaction of cellular immune effectors with cancer stem cells, leaving

unanswered questions about the sensitivity of these cells relative to bulk tumour and the

relationship between elimination of leukemic stem cells and survival. The purpose of this thesis

was to apply and develop approaches to study the interaction of natural killer cell lines with

leukemic stem cells in vitro and in vivo. Ultimately, this was to be applied to optimize a

therapeutic strategy to treat AML patients with minimal residual disease after conventional

chemotherapy who lack a suitable hematopoietic stem cell donor. There are three separate

studies in this thesis, each with hypotheses outlined below.

2.2 Hypotheses

2.2.1 Leukemic stem cells are present in cell line KG1 and are sensitive to

NK-92 mediated cytotoxicity

We hypothesized that the CD34+CD38+ cell line KG1 cell line would have a rare population of

CD34+CD38- cells that would have exclusive stem cell capacity, as determined by the ability to

self renew and differentiate into the CD34+CD38+ immunophenotype. This was based upon the

prior work demonstrating that LSCs were present in the CD34+CD38- fraction of most primary

AML samples(Lapidot, Sirard et al. 1994). Based upon one paper showing that LAK cells were

equally effective at killing primary AML LSCs and bulk leukemia, we also hypothesized that the

LSCs would be similarly sensitive to killing by NK-92. We tested this by determining the

frequency of LSCs in KG1 using several techniques, and sorted a rare CD34+CD38- fraction and

CD34+CD38+ of this cell line, then cultured over them time to determine if there was an

Page 78: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

58

exclusive stem cell compartment. We then compared the cytotoxicity of NK-92 against KG1

using the chromium release assay, flow cytometric cytotoxicity assay, proliferation assays and

clonogenic cytotoxicity assays, conducted under standardized incubation conditions to determine

impact on bulk leukemia and LSCs.

2.2.2 Primary AML leukemic stem cells have greater sensitivity to NK92

than bulk leukemia and can be targeted by CD16+NK-92 and anti-

CD123 mAb mediated ADCC in vivo

Based upon our work with cell line KG1, we hypothesized that NK-92 would preferentially kill

primary AML leukemic stem cells relative to bulk tumour. To evaluate this we utilized a

standardized incubation and simultaneous application of the chromium release assay and

methylcellulose cytotoxicity assay. We also conducted a more classical approach by cell sorting

primary AML into bulk and LSC fractions, followed by a standard chromium release assay.

Given that NK-92 is irradiated prior to administration to patients, we then addressed, for the first

time, the issue of in vivo efficacy of irradiated NK-92 in a primary AML xenograft model,

predicting that irradiated cells would have less efficacy due to lack of capacity for in vivo

expansion. We also designed an approach to target LSCs with anti-CD123 antibodies in

combination with gene modified CD16+NK-92 cells. We hypothesized that CD16+NK-92

combined with infusions of an anti-CD123 antibody would enhance cytotoxicity against LSCs in

vivo and improve survival in a primary AML xenograft model.

2.2.3 KHYG-1 has less cytotoxicity than NK-92 against leukemic targets

which can be modulated by antibody pretreatment of targets and

effectors

We then sought to compare the cytotoxicity of another NK cell line, KHYG-1, against NK-92.

We hypothesized that KHYG-1 would be less effective against leukemia than NK-92 because of

additional inhibitory KIRs expressed by KHYG-1. Previous work from our lab using RT-PCR

demonstrated more inhibitory KIR expression in KHYG-1 than NK-92. However, this mRNA

data was never confirmed by flow cytometry. We, therefore used high throughput screening

(HTS) flow cytometry to characterize a large number of markers, including activating and

inhibiting receptors on both NK-92 and KHYG-1. Subsequently, cytotoxicity against a panel of

leukemia cell line and primary AML blasts was determined, and blockade of receptors on targets

and NK cell lines conducted to elucidate mechanism of cytotoxicity of each line. Blockade of

Page 79: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

59

class I HLA on targets was conducted to determine if inhibitory KIR ligands were involved in

recognition. Unexpectedly, pretreatment of NK cell lines with putative blocking antibodies to

common NK activating receptors dramatically enhanced cytotoxicity of NK cell lines, prompting

an evaluation of the mechanism and generation of an additional hypothesis: NK cell line killing

of leukemia cells is enhanced by reverse antibody dependent cell mediated cytotoxicity via

NKp30 and NKp44 and target cell Fcγ receptor II. This hypothesis was tested using targets with

variable Fcγ receptor II expression and conducting regression analysis on target CD32

expression versus delta cytotoxicity.

Page 80: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

60

3 Chapter 3: Clonogenic assays measure leukemia stem cell

killing not detectable by chromium release and flow cytometric

cytotoxicity assays

This chapter has been published: Reprinted with minor modifications from B.A.

Williams X.-H. Wang and A. Keating. Clonogenic assays measure leukemia stem cell

killing not detectable by chromium release and flow cytometric cytotoxicity assays.

Cytotherapy 2010: 12(7);951-60. License #: 3564810548348

Contributions:

X.-H. Wang: Assisted in experimental design and execution of animal experimentation.

A. Keating: Supervised the overall research project, contributed to experimental design,

and data analysis.

Page 81: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

61

3.1 Abstract

NK-92, a permanent NK cell line, shows cytotoxicity against a variety of tumors and has been

tested in a phase I trial. We tested the toxicity of NK-92 and chemotherapy drugs against against

the stem cell capacity of the acute leukemia cell line, KG1. While the chromium release assay is

the most common method to assess cytotoxicity of immune effectors, and flow cytometry is

increasingly used, the relationship of either assay to clonogenic readouts remains unknown.

KG1 was assessed for stem cell frequency by serial dilution, single cell sorting and colony

growth in methylcellulose. KG1 was sorted into CD34+CD38+ and CD34+CD38- populations

and recultured in liquid medium or methylcellulose to determine proliferative capacity of each

fraction. Cytotoxicity of NK-92, daunorubicin and cytarabine against KG1 was measured using

the chromium release assay, flow cytometry and clonogenic assays. The culture-initiating cell

frequency of whole KG1 was between 1 in 100 to 1000 by serial dilution and single cell sorting.

Although a rare (1-3%) CD34+CD38- population could be demonstrated in KG1, both fractions

had equivalent proliferative capacity. The cumulative flow cytotoxicity assay was more

sensitive than the chromium release assay in detecting target cell killing. At a 10:1 ratio, NK-92

eliminated clonogenic capacity of KG1 that was not predicted by the chromium release assay.

Clonogenic assays provide a more sensitive means to assess the effect of a cytotoxic agent

against putative cancer stem cells within cell lines, provided that they grow well in liquid culture

medium or methylcellulose.

Page 82: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

62

3.2 Introduction

The utility of standard methodologies to assess cytotoxic agents against tumor targets have been

questioned by a growing body of literature supporting rare cancer stem cells in a number of

cancers(Lapidot, Sirard et al. 1994; Bonnet and Dick 1997; Al-Hajj, Wicha et al. 2003; Singh,

Clarke et al. 2003; Singh, Clarke et al. 2004; O'Brien, Pollett et al. 2007; Ricci-Vitiani, Lombardi

et al. 2007) The cancer stem cell hypothesis holds that only a fraction of cells within a tumour

are capable of unlimited proliferation. If true, this has profound implications for studies of

cytotoxicity against bulk tumour samples.

Acute myeloid leukemia was the first malignancy to have evidence of a stem cell hierarchy

demonstrated(Lapidot, Sirard et al. 1994; Bonnet and Dick 1997) and cancer stem cells have

subsequently been demonstrated in a variety of cancers including brain tumors(Singh, Clarke et

al. 2003; Singh, Hawkins et al. 2004), breast cancer(Al-Hajj, Wicha et al. 2003), multiple

myeloma(Matsui, Huff et al. 2004) and colon cancer(O'Brien, Pollett et al. 2007; Ricci-Vitiani,

Lombardi et al. 2007). Other work has demonstrated cancer stem cells within cell lines(Kondo,

Setoguchi et al. 2004; Matsui, Huff et al. 2004), expanding the hypothesis beyond primary

human tumours. Controversy remains, however, as to which other malignancies may be

structured hierarchically in a stem cell model, the cell of origin, as well as the frequency of

cancer stem cells and their precise immunophenotype.

KG1 is a commonly used leukemia cell line derived from a patient with erythroleukemia in

myeloblastic relapse reported as having an immunophenotype of CD34+CD38+ and to date no

cancer stem cell has been isolated or demonstrated from it. NK-92 is a cell line that has been

derived from a patient with non-Hodgkin’s lymphoma with an NK cell immunophenotype(Gong,

Maki et al. 1994) and has been demonstrated to have enhanced cytotoxicity over endogenously

derived NK cells against a variety of human leukemia cell lines and primary leukemic

blasts(Yan, Steinherz et al. 1998). The ability of these novel NK cell effectors to proliferate can

be abrogated with radiation at doses that do not affect their cytotoxicity(Yan, Steinherz et al.

1998), thereby allowing their use as clinical cellular immunotherapeutic agents which has been

evaluated in a phase I clinical trial for advanced renal cell carcinoma or melanoma(Arai,

Meagher et al. 2008).

Page 83: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

63

The implication of rare cancer stem cells potentially driving a given tumour system have

profound implications for the interpretation of assays that make an assumption of homogeneity

within a given tumour. This issue is particularly problematic for cytotoxicity assays used to

determine the effect of therapeutic agents against cancer cells. Typically, these assays focus on a

snap shot assessment of bulk tumour cells to determine whether a tumour has signs of apoptosis

(e.g. annexin V positivity), mitochondrial damage (e.g. MTT assessment of mitochondrial

activity), cell permeability (e.g. Cr release assay and PI staining in flow cytometry), or some

other readout deemed indicative of viability or cell death. Given that leukemic stem cells have

been demonstrated to have differential sensitivity to chemotherapeutic agents relative to non-

stem cells(Costello, Mallet et al. 2000), it is important to consider this as a possibility in other

systems. Where a cancer stem cell has been identified with specific cell surface markers and

validated with appropriate in vitro and in vivo assays, cell sorting can be used in conjunction

with one of the previously mentioned approaches to address toxicity against the cancer stem cell

population. However, most primary cancers and cell lines do not have a clearly identifiable

cancer stem cell population, making this approach problematic. Furthermore, even in the few

cases with well-defined cancer stem cell populations, fractionating them with cell sorting and

testing toxicity of an agent using a conventional assay such as PI and annexin V staining does

not address whether proliferative capacity has been abrogated. Although clonogenic readouts

have been used in the past to assess chemotherapydrugs(Curtis, Minden et al. 1995; Curtis,

Metcalf et al. 2000; Jacobs and Wood 2005), this approach has not been applied to the study of

immune effectors. Here, we characterize the stem cell capacity of KG1 and demonstrate

immunophenotypic heterogeneity of a putative stem cell population, which can be ablated by

NK-92 and chemotherapy drugs, as demonstrated by clonogenic assays when other conventional

cytotoxicity assays demonstrate only fractional cell kill.

Page 84: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

64

3.3 Materials and methods

3.3.1 Cell lines

KG1 and K562 was obtained from the ATCC and maintained in IMDM +20% FBS and RPMI

(RPMI, Invitrogen, Grand Island, NY, USA) +10% FBS respectively. NK-92 was originally

provided by Dr. Hans Klingemann (at the time, Rush University Medical Center, Chicago, IL,

USA), expanded and stored in liquid nitrogen and retrieved as required and maintained in GM1

medium supplemented with IL-2 (450 U/mL) (Chiron, QC, Canada). Cellular cytotoxicity

assays were done in GM1 medium and drug cytotoxicity assays were done in IMDM +20% FBS.

3.3.2 Antibodies and reagents

Anti-CD34 PE antibody (BD biosciences) and anti-CD38 APC (Becton Dickson) were used to

immunophenotype KG1 and cell sort fractions. Propidium iodide (Sigma) and Annexin V-FITC

(BD Pharmingen) were used at to stain apoptotic cells in Annexin V buffer (BD Pharmingen) for

10 minutes on ice. IMDM and RPMI (Sigma) were supplemented with 10% and 20% fetal

bovine serum (FBS) (Gibco), respectively. Cytarabine (Mayne Pharma (Canada) Montreal)

came as a premised solution and daunorubicin powder (Novopharma Toronto) was reconstituted

in 4 ml of distilled water prior to dilution in Iscoves Modified Dulbecco’s Medium to appropriate

concentrations. FACS buffer was made with PBS +2mM EGTA + 2% FBS.

3.3.3 Chromium release assay

Target cells were washed in 10 ml of IMDM and 2x10e6 cells were resuspended in 200 ul of

GM1 or IMDM +20% FBS (control) and treated with 100 µCi of Na251

CrO4 for 2 hours prior to

treatment and washed x2 in AIM-V serum free medium prior to treatment with NK-92 or drugs.

5000 radiolabelled KG1 were added to individual wells of a 96 well U bottom plate. NK-92 at

various concentrations were added to KG1 to yield 10:1 and 1:1 E:T ratios and plates were

centrifuged at 500 rpm and incubated at 37°C 5% CO2. Plates were incubated for 24 hr at 37°C

in a humidified atmosphere containing 5% CO2 and centrifuged at 400 g for 5 min, and 100 μl of

supernatant were collected from each well and transferred into collection tubes. The plates were

then centrifuged, supernatants collected and assayed in a gamma counter. The amount of 51

CrO4

present in supernatants was determined using a gamma counter. Percent lysis was calculated

Page 85: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

65

using the formula:% lysis = E-S/M-S x100% where E is the 51

Cr-release from an experimental

sample, S is the spontaneous release in the presence of complete IMDM medium and M is the

maximum release upon cell lysis with Triton X 100 10%. Data are presented as the mean

percent lysis of triplicate samples (+/-SD) from a representative experiment repeated 3 times.

3.3.4 Flow cytometry and cell sorting

Immunophenotyping was done using a BD LSR II (Becton Dickson) flow cytometer and cell

sorting using FacsAria (Becton Dickinson). 1 ul CD34 PE and 3 ul anti-CD38 APC alone and in

combination were used to stain 1x106 cells for 30 minutes prior to assessment by flow cytometry.

For cell sorting 5x106 KG1 cells were resuspended in 5 ml of FACS buffer and stained with

anti-CD38 APC (3 µl/million). Gates were set to define CD38- yielding ~0.01%. KG1 was

99.9% positive for CD34+ so was not sorted on this parameter, effectively yielding

CD34+CD38+ and CD34+CD38- fractions following sorting with the anti-CD38 APC antibody.

Cells were then resuspended in IDMD +20% FBS + antibiotics for long-term culture in T25

flasks or infused into methylcellulose (5000 per plate).

3.3.5 Flow cytometric cytotoxicity assay

The flow cytometric cytotoxicity assay was done with a BD LSR II flow cytometer and Coulter

FC500 using PI and Annexin V staining. Co-incubations of NK-92 and KG1 at a 1:1 ratio were

done in an identical manner to the chromium release assay (5000 targets per well) with

additional wells per treatment group pooled together for flow cytometric analysis. NK-92 and

KG1 were discriminated by expression of CD34 (KG1 positive, NK-92 negative) using an anti-

CD34 PE antibody. Cumulative cytotoxicity assays were done using flow count beads (Coulter)

to account for cell loss. Drug treatments were done in 10 ml of IMDM +20% in T25 flasks. All

co-incubations were at 37°C, 5% CO2 and 100% humidity. Cell counts and viability were

determined by trypan blue prior to assessment by flow cytometry using PI, Annexin V to

ascertain viability. Flow count bead were used to enumerate the number of cells to account for

cell loss relative to the untreated control. Cells were spun down in microfuge tubes at 2000 RPM

and resuspended in 1 ml of PBS, spun down, supernatant discarded and pellets resuspended in

Annexin V 1x buffer (800 ul total volume including PI and AV FITC). PI and AV-FITC were

added. Samples were split and run on two different flow cytometers. Flow cytometry was set up

with a doublet discriminator to exclude doublets or larger cell associations. A ‘vertical gate’ was

Page 86: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

66

used to exclude fragments on FS axis using operator judgement. 20 000 single whole cell events

assayed for viability. Because KG1 is CD34+ and NK-92 is CD34- a gate was established to

delineate them using a best fit quadrant strategy. A histogram was set up to assess PI and

Annexin V. Viability gates were based on cytarabine 10 µg/ml which yielded partial cell killing

with early apoptotic and dead cells used to define the midpoint between viable and early

apoptotic/dead cells. Beads were added to a second stained control and treatment sample

(CD34PE, PI and Annexiv V) to determine absolute cell counts relative to control. Calculations

for percent cytotoxicity (PC) used the formula: (% viable treated- % viable untreated)/ (% viable

untreated), as previously published (Suck G. et al. 2005). For corrected cumulative counts (CC)

using beads the formula was: (Corrected % viable treated)- (corrected viable

untreated)/(corrected % viable untreated %). Data are presented as the mean percent lysis of

triplicate samples (+/-SD) from a representative experiment repeated 3 times.

3.3.6 Methylcellulose and liquid reculturing cytotoxicity assays

5000 KG1 cells were added to wells of a 96 well U bottom in either 200 µl of IMDM + 10%

FBS or GM1 medium (for NK-92 treatments) and co-incubated with daunorubicin (1 µM),

cytarabine (0.1, 1 and 10 µg/ml) or NK-92 (1:1 or 10:1 ratio) for 24 hours. Cell co-cultures (200

µl) were suspended in 10 ml of liquid culture medium or methylcellulose base medium (R&D

Systems, Minneapolis). KG1 cell suspension +/- treatments were transferred into 10 ml of

IMDM +10% FBS + streptomycin in T25 culture flask and evaluated one month later by cell

counting using the trypan blue exclusion assay. Three 35 mm petri dishs per treatment were

plated with 1 ml of methylcellulose containing ~5000 KG1 cells +/- treatments and the three

plates were stored in a 135 mm petri dish and maintained in 5% C02, 100% humidity and 37C.

Controls for both methylcellulose and liquid re-culturing assays included 24 hour incubations of

KG1 in 96 well U-bottom plates under the following conditions: 1) no drug or cell treatment of

KG1 in IMDM +20% FBS (baseline growth) or GM1 medium (impact of GM1 on KG1 growth)

2) NK-92 alone in GM1 for 24 hours followed by infusion into methycellulose or IMDM +20%

FBS liquid culture (growth potential of NK-92 in methylcellulose or IMDM +20% FBS) 3) NK-

92 alone in GM1 for 24 hours followed by addition to KG1 in methylcellulose or IMDM +20%

liquid culture (impact of NK-92 on KG1 growth in methylcellulose or low density liquid

culture). Colonies (more than 50 cells) growing in methylcellulose were enumerated at one

Page 87: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

67

month and colony numbers were averaged for each treatment group and standard deviation

calculated.

3.3.7 Animals

NOD.CB17-prkdcscid

(NOD/SCID) mice were maintained in the Ontario Cancer Institute animal

facility according to protocols approved by the Animal Care Committee. Mice were fed

irradiated food and water ad libitum. Mice were irradiated with 325 cGy of irradiation prior to

injection with KG1 leukemia cells or PBS via tail vein (200 µl volume). Four mice were used

per treatment cohort to receive 104, 10

5 or 10

6 KG1. Mice were evaluated for signs and

symptoms of leukemia and sacrificed at appropriate humane endpoints.

Page 88: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

68

3.4 Results

3.4.1 Clonogenic capacity of KG1 in vitro and in vivo

To determine the stem cell frequency in KG1, several in vitro methods were used, assessing

growth in both liquid culture medium and semi-solid methylcellulose followed by an in vivo

assessment in NOD/SCID mice. KG1 was seeded using ten-fold dilutions into liquid medium

and assessed for recovery at 2 weeks and ability to perpetuate beyond one month yielding a stem

cell frequency between 1 in 100 to 1000 (Table 3.1). Subsequently, serial two-fold dilutions of

5000 KG1 (12 samples) in rows of 96 well flat bottom plates yielded complete confluence at two

weeks when 625 or more cells were plated, but not with 312 or less (Table 3.2). Further, cell

sorting was used to replicate serial dilutions with more numerical precision providing evidence

for a stem cell frequency between 1 in 250 and 1 in 1000 (Table 3.3).

Table 3.1: Frequency of KG1 stem cell frequency using liquid culture repopulation

# cells added Culture regrowth at 2 weeks and ability to perpetuate a

liquid culture >1 month

105 +

104 +

103 +

102 -

10 -

Table 3.2: Frequency of KG1 stem cell frequency using two fold serial dilutions in 96 well

confluence assay (5000 to 0.3 per well)

Cells/well Average % confluence @ 2 weeks

5000 100

2500 100

1250 100

625 100

312 70

156 20

78 10

<=39 0

Page 89: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

69

Table 3.3: Frequency of KG1 stem cell frequency using cell sorting and 96 well confluence

assay (1000 to 1 per well)

Cells/well Average % confluence @ 2 weeks

1000 100

500 100

250 70

100 60

75 60

50 60

20 50

1 0

Single cell sorting of KG1 into wells of ten 96 well flat bottom plates yielded a stem cell

frequency of 3 in 1000, in which only a small fraction led to confluence of a single well from one

cell─the majority of wells had no significant growth (data not shown). The methylcellulose

colony-forming assay was more variable, yielding colony-forming units ranging from 1 in 69 to

1 in 343. KG1 was infused into NOD/SCID mice at doses of 104, 10

5, 10

6 cells via tail vein to

determine the leukemia-initiating cell frequency within KG1. Mice infused with 105 and 10

6

KG1 developed symptoms of leukemia (weight loss, hindlimb tumors and paresis) by 3 months

and did not survive past 6 months. At a dose of 104 KG1 some mice appeared disease free for

over six months and survived as long as eight months after the initial infusion, but ultimately all

succumbed to progressive leukemia. Therefore, based on these observations, the leukemia-

initiating frequency of KG1 in NOD/SCID mice was less than 1 in 10 000 (Figure 3.1).

Page 90: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

70

Figure 1

Group10e410e510e6

Censor

80 100 120 140 160 180 200 220 240

100

80

60

40

20

0

Time

Surv

ival p

robabili

ty (

%)

Figure 3.1: NOD/SCID leukemia initiating frequency of KG1

Four NOD/SCID mice irradiated with 325 cGy were injected via tail vein with one of three dose

of KG1 (104, 10

5, 10

6) and monitored for signs of leukemia and survival with results being

plotted in a Kaplan Meier survival curve.

Page 91: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

71

3.4.2 Immunophenotyping and fractionation studies of KG1

To determine if KG1 had an immunophenotypically identifiable stem cell exclusively in the

CD34+CD38- fraction cell sorting combined with clonogenic assessments were performed. The

immunophenotype of KG1 was confirmed by two color flow cytometry to be predominantly

CD34+CD38+ with 99.9% CD34 positivity, but a rare (1-3%), previously unreported,

CD34+CD38- fraction was identified after multiple assessments (Figure 3.2).

10 100 1000 10000 1x105

<PE-A>: CD34/FAS

10

100

1000

10000

1x105

<A

PC

-A>

: CD

38

0.024 97.3

2.590.062

CD34 PE

CD

38

AP

C

Figure 3.2: Immunophenotype of KG1

1x106 KG1 were stained with anti-CD34 PE and anti-CD38 APC and assessed by two color flow

cytometry using a Facscalibur flow cytometer.

Page 92: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

72

Both the CD34+CD38+ and CD34+CD38- sorted fractions, however, regrew the culture from as

few as 1700 cells and propagated a suspension culture beyond 2 months. The expression of

CD38 from the CD34+CD38- sorted fraction was 30% CD38+ at 7 weeks and 98% positive at

ten weeks, reconstituting the original immunophenotypic distribution of CD38. At 7 weeks, the

CD34+CD38+ sorted fraction was 73% CD38+ and 27% CD38-, while at 10 weeks became 93%

CD38+ approximating the original culture immunophenotype which remained stable after being

passed through the cell sorter without sorting into fractions (Figure 3.3).

Page 93: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

73

CD34 PE CD34 PE

CD34 PECD34 PE

CD34 PE CD34 PE

Figure 3.3: Reconstitution of CD38 distribution following cell sorting KG1

5x106

KG1 cells were sorted into CD34+CD38- and CD34+CD38+ fractions using a FACsAria

cell sorter. As a control, KG1 cells were passed through the cell sorter without sorting

(unsorted). 1700 cells from the unsorted (A), sorted CD34+CD38- (B) and sorted CD34+CD38+

(C) fractions were added to T25 flask with IMDM +20% FBS. All samples reconstituted a

continuous growing culture after one month and cells were assessed by flow cytometry for CD34

and CD38 expression at 7 and 10 weeks.

Page 94: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

74

Further, seeding of sorted of CD34+CD38+ and CD34+CD38- KG1 cells into methylcellulose

yielded comparable colony formation (data not shown). Attempts to sort using other known

stem cell markers expressed on KG1 such as CD133 and CD123 were unsuccessful in

significantly enriching the stem cell capacity of KG1 (data not shown).

3.4.3 Chromium release assay (CRA) and flow cytometric cytotoxicity

assay of NK-92 and chemotherapy drugs versus KG1

Toxicity of NK-92 and chemotherapeutic agents were assessed using both the chromium release

assay and flow cytometric cytotoxicity assay, calculating both percent toxicity (cross-sectional)

and cumulative toxicity. Using the CRA, NK-92 was highly cytotoxic to KG1 at a 1:1

effector:target (E:T) ratio relative to K562 (% lysis: 53% versus 29%), but at 10:1 killing of each

target was comparable (Figure 3.4).

0

10

20

30

40

50

60

70

80

90

100

1:1 10:1

% L

ys

is

Effector Target ratio

KG1

K562

Figure 3.4: Chromium release assay of NK-92 against KG1 and K562

KG1 or K562 (5000 targets) were plated in 96 well plates and treated with NK-92 in ratios of

10:1 and 1:1 for 24 hours. % lysis results are the average of triplicate wells +/- SD. Results are

representative of three separate experiments.

Page 95: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

75

To compare with the CRA for cellular cytotoxicity, the flow cytometric cytotoxic assays were

conducted on the same day with KG1 in a 1:1 E:T ratio under identical co-incubation conditions,

and the percent cytotoxicity (PC) and cumulative cytotoxicity (CC) was determined to be 48%

and 85%, respectively (Figure 3.5).

Figure 3.5: Chromium release assay versus flow cytometric cytotoxicity assay

KG1 was treated with NK-92 in a 1:1 E:T ratio for 24 hours in 96 well U bottom plates in two

identical simultaneous experiments. The first sample was used in a chromium release assay and

% lysis is the average of triplicate wells +/- SD. The second sample set was used in a flow

cytometric cytotoxicity assay using CD34 to identify KG1 targets (CD34+) from effector cells

(CD34-) and staining with PI and Annexin V to determine viability of target cells. Percent

cytotoxicity (PC) was calculated from the viable cell counts of treated and untreated KG1

samples as outlined in the methods and cumulative cytotoxicity (CC) was determined by

adjusting the PC value using flow count beads to account for cell loss relative to control over the

24 hour incubation. Results of PC and CC are the average of two simultaneous independent

measurements of the same sample on different flow cytometers based on 20 000 events.

Page 96: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

76

The three methods were used to assess the toxicity of daunorubicin and cytarabine at several

dose levels. Daunorubicin 1 µM treatment of KG1 for 24 hours yielded cyotoxicity values of

23+/-4% (CRA), 36+/-3% (PC) and 64+/-2% (CC) and for cytarabine 1 µg/ml was 19+/-1%

(CRA), 19+/-3% (PC) 49+/-5% (CC) (Figure 3.6).

Figure 3.6: Chromium release assay versus flow cytometric drug assay

KG1 was treated with daunorubicin 1 µM and cytarabine 0.1, 1 and 10 µg/ml for 24 hours in

either 96 well U bottom plates (CRA) or T25 flasks (flow cytometry assays). The first sample

was used in a chromium release assay and % Lysis is the average of triplicate wells +/- SD.

Control and drug treated samples were prepared for flow cytometry and stained with PI and

Annexin V to determine viability of target cells. Percent cytotoxicity (PC) was calculated from

the viable cell counts of treated and untreated KG1 samples as outlined in the methods and

cumulative cytotoxicity (CC) was determined by adjusting the PC value using flow count beads

to account for cell loss relative to control over the 24 hour incubation. Results of PC and CC are

the average of two simultaneous independent measurements of the same sample on different

flow cytometers based on 20 000 events.

Page 97: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

77

3.4.4 Clonogenic and proliferation assays of NK-92 and chemotherapy

drugs versus KG1

Clonogenic assessment of KG1 following treatment with NK-92 or chemotherapy drugs were

performed using identical incubation conditions for the chromium release and flow cytometric

assays to facilitate a comparison of cytotoxic readouts. Duplicate samples of 5000 KG1 were

treated in 96 well plates with either NK-92 in 1:1 or 10:1 ratios, cytarabine (0.1, 1 and 10 µg/ml)

or daunorubicin (1 µM). After 24 hours, cell samples were resuspended in liquid culture or

plated in methylcellulose and assessed at one month. NK-92 was not able to proliferate

significantly in long term culture medium as it lacked IL-2. Cells in re-suspension cultures were

enumerated after one month (Figure 3.7): control 1: KG1 in IMDM medium (34.8x106),

daunorubicin 1 µM (0), cytarabine 0.1 µg/ml (0.9x106), cytarabine 1 µg/ml (0), control 2: KG1

in GM1 medium (38.5x106), NK-92 1:1 (27.7x10

6), NK-92 10:1 (0).

Figure 3.7: Liquid reculturing cytotoxicity assay

KG1 was incubated with or without NK-92 (10:1 and 1:1 E:T ratio), daunorubicin (1 µM) or

cytarabine (0.1, 1, 10 µg/ml) and cytotoxic drugs for 24 hours in 96 well U bottom plates

comparable to a chromium release assay setup. Cells were enumerated after three weeks using a

hemacytometer and trypan blue dye. Controls included KG1 cultured for 24 hours in IMDM

+10% FBS (IMDM) or Ex Vivo + 10% human AB serum + 450 U/ml IL-2 (GM1) , NK-92

alone. Results are representative of two separate experiments.

Page 98: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

78

Addition of NK-92 (10:1 ratio) to 5000 KG1 cells in long term suspension culture without prior

24 hour co-incubation in 96 well U-bottom plates did not significantly affect proliferation at 3

weeks, yielding 26.9x106 cells in the control vs 26.0x10

6 cells in the NK-92 treatment group.

The methylcellulose cytotoxicity assay for NK-92 vs KG1 at a 1:1 E:T ratio reduced colony

formation from 54.7 +/- 4.0 to 39.3 +/- 13.1 while at 10:1, colony formation was eliminated

(Figure 3.8). A comparable chromium release assay of NK-92 vs KG-1 at a 10:1 E:T ratio

yielded 78% lysis. All doses of cytarabine and daunorubicin tested eliminated colony formation

of KG1 in methylcellose (data not shown).

Page 99: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

79

Figure 3.8: Methylcellulose cytotoxicity assay KG1 was incubated alone (A) or with NK-92 at 1:1 (B) and 10:1 (C) ratio for 24 hours in 96 well

U bottom plates comparable to a chromium release assay setup. Subsequently cells were

transferred into methylcellulose and 5000 KG1 were transferred to each of three 35 mm plates.

Controls included KG1 alone, NK-92 alone, NK-92 infused at 10:1 ratio into methylcellulose

without prior 24 hour co-incubation in 96 well U bottom plates. Methylcellulose plates were

examined for colonies at one month and enumerated (colony definition >=50 cells).

Page 100: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

80

3.5 Discussion

The cancer stem cell hypothesis postulates that only a subpopulation of cells within a tumour

have unlimited proliferative capacity while the remainder become more differentiated with

limited proliferative capacity. We attempted to determine if KG1 had a stem cell-driven

hierarchy similar to the original findings for acute myeloid leukemia primary blasts(Lapidot,

Sirard et al. 1994; Bonnet and Dick 1997) and if killing of the putative cancer stem cell in the

line could be detected.

As an initial step we determined the stem cell frequency with several methods, including liquid

reculturing in a whole flask, 96 well plate liquid confluence assay using serial dilution and single

cell sorting, and the methylcellulose colony-forming assay, all of which yielded a similar stem

cell frequency of between 1 in 100 to 1 in 1000. The NOD/SCID repopulating capacity was <1

in 10,000 although it took 4-7 months for mice injected with 10,000 KG1 to develop symptoms

and ultimately a fatal disease. This frequency analysis established that only a rare sub-

population of KG1 has the potential to perpetutate the culture and grow in NOD/SCID mice.

Previous work has demonstrated cancer stem cells in brain tumour cells lines(Kondo, Setoguchi

et al. 2004) and multiple myeloma cell lines(Matsui, Huff et al. 2004). We therefore attempted

to determine if the stem cell capacity in KG1 is a stochastic process or is driven by a rare

population of immunophenotypically identifiable stem cells.

Although KG1 did contain rare CD34+CD38- cells (1-3%) similar to that reported for the

majority of primary human AML blast samples, both the CD34+CD38- and CD34+CD38+

fractions had unlimited proliferation capacity and the ability to recapitulate a culture with a

distribution of CD38+ cells similar to those of the original culture. Moreover, growth of the

CD34+CD38+ and CD34+CD38- fractions in methylcellulose was equivalent. A recent report

demonstrated that primary human AML blasts from some patient samples have stem cell

capacity in the CD34+CD38+ fraction, as shown by Lapidot(Taussig, Miraki-Moud et al. 2008),

which questioned the earliest report identifying the leukemic stem cell as being restricted to the

CD34+CD38- fraction(Lapidot, Sirard et al. 1994). The more recent study demonstrated that

the anti-CD38 antibody used in the original publication facilitated clearance of CD38+ stem cells

by the reticuloendothelial system and this effect could be blocked using IVIg or anti-CD122 (IL-

Page 101: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

81

2Rβ) treated immunodeficient mice(Taussig, Miraki-Moud et al. 2008). Specifically, they

showed that in seven AML patient samples, leukemic stem cells could be detected in the

CD34+CD38+ fraction, as determined by engraftment in IVIG or anti-CD122 treated

immunodeficient mice. Although, this has reopened debate about the validity of the cancer stem

cell hypothesis in AML, significant evidence remains that in primary AML, stem cells can be

enriched with anti-CD34, anti-CD123 or other antibodies that are not susceptible to the same

form of immune-mediated clearance. Other tumour systems can be enriched for cancer stem

cell populations based on the presence, rather than absence, of cell surface markers, indicating

that this clearance mechanism does not apply in all systems. However, in our studies of KG1 it

seems that the putative stem cell is contained in both the CD38+ and CD38- fractions, an

observation consistent with the recent findings of Taussig et al.(Taussig, Miraki-Moud et al.

2008). Attempts to sort other sub-populations within KG1 using anti-CD133 and anti-CD123

markers did not significantly enhance proliferative capacity.

Only one study to date has looked at the in vitro sensitivity of CD34+CD38- AML cancer stem

cells to immune effector cell killing. In that study, lymphokine activated killer cells and

allogeneic lymphocytes exerted a modest cytotoxic effect on AML cancer stem cells that were

intrinsically resistant to the chemotherapeutic agent, daunorubicin.(Costello, Mallet et al. 2000)

Given that no cell surface marker could significantly enrich stem cell capacity in KG1, to address

the effect on the cancer stem cell fraction, we turned to functional readouts of cellular

proliferation such as methylcellulose colony formation and liquid culture reconstitution, not

typically used in assessing immune effector cell cytotoxic function.

The study of immune effector cytotoxicity has predominantly focused on the chromium release

assay by measuring cell permeabilization since its initial use in the 1960s.(Brunner, Mauel et al.

1968) Previous work showed that the cross sectional assay approximates the chromium release

assay and the cumulative assay is more sensitive at detecting cell kill(Ozdemir, Ravindranath et

al. 2003). Our comparison of the chromium release assay to flow cytometric methods further

demonstrated that the cumulative cytotoxicity readout was significantly greater than both the

chromium release assay and percent cytotoxicity methods for all drugs used as well as for NK-

92. Neither of these assays however, addresses the issue of proliferative capacity, the most

Page 102: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

82

important aspect of assessing whether a cytotoxic intervention has had a true impact on the cell

population of interest.

There was partial killing of KG1 by NK-92 in 24 hr co-incubations at a 10:1 ratio as measured

by the chromium release assay, but complete ablation of the ability of 5000 cells to regrow a

liquid suspension culture or grow colonies in methylcellulose when assessed after one month.

This methodologic discrepancy is important as it demonstrates that the chromium release assay

significantly underestimated the cytotoxicity of NK-92 against the stem cell fraction in KG1.

The toxicity against the stem cell capacity of KG1 was 100% by the clonogenic and liquid

reculturing cytotoxicity assay at an E:T of 10:1 over 24 hrs while toxicity against the bulk

tumour was 78% as determined by the chromium release assay. Given that other studies of

cancer stem cells show them to be resistant to chemotherapy, this finding is important in

demonstrating that an immune effector can preferentially kill a leukemic stem cell. The

clonogenic readouts for daunurubicin 1 µM and cytarabine 0.1 µg/ml also ablated the growth of

colonies or liquid reculturing capacity. These doses are therapeutically achievable and indicate

that NK-92 can exert a powerful impact relative to agents used in standard-of-care protocols that

are potentially curative.

Here, we show that that conventional chromium release assay can be approximated with the flow

cytometric cytotoxicity assay to evaluate immune effector cell killing and drug cytotoxicity. The

sensitivity is increased when Annexin V is used in addition to propidium iodide readouts to

determine viability as early apoptotic cells can be detected as well. When counting beads are

used to convert a cross sectional assay to a cumulative one, a significantly greater amount of

killing is detected as this method accounts also for disintegrated cells that would otherwise not

be detected. Over longer co-incubations at 24 hours, the cumulative assay also measures

differential proliferation, as underscored by the low percent cytotoxicity, but high cumulative

cytotoxicity readouts observed with the cytostatic agent, cytarabine.

Although the cumulative flow cytotoxicity assay detects greater cytotoxicity compared with the

chromium release assay for both drugs and NK-92, neither method predicts the elimination of

leukemia stem cells as determined by the lack of regrowth in liquid culture or colony formation

Page 103: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

83

in methylcellulose. Thus, KG1 stem cell capacity appears to be more sensitive to immune

effector cell killing and drugs than are the majority of cells in the culture.

Although we were unable to isolate a cancer stem cell by immunophenotypic profile, we provide

evidence that clonogenic cell readouts enable the proliferative capacity of target cells to be

assessed regardless of whether or not an immunophenotypically identifiable stem cell can be

isolated. We conclude that colony-forming assays and liquid culture methods provide a more

important parameter of cytotoxic readout than the chromium release assay or flow cytometric

cytotoxicity assays.

Page 104: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

84

4 Chapter 4: Irradiated CD16+NK-92 prolongs survival

in an AML xenograft model in combination with anti-

CD123 monoclonal antibody therapy by targeting

leukemic stem cells through antibody dependent cell

mediated cytotoxicity (ADCC)

The results of this chapter are unpublished.

Contributions:

X.-H. Wang: Assisted in experimental design and execution of animal experimentation.

J. V. Leyton: Assisted in experimental design and flow cytometry of leukemic stem cell

fractions in mouse bone marrow for secondary transplant experiments.

S. Maghera: Assisted in clonogenic assays.

B. Dief: Assisted in clonogenic assays.

Page 105: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

85

4.1 Abstract

Patients with acute myeloid leukemia (AML) often relapse after initial therapy, because of the

presence of minimal residual disease containing leukemic stem cells (LSCs) that express the IL-3

receptor alpha chain CD123. We evaluated the use of NK-92, an infusible CD16- NK cell line,

against leukemic cell lines and primary AML, using bulk and clonogenic assays followed by

testing in a murine AML xenograft model. NK-92 could preferentially target LSCs over bulk

leukemia. Further, both irradiated and non-irradiated NK-92 infusions improved survival in a

primary AML murine xenograft model. To enhance therapeutic efficacy, a gene modified

CD16+NK-92 cell line expressing the high affinity Fc gamma receptor was utilized in

combination with an anti-CD123 monoclonal antibody used to coat AML targets. Cytotoxicity

of CD16+NK-92 against CD123+ AML targets by could be enhanced with the anti-CD123 mAb

7G3 in vitro. Further, combination therapy with irradiated CD16+NK-92 and 7G3 resulted in

improved survival in an AML xenograft model of leukemia relative to controls. We demonstrate

for the first time that NK-92 targets LSCs and that irradiated NK-92 treatment can improve

survival in an AML xenograft model. Finally, we demonstrate that an irradiated CD16+NK-92

cell line can be redirected to kill LSCs more effectively by ADCC both in vitro and in vivo,

leading to prolonged survival of AML xenografted mice.

Page 106: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

86

4.2 Introduction

AML accounts for a significant proportion of acute leukemias in both adults (90%) and children

(15-20%)(Hurwitz, Mounce et al. 1995; Lowenberg, Downing et al. 1999). While 70-85% of

AML patients treated with current chemotherapy protocols are able to achieve a morphologic

remission(Hurwitz, Mounce et al. 1995; Ribeiro, Razzouk et al. 2005), many relapse because of

recurrence from minimal residual disease (MRD) containing leukemic stem cells (LSCs) leading

to an overall five year survival of approximately 40%(Lowenberg, Downing et al. 1999). Acute

myeloid leukemia (AML) was the first malignancy to have evidence of a stem cell hierarchy

with enrichment of LSCs demonstrated in the CD34+CD38- fraction(Lapidot, Sirard et al. 1994;

Bonnet and Dick 1997). These LSCs express the IL-3 receptor alpha chain (CD123), a marker

not found on normal hematopoietic stem cells(Jordan, Upchurch et al. 2000). When standard

chemotherapy fails, AML can be treated successfully with bone marrow transplantation due to

the administration of higher doses of myeloablative chemotherapy and the graft-versus-leukemia

effect (GVL) derived from the donor immune system. GVL has been shown to be mediated

primarily by T-cells(Horowitz, Gale et al. 1990), but there is evidence to show that natural killer

(NK) cell mediated GVL can prolong survival in AML patients treated with haplotype bone

marrow transplantation(Ruggeri, Capanni et al. 2002; Ruggeri, Mancusi et al. 2005). Several

permanent NK cell lines have been generated with therapeutic potential and the most notable of

these is NK-92.

NK-92 is a CD16- cell line that has been derived from a patient with non-Hodgkin’s lymphoma

with an NK cell immunophenotype(Gong, Maki et al. 1994) and has been demonstrated to have

enhanced cytotoxicity over endogenously-derived NK cells against a variety of human leukemia

cell lines and primary leukemic blasts(Yan, Steinherz et al. 1998). The ability of these novel NK

cell effectors to proliferate can be abrogated with radiation at doses that do not affect their in

vitro cytotoxicity significantly(Yan, Steinherz et al. 1998). NK-92 has been tested in a two

published phase I clinical trials (Arai, Meagher et al. 2008; Tonn, Schwabe et al. 2013) and in an

ongoing clinical trial by our group for relapsed and refractory hematologic cancers (lymphoma

and multiple myeloma) with minimal toxicities. However, to prevent potential engraftment of

NK-92 in patients it is irradiated with 1000 cGy to prevent proliferation. While this dose of

irradiation does not significantly decrease in vitro cytotoxicity (Klingemann, Wong et al. 1996;

Page 107: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

87

Tam, Miyagawa et al. 1999; Tonn, Becker et al. 2001) it renders the cells incapable of in vivo

expansion. The original preclinical animal study utilizing NK-92 to treat cell line and primary

cell-induced leukemia have shown efficacy using non-irradiated NK-92 cells, which would be

capable of in vivo expansion.(Yan, Steinherz et al. 1998) Therefore, understanding the efficacy

of irradiated NK-92 cells in vivo is an open question highly relevant to clinical translation of this

approach, given that these cells are irradiated in all ongoing and completed clinical trials

utilizing NK-92 (Arai, Meagher et al. 2008).

NK cells typically express CD16 and are able to mediate antibody-dependent cell-mediated

cytotoxicity (ADCC) against antibody-coated targets, allowing for a bridge between the adaptive

and innate immune response. As NK-92 lost CD16 expression during malignant transformation,

a gene-modified variant of NK-92 was generated whereby the high-affinity allelic variant (valine

at position 176 instead of phenylalanine) of the CD16A Fc gamma receptor has been transduced

into parent line NK-92. These CD16-NK-92 cells (NK-92.176V and NK-92.176V.GFP) allow

for antibody-dependent cell-mediated cytotoxicity (ADCC) with proven efficacy in

vitro(Binyamin, Alpaugh et al. 2008), but with no testing of this gene modified cell line in vivo.

In this report, we show that NK-92 preferentially kills leukemic stem cells relative to bulk

leukemia and can prolong survival with and without radiation pre-treatment. Gene modified NK-

92 expressing the high affinity CD16 receptor (NK-92.176V.GFP) more effectively killed

CD123+ targets in vitro demonstrating a means to enhance the innate ability of NK-92 to target

LSCs. Finally, irradiated CD16+NK-92 therapy combined with 7G3 antibody therapy enhanced

survival in a primary AML xenograft model.

Page 108: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

88

4.3 Methods

4.3.1 Cell lines and primary samples

K562 was obtained from the ATCC and maintained in IMDM + 10% FBS respectively. NK-92

was originally provided by Dr. Hans Klingemann, expanded and stored in liquid nitrogen and

retrieved as required and maintained in Ex Vivo medium supplemented with 450 U/ml of IL-2

and 10% human AB serum (GM1). Five primary AML samples were obtained from the

Princess Margaret Hospital Leukemia Tissue Bank as per institutional protocol. For in vivo

studies, primary AML from a single patient derived from one of two separate visits

(080179/080315) was obtained. The AML sample was M4 with cytogenetic abnormalities

(45XY inv3 -7) with an immunophenotype positive for CD4, CD7, CD11, CD13, CD15, CD34,

HLA-DR obtained at diagnosis (080719) or first relapse (080315). NK-92 and NK-92.176V

GFP (expressing high affinity Fc gamma receptor) was obtained from Conkwest under a

Material Transfer Agreement and maintained as described for NK-92.

4.3.2 Chromium release assay

Target cells (1x106) were resuspended in 200 ul of MEM-Alpha medium and treated with 100

µCi of Na251

CrO4 for 2 hours prior to treatment and washed x2 in AIM-V serum free medium

prior to treatment with NK-92. Radiolabelled targets (1x104) were added to individual wells of a

96 well U bottom plate. NK-92 was added at various concentrations to yield 25:1, 10:1, 5:1 and

1:1 effector:target (E:T) ratios and plates were centrifuged at 500 rpm and incubated at 37°C 5%

CO2 x24 hrs. Plates were incubated for 4 hr at 37°C in 5% CO2 and centrifuged at 400 g for 5

min, and 100 μl of supernatant were collected from each well and transferred into collection

tubes. The amount of 51

CrO4 present in supernatants was determined using a gamma counter.

Percent lysis was calculated using the formula:% lysis = E-S/M-S x100% where E is the 51

CrO4-

release from an experimental sample, S is the spontaneous release in the presence of target cell

growth medium and M is the maximum release upon cell lysis with Triton X 100 10%. To

inhibit granule exocytosis calcium was chelated from medium during four hour incubations using

ethylene glycol tetraacetic acid (EGTA) and MgCl2 at a final concentration of 4 mM with 3 mM

respectively. For ADCC assays target cells were labelled with 100 µCi of Na251

CrO4 for 2 hours

prior to treatment +/-10 µg/ml of mAb prior to treatment with CD16+NK-92 in 96 well plates.

Page 109: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

89

4.3.3 ADCC chromium release assay

Target cells were labelled with 100 µCi of Na251

CrO4 for 2 hours with simultaneous incubation

with +/- 10 µg/ml of mAb (anti-class I HLA A, B, C or 7G3 or isotype control) prior to treatment

with CD16+NK-92 in 96 well plates in a standard chromium release assay as previously

described. Additional spontaneous and maximal release controls were conducted with and

without antibody pretreatment. Plates were centrifuged at 400 g and 100 μl of supernatant were

collected and assayed in a gamma counter.

4.3.4 Flow cytometry

Immunophenotyping of BM was done using an FC500 or Facscalibur flow cytometer. FACS

buffer was made with PBS + 2mM EGTA + 2% FBS. Primary AML and leukemic stem cell

fractions were detected using the following antibodies: anti-CD34 PE (BD biosciences), anti-

CD34 FITC (BD biosciences), anti-CD38 APC (Becton Dickson) and anti-CD123 PE (BD

Biosciences), anti-CD45 APC (BD Biosciences) and anti-class I HLA A, B, C (BD Biosciences)

NK-92 cells lines were assessed for CD16 expression using CD16 PE (BD Biosciences).

Leukemia cell lines were evaluated using anti-CD123 PerCy5.5 (BD Biosciences).

4.3.5 Cell sorting

Cell sorting was done using a FacsAria cell sorter. Primary AML samples were stained with

anti-CD34 PE and anti-CD38 APC (BD Biosciences). 10 million fresh primary AML blasts

were thawed, washed and resuspended in alpha-MEM +20%FBS +10% 5637 conditioned

medium. Viability was assessed using trypan blue exclusion test. Samples were then washed in

10 ml of FACS buffer (PBS, 1% FBS, 2 mM EDTA), supernatant discarded and resuspended in

2 ml FACS buffer (~5x106 cells/ml). Cells were then filtered using 5ml polystyrene round tube

with cell-strainer caps (BD Falcon). Samples were split samples into five groups to use in

setting gates: not sorted control, untreated control (ie no antibody treatment, but run through

sorter), CD34 PE control, CD38 APC control, CD34 PE + CD38 APC control. Following

antibody addition, samples were incubated at room temperature for 20-30 minutes in the dark,

washed with 10 ml of FACS buffer and resuspended in 1-2 ml of FACS buffer to yield 5 million

cells/ml. Following establishment of the CD34+CD38+ and CD34+CD38- gates, remaining

primary AML cells with double staining were sorted into these two fractions for later use a

chromium release assay as outlined previously. Purity checks were conducted to confirm the

Page 110: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

90

efficacy of the sorting procedure and viability of cells were again determined with trypan blue

exclusion assay prior prior to use in chromium release assay.

4.3.6 Methylcellulose cytotoxicity assay

Effectors and targets were incubated together in 96 well U bottom plates for four hours and spun

at 500 rpm (high density conditions), and then infused into methylcellulose (low density

conditions) and evaluated at two weeks for cell lines and four weeks for primary AML samples.

Controls included tumour alone and tumour and effector incubated in separate wells for four

hours and co-infused into methylcellulose (low density control) to determine the impact of

effectors on targets in methycellulose under low density conditions. Five primary AML samples

(10 000 cells/plate) were incubated with or without NK-92 at 25:1 E:T ratio for four hours in 96

well U bottom plates comparable to a chromium release assay setup. Subsequently cells were

transferred into methylcellulose and 10 000 whole AML blasts from each treatment group were

transferred to each of three 35 mm plates. Methylcellulose plates were examined for colonies at

2-4 weeks and enumerated (colony definition >=50 cells) and number of colonies on each of the

three plates per group were averaged and presented +/- SD. Percent clonogenic inhibition was

calculated according to the following equation:

% Colony inhibition = [(#ColoniesLDC)- (#ColoniesTreatment)] x100%

[#ColoniesLDC]

#ColoniesLDC=the number of colonies in the low density control.

4.3.7 Animals

NOD/SCID gammanull

(NSG) mice from The Jackson Laboratory were bred and maintained in

the Ontario Cancer Institute animal facility according to protocols approved by the Animal Care

Committee. Mice were fed irradiated food and Baytril containing water ad libitum during

experimental periods. Prior to infusion with AML NSG mice were irradiated with 325 or 225

cGy to facilitate engraftment. We developed a primary AML xenograft model utilizing a patient

derived AML sample outlined in the results.

4.3.8 Statistics

Survival analysis was done with Kaplan Meier survival curves using the log rank rest with

Medcalc software. Comparison of cytotoxicity data was done using two tailed student’s t-test

were done to compare in vitro cytotoxicity and engraftment data using Medcalc software.

Page 111: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

91

4.4 Results

4.4.1 NK-92 is cytotoxic against primary AML by granule exocytosis

We initially set out to determine cytotoxicity of NK-92 against primary AML using a standard

chromium release assay as a measure of bulk tumour cell kill. Primary AML was killed in a

dose-dependent manner, and in the presence of the calcium chelator EGTA, killing was

abrogated (Figure 4.1). A larger panel of five primary AML blast samples treated with NK-92

yielded a dose-dependent response and moderate degrees of cytotoxicity against 4 samples at a

25:1 E:T (% lysis): 080179 (42.3+/-3.6%), 080078 (29.8+/-3.6%), 43.9+/- 1.47%, 42.6+/-0.1%

(Figure 4.2).

-10

0

10

20

30

40

50

60

70

80

90

100

1:1 5:1 10:1 25:1

% L

ysis

Effector:Target ratio

AIM-V

EGTA

Figure 4.1: Chromium release assay of NK-92 a against a primary AML sample at a range

of Effector:Target ratios with and without calcium chelation

AML blasts (sample 080078) were labelled with 100 µCi of Na251

CrO4 prior to treatment with

NK-92 at four E:T ratios with and without calcium chelator EGTA 4 mM and MgCl2 3 mM.

Data are presented as the mean percent lysis of triplicate samples (+/-SD) from a representative

experiment done three times.

Page 112: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

92

0

10

20

30

40

50

60

70

80

90

100

080179 080078 080008 0909

% L

ys

is

Patient sample ID

1:1

5:1

10:1

25:1

Figure 4.2: Chromium release assay of NK-92 against a panel of primary AML patient

samples at a range of Effector:Target ratios

Freshly thawed primary AML blast samples were labelled with 100 µCi of Na251

CrO4 prior to

treatment with NK-92 at four E:T ratios. Data are presented as the mean percent lysis of

triplicate samples (+/-SD) from a representative experiment done three times.

Page 113: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

93

4.4.2 Chromium release assay versus methylcellulose cytotoxicity assay

We tested NK-92 against two AML cell lines in a simultaneous chromium release assay using

OCI/AML2 and OCI/AML3 as targets. As demonstrated previously with KG1 (Chapter 3),

OCI/AML3 clonogenic cells were more sensitive to NK-92 than bulk tumour. However,

OCI/AML2 bulk and clonogenic cells were equivalently sensitive to NK-92 (Figure 4.3). K562

and OCI/AML5 also showed less sensitivity in the CRA than the MCA (data not shown), making

OCI/AML2 the only exception to this phenomenon. This demonstrates that the enhanced

cytotoxicity measured in the MCA relative to the CRA for most targets is not intrinsically related

to the method of data comparison.

0

10

20

30

40

50

60

70

80

90

100

OCI/AML2 OCI/AML3

% ly

sis

or

colo

ny

inh

ibit

ion

Cell line

CRA

MCA

*

Figure 4.3: Clonogenic cytotoxicity assay of NK-92 against OCI/AML2 and OCI/AML3

NK-92 cytotoxicity was determined against cell lines OCI/AML2 and OCI/AML3 using a

methylcellulose cytotoxicity assay (MCA) and chromium release assay (CRA) both with four

hour incubations. For the CRA, OCI/AML2 and OCI/AML3 were labelled with 100 µCi of

Na251

CrO4 for 2 hours prior to treatment with NK-92 at a 25:1 E:T in 96 well U bottom plates.

For the MCA, targets cells, target cell + effector cells incubated separately and target cells and

effector cells incubated together were infused into methylcellulose. Colonies were quantified by

inverted microscopy following 10 days at 37°C incubation. CRA is presented as % lysis and

MCA results are presented as % colony inhibition. Data are presented as the mean of triplicate

samples (+/-SD) from a representative experiment repeated 3 times. *= p<0.05.

Page 114: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

94

4.4.3 NK-92 preferentially kills leukemic stem cells relative to bulk

leukemia

To determine the impact of NK-92 on LSCs we sorted a primary AML sample into

CD34+CD38- LSCs and CD34+CD38+ fractions for further testing using a CRA. Primary

AML-derived CD34+CD38- sorted LSCs were more sensitive to killing than CD34+CD38+

blasts by NK-92 in a 4 hour CRA at E:T ratios of 1:1 (58.9+/-11.5%, 20.3+/-1.7%), 5:1 (78.3+/-

9.7%, 43.5+/-11.1%) and 10:1 (72.9+/-5.6%, 38.5+/-2.4%), but this difference was not

significant at a 25:1 E:T ratio (Figure 4.4).

0

10

20

30

40

50

60

70

80

90

100

1:1 5:1 10:1 25:1

% L

ysis

Effector:Target ratio

CD34+CD38+

CD34+CD38-

Figure 4.4: NK-92 cytotoxicity against sorted leukemic stem cells (CD34+CD38-) and bulk

leukemia (CD34+CD38+)

Primary AML samples were stained with anti-CD34PE and anti-CD38APC and sorted into

CD34+CD38- and CD34+CD38+ fractions for subsequent testing in a chromium release assay.

Sorted primary AML cells were labelled with Na251

CrO4 and 10 000 labelled cells were plated

into wells of a 96 well U bottom plate and treated with NK-92 at four E:T ratios. Data are

presented as the mean percent lysis of triplicate samples (+/-SD). Data are representative of two

separate experiments.

Page 115: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

95

To test the impact of NK-92 against LSCs relative to bulk tumour, we performed a CRA with a

methylcellulose cytotoxicity assay (MCA) designed to measure the killing during the four-hour

co-incubation. To correct for the effect of NK-92 against targets during the 4 week incubation in

methylcellulose we also utilized a ‘low density control’, whereby NK-92 and targets that were

co-infused into methylcellulose without prior incubation together in the 96 well plate under

‘high density’ conditions (Figure 4.5). There was a minor inhibitory effect of colony formation

from low density incubation with NK-92 on sample 0909. Therefore, all calculations of

cytotoxicity were done relative to the low density control rather than baseline control. The MCA

showed that NK-92 at 25:1 E:T eliminated clonogenic growth of 3/5 primary AML blast samples

with minimal colony growth in 2/5 yielding % colony inhibition values of: 100+/-0%, 86.3+/-

2.3%, 98.4+/-2.8%, 100+/-0% and 100+/-0%, demonstrating much higher cytotoxicity than

obtained with the CRA, which was done on the same day (Figure 4.6).

15 ml conical tube 33 mm petri dish96 well U bottom plate

4 hrs 4 weeks

4 hrs 4 weeks

4 weeks

4 hrs 4 weeks

Tumour cell target NK-92 effector cell

Cancer stem cell target

Dead cell Colony

Control

Treatment

Low density control

Figure 4.5: Schematic of methylcellulose cytotoxicity assay

Page 116: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

96

A

Control (i) Low density control (ii) Treatment (iii)

B

0

20

40

60

80

100

120

080179 080078 08008 0909

% L

ysi

s o

r %

co

lon

y in

hib

itio

n

Sample #

CRA

MCA

Figure 4.6: NK-92 against primary AML blasts using the methylcellulose cytotoxicity assay

compared to the chromium release assay

An example of the MCA (A) shows a representative assay for one sample (080179) with a

control (AML only) (i), low density control (AML + NK-92 in methylcellulose only) (ii) and

treatment group (AML + NK-92) (iii). Four primary AML samples were incubated with or

without NK-92 at a 25:1 E:T ratio for 4 hours in 96 well U bottom plates and utilized in a

chromium release assay setup and methylcellulose cytotoxicity assay (MCA) and % lysis values

and % colony inhibition compared (B). Methylcellulose plates were examined for colonies at

two weeks and enumerated (colony definition >=50 cells) and number of colonies on each of the

three plates per group were averaged and presented +/- SD.

Page 117: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

97

4.4.4 Primary AML xenograft model

We developed a primary AML xenograft model utilizing a patient derived M4 sample with

aggressive engraftment features infused into irradiated NSG mice. The primary AML sample

was CD34+ and contained a small fraction of CD34+CD38- cells that were predominantly

CD123+ (Figure 4.7). NSG mice were irradiated with 200 or 325 cGy of irradiation followed by

immediate injection with 3x106 of freshly thawed primary AML cells or in vivo passaged

primary AML derived from the spleens of mice with end stage leukemia. This led to a

symptomatic leukemia by week 6. Leukemic progression led to significant weight loss, bone

marrow infiltration with human leukemia, anemia and splenomegaly. Further, potency of

serially transplanted BM or SPL cells from NSG with end stage leukemia was maintained and

could lead to leukemia in mice given secondary, tertiary or quaternary transplants with the same

kinetics of freshly thawed primary AML samples from the leukemia bank (Figure 4.8). Spleen

derived AML was as potent as that derived from BM at inducing leukemia (data not shown).

Figure 4.7: Primary AML immunophenotype

Primary AML 080078 was immunophenotyped using CD34 PE and CD38 APC antibodies and

CD34+CD38+ and CD34+CD38- subfractions were identified.

Page 118: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

98

Group1st passage AMLprimary AML

Survival

0 20 40 60

100

80

60

40

20

0

Time

Surv

ival pro

babili

ty (

%)

Figure 4.8: Survival of NSG mice with primary AML versus 1

st passage AML derived from

BM

NSG mice were infused with 3x10e6 freshly thawed primary AML (080791) or spleen derived

first passage AML. Mice were monitored until humane endpoint and sacrificed. Survival was

analyzed using Kaplan Meier curves and log rank test (p= NS).

Page 119: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

99

4.4.5 In vitro treatment of primary AML cells by irradiated NK-92 reduce

engraftment potential

In vitro cytotoxicity against LSCs was assessed by treating 1x10e6 first-passage BM-derived

primary AML with or without 5x10e6 iNK-92 and injecting into two cohorts mice (Figure 4.9).

At 6 weeks, mice were sacrificed and bone marrows harvested. Average leukemic engraftment in

the iNK-92 group (79.8, 3.48, 92.1, 81.3, 86.1, Av= 68.6%) was less than untreated AML

inoculated group (95.0, 93.4, 19.4, 95, 97.3, Av=80.0%), but not statistically significant

(p=0.62). Each group had a poor engrafting mouse, possibly related to an interstitial injection of

AML cells rather than directly into the tail vein, which contributed to a very high variance for

each group. Removing one poorly engrafted outlier mouse from each group yielded a higher

engraftment in the control (Average=95.1%) versus the treatment group (Av=84.8%) and was

significant (p=0.011) (Figure 4.10).

Engraftment levels were determined by proportion of cells expressing human class I HLA and

subgated on to assess the CD34+CD38- LSC fraction (Figure 4.10). iNK-92 (HLA+CD34-)

could be differentiated from AML (HLA+CD34+) using this panel and iNK-92 did not

significantly engraft in the BM (<1%) (data not shown).

Day 1

NOD/SCID IL-2Rgc-/- mice

Day X

225 cGy

+/-1000 cGy

iNK-92 Primary AML

+spin 4 hrs

Sacrifice

Inject

Figure 4.9: Schematic of in vitro cytotoxicity assay with in vivo engraftment readout

Primary AML cells (5x106) were incubated with and without iNK-92 (25x10

6) in 15 ml conical

tubes, spun down at 400 g, and incubated a 4 hours at 37° C in 5% CO2. 1x10e6 primary AML

cells +/- 5x106 iNK-92 were injected via tail vein into cohorts of five mice. At six weeks mice

were sacrificed to assess BM engraftment.

Page 120: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

100

A B

HLA +

95.0

1 0- 1

1 00

1 01

1 02

1 03

HLA

0

2 0

4 0

6 0

8 0

100

Co

un

t

Q1

0.10

Q2

96.5

Q3

3.23

Q4

0.17

1 0- 1

1 00

1 01

1 02

1 03

CD34

1 0- 1

1 00

1 01

1 02

1 03

CD

38

HLA +

79.8

1 0- 1

1 00

1 01

1 02

1 03

HLA

0

2 0

4 0

6 0

8 0

100

Co

un

t

Q1

0.19

Q2

96.6

Q3

3.02

Q4

0.19

1 0- 1

1 00

1 01

1 02

1 03

CD34

1 00

1 01

1 02

1 03

CD

38

C p=0.62

Engraftment of AML

Control Treatment0

50

100

150

% E

ngra

ftm

ent

Figure 4.10: In vitro cytotoxicity assay with in vivo engraftment readout NSG mice were sacrificed 6 weeks after injection of primary AML +/- iNK-92 BM assayed for

leukemic engraftment using anti-class I HLA antibody followed by subgating of CD34+CD38+

and CD34+CD38- subfractions. Plots are of a typical control mouse with 95.0% engraftment

(top) and therapy mouse with 79.8% engraftment (bottom) (A). Spleen sizes were enlarged in all

mice except one mouse in control and treatment groups with very low BM engraftment (B).

Engraftment of mouse bone marrow in controls versus treatment is shown (C).

Page 121: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

101

4.4.6 Irradiated NK-92 reduce leukemic stem cell fraction in secondary

transplantation assay

To assess the cytotoxic effect of irradiated NK-92 (iNK-92) on LSCs in the in vivo setting,

secondary transplantation experiments were conducted to evaluate total engraftment and fraction

of LSCs in secondary recipients. Primary AML cells (3x10e6) were also infused into two

cohorts of four mice and treated with or without iNK-92 from day 2 and given 15x10e6 cells

twice weekly to a total dose of 75x10e6. At six weeks mice were sacrificed and bone marrow

(1x10e6 cells) from each of 4 primary recipients in control or treatment were serially

transplanted 1:1 into four new NSG mice. Evaluation of BM from secondary recipients

inoculated with AML only revealed a high proportion of human CD45+ cells (80.8, 93.3, 80.4,

96.4 Av=87.7%), while one mouse from iNK-92 group was leukemia free with engraftment at

background levels of non-injected mice (96.4, 94.7, 1.8, 95.7 Av=72.2%). The difference

between average leukemic secondary engraftment between groups was not significant, because

of high sample variance in the treatment group. However, the proportion of CD34+CD38-

CD123+ cells in secondary transplanted mice for the AML group was 7.85% (8.01, 9.48, 8.66,

5.25) and for the AML + iNK-92 group was 3.66% (7.13, 3.46, 0.03, 4.00), which was

significantly lower (p=0.05) (Figure 4.11).

Page 122: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

102

A

Day 1 2

NOD/SCID IL-2Rgc-/- mice

Week 6

AML 3x106 i.v.

Sacrifice

5

225 cGy

NK-92

15x106

i.v.

12 16

+/-1000 cGy

9 19

1x106 BM cells infused into two

new cohorts of four iNSG (1:1)

Week 6

BM flow

B C

Control Treatment

0

50

100

150

% E

ng

rag

tme

nt

Control Treatment

0

2

4

6

8

10

% L

eu

ke

mic

Ste

m C

ells *

Figure 4.11: In vivo cytotoxic impact of iNK-92 on secondary BM engraftment of AML

cells and LSCs

3x10e6 AML cells were also infused into two cohorts of four mice and treated with and without

iNK-92 from day 2 and given 15x10e6 cells twice weekly to a total dose of 75x10e6 (A). BM

(1x10e6 cells) from each of 4 primary recipients in control and treatment was serially

transplanted 1:1 into four new NSG mice. These mice were sacrificed at 6 weeks and bone

marrow assayed for overall leukemic engraftment (B =% human CD45+) and LSC engraftment

(C=% human CD34+CD38-CD123+) (* p=0.05).

Page 123: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

103

4.4.7 NK-92 prolongs survival in a primary AML xenograft model

We next sought to assess the impact of NK-92 on survival of mice inoculated with primary

human AML. NSG mice injected with 3x106 primary AML cells received 10x10

6 non-irradiated

NK-92 weekly for three doses (Figure 4.12). This treatment increased median survival from 57

to 72 days (log rank test p<0.01) although most succumbed to disease ultimately (Figure 4.13A).

Autopsy revealed enlarged spleens and pale fragile bones relative to controls. Flow cytometry of

bone marrow from NSG mice innoculated with AML only (Figure 4.13B), or AML + NK-92

treatments (Figure 4.13C) that became symptomatic, had human 99% engraftment of human

leukemia detected in bone marrow, while the mouse that survived long term (~9 months) was

healthy at sacrifice and did not have evidence of leukemic infiltration in BM (Figure 4.14D) or

splenomegaly. To determine the impact of irradiation on the in vivo activity of NK-92, the cells

were irradiated with 1000 cGy prior to infusion into NSG mice inoculated 10 days before with

3x10e6 primary AML cells. NSG mice were administered 20x10e6 iNK-92 ip weekly x 5 doses

and monitored for signs of leukemia. Survival was improved in the treatment group (26 to 48

days) to near statistical significance (p=0.0566), but all mice ultimately succumbed to disease

(Figure 4.14).

NK-92 10x106 i.v.

Day 1 Day 8

NOD/SCID IL-2Rgc-/- mice

Day X

AML 3x106 i.v.

Humane endpoint sacrifice

Day 15

325 cGy

Figure 4.12: Schematic of NK-92 therapy for primary AML xenografted NSG mice

Page 124: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

104

A

GroupAMLAML + NK-92

Survival

0 20 40 60 80 100 120 140 160 180 200

100

80

60

40

20

0

Time (days)

Surv

ival p

robabili

ty (

%)

B C D

Anti-Class I HLA FITC

Figure 4.13: NK-92 therapy of primary AML xenografted NSG mice

3x106 primary AML cells were injected via tail vein into irradiated NOD/SCID gamma null

mice. 10x106 NK-92 were infused via tail vein weekly for three weeks starting on the day of

AML innoculation. Mice were monitored for signs of leukemia and sacrificed at humane

endpoints. Kaplan Meier survival curves were generated to compare survival in control and

treatment groups (P<0.01) (A). Autopsies were performed on select mice from each cohort,

including flow cytometry of BM to determine leukemic engraftment. Unstained (blue) and anti-

Class I FITC stained (red) specimens are presented from one mouse with symptomatic leukemia

(B), a mouse with AML in the treatment group that developed symptomatic leukemia (C) and a

healthy long term survivor (D).

*

Page 125: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

105

Day 0

NOD/SCID IL-2Rgc-/- mice

Day X

AML 3x106 i.v.

Death/sacrifice

10

325 cGy

iNK-92

20x106

i.p.

14 17

+1000 cGy

12 15 19

GroupAMLAML + iNK-92 IP

Survival

0 10 20 30 40 50 60 70 80

100

80

60

40

20

0

Time

Surv

ival p

robabili

ty (

%)

Figure 4.14: iNK-92 therapy of primary AML xenografted NSG mice

3x106 primary AML cells were injected via tail vein into irradiated NOD/SCID gamma null

mice. iNK-92 given ip 20x20e6 weekly x 6 were used to treat of AML xenografted mice starting

10 days after inoculation. Mice were monitored for signs of leukemia and sacrificed at humane

endpoints. Kaplan Meier survival curves were generated to compare survival in control and

treatment groups (p=0.0566)

CD

38

AP

C

Page 126: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

106

4.4.8 iCD16+NK-92 can mediate ADCC against bulk and stem cell

antigens in vitro

To develop a strategy to enhance killing of LSCs we utilized a gene-modified CD16+NK-92

transduced with the high affinity CD16 receptor (NK-92.176V GFP), which is capable of

mediating ADCC against antibody coated targets. Expression level of CD16 was 2.3% on NK-

92 and 27.9% on CD16+ NK-92 (Figure 4.15A and B). Three AML cell lines were assessed for

levels of CD123 expression by flow cytometry which demonstrated: OCI/AML2 (69.9%),

OCI/AML3 (11.3%) OCI/AML5 (31.1%) (Figure 4.14C, D and E). We opted to evaluate

OCI/AML5 further because of its considerable degree of CD123 expression, fast cycling time,

excellent colony growth in methylcellulose and its rapid engraftment capability in

immunodeficient mice allowing for in vivo studies in the future.

A B

CD16+

2.26

1 0- 1

1 00

1 01

1 02

1 03

CD16

0

2 0

4 0

6 0

8 0

100

Co

un

t

CD16+

27.9

1 0- 1

1 00

1 01

1 02

1 03

CD16

0

2 0

4 0

6 0

8 0

100

Co

un

t

C D E

CD 123+

31.1

1 0- 1

1 00

1 01

1 02

1 03

CD123

0

2 0

4 0

6 0

8 0

100

Co

un

t

CD123+

11.3

1 0- 1

1 00

1 01

1 02

1 03

CD123

0

2 0

4 0

6 0

8 0

100

Co

un

t

CD123+

69.9

1 0- 1

1 00

1 01

1 02

1 03

CD123

0

2 0

4 0

6 0

8 0

100

Co

un

t

Figure 4.15: Immunophenotyping of CD16+NK-92, NK-92 and OCI/AML 2, 3 and 5

Immunophenotyping of NK-92 (A) and CD16+NK-92 (B) after staining with with CD16 PE and

OCI/AML 2, 3 and 5 (C, D and E respectively) after staining with CD123 PerCy5.5 PE.

Samples were run on an FC500 flow cytometer. Unstained (red) and stained (blue) populations

are presented in the histogram. Gates were set to exclude 99% of unstained events for each cell

line to define positive and negative populations.

Page 127: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

107

The chromium release assay was modified to measure ADCC by coating targets with antibodies

during the 2 hour chromium incubation. Isotype control antibody (DNP) and anti-class I HLA

antibodies at 10 µg/ml were utilized to coat primary AML targets known to express high levels

of class I. CD16+NK-92 killed primary AML at E:T ratios of 25:1, 10:1, 5:1 and 1:1 (% lysis

+/-SD: 34.7 +/-4.6, 15.6 +/- 4.7, 11.5 +/- 2.0, 7.7 +/-1.6) which was significantly enhanced (2-

3x) by coating with anti-Class I monoclonal antibodies relative to isotype control (% lysis +/-SD:

64.8 +/-10.4, 31.1 +/- 8.9, 30.2 +/- 9.4, 23.9+/-2.8) (Figure 4.16).

0

10

20

30

40

50

60

70

80

90

100

25:1 10:1 5:1 1:1

None

TNP

HLA I

Figure 4.16: CD16+NK-92 in vitro ADCC assay against primary AML

Primary AML cells were labelled with 100 µCi of Na251

CrO4 for 2 hours +/- 10 µg/ml of mAb

(isotype control-DNP or anti-class I HLA) and washed x2 in AIM-V serum free medium prior to

treatment with CD16+NK-92 in 96 well plates in a standard chromium release assay. Primary

AML pre-treated with anti-Class I HLA antibody or isotype control (clone MG1-45 specific to

TNP) prior to incubation with CD16+NK-92. Data are presented as the mean percent lysis of

triplicate samples (+/-SD) from a representative experiment done two times.

Page 128: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

108

Isotype control antibody (DNP) and anti-CD123 (7G3) antibodies at 10 µg/ml were utilized to

coat OCI/AML5 targets shown to express CD123 previously. CD16+NK-92 was cytotoxic

against OCI/AML5 cells at E:T ratios of 25:1, 10:1, 5:1 and 1:1 (% lysis +/-SD: 35.0 +/-4.0,

9.0+/-6.6, -2.0 +/-0.1, 1.7 +/-3.3) and was significantly enhanced (2-6x) when targets were

coated with anti-CD123 mAb (% lysis +/-SD: 64.3 +/-3.1, 48.5 +/-4.1, 20.9 +/-0.1, 10.1+/-3.3)

(Figure 4.17).

-10

0

10

20

30

40

50

60

70

80

90

100

25:1 10:1 5:1 1:1

% L

ysis

Effector:Target ratio

None

7G3

Figure 4.17: CD16+NK-92 in vitro ADCC assay against OCI/AML5

OCI/AML 5 cells were labelled with 100 µCi of Na251

CrO4 for 2 hours +/- 10 µg/ml of mAb and

washed x2 in AIM-V serum free medium prior to treatment with CD16+NK-92 in 96 well plates

in a standard chromium release assay. OCI/AML5 was treated with anti-CD123 antibody or

isotype control prior to incubation with CD16+NK-92. Data are presented as the mean percent

lysis of triplicate samples (+/-SD) representative of three experiments.

Page 129: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

109

4.4.9 CD16+NK-92 improves survival in an AML xenograft model with

enhancement by anti-CD123 mAb therapy

To enhance the therapeutic approach of irradiated NK-92 against AML-xenografted NSG mice,

we treated mice with a CD16+NK-92 cell line, in combination with an anti-CD123 mAb (7G3),

given on the same days to facilitate targeting of leukemic stem cells by antibody-dependent cell-

mediated cytotoxicity. Due to known splenic sequestration of 7G3 in the spleen, a blocking dose

of isotype control antibody BM4 (200 µg) was given prior to the administration of 7G3 at a low

dose (8 µg) (Figure 4.18). In this pilot experiment, we demonstrated that 7G3 (8 µg) could

improve therapeutic efficacy of iCD16+NK-92, as determined by improvement of median

survival by 13 days (p=0.0173) (Figure 4.19).

Day 1 4

NOD/SCID IL-2Rgc-/- mice

Day X

AML3x106 i.v.

Death/sacrifice

5

225 cGy

iCD16+NK-92

20x106

i.p.

10 12

+/-1000 cGy

8 15

8 µg 7G3 mAb

200 µg BM4 mAb

2

Figure 4.18: Schematic of iCD16+NK-92 +/- a single anti-CD123 mAb dose for primary

AML xenografted NSG mice

Page 130: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

110

Group

CD16+NK-92

CD16+NK-92 + 7G3

Survival

0 10 20 30 40 50 60 70 80

100

80

60

40

20

0

Time (days)

Su

rviv

al p

rob

ab

ility

(%

)

Figure 4.19: iCD16+NK-92 +/- a single anti-CD123 mAb dose for primary AML

xenografted NSG mice

NSG mice were inoculated with 3x106 passage human AML spleen derived cells (day 0) and

treated with iCD16+NK-92 x 5 doses +/- 8 µg 7G3 starting on day 3. Survival was determined

using Kaplan Meier survival analysis with a log rank test (p=0.0173).

Page 131: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

111

We conducted a more rigorously controlled experiment of CD16+NK-92 and anti-CD123 mAb

therapy utilizing an isotype control antibody (BM4) in control arms (Figure 4.20). The cohorts

included: no therapy, 7G3, BM4, iCD16+NK-92, iCD16+NK-92 + 7G3, iCD16+NK-92 + BM4.

The dosing schedule was iCD16+NK-92, with or without 7G3 or BM4, given on day 3, 5, 7, 10,

and 12 after AML inoculation (day 0). NSG mice without therapy had a median survival of 32

days. iCD16+NK-92 alone improved survival to a median of 37 days which was significant

(p<0.001). Treatment with BM4 did not enhance survival with a median survival of 32 days

(p=0.619), but 7G3 improved median survival to 35 days which was significantly increased

relative to the AML only control (p<0.001), but not relative to BM4 isotype control (p=0.1509).

Combination of 7G3 and iCD16+NK-92 produced the best survival outcome, with median

survival of 42 days, which was significantly enhanced over AML only (p<0.001), 7G3

(p<0.0025) and iCD16+NK-92 + BM4 (p<0.0025) (Figure 4.21B). An identically repeated

experiment using an alternative source of 7G3 and a different isotype control (MG2a-53) was

conducted (data not shown). MG2a-53 or 7G3 alone did not have any survival benefit.

iCD16+NK-92 combined with 7G3 enhanced median survival by 8 days relative to iCD16+NK-

92 combined with isotype control (p=0.69) which was statistically significant if one long-term

survivor outlier in the control group was removed from analysis (p=0.03).

Day 0

NOD/SCID IL-2Rgc-/- mice

Day X

AML3x106 i.v.

Death/sacrifice

3

225 cGy

iCD16+NK-92

20x106

i.p.

7 10

+1000 cGy

5 12

100 µg 7G3 mAb i.v. Isotype control BM4

Figure 4.20: Schematic of CD16+NK-92 +/- five doses of anti-CD123 mAb therapy for

primary AML xenografted NSG mice

Page 132: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

112

A

Group

iCD16+NK-92

iCD16+NK-92 +7G3

iCD16+NK-92 +BM4

7G3

BM4

No Therapy

Survival

0 5 10 15 20 25 30 35 40 45 50

100

80

60

40

20

0

Time (days)

Surv

ival pro

babili

ty (

%)

B

Group

iCD16+NK-92 +7G3

iCD16+NK-92 +BM4

Survival

0 5 10 15 20 25 30 35 40 45 50

100

80

60

40

20

0

Time (days)

Surv

ival pro

babili

ty (

%)

Figure 4.21: iCD16+NK-92 with and without 7G3 or isotype control treatment for primary

AML xenograft model NSG mice were inoculated with 3x10

6 passage human AML spleen derived cells (day 0) and

treated with iCD16+NK-92 +/- 7G3 or BM4 x 5 doses (3x/week) starting on day 3. Controls

included no therapy and antibodies alone. Survival was determined using Kaplan Meier survival

analysis with a log rank test. All curves are presented (A) with a subgroup comparison (B) of

iCD16+NK-92 +7G3 versus iCD16+NK-92 +BM4 (p=0.0015).

Page 133: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

113

4.5 Discussion

While the overall long-term survival for AML is around 40% in adults (Lowenberg, Downing et

al. 1999), subgroups of AML continue to fare very poorly, including patients with the recurrent

cytogenetic abnormalities inv(3)/(t3;3), -5/del(5q) and 7/del(7q), where the 10 year survival is 3,

6 and 10% respectively(Grimwade, Hills et al. 2010). Therefore, there is a need for novel

therapeutic approaches in treating AML, particularly those who have completed therapy, but

have detectable minimal residual disease. We have evaluated the mechanism and efficacy of

NK-92 against primary AML samples, with a focus on using methods that evaluate the impact on

leukemic stem cells and overall survival in an AML xenograft model. We confirmed initial

reports that NK-92 could mediate cytotoxicity in vitro against primary AML(Yan, Steinherz et

al. 1998), and were further able to demonstrate killing was mediated primarily by granule

exocytosis rather than ligand-mediated cytotoxicity (e.g. via Fas ligand). This was evidenced by

minimal killing in the presence of the calcium chelator, EGTA, which prevents calcium influx

required for degranulation of NK and T cells(Ostergaard, Kane et al. 1987; Trenn, Takayama et

al. 1987).

We noted that classically defined sorted CD34+CD38- LSCs(Lapidot, Sirard et al. 1994) were

more sensitive to NK-92 killing than bulk leukemia at low E:T ratios utilizing a standard

chromium release assay. Given the conflicting reports in the literature of the definitive

immunophenotype of the leukemic stem cell in AML(Lapidot, Sirard et al. 1994; Bonnet and

Dick 1997; Taussig, Miraki-Moud et al. 2008; Goardon, Marchi et al. 2011), we opted to use a

clonogenic assay to assess the impact of immune effectors against leukemic stem cells in a larger

set of samples. Primary AML grows well in methylcellulose, allowing for measurement of

colony forming units, which is a measure of stem and progenitor leukemic cells capable of

sufficient divisions to form a colony. Specifically, we used a methylcellulose cytotoxicity assay

(MCA) established previously by our lab(Williams, Wang et al. 2010) that is designed to have

comparable four-hour co-incubation conditions and a baseline control for effects of immune

effectors over the two week incubation in methylcellulose (termed low density control). This

allows for a comparison of the degree of bulk killing versus colony inhibition in a four-hour

period, effectively providing another means to assay differential cytotoxicity against bulk and

LSC populations. An additional benefit of this approach is that it does not require the

Page 134: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

114

confirmation of the cell surface markers defining the LSC population in each patient sample,

which requires significant cost and time to assay in immunodeficient mice. The MCA showed

that NK-92 at a 25:1 E:T eliminated clonogenic growth of 2/4 primary AML blast samples with

minimal colony growth in the remaining two, demonstrating a 2-3 fold higher % colony

inhibition than the % lysis measured by the CRA. These results support our initial finding that

NK-92 can preferentially recognize and kill LSCs over bulk leukemia.

Only two studies to date have looked at the in vitro sensitivity of CD34+CD38- LSCs to immune

effector cell killing. In the first, lymphokine-activated killer (LAK) cells and allogeneic

lymphocytes exerted a modest cytotoxic effect on AML LSCs comparable to the effect on the

non-stem cell fraction(Costello, Mallet et al. 2000). In a more recent study, endogenous single

killer immunoglobulin-like receptor (KIR)-expressing NK cells, mismatched for the HLA of

primary AML targets, showed killing against LSCs using both the chromium release assay and a

methylcellulose-based cytotoxicity assay, demonstrating equivalent killing of LSCs and

blasts(Langenkamp, Siegler et al. 2009).

Here, we demonstrate preferential killing of LSCs by NK-92 relative to bulk leukemia, not

shown by these other studies, and utilize a better controlled methylcellulose cytotoxicity assay

than Lankencamp et al. to demonstrate differential cytotoxicity relative to the CRA. This is

consistent with our work on NK-92 treatment of multiple myeloma (MM) cell lines. Using the

MCA, NK-92 showed preferential killing of clonogenic MM cells over bulk tumour as

determined by both a flow cytometric cytotoxicity assay and the CRA (Swift, Williams et al.

2012).

To pursue more in depth studies of the effectiveness of NK-92 in killing LSCs, we developed an

animal model of primary human AML by using NSG mice infused with a primary AML sample

containing a small fraction of CD34+CD38- cells. Primary AML, or in vivo passaged primary

AML cells induced leukemia in NSG mice and maintained comparable potency beyond

quaternary transplantations with a stable immunophenotype. There was no difference in potency

of BM or spleen derived cells or delivery by ip or iv injection sites. In using this model we

demonstrate the ability to test a single-patient primary AML sample over multiple in vivo

experiments independent of the quantity of original samples obtained, effectively generating an

‘in vivo’ primary cell line.

Page 135: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

115

Secondary transplantation assays are the current gold standard assay to determine the impact of

small molecule therapies against LSCs(Skrtic, Sriskanthadevan et al. 2011), and have not been

used to evaluate cellular therapies for leukemia. We attempted this by treating mice and

transplanting bone marrow from control and treated mice into new mice in a one-to-one manner,

so as to assess the impact on individual mice rather than a pooled outcome. BM engraftment

occurred in all AML-only cohort secondary mice, while one mouse from iNK-92 group was

leukemia free, with engraftment at background levels of non-injected mice, suggesting that the

primary animal receiving treatment was cured. While the average BM engraftment of secondary

transplant mice in the therapy groups was less than the control, this was not statistically

significant. However, the LSC fraction was significantly decreased in secondary recipients,

providing some evidence of NK-92 cytotoxicity against LSCs in the secondary transplant assay.

AML-xenografted NSG mice were effectively treated with NK-92 infusions, leading to

improvement in survival relative to controls, confirming previous work (Yan, Steinherz et al.

1998). We accomplished this with lower doses of NK-92 on a less compressed schedule

(10x10e6 weekly for three doses) than the original study (20x10e6 every other day for five

doses), and without the use of IL-2 in the regimen. Irradiated NK-92 could prolong survival in

mice, but was less effective than the non-irradiated cells. We postulate that the reason for this

reduction in therapeutic efficacy is the lack of ability to expand in vivo.

We have demonstrated for the first time that irradiated NK-92 improves survival in an AML

xenograft model, which has translational relevance given that this is the cellular preparation

given to patients in the phase I setting. However, given the modest effects of iNK-92 on

improving survival in vivo, we recognized the need to enhance its efficacy to achieve better

therapeutic outcomes. We therefore attempted to use a gene modified CD16+NK-92 in

combination with monoclonal antibodies to enhance killing of leukemia by ADCC. CD16+NK-

92 have been utilized in combination with Rituximab to enhance killing of CD20+ malignant

targets, showing their potential to enhance killing of cells expressing a tumour-associated

antigen(Binyamin, Alpaugh et al. 2008). CD16+NK-92 cytotoxicity against primary AML was

enhanced when target were coated with anti-class I antibodies via ADCC, demonstrating that

CD16+NK-92 can be redirected against bulk primary leukemia using a highly expressed cell

surface marker. More importantly, coating of OCI/AML5 with a murine anti-human CD123

Page 136: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

116

mAb (7G3) was able to facilitate ADCC indicating the ability to redirect CD16+NK-92 against a

leukemia stem cell-specific antigen.

We then sought to combine iCD16+NK-92 and 7G3 therapy in our AML xenograft model.

Systemic treatment with 7G3 alone has been tested in an AML NOD/SCID xenograft model with

evidence of impact on primary bone marrow engraftment(Jin, Lee et al. 2009). The dosage

regimen used in one experiment in this study was 300 µg IV give three times weekly for 4 doses,

which reduced primary engraftment of 1/3 primary AML samples relative to isotype control. In

this study, survival could only be improved by ex vivo coating of AML cells prior to

administration, or passive immunization of mice with 7G3 prior to injection with AML, which is

not a clinically relevant model.

We initially used a very small quantity of 7G3 in a single dose, preceded by blocking with a non-

specific isotype-matched antibody to block Fc receptors, and improve circulation and binding of

7G3 to CD123 as established by Leyton et al. (Leyton, Hu et al. 2011). This approach worked in

increasing the efficacy of the iCD16+NK-92 cells and improving survival. In a follow-up

experiment, we did not use an Fc blocking pre-dose strategy, but used 100 µg of 7G3 or isotype

control antibody BM4 for five doses given with or without the iCD16+NK-92 cells. This was at

a dose level comparable to that used by initial studies by Jin et al. that would likely have

minimal impact on survival, but lead to circulating levels of antibody capable of facilitating

ADCC from infused cells. Doses of 7G3 were given just after administration of cells to optimize

ADCC, given the 7 hour half-life of 7G3(Leyton, Hu et al. 2011).

In this experiment, iCD16+NK-92 alone prolonged survival over control. The BM4 antibody

had no therapeutic effect and did not enhance iCD16+NK-92, while 7G3 alone had a modest

survival benefit above control, which was not quite statistically significant above BM4 isotype

control group. Of note, the best outcome was in the iCD16+NK-92 + 7G3 treated group which

had a ten day improvement in median survival relative to the iCD16+NK-92 + BM4 treatment

group. Therefore, this demonstrates that the combination of iCD16+NK-92 with 7G3 can

improve survival by antibody-dependent cell-mediated cytotoxicity. Further, this represents the

first demonstration of in vivo efficacy of the CD16+NK-92 cell line alone, and in combination

with antibody which has only previously been tested in vitro(Binyamin, Alpaugh et al. 2008).

Recently, an Fc optimized anti-CD123 humanized monoclonal antibody CSL362 (derived from

Page 137: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

117

7G3) was shown to facilitate ADCC from peripheral blood-derived allogeneic NK cells against

primary AML and CD123-expressing cell line targets.(Busfield, Biondo et al. 2014) Further,

CSL362 is in clinical trials and would make an ideal combination therapy with CD16-NK-92.

In summary, we demonstrate that NK-92 can preferentially target LSCs over bulk leukemia in

vitro and irradiated NK-92 can impact survival in an AML xenograft model, which can be

enhanced using CD16+NK-92 in combination with anti-CD123 monoclonal antibodies. This

provides the first proof-of-principle for the targeting of leukemic stem cells by combining an

antibody and a standardized cellular therapy. A humanized version of 7G3 (CDSL360 and

CLS362) has been developed which would allow for therapeutic translation of this approach into

a clinical trial in the future. The approach we demonstrate can be readily applied to enhance

targeting of any antigen and is of particular novelty here, because we are targeting a cancer stem

cell marker and improving survival.

Page 138: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

118

5 Chapter 5: NK cell line killing of leukemia cells is

enhanced by reverse antibody dependent cell mediated

cytotoxicity (R-ADCC) via NKp30 and NKp44 and

target cell Fcγ receptor II (CD32)

Contributions:

X.-H. Wang: Assisted in experimental design, chromium release assays and animal

experimentation.

S. Maghera: Assisted in clonogenic assays, flow cytometery and data analysis.

R. Cheng: Assisted in clonogenic assays.

J. Patterson: Conducted HTS flow cytometry on NK-92, KHYG-1, OCI/AML3 and

OCI/AML5.

B. Routy: Assisted in assays involving esophageal cancer cell lines and data analysis.

Page 139: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

119

5.1 Abstract

NK-92 and KHYG-1 are natural killer cell lines with the potential to treat cancer. Phase I NK-92

trials show minimal toxicity while KHYG-1 has not been tested in humans. We evaluated their

cytotoxicity against leukemia and esophageal cell lines and primary acute myeloid leukemia

(AML) and modulated efficacy with monoclonal antibodies against NK activating receptors.

Pretreatment of NK-92 and KHYG-1 with antibodies (0.01-10 µg/ml) against the activating

receptors NKp30 and NKp44 led to enhancement of cytotoxicity against a panel of leukemic cell

lines and primary AML samples, but not esophageal cancer cell lines. Immunophenotyping

cancer cell lines showed high expression of Fcγ receptor II (CD32) on leukemia cell lines, which

was absent on esophageal cancer cell lines. There was a significant correlation with CD32 target

expression and anti-NKp30 or anti-NKp44-mediated cytotoxic enhancement for KHYG-1

(p<0.01), implicating reverse antibody-dependent cell-mediated cytotoxicity (R-ADCC) as the

mechanism of enhancement. Clonogenic OCI/AML5 were more sensitive to NK-92 than

KHYG-1. However, anti-NKp30 pretreatment of KHYG-1 enhanced percent colony inhibition

by three-fold with minimal enhancing effect on NK-92. NOD-SCID gamma null mice injected

ip with OCI/AML5 co-incubated in vitro with anti-NKp30-pretreated, irradiated KHYG-1

(iKHYG-1) showed improved survival over control and iKHYG-1 treatments (p<0.05).

Treatment of OCI/AML5-xenografted mice with iKHYG-1 infusions without or with anti-

NKp30 pretreatment improved median survival 35 or 37 days over control, respectively. In

summary, we demonstrate that KHYG-1 cytotoxicity can be enhanced by R-ADCC, leading to

enhanced killing of leukemic targets in both bulk and clonogenic in vitro cytotoxicity assays.

Finally, we demonstrate for the first time, the efficacy of KHYG-1 in improving survival using

an in vivo model of cancer.

Page 140: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

120

5.2 Introduction

Acute myeloid leukemia (AML) is a hematopoietic malignancy involving precursor cells

committed to myeloid development, and accounts for a significant proportion of acute leukemias

in both adults (90%) and children (15-20%).(Hurwitz, Mounce et al. 1995; Lowenberg, Downing

et al. 1999) Despite 80% of patients achieving remission with standard chemotherapy (Hurwitz,

Mounce et al. 1995; Ribeiro, Razzouk et al. 2005), survival remains unsatisfactory because of

high relapse rates from minimal residual disease (MRD). The five-year survival is age-

dependent; 60% in children(Rubnitz 2012), 40% in adults under 65(Lowenberg, Downing et al.

1999) and 10% in adults over 65 (Ferrara and Schiffer 2013). These outcomes can be improved

if patients have a matched hematopoietic cell donor, but most do not, highlighting the need for an

alternative approach to consolidation treatment, such as NK cell therapy.

In haplotype transplantation, the graft-versus-leukemia effect is mediated by NK cells when

there is a KIR receptor-ligand mismatch, which can lead to improved survival in the treatment of

AML (Ruggeri, Capanni et al. 2002; Ruggeri, Mancusi et al. 2005). Further, rapid NK recovery

is associated with better outcome and a stronger GVL effect in patients undergoing haplotype T-

depleted hematopoietic cell transplantation (HCT) in AML.(Savani, Mielke et al. 2007) Other

trials have used haploidentical NK cells expanded ex vivo to treat AML in adults(Miller, Soignier

et al. 2005) and children(Rubnitz, Inaba et al. 2010), supporting a therapeutic role for NK cells in

AML therapy. However, all current adoptive immunotherapy protocols are affected by donor

variability in the quantity and quality of effector cells, variables that could be eliminated if

effective cell lines were available to provide more standardized therapy.

Several permanent NK cell lines have been established, and the most notable is NK-92, derived

from a patient with non-Hodgkin’s lymphoma expressing typical NK cell markers except for

CD16 (Fc gamma receptor), making it incapable of antibody-dependent cell-mediated

cytotoxicity (ADCC). NK-92 has undergone extensive preclinical testing and exhibits superior

lysis against a broad range of tumours compared with activated NK cells and lymphokine-

activated killer (LAK) cells.(Gong, Maki et al. 1994) Further, NK-92 has been evaluated in

three phase I clinical trials for: renal cell carcinoma and melanoma (Arai, Meagher et al. 2008),

solid tumours and lymphoid malignancies(Tonn, Schwabe et al. 2013), and lymphoma and

Page 141: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

121

multiple myeloma (in progress at our center). There have been minimal cytotoxicities reported

in these trials.

Another NK cell line with therapeutic potential is KHYG-1, derived from a patient with an NK

cell leukemia with a p53 mutation.(Yagita, Huang et al. 2000), with the unique features of

constitutively phosphorylated ERK2 (Suck, Branch et al. 2005) and polarized granules(Suck,

Branch et al. 2006). Like NK-92, irradiation of KHYG-1 prevents proliferation, but preserves

cytotoxicity in vitro (Suck, Branch et al. 2006), thereby making it safe for human administration.

Here, we demonstrate that pretreatment of NK cell lines with monoclonal antibodies to activating

receptors causes several-fold enhancement of cytotoxicity against leukemic cell lines and

primary AML blasts. This effect is most prominent with anti-NKp30 and anti-NKp44

pretreatment of KHYG-1 against CD32-expressing targets implicating reverse antibody-

dependent cell-mediated cytotoxicity (R-ADCC) as the mechanism of enhancement. We further

demonstrate an impact of NKp30 pretreated KHYG-1 in an in vivo model.

Page 142: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

122

5.3 Methods

5.3.1 Cell lines and primary samples

K562 was obtained from the ATCC and maintained in IMDM + 20% FBS and 10% fetal bovine

serum (FBS), respectively. KG1 and KG1a was obtained from the ATCC and maintained in

IMDM + 20% FBS and 10% FBS, respectively. OCI/AML 2, 3 and 5 were derived at the

Ontario Cancer Institute (OCI). OCI/AML 2 and 3 were cultured in MEM alpha + 10% FBS and

OCI/AML5 was cultured in MEM alpha + 10% FBS and 10% 5637 bladder carcinoma condition

medium. KHGY-1 was purchased from The Human Science Research Resources Bank

(JCRB0156; Tokyo, Japan) and cultured in GM1 (Ex Vivo medium with 450 U/ml and human

A/B serum). NK-92 was obtained from Dr. Hans Klingemann and also cultured in Ex Vivo with

human A/B serum and 450 U/ml of IL-2 (GM1). KHYG-1 was irradiated (iKHYG-1) with 1000

cGy prior to use in in vivo experiments. Four primary AML samples were obtained from the

Princess Margaret Hospital Leukemia Tissue Bank as per institutional protocol (5890, 080179,

080078, 080008, 0909). The University Health Network HLA database was linked with the

leukemia bank with REB approval to identify the HLA class I type of leukemia specimens.

5.3.2 Chromium release assay

We utilized a standard chromium release assay as previously described by our group (Williams,

Wang et al. 2010) and detailed in the Chapter 2 methods section. Briefly, 1x106 target cells were

labelled with 100 µCi of Na251

CrO4 for 2 hours prior to plating 10 000 cells per wells followed

by treatment with NK-92 at various concentrations. The amount of 51

Cr present in supernatants

was determined using a gamma counter and percent lysis calculated.

5.3.3 Antibody pretreatment of NK cell effectors

All antibodies used were from Biolegend. For NK pretreatment experiments, antibodies against

the following NK receptors were utilized (clone; product #): NKp30 (clone P30-15; 325204),

NKp44 (clone P44-8; 325104), NKp46 (clone 9E2; 331904), DNAM-1 (clone DX-11; 316802),

NKG2D (clone 1D11; 320810), CD7 (CD7-6B7; 343102). Isotype controls specific to trinitrol

phenol + KLH were utilized: MG1-45 (clone MG1-45; 401404) and MG2a-52 (clone MG2a-53,

Page 143: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

123

401502). Briefly, 1.5x106 NK cells (NK-92 or KHYG-1) were treated with in 1 ml of AIM-V

serum free medium for 1 hour, washed in 10 ml of AIM-V medium and resuspended in 1.5 ml of

AIM-V medium (1x106/ml). Concentration of antibodies ranged from 10 µg/ml to 0.01 µg/ml.

0.1 ul (105 cells) of NK cell suspension were added to 10 000 tumour targets also in AIM-V

medium in 96 well U bottom plates to yield a 10:1 E:T ratio.

5.3.4 Flow cytometry

Immunophenotyping of BM was done using an FC500. FACS buffer was made with PBS

+2mM EGTA + 2% FBS. For routine flow cytometry of leukemia and esophageal cancer cell

lines the following antibodies to Fcγ receptors were utilized: CD16 APC (clone 3G8, 302011),

CD32 PE (clone FUN-2, 303205), CD64 FITC (clone 10.1; 305005). Antibody concentrations

were utilized at ~1 µg/ml in a 50 µl reaction volume with 200 000 to 1,000 000.

5.3.5 High throughput sampling flow cytometry

Commercially validated FITC, PE or APC conjugated antibodies (374) to cell surface markers

(BD Pharmingen, eBioscience, Abcam, AbD Serotech, BioLegend, Lifespan Biosciences,

Miltenyi, R&D Systems, Beckman-Coulter, and Imgenex) were aliquotted into individual wells

of 96-well plates in Hanks Balanced Salt Solution supplemented with 1% bovine serum albumin

and 2 mM EDTA (FACS buffer) at a dilution of 1:25 (Supplemental Table 1, 2). NK-92 or

KHYG-1 cells (30x10e6) were prepared in 10 ml of PBS, spun down and resuspended in HBSS

+ 1% BSA, 2mM EDTA and volume adjusted to 1x106/ml. 50 ul of cell suspension (50 000

cells) suspension was added to each well to yield a final antibody dilution of 1:50. Cells were

stained for 30 minutes on ice at a concentration of 0.25-1.0x106/mL, washed once with cold

FACS buffer, and resuspended in FACS buffer with 0.1 µg/mL DAPI to allow for dead cell

exclusion. Flow cytometry was performed using a High Throughput Sampler-equipped Becton-

Dickinson LSRII flow cytometer. Plates were placed into an automated flow cytometry plate

reader. Data was acquired and analyzed on FlowJo 9. Gating strategy utilized both a FS and SS

plot and subsequent DAPI staining to exclude non-viable cells, followed by FSC-H and FSC-W

to exclude doublets. Final gate was contoured around viable, unstained cells. Percentage positive

cells and mean fluorescence intensity were quantitated for each marker

Page 144: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

124

5.3.6 Animals

NOD/SCID gammanull

(NSG) mice from The Jackson Laboratory were bred and maintained in

the Ontario Cancer Institute animal facility according to protocols approved by the Animal Care

Committee. Mice were fed irradiated food and Baytril containing water ad libitum during

experimental periods. Prior to infusion with AML NSG mice were irradiated with 200 cGy to

facilitate engraftment. We developed ip and iv injection route OCI/AML5 NSG xenograft

models utilizing a dose of 2x106 cells. To determine the impact of in vitro incubation with

iKHYG-1 on proliferative capacity of OCI/AML5, the ip route of injection was utilized, with

sacrifice at humane endpoints. Briefly, OCI/AML5 cells were incubated in 15 ml conical tubes

with or without iKHYG-1 (+/- 1 µg/ml anti-NKp30 pretreatment x 1 hour), at a 10:1 E:T ratio,

spun at 1200 rpm to pellet, and incubated for four hours at 37°C. Cell mixtures were then

washed and resuspended in PBS and 2x10e6 OCI/AML5 cells with or without 20x106 iKHYG-1

or NKp30 iKHYG-1 cells in 200 µl of PBS were injected ip into three cohorts of five NSG mice.

To determine in vivo effect of NK cell line therapy OCI/AML5 or primary AML were injected iv

on day 0 with and without iKHYG-1 or anti-NKp30 pretreated iKHYG-1 treatment started on

day 3 (10x106 x 6 doses; days 3, 5, 7, 10, 12, 14). The primary AML sample (080179) was

derived from an M4 leukemia with aggressive engraftment features and passaged through NSG

mice prior to use in these experiments.

5.3.7 Statistics

Survival analysis was done with Kaplan Meier survival curves using the log rank rest with

Medcalc software. Comparison of cytotoxicity and engraftment data was done using a two tailed

student’s t-test performed on Medcalc software. Linear regression analysis was done using

Medcalc software and used to generate scatter plots with best fit line, coefficients of

determination (R2), F test and degree of significance.

Page 145: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

125

5.4 Results

5.4.1 NK-92 and KHYG-1 cytotoxicity against leukemia cell lines

NK-92 and KHYG-1 were tested against a panel of leukemic cell lines (K562, KG1, OCI/AML2,

3 and 5) at a 10:1 E:T ratio using the chromium release assay. Both cell lines demonstrated

cytotoxicity against these targets, with NK-92 showing overall better cytotoxicity than KHYG-1

(Figure 5.1). OCI/AML5 was particularly sensitive to NK-92 killing with percentage lysis of

68%, exceeding that for K562. OCI/AML2 was relatively resistant to killing by both cell lines

with mimimal cytotoxicity demonstrated.

0

10

20

30

40

50

60

70

80

90

100

K562 KG1 OCI/AML2 OCI/AML3 OCI/AML5

% L

ysis

Cell line targets

NK-92

KHYG-1

Figure 5.1: NK-92 and KHYG-1 cytotoxicity against a panel of leukemia cell lines K562, KG1, OCI/AML2, 3, 5 cells were labelled with 100 µCi of Na2

51CrO4 prior to treatment

with NK-92 (A) and KHYG-1 (B) at a 10:1 E:T ratios. Data are presented as the mean percent

lysis of triplicate samples (+/-SD) from a representative experiment. Data are presented as the

mean percent lysis of triplicate samples (+/-SD) from a representative experiment repeated 3

times.

Page 146: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

126

NK-92 cytotoxicity against K562 was completely abrogated by calcium chelation at all effector

targets ratios, indicating that granule exocytosis was the primary means of cytotoxicity (Figure

2A). However, there was a small amount of residual killing of K562 by KHYG-1 particularly at

effector:target ratios of 1:1 and 5:1 (Figure 5.2).

A

B

0

10

20

30

40

50

60

70

80

90

100

1:1 5:1 10:1 25:1

% L

ys

is

Effector:Target ratio

Medium

EGTA

Figure 5.2: NK-92 and KHYG-1 cytotoxicity against K562 with and without calcium

chelation K562 cells were labelled with 100 µCi of Na2

51CrO4 prior to treatment with NK-92 (A) and

KHYG-1 (B) at four E:T ratios in a standard four hour chromium release assay. Data are

presented as the mean percent lysis of triplicate samples (+/-SD) from a representative

experiment. Data are presented as the mean percent lysis of triplicate samples (+/-SD) from a

representative experiment repeated 3 times.

Page 147: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

127

5.4.2 High throughput screening flow cytometry of NK-92 and KHYG-1

surface receptors

To better understand the differences in cytotoxicity of NK-92 and KHYG-1, we conducted high

throughput screening flow cytometry and identified the expression level of 374 cell surface

markers on NK-92 and KHYG-1 (Appendix I and II). Relevant subsets of activating, inhibiting

and apoptosis inducing cell surface proteins were more closely evaluated, as these are relevant to

recognition of malignant targets and cytotoxic effector function (Table 5.1). Cell surface

expression was categorized into quartiles and labelled tables with a colored heat map as follows:

Minimal-no color (0-1%), low-green (1-25%), intermediate-yellow (25-50%), high-orange (50-

75%) and very high-red (75-100%).

On KHYG-1 we identified the presence of very high levels of NKp30, NKp44, NKG2D and

DNAM-1 and high levels of NKp46. Further, other receptors known to mediate adhesion and

also transmit an activating signal to NK cells were also detected on KHYG-1 to variable degrees:

CD2 (LFA-2), CD7 (LEU-9), CD11a (LFA-1 component), CD11b (Mac-1), CD27 (TNFRSF7),

CD44 (Hyaluronate receptor), CD59 (Protectin), CD96 (TACTILE), CD160 (BY55), CD158i

(KIR2S4), CD161 (NKR-P1), CD223 (Lag3), and CD319 (CRACC). KHYG-1 expressed low

levels of two inhibitory KIRs, CD158b2 (KIR2DL3) and CD158f (KIR2DL5), high levels of

CD158d (KIR2DL4), and very high levels of CD85j (LIR-1). KHYG-1 had some degree of

expression of all ten tumour necrosis family members assayed, with intermediate and high levels

of expression of Fas Ligand CD178 (Fas Ligand) and CD253 (TRAIL), respectively.

NK-92 had a lesser degree of expression of natural cytotoxicity receptors: NKp44 (low), NKp46

(intermediate) and NKp30 (intermediate). DNAM-1 and NKG2D had very high expression.

NK-92 expressed low levels of KIR2DL4 and intermediate levels of KIR2DL5. However, LIR-1

was highly expressed by NK-92. NK-92 expressed nine of ten tumour necrosis family members

to variable degrees. Most notably, CD120b (TNF-β/lymphotoxin-α) was very highly expressed

and CD178 (Fas Ligand) and CD253 (TRAIL) had intermediate expression.

Page 148: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

128

Table 5.1: Differential expression of cell surface activating, inhibiting and apoptosis

inducing molecules on NK-92 and KHYG-1

CD

Marker

Name Ligand % expression*

NK-92 KHYG-1 Activating

CD2 LFA-2 CD58 (LFA-3) 100 100

CD7 LEU-9 SECTM1, Galectin 99.9 100

CD11a LFA-1 component ICAM-1,-2,-3,-4,-5 100 100

CD11b Mac-1 ICAM-1, Fibrinogen 5.67 52.7

CD16 FcγRIII IgG 1.78 16.5

CD44 Hyaluronate receptor Hyaluronan 99 100

CD59 Protectin C8, C9 13.7 85.2

CD69 CLEC2C Unknown 0.946 93.7

CD96 TACTILE CD155 99.5 98.6

CD158i KIR2DS4 HLA-C 6.01 18.8

CD159c NKG2C HLA-E 0.971 47.9

CD160 BY55 HLA-C 73.2 28.6

CD223 Lag3 HLA Class II 10.4 99

CD226 DNAM-1 CD112, CD155 75.1 82.6

CD244 2B4 CD48 6.79 3.64

CD314 NKG2D MICA, MICB, ULB-1, -2, -3, -4, -5, -6 78.9 94.7

CD319 CRACC CRACC 92.8 82.2

CD335 NKp46 Influenza hemaglutinins, HSPs 36.9 50.8

CD336 NKp44 Influenza hemaglutinins 4.55 99.5

CD337 NKp30 BAT3, B7-H6 41.6 99.3

CD352 NTB-A CD352 (NTB-A) 100 100

Inhibiting

CD85d LIR-2, ILT-4 HLA-G 12.2 38.3

CD85j LIR-1, ILT-2 HLA-A, -B, -G 95.7 81.8

CD158a KIR2DL1 HLA-C2 0.243 0.175

CD158b KIR2DL2 HLA-C1 0.263 0.193

CD158b2 KIR2DL3 HLA-C1 0.395 14.5

CD158d KIR2DL4 HLA-G 37.3 73.5

CD158e2 KIR3DL1 HLA-Bw4 0.225 0.162

CD158f KIR2DL5 Unknown 6.73 13.5

CD159a NKG2A HLA-E 1.82 17

CD161 NKR-P1 LLT1 8.76 30.5

Apoptosis

inducing

CD120a TNF-alpha 1.99 19.2

CD120b TNF-β/lympotoxin-α 98.7 99.4

CD137L 4-1BB ligand 4-1BB ligand 29.5 76.6

CD153 CD30 ligand CD30 4.88 29.3

CD154 CD40 ligand CD40 0.428 4.4

CD178 Fas Ligand CD95 (Fas) 30.4 31.9

CD252 OX40 Ligand CD134 (OX40) 15.8 17.2

CD253 TRAIL TRAIL R1, TRAIL R2 36.3 59.9

CD256 APRIL TACI 25.3 18

CD257 BAFF BAFF-R, TACI and BCMA 49.5 73.4

CD258 LIGHT HVEM 4.51 36.0

*Percent positivity of cell surface expression was categorized into quartiles and tables labelled

with a colored heat map as follows: Minimal-no color (0-1%), Low-green (1-25%), Intermediate-

yellow (25-50%), High-orange (50-75%) and Very High-red (75-100%).

Page 149: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

129

5.4.3 Anti-Class I HLA blockade of AML targets

To explore the role of inhibitory killer immunoglobulin-like receptors (KIRs), we blocked the

class I HLA molecules on target cells, which serve as the ligand for this receptor group. The

antibody used was a pan-HLA blocking antibody with capability to block HLA- A, B and C

groups by binding to generic epitopes on each of these molecules. NK-92 was incubated at a

25:1 E:T ratio with four primary AML blast samples (080179, 080078, 080008 and 0909),

yielding moderate degrees of cytotoxicity by the CRA: 42.3, 29.8, 43.9, 42.6 % lysis. Blockade

of class I HLA on primary AML targets with antibody did not affect cytotoxicity of NK-92 (data

not shown). KHYG-1 at a 25:1 E:T had lower cytotoxicity against this panel: 9.8, 5.1, 17.1 and

8.5 % lysis. However, following class I HLA blockade, sample 080008 was rendered more

sensitive to killing by KHYG-1 increasing from 17.1 +/-2.8% to 35.8 +/-1.2 % lysis (p<0.001)

(Figure 5.3). We obtained the HLA typing on the four primary AML specimens to identify

potential HLA class I ligands that could interact with inhibitory KIRs and noted the following

HLA-C groups: 080179 (C1/C1), 080078 (C2/C2), 080008 (C2/C2), 0909 (C1/C1) (Table 5.2)

No relation between KIR ligand expression and cytotoxicity could be made. Further, predictions

of cytotoxicity enhancement based upon the KIR-ligand receptor mismatch hypothesis were not

confirmed. Finally, blockade of KIR2DL3 (specific for HLA-C1) on KHYG-1 did not enhance

cytotoxicity against primary AML targets 08008 and 0909 (data not shown).

Page 150: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

130

0

10

20

30

40

50

60

70

80

90

100

080179 080078 080008 0909

% L

ys

is

Sample ID

Control

1 ug/ml α MHC I

10 ug/ml α MHC I

*

Figure 5.3: KHYG-1 cytotoxicity against 4 primary AML samples +/- class I HLA blockade

AML blasts were labelled with 100 µCi of Na251

CrO4 for 2 hours prior to treatment with KHYG-

1 at a 25:1 E:T in 96 well U bottom plates. Some samples were also incubated with 1 or 10

µg/ml of anti-Class I A, B, C blocking mAb for 1 hour prior to effector cell addition. Plates

were centrifuged at 200 g and incubated at 37°C 5% CO2 x4 hours. KHYG-1 against primary

AML blasts +/- 1 or 10 µg/ml class I blockade. Data are presented as the mean percent lysis of

triplicate samples (+/-SD) representative of two separate experiments (*= p<0.001).

Table 5.2: HLA type of primary AML panel and sensitivity to NK-92 and KHYG-1 +/-

HLA blockade

Sample

ID #

HLA C type

(C1 or C2)

Sensitivity to cytolysis

Effect of HLA class I

blockade on

cytotoxicity

Consistent with KIR

ligand mismatch

hypothesis?

NK-92 KHYG-1 NK-92 KHYG-1 NK-92 KHYG-1

080179 C1 (C*07:01)

C1 (C*07:02)

Intermediate Low None None Yes No

080078 C2 (C*05:01)

C2 (C*15:02)

Intermediate Low None None Yes Yes

080008 C2 (C*02:xx)

C2 (C*06:02)

Intermediate Low None Increase Yes No

0909 C1 (C*07:01)

C1 (C*16:01)

Intermediate Low None None Yes No

Page 151: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

131

5.4.4 Effect of pretreating NK-92 and KHYG-1 with activating receptor

specific antibodies

We attempted to address the role of common activating receptors in NK cell line-mediated

recognition of leukemic targets by pretreating NK-92 and KHYG-1 with of antibodies specific

to NKp30, NKp44, NKp46, DNAM-1, NKG2D, and CD7 (10 µg/ml), prior to co-incubation

with the target cells K562, KG1a and OCI/AML5. An off-target antibody was used as an isotype

control. Pretreatment of NK-92 with antibodies to NKp30, NKp44 and NKp46 unexpectedly

increased killing of K562 above isotype control [(1.3X (p<0.0001), 1.2X (p<0.05), 1.2X

(p=0.11)], while anti-NKp30 treatment enhanced killing of KG1a [+1.8X (p<0.0001)] and

OCI/AML5 [1.2X (p<0.01)] (Figure 5.4A). Treatment of KHYG-1 with antibodies to NKp30,

NKp44 and NKp46 increased killing of K562 above isotype control [1.4X (p<0.001), 1.5X

(p<0.01), 1.2X (p<0.05)], while anti-NKp30 treatment increased killing of KG1a [2.6X

(p<0.01)] and anti-NKp30, anti-NKp44 and anti-NKp46 treatment increased killing of

OCI/AML5 [3.4X (p<0.00001), 3.2X (p<0.0001), 1.8X (p<0.0001)] (Figure 5.4B). Pretreating

NK cell lines with antibodies to DNAM-1 and NKG2D had minimal effects on cytotoxicity.

A

0

10

20

30

40

50

60

70

80

90

100

K562 KG1a OCI/AML5

% L

ysis

Cell line

None

MG1-45

NKp30

NKp44

NKp46

NKG2D

DNAM-1

* *

*

*

*= <0.05

*

Page 152: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

132

B

0

20

40

60

80

100

120

140

K562 KG1a OCI/AML5

% L

ysis

Cell line

None

MG1-45

NKp30

NKp44

NKp46

NKG2D

DNAM-1

* *

*

*

**

*

Figure 5.4: Effect of antibody pre-treatment of activating receptors on NK-92 and KHYG-

1 cytotoxicity against leukemia cell lines NK-92 (A) and KHYG-1 (B) were pretreated with a panel of antibodies (10 µg/ml) against

receptors with activating function for NK cells including an untreated control and isotype control

as follows: none, MG1-35 (isotype control), NKp30, NKp44, NKp46, DNAM-1, NKG2D, CD7.

NK-92 and KHYG-1 were incubated for one hour with antibodies and then washed in AIM-V

medium to remove residual antibody. Effector cells were utilized in a standard chromium

release assay against K562, KG1a and OCI/AML5 labelled with 100 µCi of Na251

CrO4 at an E:T

ratio of 10:1. Statistical comparisons were done using a student’s test between isotype control

(MG1-45) and antibody pretreated groups (* =p<0.05).

We then attempted a similar experiment with K562 and two primary AML cell lines. NK-92 and

KHYG-1 were pre-treated with antibodies specific to NKp30, NKp44, NKp46, DNAM-1,

NKG2D and CD7 (10 µg/ml) prior to co-incubation with the leukemic target cells K562, and the

primary AML specimens 080078 and 0909. NK-92 cytotoxicity against primary AML samples

demonstrated prominent increases above isotype control when pretreated with anti-NKp30 [7.1X

(p<0.001) and 3.0X (p<0.0001)] (Figure 5.5A).

Pretreatment of KHYG-1 with either anti-NKp30 or anti-NKp44 led to dramatic increases of

cytotoxicity relative to isotype control against primary AML samples 080078 [16.9X (p<0.0001)

and 17.6X (p<0.001)] and 0909 [2.8X (p<0.0001) and 2.9X (p<0.001)], (Figure 5.5B). The dose

dependence of anti-NKp30 and anti-NKp44 mediated enhancement of killing was then explored

by testing several dose ranges.

Page 153: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

133

A

0

10

20

30

40

50

60

70

80

90

100

K562 080078 0909

% L

ysis

Cell line or primary AML sample

None

MG1-45

NKp30

NKp44

NKp46

NKG2D

DNAM-1

CD7

*

*

*

*

*

*

B

0

10

20

30

40

50

60

70

80

90

100

K562 080078 0909

% L

ysis

Cell line or primary AML sample

None

MG1-45

NKp30

NKp44

NKp46

NKG2D

DNAM-1

CD7

**

*

**

**

Figure 5.5: Effect of antibody pre-treatment of activating receptors on NK-92 and KHYG-

1 cytotoxicity against primary AML samples

NK-92 (A) and KHYG-1 (B) were pretreated with a panel of antibodies (10 µg/ml) against

receptors with activating function for NK cells (NKp30, NKp44, NKp46, DNAM-1, NKG2D,

CD7), including an untreated control, and isotype control (MG1-45). NK-92 and KHYG-1 were

incubated for one hour, and then washed in AIM-V medium to remove residual antibody.

Effector cells were utilized in a standard chromium release assay against K562 and two primary

AML samples labelled with 100 µCi of Na251

CrO4 at an E:T ratio of 10:1. Data are presented as

the mean percent lysis of triplicate samples (+/-SD) representative of two similar experiments.

Statistical comparisons were done using a student’s test between isotype control (MG1-45) and

antibody pretreated groups (* =p<0.05).

Page 154: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

134

In an attempt to determine the linear portion of the dose response curve and approximate the

EC50%, pretreatment of NK cell lines was done with a range of doses of anti-NKp30 and anti-

NKp44 (1, 5 and 10 µg/ml) against K562, OCI/AML3 and OCI/AML5 and primary AML

080078. A dose response was seen with anti-NKp30 pretreatment of NK-92 against OCI/AML3

and primary AML sample 080078 however the EC50% was less than the lowest dose used (1

µg/ml) (data not shown). KHYG-1 had a similar degree of enhancement seen at all dose ranges

of both anti-NKp30 and anti-NKp44 antibody pretreatments (data not shown).

A lower dose range was then selected utilizing NK cell lines pretreated with isotype control, anti-

NKp30 and anti-NKp44 at 0.1, 0.5 and 1 µg/ml. Isotype-control pretreated NK-92 and KHYG-1

had minimal effects on cytotoxicity against K562, OCI/AML3, OCI/AML5 and KG1, with no

dose response (Figure 5.6). Anti-NKp30 pretreatment enhanced NK-92 cytotoxicity against

OCI/AML3 (EC50% ~0.1) and KG1 (EC50%<0.1 µg/ml) only. Combined anti-NKp30 and anti-

NKp44 pretreatment (0.1 µg/ml) of NK-92 did not have additive or synergistic effects on

cytotoxicity against any targets (Figure 5.7A).

Anti-NKp30 pretreatment enhanced KHYG-1 cytotoxicity against all targets with a dose

response seen for K562 (EC50% ~0.1 µg/ml), OCI/AML3 (EC50% <0.1 µg/ml), and KG1

(EC50% <0.1 µg/ml), while OCI/AML5 showed a plateau from the lowest dose (EC50% <0.1

µg/ml). Anti-NKp44 pretreatment enhanced KHYG-1 cytotoxicity against all targets, with a

dose response seen for K562 only (EC50% ~0.1 µg/ml), while OCI/AML3, KG1 and

OCI/AML5 showed a plateau from the lowest dose (EC50% <0.1 µg/ml) (Figure 5.7B).

Page 155: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

135

A

0

10

20

30

40

50

60

70

80

90

100

K562 OCI/AML3 OCI/AML5 KG1

% L

ysis

Cell line

None

MG2a-53 (0.1)

MG2a-53 (0.5)

MG2a-53 (1)

B

0

10

20

30

40

50

60

70

80

90

100

K562 OCI/AML3 OCI/AML5 KG1

% L

ysis

Cell line

None

MG2a-53 (0.1)

MG2a-53 (0.5)

MG2a-53 (1)

Figure 5.6: Effect of antibody pre-treatment with isotype control on NK-92 and KHYG-1

cytotoxicity against leukemia cell lines

NK-92 and KHYG-1 were pretreated with isotype control MG2-53, incubated for one hour and

then washed in AIM-V medium to remove residual antibody. Effector cells were utilized in a

standard chromium release assay against K562, OCI/AML3, 5 and KG1 labelled with 100 µCi of

Na251

CrO4at an E:T ratio of 10:1. Data are presented as the mean percent lysis of triplicate

samples (+/-SD).

Page 156: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

136

A

0

10

20

30

40

50

60

70

80

90

100

K562 OCI/AML3 OCI/AML5 KG1

% L

ysis

Cell line

None

NKp30 (0.1)

NKp30 (0.5)

NKp30 (1)

NKp44 (0.1)

NKp44 (0.5)

NKp44 (1)

NKp30/NKp44 (0.1)

B

0

10

20

30

40

50

60

70

80

90

100

K562 OCI/AML3 OCI/AML5 KG1

% L

ysis

Cell line

None

NKp30 (0.1)

NKp30 (0.5)

NKp30 (1)

NKp44 (0.1)

NKp44 (0.5)

NKp44 (1)

NKp30/NKp44 (0.1)

Figure 5.7: Effect of antibody pre-treatment with anti-NKp30 and anti-NKp44 on NK-92

and KHYG-1 cytotoxicity against leukemia cell lines

NK-92 (A) and KHYG-1 (B) were pretreated with anti-NKp30 and anti-NKp44 antibodies at 0.1,

0.5, 1 µg/ml incubated for one hour and then washed in AIM-V medium to remove residual

antibody. Effector cells were utilized in a standard chromium release assay against K562,

OCI/AML3, 5 and KG1 labelled with 100 µCi of Na251

CrO4at an E:T ratio of 10:1. Data are

presented as the mean percent lysis of triplicate samples (+/-SD).

Page 157: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

137

Combined anti-NKp30 and anti-NKp44 pretreatment (0.1 µg/ml) of KHYG-1 demonstrated

greater enhancement on cytotoxicity than each antibody alone against K562, OCI/AML3 and

KG1, but not OCI/AML5. Not only did the combined dosing of 0.1 µg/ml anti-NKp30 and anti-

NKp40 exceed the effect of each antibody alone at this dose level, it also matched or exceeded

the effect of a 10-fold higher dose (1 µg/ml) of each antibody alone for K562, OCI/AML3 and

KG1, demonstrating true synergy. The % lysis values for untreated, isotype control, anti-

NKp30, anti-NKp40 and combined anti-NKp30 and anti-NKp44 groups (0.1 µg/ml) and anti-

NKp30 and anti-NKp44 (1 µg/ml) for the cell line targets treated with KHYG-1 from Figure 5.6

and Figure 5.7 are tabulated for comparison (Table 5.3).

Table 5.3: Comparison of antibody pretreatment effects on KHYG-1 cytotoxicity and

synergy assessment at 0.1 µg/ml

Cell line % Lysis (mean +/- SD)

Pretreatment

0.1 µg/ml 1.0 µg/ml

None MG2a-

53

Anti-

NKp30

Anti-

NKp44

Anti-

NKp44/

NKp30

Anti-

NKp30

Anti-

NKp44

K562 26

+/- 3.2

37

+/- 2.9

41

+/-6.0

46

+/-11.1 *61

+/- 5.9

54

+/-16.7

59

+/-2.6

OCI/AML3 0

+/- 0.8

9

+/-7.8

20

+/- 2.0

25

+/- 1.5 *39

+/-1.7

34

+/-1.3

26

+/-4.8

OCI/AML5 11

+/- 0.5

19

+/- 3.1

68

+/- 2.7

62

+/- 3.5

69

+/- 3.8

74

+/-42

67

+/- 7.0

KG1 1

+/- 0.4

9

+/- 1.7

16

+/- 1.7

10

+/- 2.2 *24

+/- 1.1

25

+/-0.5

11

+/- 0.7

*Combined anti-NKp30 and anti-NKp44 regimens that yielded statistically significant (p<0.05)

increases above each antibody alone in separate comparisons are in bold font.

While dose responses could be seen in the range of 0.1 to 1 µg/ml in many cases, the lowest dose

exceeded the EC50%. Therefore, an additional experiment testing a dose range one log lower

was conducted (0.01, 0.1 and 1 µg/ml). There was minimal effect of the isotype control (MG2a-

53) antibody in this range, with no dose response seen (data not shown). Pretreatment of NK-92

with 0.01, 0.1 and 1 µg/ml of anti-NKp30 enhanced cytotoxicity of OCI/AML3 only (ED50%

0.01 to 0.1 µg/ml) and there was no effect of anti-NKp44 pretreatment (Figure 5.8A).

Page 158: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

138

Anti-NKp30 and anti-NKp44 pretreatment enhanced KHYG-1 cytotoxicity against all targets

with a dose response seen for K562, OCI/AML3 and OCI/AML5 (EC50% 0.01-0.1

µg/ml)(Figure 5.8B). Combined anti-NKp30 and anti-NKp44 pretreatment of KHYG-1 (0.01

µg/ml) demonstrated synergistic effects on cytotoxicity against OCI/AML3 only at this dose

level. The combined dosing of 0.01 µg/ml anti-NKp30 and anti-NKp40 exceeded the effect of

each antibody alone (2X) and the absolute cytotoxicity was comparable to a 10-fold higher dose

of each antibody (0.1 µg/ml). The % lysis values for untreated, isotype control, anti-NKp30,

anti-NKp40 and combined anti-NKp30 and anti-NKp44 groups (0.01 µg/ml) and anti-NKp30

and anti-NKp44 (1 µg/ml) for the cell line targets treated with KHYG-1 from Figure 5.8 and its

isotype controls are tabulated for comparison (Table 5.4).

Table 5.4: Comparison of antibody pretreatment effects on KHYG-1 cytotoxicity and

synergy assessment at 0.01 µg/ml

Cell line % Lysis (mean +/- SD)

Pretreatment

0.01 µg/ml 0.1 µg/ml

None MG2a-

53

Anti-

NKp30

Anti-

NKp44

Anti-

NKp44/

NKp30

Anti-

NKp30

Anti-

NKp44

K562 21

+/- 1.6

29

+/- 2.4

28

+/- 6.0

26

+/-2.5

32

+/- 2.0

35

+/- 0.5

41

+/- 3.4

OCI/AML3 4

+/- 0.1

7

+/- 1.8

7

+/-0.8

7

+/- 0.6 13*

+/- 3.8

13

+/- 0.5

19

+/- 3.4

OCI/AML5 12

+/- 0.8

16

+/- 5.0

38

+/- 1.9

30

+/- 3.8

36

+/- 7.6

65

+/- 1.5

63

+/- 5.4

KG1 9

+/- 7.4

9

+/- 1.2

6

+/- 1.0

9

+/- 4.7

11

+/- 1.0

9

+/- 1.1

15

+/- 4.5

Combined anti-NKp30 and anti-NKp44 regimens that yielded statistically significant (p<0.05)

increases above each antibody alone in separate comparisons are in bold font.

Page 159: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

139

A

0

10

20

30

40

50

60

70

80

90

100

K562 OCI/AML3 OCI/AML5

% L

ysis

Cell line

None

NKp30 (0.01)

NKp30 (0.1)

NKp30 (1)

NKp44 (0.01)

NKp44 (0.1)

NKp44 (1)

NKp30/NKp44 (0.01)

B

0

10

20

30

40

50

60

70

80

90

100

K562 OCI/AML3 OCI/AML5

% L

ysis

Cell line

None

NKp30 (0.01)

NKp30 (0.1)

NKp30 (1)

NKp44 (0.01)

NKp44 (0.1)

NKp44 (1)

NKp30/NKp44 (0.01)

Figure 5.8: Effect of antibody pre-treatment with anti-NKp30 and anti-NKp44 on NK-92

and KHYG-1 cytotoxicity against leukemia cell lines and primary AML

NK-92 (A) and KHYG-1 (B) were pretreated without or with anti-NKp30 and anti-NKp44

antibodies at 0.01, 0.1, 1 µg/ml (C and D), incubated for one hour and then washed in AIM-V

medium to remove residual antibody. Effector cells were utilized in a standard chromium

release assay against K562, OCI/AML3 and OCI/AML5 with 100 µCi of Na251

CrO4at an E:T

ratio of 10:1. Data are presented as the mean percent lysis of triplicate samples (+/-SD).

Page 160: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

140

To determine if anti-NKp30 and anti-NKp44 pretreatment of NK cell lines could enhance

cytotoxicity against a solid tumour, we performed the same experiment with esophageal cancer

cell lines (FLO-1, OE-33, SKGT-4, KYAE-1). However, pretreatment of NK-92 and KHYG-1

with 0.1 µg/ml of anti-NKp30 and anti-NKp44 mAb did not enhance cytotoxicity against four

esophageal cancer cells lines relative to the isotype control (Figure 5.9). This suggested the

presence of a unique cell surface marker present on leukemia cells, but not esophageal cancer

cells, that was mediating the enhancing effect of antibody-pretreated NK cell lines.

Page 161: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

141

A

0

10

20

30

40

50

60

70

80

90

100

K562 FLO-1 OE-33 SKGT-4 KYAE-1

% ly

sis

Cell line

None

MG1-45 (0.1)

NKp30 (0.1)

NKp44 (0.1)

B

0

10

20

30

40

50

60

70

80

90

100

K562 FLO-1 OE-33 SKGT-4 KYAE-1

% ly

sis

Cell line

None

MG1-45 (0.1)

NKp30 (0.1)

NKp44 (0.1)

Figure 5.9: Impact of antibody pre-treatment with anti-NKp30 and anti-NKp44 on NK-92

and KHYG-1 cytotoxicity against esophageal cancer cell lines

NK-92 (A) and KHYG-1 (B) were pretreated with isotype control MG1-45 or anti-NKp30 and

anti-NKp44 antibodies at 0.1 µg/ml, incubated for one hour and then washed in AIM-V medium

to remove residual antibody. Effector cells were utilized in a standard chromium release assay

against K562 and esophageal cancer cell lines FLO-1, OE-33, SKGT-4, KYAE-1 and labelled

with 100 µCi of Na251

CrO4at an E:T ratio of 10:1. Data are presented as the mean percent lysis of

triplicate samples (+/-SD).

Page 162: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

142

5.4.5 Relationship of Fcγ receptor expression and enhancement of

cytotoxicity

We conducted HTS flow cytometry on two representative leukemic cell line targets (OCI/AML3

and OCI/AML5) to assess for potential cell surface markers that might be involved in enhancing

the cytotoxicity of anti-NKp30 or anti-NKp44 coated NK cell lines (Appendix III and IV)I). We

noted a high degree of CD32 (FcγRII) expression on both cell lines. Subsequently, we

conducted routine flow cytometry on all leukemic and esophageal cancer cell lines for

expression all Fcγ receptors (CD16, CD32, CD64). The leukemia cell lines showed relatively

high expression of Fcγ receptor II (CD32), but very low expression of Fcγ receptor I (CD64) or

Fcγ receptor III (CD16) on leukemia lines (K562, KG1, KG1a OCI/AML3, OCI/AML5) (Figure

5.10). The histogram shape for K562 suggested the presence of both intermediate and high

CD32 expressing subpopulations. KG1a appeared to have a dual population of negative and low

CD32 expressing subpopulations. OCI/AML3 had a clear dual peak representing two high CD32

expressing subpopulations. There were clear single populations of CD32 expressing cells in

KG1 (moderate) and OCI/AML5 (high). There was no significant expression of Fcγ receptors

on esophageal cancer lines (OE-33, FLO-1, KYAE-1, SKGT-4) (Figure 5.11).

Page 163: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

143

A

B

C

D

E

Figure 5.10: Fc gamma receptor expression on leukemia cell lines (K562, KG1, KG1a,

OCI/AML3, OCI/AML5)

Leukemia cell lines K562 (A), KG1 (B), KG1a (C), OCI/AML3 (D), OCI/AML5 (E) were tested

for CD16, CD32 and CD64 expression by flow cytometry. Unstained (red) and stained (blue)

populations are presented in the histogram.

Page 164: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

144

A

B

C

D

Figure 5.11: Fc gamma receptor expression on esophageal cancer cell lines (OE-33, FLO-1,

KYAE-1, SKGT-4)

Esophageal cancer cell lines were tested for CD16, CD32 and CD64 expression by flow

cytometery. Unstained (red) and stained (blue) populations are presented in the histogram.

Page 165: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

145

The percent positivity of each leukemic and esophageal cancer cell line was determined from

flow cytometry plots (Figure 5.10 and Figure 5.11). Data from prior cytotoxicity assays

measuring the enhancement of cytotoxicity of NK-92 and KHYG-1 when pretreated with 10

µg/ml of either anti-NKp30 or anti-NKp44 antibody were compared to isotype controls and delta

cytotoxicity calculated. The delta cytotoxicity relative to isotype control was correlated with the

degree of CD32 expression using regression analysis to create best fit lines, calculate co-efficient

of determination (R2) and statistical significance. Regression analysis of the relationship of delta

cytotoxicity of antibody-pretreated NK-92 with CD32 expression of targets did not reveal a

correlation for anti-NKp30 (R2=0.13; p=0.34) and anti-NKp44 (R

2=0.22; p=0.20) pretreatments

(Figure 5.12A and B). However, regression analysis of the relationship of delta cytotoxicity of

antibody pretreated KHYG-1 with CD32 expression of targets revealed a strong correlation for

both anti-NKp30 (R2=0.71; p<0.01) and anti-NKp44 pretreatments (R

2=0.64; p<0.01) (Figure

5.12C and D).

Page 166: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

146

Figure 5.12: Regression analysis of CD32 expression and delta cytotoxicity of NKp30 and

NKp44 pretreated NK-92 and KHYG-1

Relation between delta cytotoxicity of isotype control versus NKp30 or NKp44 pretreated NK-

92 (A and B) and KHYG-1 (C and D) against leukemic and esophageal cancer cell lines was

determined by using linear regression analysis. Scatter plots with best fit line are presented with

coefficient of determination (R2) calculated and F test for significance performed. NK-92

pretreatments: anti-NKp30 (A) (R2=0.13; p=0.34) and anti-NKp44 (B) (R

2=0.22; p=0.20).

KHGY-1 pretreatments: anti-NKp30 (C) (R2=0.71; p<0.01) and anti-NKp44 (D) (R

2=0.64;

p<0.01).

Page 167: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

147

5.4.6 Effect of anti-NKp30 pretreatment on NK cell line cytotoxicity

against clonogenic OCI/AML5

To determine the effect of anti-NKp30 and anti-NKp44 pretreated NK cell lines against

clonogenic leukemic cells, we utilized our previously established clonogenic cytotoxicity assay

utilizing OCI/AML5 as the target. Comparison of killing was made with untreated, isotype

control (MG1-45) and anti-CD7 pretreated NK cell lines. CD7 is highly expressed on NK-92

and KHYG-1, with no confirmed activating capacity in these cell lines. Therefore, anti-CD7

antibody pretreatment was chosen as an additional control. There was no difference between

isotype control and CD7-pretreated NK-92. Pretreatment of NK-92 with 0.1 µg/ml anti-NKp30

had only a slight impact (+3.7%; p<0.05) on OCI/AML5 clonogenic capacity relative to baseline

and isotype control (Figure 5.13A). However, pretreatment of KHYG-1 with anti-NKp30

enhanced % colony inhibition 3-fold (+63.1%; p<0.0001) relative to baseline and isotype

control. While the baseline % colony inhibition of NK-92 (61.9 %) was greater than KHYG-1

(32.0%), anti-NKp30 pretreated KHYG-1 (90.7%) had the greater inhibition relative to NK-92

(+28.8%; p<0.0001).

A

Page 168: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

148

B

*

Figure 5.13: Methylcellulose cytotoxicity assay of NK-92 and KHYG-1 +/- pretreatment

with antibodies against OCI/AML5

NK-92 (A) and KHYG-1 (B) were pretreated without (no pretreatment) and with monoclonal

antibodies (MG1-45, anti-CD7, anti-NKp30) at 0.1 µg/ml for 1 hour prior to use in

methylcellulose cytotoxicity assay (MCA) at a 10:1 E:T ratio. OCI/AML5 cells were incubated

with or without effectors prior to infusion into methylcellulose. Plates were incubated at 37°C

with 5% CO2 for two weeks. Colonies were then enumerated by using an inverted microscope

defining a colony as a cell cluster of >50 cells. Cytotoxicity was measured relative to the low

density control (LDC) using untreated effector cells which yielded similar numbers of colonies

to OCI/AML5 cells incubated alone. % colony inhibition was calculated using the following

formula: [(#ColoniesLDC)- (#ColoniesTreatment)] x100% [#ColoniesLDC]. Data are presented

as the mean percent lysis of triplicate samples (+/-SD) representative of two separate

experiments. Statistical comparison was done using a student’s test between no pretreatment, or

isotype control (MG1-45), and anti-NKp30 pretreated groups (* =p<0.0001).

Page 169: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

149

5.4.7 In vitro effect of anti-NKp30 pretreated iKHYG-1 against

OCI/AML5 capacity for leukemic progression in an NSG xenograft

model

We tested the in vitro cytotoxic effect of KHYG-1 on in vivo progression of leukemia in an

OCI/AML5 xenograft model. KHYG-1 proliferation was prevented by irradiation with 1000

cGy prior to use. OCI/AML5 cells were co-incubated with irradiated KHYG-1 (iKHYG-1) +/-1

µg/ml anti-NKp30 pretreatment for one hour prior to a 4 hour co-incubation at a 10:1 E:T with

OCI/AML5 cells. Subsequently, cell mixtures were injected ip into three cohorts of NSG mice

with survival as an endpoint. Individual NSG mice were injected with 2x106 OCI/AML5 cells

+/- 20x106 viable effector cells (iKHYG-1 or anti-NKp30 pretreated iKHYG-1). At 9 weeks,

control mice developed progressive malignant ascites with minimal splenomegaly and imbedded

vascular tumours in the omentum (Figure 5.14). Co-incubation with iKHYG-1 did not improve

survival (p=0.92). However, anti-NKp30 pretreated iKHYG-1 improved survival compared to

the no therapy (p<0.05) or iKHYG-1 (p<0.05) cohorts (Figure 5.15).

Page 170: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

150

A B

DC

Figure 5.14: OCI/AML5 induced malignant ascites

2x10e6 OCI/AML5 cells were injected ip into NSG mice and monitored for progression of

leukemia. NSG mice developed massive malignant ascites (A and B) with development of

inflamed omental tumours (C red arrow), but minimal increase in spleen size (D red arrow).

Page 171: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

151

Group

OCI/AML5

OCI/AML5 + iKHYG1

OCI/AML5 + NKp30 iKHYG1

Survival

0 20 40 60 80 100

100

80

60

40

20

0

Time (days)

Surv

ival p

robability

(%

)

Figure 5.15: In vitro incubation of OCI/AML5 with iKHYG-1 +/- anti-NKp30 and in vivo

proliferation in NSG mice

OCI/AML5 cells were incubated in 15 ml conical tubes with or without iKHYG-1 (+/- 1 ug/ml

anti-NKp30 pretreatment x 1 hour) at a 10:1 E:T ratio, spun at 1200 rpm to pellet and incubated

for four hours at 37°C. Cell mixtures were then washed and resuspended in PBS and 2x10e6

OCI/AML5 cells with or without 20x106 iKHYG-1 or NKp30 iKHYG-1 cells were injected ip

into three cohorts of five NSG mice. Mice were sacrificed at humane endpoint and Kaplan

Meier curves generated and compared with the log rank test (p<0.05).

Page 172: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

152

5.4.8 Effect of anti-NKp30 pretreatment of iKHYG-1 on therapeutic

efficacy for OCI/AML5 or primary AML xenografted mice

We evaluated OCI/AML5 engraftment potential in NSG mice by infusing 5x106 OCI/AML5

cells via tail vein and measured bone marrow engraftment at two weeks. Bone marrow

engraftment of OCI/AML5 was detected by measuring human CD33 expression in bone marrow

samples (Figure 5.16) and revealed relatively rapid, but variable bone marrow engraftment (13.0,

52.3, 29.9, 63.5%). In a subsequent survival endpoint experiment, 2x106 OCI/AML5 cells were

injected iv into cohorts of five mice. OCI/AML5-xenografted mice were then treated without

and with iKHYG-1 or anti-NKp30 pretreated iKHYG-1 (10x106 x 6 doses ip). There was

significant improvement in survival of mice treated with either iKHYG-1 (+35 days median

survival; p<0.05) or anti-NKp30 pretreated iKHYG-1 (+37 day median survival p<0.05) above

control. The two day difference in median survival of mice treated with anti-NKp30 pretreated

iKHYG-1 over iKHYG-1 alone was not significant, although the longest surviving mouse was in

the anti-NKp30 pretreated iKHYG-1 group (Figure 5.17).

Figure 5.16: Bone marrow engraftment of OCI/AML5 injected iv into NSG mice

5x106 OCI/AML5 cells were injected via tail vein into four NSG mice. Mice were sacrificed at 3

weeks and bone marrow harvested, stained with anti-CD33 antibody and measured using flowing

cytometry. Flow cytometry plots represent bone marrow from each mouse showing unstained

(red) and anti-CD33 stained (blue) samples.

CD33+

13.0

1 0- 1

1 00

1 01

1 02

1 03

FL4

0

2 0

4 0

6 0

8 0

100

Co

un

t

CD33+

52.3

1 0- 1

1 00

1 01

1 02

1 03

FL4

0

2 0

4 0

6 0

8 0

100

Co

un

t

CD33+

29.9

1 0- 1

1 00

1 01

1 02

1 03

FL4

0

2 0

4 0

6 0

8 0

100

Co

un

t

CD33+

63.5

1 0- 1

1 00

1 01

1 02

1 03

FL4

0

2 0

4 0

6 0

8 0

100

Co

un

t

Page 173: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

153

GroupiKHYG-1iKHYG-1 NKp30OCI/AML5 only

Survival

0 10 20 30 40 50 60 70 80 90

100

80

60

40

20

0

Time

Surv

ival p

robabili

ty (

%)

Figure 5.17: Treatment of OCI/AML5 leukemia in NSG mice with iKHYG-1 +/- NKp30

pretreatment

2x106 OCI/AML5 were injected iv on day 0 into three cohorts of mice that were subsequently

treated as follows: no treatment, iKHYG-1 or anti-NKp30 pretreated iKHYG-1. Treatment

started on day 3 (10x106 x 6 doses ip; days 3, 5, 7, 10, 12, 14). Mice were sacrificed at humane

endpoints and Kaplan Meier curves generated and compared with the log rank test.

Page 174: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

154

We utilized a primary AML sample (080179) known to engraft and cause leukemia in NSG mice

as a model to test the efficacy irradiated NK-92 and KHYG-1 pretreated with and without anti-

NKp30 (1 ug/ml) prior to injection into NSG mice inoculated with either primary AML.

iKHYG-1 and iNK-92 did not prolong survival in the primary AML model, although iKHYG-1

pretreated with anti-NKp30 showed some longer term survivors (3-4 weeks above control

median) with a trend toward significance (p=0.20) versus iKHYG-1 alone (Figure 5.18).

A

GroupControliKHYG-1iKHYG-1 NKp30

Survival

0 10 20 30 40 50 60 70

100

80

60

40

20

0

Time (days)

Surv

ival p

robabili

ty (

%)

Figure 5.18: Treatment of primary AML xenografted NSG mice with iKHYG-1 +/- NKp30

pretreatment

Primary AML cells (3x106) were injected iv on day 0 with and without iKHYG-1 or NKp30

iKHYG-1 ip treatment started on day 3 (10x106 x 6 doses; days 3, 5, 7, 10, 12, 14). Mice were

sacrificed at humane endpoint and Kaplan Meier curves generated and compared with the log

rank test.

Page 175: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

155

5.5 Discussion

Adoptive immunotherapy with NK cells represents a novel emerging treatment modality for

patients with AML with several established protocols for allogeneic NK cell therapy for

AML.(Miller, Soignier et al. 2005; Rubnitz, Inaba et al. 2010) Anti-cancer cell-based

immunotherapy with a standardized, highly cytotoxic NK cell line is an attractive alternative to

autologous or allogeneic NK cells with their attendant variability in cytotoxicity and cell

manufacturing characteristics. The permanent, malignant NK line, NK-92 has been tested in

phase I trials and shown to have minimal toxicities (Arai, Meagher et al. 2008; Tonn, Schwabe et

al. 2013), while KHYG-1 has only been assessed in preclinical studies. Here, we have

conducted all experiments with KHYG-1 using a clinical grade medium (GM1) that lacks fetal

bovine serum and may be suitable for future clinical application. To further characterize both

NK cell lines and provide insight into mechanisms of cytotoxicity, high throughput screening

flow cytometry assaying for 374 distinct cell surface markers was conducted and provides the

most extensive immunophenotypic assessment of these cell lines to date. Prior to this study,

only ~30 receptors were reported for NK-92(Maki, Klingemann et al. 2001) and several for

KHYG-1 (Suck, Branch et al. 2005).

We confirmed previous findings that NK-92 and KHYG-1 are cytotoxic to leukemic cell lines

(Yan, Steinherz et al. 1998; Tonn, Becker et al. 2001; Suck, Branch et al. 2005; Williams, Wang

et al. 2010). We sought initially to determine basic mechanisms of cytotoxicity of these cell

lines and determined that NK-92 and KHYG-1 cytotoxicity was mediated primarily by granule

exocytosis as evidenced by minimal killing of leukemic cell line or primary AML blasts in the

presence of the calcium chelator EGTA. Calcium influx is a key step in the process of granule

exocytosis (Maul-Pavicic, Chiang et al. 2011), which can be specifically blocked by chelation of

calcium from the medium using EGTA without altering other cellular pathways. We observed

however, a small amount of residual killing after EGTA treatment of KHYG-1 at low E:T ratios

implicating ligand-mediated killing not seen with NK-92. Our high throughput flow cytometry

screen of KHYG-1 demonstrated the presence of all ten tumour necrosis factor family members

tested for with high expression of Fas ligand and TRAIL, both major ligands involved in

apoptosis induction by T and NK cells. NK-92 expressed nine tumour necrosis factor receptors

with equivalent Fas Ligand expression to KHYG-1, but lower TRAIL expression.

Page 176: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

156

While cytotoxicity of NK-92 against primary AML has been established (Yan, Steinherz et al.

1998), KHYG-1 cytotoxicity against primary AML has not been previously reported. We noted

that KHYG-1 was less effective than NK-92 at killing both leukemia cell lines and primary AML

samples and sought to determine the basis for this finding. KIRs present on NK cells recognize

class I HLA and can be either activating or inhibiting (Velardi 2008). Presence of an inhibitory

KIR ligand, particularly HLA-C alleles can suppress NK cytolytic function. Further, KIR

receptor-ligand mismatch has been a factor associated with improved survival outcomes in

haplotype hematopoietic marrow transplantation for AML in some, indicating that NK cells can

mediate a powerful graft-versus-leukemia effect(Ruggeri, Capanni et al. 2002; Ruggeri, Capanni

et al. 2005).

It was initially reported that NK-92 lacked inhibitory killer immunoglobulin-like receptors

(KIRs)(Tonn, Becker et al. 2001), but we had previously demonstrated that NK-92 expresses

mRNA for KIR- 2DL4, 2DL5, 3DL1 and 3DL3 and KHYG-1 expresses mRNA for KIR- 2DL3,

2DL4, 2DL5, 3DL1, 3DL2, 3DL3(Suck, Branch et al. 2005). Among these receptors, all are

inhibitory, with the exception of KIR2DL4, which has been shown to have both activating and

inhibiting function (Faure and Long 2002). However, RT-PCR measures mRNA levels which

may not be translated to protein or be transported to the cell surface, so we therefore utilized

HTS flow cytometry to analyze the receptor profiles of NK-92 and KHYG-1, to assist in

determining the reason for differential killing against AML. Our HTS flow cytometry did not

detect significant levels of inhibitory KIRs on the surface of NK-92 consistent with previous

work(Tonn, Becker et al. 2001) with the exception of KIR2DL4. HTS flow cytometry of

KHYG-1 revealed expression of low levels of KIR-2DL3, 2DL5, high levels of KIR2DL4, but

no 3DL1, demonstrating that presence of KIR mRNA does not always translate into cell surface

expression of the protein. We did not detect significant levels of KIR2DL1 and KIR2DL2 on

KHYG-1 as previously reported (Matsuo 2003), which may have been related to differing culture

conditions.

The ligands for the inhibitory KIR receptors expressed on KHYG-1 are: KIR2DL3 (HLA-C1),

KIR2DL4 (HLA-G), KIR2DL5 (unknown). We postulated that exclusive KHYG-1 expression

of KIR2DL3 binding to HLA-C1 might explain the poor baseline cytolytic function of KHYG-1

relative to NK-92. We therefore blocked class I HLA on AML cell lines to determine if this or

other undefined receptors involved in HLA recognition might be involved in recognition. Class I

Page 177: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

157

HLA blocking was done using antibody from clone W6/32, which results in a pan-HLA class I

blockade including HLA-E(Wooden, Kalb et al. 2005). Blockade of class I HLA of primary

AML target cells did not enhance NK-92 killing, consistent with the minimal inhibitory KIR

expression. However, class I blockade resulted in improved killing by KHYG-1 of one primary

AML sample tested (080008), suggesting blockade of an inhibitory KIR receptor on KHYG-1.

We obtained the HLA typing of these primary AML samples to determine consistency with the

KIR receptor-ligand hypothesis. Sample 080008 was homozygous for HLA-C2, which would

typically engage the inhibitory receptor KIR2DL1, but this receptor is not expressed on KHYG-

1. Therefore, the enhancement following blockade of class I HLA cannot be explained by

interference with an inhibitory signal from KIR2DL1. Sample 080179, 5890 and 0909 were

homozygous for HLA C1 and would be expected to mediate inhibition of KHYG-1 via KIR

2DL3, which could potentially be impacted by HLA class I blockade, but there was no change in

cytotoxicity with target class I blockade. KHYG-1 did have a low expression of KIR2DL3, but

antibody blockade of this receptor did not result in an increase in cytotoxicity against primary

AML sample 0909 which was homozygous for the HLA-C1 group, the natural ligand of this

KIR. KHYG-1 also does not express KIR3DL1 that engages HLA-Bw4 group. However,

KHYG-1 does express mRNA for KIR3DL2, which was not included in the HTS flow cytometry

screen and therefore may be expressed by KHYG-1. However, KIR3DL2 only engages with

HLA-A3 and -A11, making it an unlikely candidate receptor to explain the results with sample

080008.

HLA-G could in theory inhibit KHYG-1 via KIR2DL4, but is predominately expressed on the

placenta and serves to down modulate NK cell cytotoxicity at the materno-fetal interface. HLA-

G was not been detected on acute leukemias in one study (Polakova, Krcova et al. 2003), but was

reported on 68% of primary AML samples in another study.(Locafaro, Amodio et al. 2014)

NKG2A, when engaged by HLA-E can mediate an inhibitory signal.(Lee, Llano et al. 1998)

KHYG-1 does express NKG2A, and blockade of HLA E using W6/32 monoclonal antibody

could impact cytotoxicity if 080008 expresses HLA-E. HL-60 cells (an acute promyelocytic cell

line) express HLA-E (Marin, Ruiz-Cabello et al. 2003), while Majumber et al. demonstrated a

lack of expression of HLA-E on primary leukemic cells (Majumder, Bandyopadhyay et al.

2006). Another study demonstrated that interferon-γ was required to induce primary AML cells

to express HLA-E, leading to inhibition of NK cell-mediated cytolysis.(Nguyen, Beziat et al.

Page 178: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

158

2009). Therefore, it is conceivable that sample 080008 expresses HLA-E, which could mediate

an inhibitory effect that when blocked by anti-class I HLA monoclonal antibody, could enhance

cytotoxicity.

We then sought to determine the effect of pretreating NK-92 and KHYG-1 with antibodies

against a panel of activating receptors commonly associated with NK cell cytotoxicity (natural

cytotoxicity receptors, NKG2D and DNAM-1). While we anticipated potential blocking of

cytotoxicity at 10 µg/ml against a panel of leukemia cell line targets, we observed stimulation of

cytotoxicity from some of the antibodies and no blocking of cytotoxicity. Treatment of NK-92

with antibodies to NKp30, NKp44, NKp46 increased killing of K562 by approximately 10%,

while only NKp30 treatment enhanced killing of KG1a and OCI/AML5. Further, anti-NKp30,

but not anti-NKp44 pretreated NK-92 had a large degree of enhancement in killing of primary

AML. Previously, we detected expression of NKp30 on NK-92 and found it to mediate

recognition of multiple myeloma cells (Swift, Williams et al. 2012), which was reduced when

NK-92 was blocked by this anti-NKp30 antibody (P30-15 clone). The discrepancy of activity of

this antibody prompted us to consider the mechanism of enhancement against leukemic targets.

HTS flow cytometery demonstrated higher expression of NKp30, NKp44 and NKp46 on

KHYG-1 compared with NK-92. Treatment of KHYG-1 with antibodies to all the natural

cytotoxicity receptors increased killing of K562 and OCI/AML5 to a greater degree than NK-92.

KHYG-1 cytotoxicity against primary AML was enhanced to a greater degree than NK-92 with

pretreatment with anti-NKp30 or anti-NKp44. Pretreatment of NK cell lines with antibodies

against DNAM-1, NKG2D (both commonly involved in NK cell recognition) induced small

statistically significant inhibitory effects against target K562 in some experiments, indicating a

possible role for these molecules in recognition. However, anti-DNAM-1 and anti-NKG2D were

not able to facilitate reverse ADCC despite high expression of DNAM-1 and NKG2D on both

cell lines.

We sought to determine the dose response curve of antibody-mediated cytotoxic enhancement,

which was most prominent with KHYG-1 against OCI/AML5. Increases in KHYG-1

cytotoxicity against OCI/AML5 was seen as low as 0.01 µg/ml, which is near the EC50 of the

stimulatory effect. A plateau in enhancement started to occur at 0.1 µg/ml with slight increase in

efficacy at 1 µg/ml. NK-92 had lesser enhancement than KHYG-1 with this approach, but

Page 179: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

159

effects could be observed in the 0.01 µg/ml dose range particularly against OCI/AML3, which is

somewhat resistant to NK-92 cytotoxicity.

While the antibodies we used were blocking antibodies, based on studies of endogenous NK cell

cytotoxicity(Markel, Seidman et al. 2009), we postulated that the increased killing was mediated

by reverse-antibody dependent cell-mediated cytotoxicity (R-ADCC). R-ADCC results when an

antibody fixed via the Fab fragment to an effector cell activating receptor can become

crosslinked by engagement with Fc gamma receptors on the target cell. We further tested this

approach with a panel of esophageal lines, but did not observe any significant increase in

cytotoxicity after pretreatment of NK-92 or KHYG-1 with anti-NKp30 or anti-NKp44.

Regression analysis of the delta cytotoxicity with CD32 expression of the cell line targets yielded

highly significant correlation for KHYG-1, but not for NK-92. However, NK-92 had higher

baseline killing against some cell line targets relative to KHYG-1 and had less potential for

enhancement, particularly for OCI/AML5. Also, NK-92 had lower expression of NKp30 than

KHYG-1, and minimal expression of NKp44, which may have affected the degree of

enhancement possible by R-ADCC against leukemia cell lines. However, there was a high

degree of enhancement of NK-92 pretreated with anti-NKp30 against primary AML targets. The

regression analysis supported reverse ADCC via CD32 as the mechanism of the enhancement for

KHYG-1 cytotoxicity against the cancer cell line panel. Given that the enhancing effect of anti-

NKp30 and anti-NKp44 was not generic in nature makes another mechanism such as co-

activation or co-stimulation unlikely. Further, the low concentrations of antibody that could

elicit this effect and the high magnitude of enhancement were consistent with R-ADCC which

can occur <0.1 µg/ml and lead to several- fold increases in cytotoxicity against resistant targets.

NK cells were first implicated in mediating reverse ADCC by Saxena et al.(Saxena, Saxena et al.

1982) The cell lines and primary AML samples we tested were predominantly FAB M4

(myelomonocytic) and M5 (monocytic), which typically express Fc gamma receptors. This

enables them to bind the Fc portions of antibodies, which in turn allows for reverse ADCC to

occur in the context of our assay. Reverse ADCC has been primarily demonstrated using P815

murine mastocytoma cell lines in combination with NK cells pretreated with antibodies against

unknown or known NK cell antigens, to determine novel receptors, and this method allowed for

the discovery of NKp30. (Pende, Parolini et al. 1999). Subsequently, the approach was used to

discover and characterize the NKp44(Vitale, Bottino et al. 1998) and NKp46(Pessino, Sivori et

Page 180: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

160

al. 1998) receptors. Antibody pretreatment of NK cells with antibodies directed at activating

receptors can allow them to become crosslinked followed binding to an Fcγ receptor positive

target cell. The effect can be very potent in facilitating lysis against resistant P815 cells, with

effects as low as 0.001 µg/ml reported.(Pende, Parolini et al. 1999) While reverse ADCC has

been described in this context, it has never been shown to function in vivo. Ball et al. conducted

a study of the expression of Fcγ receptors on primary AML and noted the following frequencies:

FcγRI (58%); FcγII, (67%); and Fcγ III, (26%). FcγI and II receptor expression was highly

correlated with FAB M4 and M5 morphology. Notter et al. demonstrated that precoating

lymphokine activated killer (LAK) cells with anti-CD3 antibodies could enhance killing of

autologous AML blasts through reverse ADCC via FcγRI (CD64).(Notter, Ludwig et al. 1993)

This was established by showing a 1.5-9.3 fold increase in killing that was dependent on CD64

expression of targets and could be partially reduced by blocking FcγRs with very high doses of

murine IgG2a or human monomeric IgG. In this study, CD34+ hematopoietic stem cells did not

express FcγRs and were not subject to enhanced killing by anti-CD3 coated LAK cells. R-

ADCC was not tested in vivo, but the authors proposed a combination of IL-2, IFN-γ and anti-

CD3 monoclonal antibody as a potential treatment for AML. However, here we demonstrate R-

ADCC as a means of enhancing NK cell line cytotoxicity against leukemic cell lines and, more

importantly, primary AML cells. The combination of antibody and NK cell line together in this

manner provides a potentially novel leukemia therapy.

While this is the first time NK-92 and KHYG-1 have been shown to mediate reverse ADCC,

gene modified CD16+NK-92(Binyamin, Alpaugh et al. 2008) and CD16+KHYG-1(Kobayashi,

Motoi et al. 2014) have been generated and are capable of mediating ADCC against CD20+

targets in combination with Rituximab. However, for CD16+KHYG-1, the lowest effective

concentration of Rituximab was 0.1 µg/ml, while for anti-NKp30 or anti-NKp44 mediated

reverse ADCC the dose was less than 0.01 µg/ml, demonstrating the relative potency of reverse

ADCC.

We then sought to determine if the enhancing effect of pretreating NK cell lines with anti-

NKp30 and anti-NKp44 held against clonogenic cells by using our established methylcellulose

cytotoxicity assay. We noted that NK-92 was relatively effective at inhibiting OCI/AML5

colony formation, but this could not be enhanced by pretreatment with anti-NKp30 over isotype

control. However, KHYG-1 was less effective at inhibiting OCI/AML5 colony formation, but

Page 181: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

161

this could be enhanced three-fold by pretreatment with anti-NKp30 antibody. The inhibition of

colonies indicates a cytotoxic or cytostatic effect on clonogenic cells within the cell line

populations that represent leukemic stem and progenitor cells. Therefore, this provides indirect

evidence that reverse ADCC can facilitate cytotoxicity against leukemic stem cells.

To evaluate the impact of NK cell line therapy in vivo, we established an OCI/AML5 xenograft

model in NSG mice. OCI/AML5 was derived from a patient with M4 leukemia, and highly

expresses CD33, which is useful for tracking in vivo.(Wang, Koistinen et al. 1991) This cell line

was used previously in a NOD/SCID mouse model to evaluate Indium111

conjugated anti-CD123

antibody therapy.(Leyton, Hu et al. 2011) We confirmed injection of OCI/AML5 iv led to

leukemia with splenomegaly and bone marrow engraftment. However, ip injection tended to

cause progressive malignant ascites over a longer timeframe, rather than classic leukemia.

To determine the effect of in vitro cytotoxicity on in vivo proliferation, we incubated OCI/AML5

with or without iKHYG-1 (+/-anti-NKp30 pretreatment) and injected the cells ip. We utilized

this injection route instead of iv, because of the high cell load (2x106 OCI/AML5 + 20x10

6

viable iKHYG-1 + ~3x106 non-viable iKHYG-1) and relatively larger KHYG-1 cells, which

might have caused pulmonary stress to the mice if injected via tail vein. Co-incubation with

iKHYG-1 had no impact, while anti-NKp30 pretreated iKHYG-1 improved median survival by

10 days.

We subsequently utilized the iv injection OCI/AML5 NSG xenograft model to test therapeutic

efficacy of iKHYG-1 with or without anti-NKp30 pretreatment. Unexpectedly, iKHYG-1 was

able to improve the median survival by 35 days, despite its poor cytotoxicity in the CRA against

bulk OCI/AML5. However, KHYG-1 had three-fold better cytotoxicity against clonogenic

OCI/AML5 than bulk OCI/AML5, as determined by the CRA, providing some basis for this

finding. This is the first demonstration of efficacy by KHYG-1 in an in vivo cancer model and

confirms that the irradiated cells can persist and reduce tumour burden. While it has been shown

that irradiation has minimal effects on cytotoxicity in vitro (Suck, Branch et al. 2006), this is the

first evidence that the irradiated cells can function in vivo. However, pretreatment of iKHYG-1

with anti-NKp30 did not enhance survival over iKHYG-1-treated OCI/AML5-xenografted NSG

mice. We postulate that this is due to antibody dissociating in vivo, given that no iv doses of

antibody were given to the mice. We also used a primary AML xenograft model to test iKHYG-

Page 182: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

162

1 and anti-NKp30 iKHYG-1, demonstrating lack of efficacy of iKHYG-1, but a trend to

improved survival in the anti-NKp30 iKHYG-1 treated group.

In summary, we have conducted an extensive characterization of the cell surface expression of

the two most potent NK cell lines with therapeutic potential, using high throughput screening

flow cytometry identifying key molecules involved in adhesion, activation and inhibition of

cytotoxicity as well as apoptosis inducing ligands. We demonstrate that NK-92 and KHYG-1

have cytotoxicity against a broad range of leukemic targets that can be enhanced several-fold by

by anti-NKp30 and anti-NKp44 antibodies. For KHYG-1, reverse ADCC has been implicated at

the mechanism of enhancement via interaction of antibody coated effectors with FcγRII (CD32)

on the target cells. Furthermore, NKp30 mediated reverse ADCC can enhance cytotoxicity of

KHYG-1 against clonogenic leukemic cells and affect in vivo proliferation of leukemia. Further

studies of combination of anti-NKp30 antibody pretreatment and NK cell line therapies are

warranted.

Page 183: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

163

6 Chapter 6: General Discussion

6.1 Overview

Cancer immunology has focused primarily on the interaction of the immune system with bulk

tumour cells. This has related to both a historical conceptual understanding of tumour biology,

and limitations in methodologies to address heterogeneity within tumours, as it relates to immune

cell recognition and cytotoxicity. While tumour heterogeneity was noted in the 19th

century by

Rudolph Virchow,(Brown and Fee 2006) the connection with stem cell biology occurred much

later. However, as early as 1937, leukemia was demonstrated as clonal, driven by rare leukemia-

initiating cells.(Furth, Kahn et al. 1937) The discovery of hematopoietic stem cells(McCulloch

and Till 1960) led to a paradigm shift in thinking about cell biology and initial work supported

that cell fate decisions were stochastic(Till, McCulloch et al. 1964). Around this time, these

concepts were applied to cancer progression, with a cogent leukemia stem cell hypothesis being

postulated as an alternative to a stochastic model(Bruce and Ash 1963). The formal

demonstration of the existence of rare leukemic stem cells(Lapidot, Sirard et al. 1994) opened

the door to questions about how the immune system interacts with these rare cancer stem cells.

This work also called into question the conclusions made from work done using bulk tumour

cells in cancer immunology (Williams, Swift et al. 2013), and other fields of cancer research

such as drug discovery(Williams, Anderson et al. 2013). Here we have investigated the natural

killer cell lines, NK-92 and KHYG-1, against leukemia utilizing techniques to address the impact

on bulk and leukemic stem cells. Both of these cell lines can preferentially target leukemic stem

cells as demonstrated by comparing a novel in vitro clonogenic cytotoxicity assay with the

standard chromium release assay. These findings can be extended in vivo, with NK-92 and

KHYG-1 capable of prolonging survival in AML xenograft models. Using gene-modified NK-

92 expressing the high affinity Fc gamma receptor, LSCs can be targeted by ADCC in vitro

using anti-CD123 monoclonal antibodies, which also can be combined in vivo to improve

survival in a primary AML xenograft model. While KHYG-1 has a lower baseline ability to kill

leukemic targets than NK-92, it can be significantly enhanced when pretreated with anti-NKp30

and NKp44 monoclonal antibodies, via reverse ADCC. These data support the notion that NK

cell lines can target leukemic stem cells, be enhanced by antibodies via ADCC or reverse ADCC,

and lead to enhanced survival in translationally relevant leukemia models. These results are on

Page 184: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

164

par with recent pre-clinical studies of CD123 chimeric antigen receptor (CAR) T-cell therapy of

AML(Mardiros, Dos Santos et al. 2013; Tettamanti, Marin et al. 2013; Gill, Tasian et al. 2014;

Pizzitola, Anjos-Afonso et al. 2014), a rapidly emerging cellular immunotherapy with

unprecedented clinical success in the context of CD19 CAR T-cell therapy for relapses and

refractory B-cell malignancies(Kalos, Levine et al. 2011; Porter, Levine et al. 2011; Grupp,

Kalos et al. 2013). Therefore, cell line therapy, with its more standardized expansion and

functional characteristics, represents a viable cellular therapeutic platform, with a strong

rationale to test combined cell line and antibody therapy in a clinical trial for AML patients with

minimal residual disease lacking a suitable allogeneic transplant donor.

6.2 Methodologic approaches to measuring the impact of immune

effectors on leukemic stem cell and bulk leukemia

Our initial focus was to develop a cell line model of leukemic stem cells to facilitate a better

understanding of how immune effectors recognize and kill this subpopulation of cells relative to

bulk leukemia. We selected KG1, a CD34+CD38+ cell line, which immunophenotypically

represented a typical AML primary sample, and postulated that it might contain a stem cell-

driven hierarchy, similar to the original findings for acute myeloid leukemia primary

blasts(Lapidot, Sirard et al. 1994; Bonnet and Dick 1997) The stem cell frequency was

determined with liquid reculturing in combination with serial dilution and single cell sorting, and

the methylcellulose colony-forming assay, all of which yielded a similar stem cell frequency

(1:100 to 1:1000). Therefore, we established that KG1 had a relatively rare tumour-initiating

cell, and sought to demonstrate if this was driven by an identifiable cancer stem cell, as had been

shown for some brain tumour(Kondo, Setoguchi et al. 2004), and multiple myeloma cell

lines(Matsui, Huff et al. 2004). KG1 did contain rare CD34+CD38- cells (1-3%), however, both

the CD34+CD38- and CD34+CD38+ fractions had unlimited proliferation capacity and the

ability to recapitulate the original culture immunophenotypic distribution of CD38. While these

findings were at odds with the original report from John Dick’s research group(Lapidot, Sirard et

al. 1994), they were consistent with a follow-up study demonstrating that CD34+CD38+ AML

cells had stem cell capacity. This was masked in the original study because the anti-CD38 mAb

used facilitated clearance by the NOD/SCID reticuloendothelial system, which could be blocked

by pretreating the immunodeficient mice with either IVIg or anti-CD122 (IL-2Rβ).(Taussig,

Page 185: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

165

Miraki-Moud et al. 2008). This work questioned the earliest report identifying the leukemic

stem cell as being restricted to the CD34+CD38- fraction(Lapidot, Sirard et al. 1994). However,

the experiments supporting CD34 as a LSC marker were not subject to the same artifactual

clearance issue, and it remains a reliable functional LSC marker for the majority of primary

AML samples. In our in vitro studies of sorted KG1, the putative stem cell was contained in

both the CD38+ and CD38- fractions, consistent with Taussig et al.(Taussig, Miraki-Moud et al.

2008).

Prior to initiation of our studies, only one group had looked at the in vitro sensitivity of

CD34+CD38- AML cancer stem cells to immune effector cell killing. In that study, lymphokine

activated killer cells and allogeneic lymphocytes exerted a modest cytotoxic effect on AML

cancer stem cells that were intrinsically resistant to the chemotherapeutic agent,

daunorubicin.(Costello, Mallet et al. 2000) Since KG1 had no identifiable stem cell fraction, cell

sorting could not be used, so we utilized functional stem cell readout- the methylcellulose

colony formation assay. This approach is blind to whether a tumour population is driven by a

stem cell hierarchy or a stochastic process, and there is no need to know the molecular features

of a putative stem cell population. To address the question of relative cytotoxicity of immune

effectors to LSCs and bulk leukemia required the development of a properly controlled

methylcellulose assay to facilitate this comparison. We demonstrated that the chromium release

assay significantly underestimated the cytotoxicity of NK-92 against the stem cell fraction in

KG1. At a 10:1 E:T NK-92 had 100% colony inhibition against KG1, but only 78 % lysis by the

chromium release assay. Given that other studies of cancer stem cells show them to be resistant

to chemotherapy(Costello, Mallet et al. 2000), this finding is important in demonstrating that an

immune effector can preferentially kill a leukemic stem cell.

Despite being unable to identify cancer stem cell by immunophenotypic profile within KG1,

using a clonogenic assay we have by-passed this requirement to obtain valuable information on

the cytotoxicity of NK-92 against putative leukemic stem cells within this cell line. This

provides a method for assessment of the impact of any immune effector cell on putative cancer

stem cells of numerous cell lines and primary samples extending beyond leukemia. Further, by

standardizing the methylcellulose cytotoxicity assay as closely as possible with the CRA and by

utilizing the appropriate controls, it is possible to determine if there is differential killing of an

Page 186: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

166

effector cell against bulk or leukemic stem cells. This approach is relatively simple, and can be

applied to any tumour that can grow well in semi-solid medium such as methylcellulose.

6.3 Natural cytotoxicity of NK cell lines with and without ADCC

enhancement against leukemic stem cells

While the overall long-term survival for AML is around 40% in adults (Lowenberg, Downing et

al. 1999) and 60% for children, subgroups of AML continue to fare very poorly, including

patients with the recurrent cytogenetic 10 year survival is less than 10% respectively(Grimwade,

Hills et al. 2010). The induction rates for most protocols is 80%, however, the majority still

harbor minimal residual disease, leading to relapse, often with chemotherapy resistant disease.

Therefore, there is a need for novel, non-cross resistant therapies to treat MRD, which contains

LSCs. Immune based therapy, such as NK-92, is an excellent experimental therapeutic option

for AML patients with MRD.

Therefore, we evaluated NK-92 with a focus on using methods that evaluate the impact on

leukemic stem cells and overall survival in an AML xenograft model. We confirmed initial

reports that NK-92 could mediate cytotoxicity in vitro against primary AML(Yan, Steinherz et

al. 1998), and demonstrated that this is done via granule exocytosis. Classically defined sorted

CD34+CD38- LSCs(Lapidot, Sirard et al. 1994) were more sensitive to NK-92 killing than bulk

leukemia at low E:T ratios utilizing a standard chromium release assay. Given the conflicting

reports in the literature of the definitive immunophenotype of the leukemic stem cell in

AML(Lapidot, Sirard et al. 1994; Bonnet and Dick 1997; Taussig, Miraki-Moud et al. 2008;

Goardon, Marchi et al. 2011), we opted to use a clonogenic assay to assess the impact of immune

effectors against leukemic stem cells in a larger set of samples. The comparison of the MCA

with the CRA demonstrated a 2-3 fold higher % colony inhibition than the % lysis, supporting

our initial finding with cell line KG1, that NK-92 can preferentially recognize and kill LSCs over

bulk leukemia. Langencamp et al. demonstrated that single KIR-expressing NK cells,

mismatched for the HLA of primary AML targets, had equivalent killing of LSCs and blasts

using a chromium release assay and a methylcellulose-based cytotoxicity assay,(Langenkamp,

Siegler et al. 2009). By contrast, we demonstrate preferential killing of LSCs by NK-92 relative

to bulk leukemia. This suggests that NK-92 has enhanced ability to kill LSCs relative to LAK

Page 187: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

167

cells(Costello, Mallet et al. 2000) or endogenously derived single-KIR expressing NK

cells(Langenkamp, Siegler et al. 2009). This phenomenon is also seen with NK-92 treatment of

multiple myeloma (MM) cell lines which also show preferential killing of clonogenic MM cells

over bulk tumour (Swift, Williams et al. 2012).

Using a secondary transplantation assay, iNK-92 therapy of primary engrafted mice was shown

to impact the LSC fraction in the secondary transplant mice, providing some evidence for in vivo

LSC cytotoxicity using a gold standard assay. AML-xenografted NSG mice were effectively

treated with NK-92 infusions, leading to improvement in survival relative to controls, confirming

previous work (Yan, Steinherz et al. 1998). However, we accomplished this with lower doses of

NK-92 than the original and without the use of IL-2 infusions. Irradiated NK-92 were able to

prolong survival in mice, but were less effective than the non-irradiated NK-92 cells, as expected

due to their inability to in vivo expand. Given the modest impact of iNK-92 on improving

survival in vivo, we attempted to enhance efficacy of this approach using a gene-modified

ADCC-capable NK-92 cell line (Figure 6.1). CD16+NK-92 have been successfully redirected to

kill CD20+ malignant targets using Rituximab, (Binyamin, Alpaugh et al. 2008). CD16+NK-92

cytotoxicity could be enhanced against the CD123+ leukemia line OCI/AML5 using an anti-

CD123 mAb through ADCC.

We then sought to combine iCD16+NK-92 and 7G3 therapy in our AML xenograft model.

Systemic treatment with 7G3 alone had been tested in an AML NOD/SCID xenograft model,

with evidence of impact on primary bone marrow engraftment, but not on survival in

therapeutically relevant animal models(Jin, Lee et al. 2009), indicating a need to optimize this

therapeutic strategy. We were able to increase the efficacy of the iCD16+NK-92 cells and

improve survival by administering 100 µg of 7G3 antibody for five doses relative to use with

isotype control. This demonstrates that the combination of iCD16+NK-92 with 7G3 can

improve survival by ADCC. This is the first demonstration of in vivo efficacy of the CD16+NK-

92 cell line in combination with antibody against any cancer. Recently, CSL362, an Fc-

optimized anti-CD123 humanized monoclonal antibody, was shown to facilitate ADCC from

peripheral blood-derived allogeneic NK cells against primary AML and CD123-expressing cell

line targets.(Busfield, Biondo et al. 2014) Further, CSL362 is in clinical trials as single agent for

relapsed and refractory AML and would make an ideal combination therapy with CD16-NK-92.

Page 188: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

168

Figure 6.1: Antibody-dependent cell-mediated cytotoxicity (ADCC)

CD16+NK-92 can mediate ADCC via the high affinity CD16A receptor in combination with

anti-CD123 monoclonal antibody (7G3) against CD123 (black antigen) expressing AML cells.

Page 189: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

169

6.4 Natural cytotoxicity of NK cell lines with and without reverse-

ADCC enhancement against leukemic stem cells

While NK-92 has been tested in phase I trials with minimal toxicities (Arai, Meagher et al. 2008;

Tonn, Schwabe et al. 2013), KHYG-1 has only been assessed against leukemia in vitro,

warranting further study of its potential clinical utility. KHYG-1 cytotoxicity against primary

AML was unreported, and we wished to compare its efficacy to NK-92. We noted that KHYG-1

was less effective than NK-92 at killing both leukemia cell lines and primary AML samples, and

sought to determine the basis for this finding. While we demonstrated that KHYG-1 expressed

more inhibitory KIRs than NK-92, we were unable to implicate them in the role of reduced

relative cytotoxicity.

We pretreated NK-92 and KHYG-1 with antibodies against a panel of activating receptors with

the objective of blocking receptors involved in AML recognition. However, we observed

predominantly stimulation of cytotoxicity from several of these antibodies, with minimal

inhibitory effects. Treatment of NK-92 with antibodies to NKp30 most notably enhanced

cytotoxicity to a modest degree against AML cell lines and to a great degree against primary

AML samples. Flow cytometry demonstrated higher expression of the natural cytotoxicity

receptors (NKp30, NKp44 and NKp46) on KHYG-1 compared with NK-92. Pretreatment of

KHYG-1 with antibodies to all the natural cytotoxicity receptors increased killing of cell line

targets, while anti-NKp30 and anti-NKp44 pretreatment enhanced cytotoxicity against primary

AML. KHYG-1 cytotoxicity against OCI/AML5 was enhanced with effector antibody

pretreatments with an EC50% of 0.01 µg/ml, indicating the potency of this approach.

We postulated that the increased killing was mediated by reverse antibody-dependent cell-

mediated cytotoxicity (R-ADCC), which involves the Fab of an antibody bridging an activating

receptor on the NK cell with an Fcγ receptor on the target (Figure 6.2), leading to NK signal

transduction and degranulation. We did not observe any significant increase in cytotoxicity after

pretreatment of NK-92 or KHYG-1 with anti-NKp30 or anti-NKp44 against Fc gamma negative

esophageal cancer cells. This is consistent with R-ADCC as the mechanism, and not co-

stimulation or co-activation, which would have led to generic enhancement. Regression analysis

Page 190: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

170

of the delta cytotoxicity with CD32 expression of the cell line targets yielded highly significant

correlations for KHYG-1, further supporting that the enhancement was from R-ADCC.

Figure 6.2: Reverse antibody-dependent cell-mediated cytotoxicity (R-ADCC)

KHYG-1 can mediate reverse ADCC in combination with anti-NKp30 monoclonal antibody

against Fcγ receptor expressing AML cells.

NK cells were first demonstrated to mediate non-ADCC, antibody-induced redirected lysis by

Saxena et al.,(Saxena, Saxena et al. 1982), where the term R-ADCC was first utilized to describe

the phenomenon. R-ADCC has been demonstrated using FcRγ+ P815 murine mastocytoma cell

lines in combination with NK cells pretreated with novel anti-NK cell antibodies to discover the

natural cytotoxicity receptors (NKp30, NKp44 and NKp46). (Vitale, Bottino et al. 1998; Pende,

Parolini et al. 1999) (Pessino, Sivori et al. 1998). Reverse ADCC is a very potent form of

redirected lysis, with an EC50 of ~0.01 µg/ml reported for anti-NKp30 redirected NK cell lysis

against P815 targets(Pende, Parolini et al. 1999), similar to our findings. However, reverse

ADCC has never been demonstrated in vivo.

Page 191: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

171

A thorough study of the expression of Fcγ receptors on 146 primary AML samples demonstrated

considerable expression of FcγRI (58%), FcγII (67%), and FcγIII (26%). The actual percentage

of samples expressing at least one Fcγ receptor was not presented in this work, but would be

expected to be 90%, assuming independent segregation of receptors, indicating they are relevant

therapeutic targets for AML. One study has demonstrated that lymphokine-activated killer

(LAK) cells could be redirected to kill autogolous AML blasts with anti-CD3 antibodies via

reverse ADDC. Further, this effect was demonstrated to occur through FcγRI (CD64).(Notter,

Ludwig et al. 1993) Precoating LAK cells with anti-CD3 antibodies led to many-fold increases

in cytotoxicity against CD64-expressing targets. Of note, this group also demonstrated that

CD34+ hematopoietic stem cells do not express FcγRs, and are insensitive to anti-CD3

pretreated LAK cells, suggesting that this approach would not be toxic to bone marrow HSCs in

vivo. However, no work was done to assess the impact of R-ADCC on leukemic stem cells. We

show that KHYG-1 was moderately effective at inhibiting OCI/AML5 colony formation, which

could be enhanced three-fold by pretreatment with anti-NKp30 antibody. This provides the first

evidence that R-ADCC can be used to target clonogenic cells in vitro which includes leukemic

stem and progenitor cells. Further, co-incubation of OCI/AML5 with anti-NKp30 pretreated

iKHYG-1 and subsequent infusion in NSG mice improved median survival by 10 days over

control or iKHYG-1 treatment alone, further supporting that reverse ADCC can target leukemic

stem cells within OCI/AML5 cells that have leukemogenic potential. In a therapeutically

relevant, OCI/AML5 NSG xenograft model, we tested the therapeutic efficacy of iKHYG-1 with

or without anti-NKp30 pretreatment. iKHYG-1 was able to improve the median survival by 35

days, which could not be predicted from the results of the chromium release assay which showed

poor cytotoxicity, but was predicted by the clonogenic assay which showed a moderate cytotoxic

effect. This represents the first demonstration of efficacy by KHYG-1 in an in vivo cancer

model, and confirms that the irradiated cells can persist and reduce tumour burden. This builds

on prior work from our group that demonstrated that irradiation has minimal effects on

cytotoxicity in vitro (Suck, Branch et al. 2006). However, while pretreatment of iKHYG-1 with

anti-NKp30 at slight enhanced survival over iKHYG-1-treated OCI/AML5 or primary AML-

xenografted NSG mice, the results were not statistically significant, falling short of a formal

demonstration of in vivo R-ADCC. Given that no antibody was injected directly into these

mice, the antibody likely dissociated in vivo, reducing its potential efficacy.

Page 192: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

172

We demonstrate that NK-92 and KHYG-1 have cytotoxicity against a broad range of leukemic

targets that can be enhanced several-fold by pretreatment with anti-NKp30 and anti-NKp44

antibodies. For KHYG-1, reverse ADCC has been formally demonstrated as the mechanism of

enhancement via interaction of antibody coated effectors with FcγRII (CD32) on the target cells.

Furthermore, NKp30 mediated reverse ADCC can enhance cytotoxicity of KHYG-1 against

clonogenic leukemic cells, and affect in vivo proliferation of leukemia. Additional

experimentation is required to fully demonstrate if reverse ADCC can occur in vivo in a

therapeutically relevant model.

6.5 Translational relevance

Adoptive immunotherapy with NK cells represents a novel emerging treatment modality for

patients with AML with several established protocols for allogeneic NK cell therapy for

AML.(Miller, Soignier et al. 2005; Rubnitz, Inaba et al. 2010) Anti-cancer cell-based

immunotherapy with a standardized, highly cytotoxic NK cell lines,(Arai, Meagher et al. 2008;

Tonn, Schwabe et al. 2013) is an attractive alternative to autologous or allogeneic NK cells

which have attendant variability in cytotoxicity and cell manufacturing characteristics. Further,

gene modifications can be done once with cell lines, rather than for each therapeutic attempt with

autologous, patient-derived immune effector cells. As an example, high affinity CD16-

transduced NK-92 cells can be utilized in combination with any humanized monoclonal

antibody, providing a flexible platform for combined cellular and humoral immune therapeutic

approaches. We have demonstrated, proof-in-principle, that this approach can be used to target

LSCs by anti-CD123-facilitated ADCC in vivo and enhance survival in a primary AML

xenograft model. The improvement in survival with this approach was approximately 2 week

extension of median survival. This is consistent with results of CD123 CAR T-cell therapy in a

an AML NSG xenograft model(Mardiros, Dos Santos et al. 2013), however, this model utilized

cell line KG1a, not primary AML cells. Using a different CAR vector, another group tested

CD123 CAR T-cell in a primary AML model with no improvement of median survival, but had

approximately 40% long term survivors at day 100 (~70 days after control median survival),

which was statistically significant.(Gill, Tasian et al. 2014) One other study of CD123 CAR T-

cells demonstrated efficacy against LSCs using primary and secondary engraftment models, but

Page 193: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

173

did not study impact on survival(Pizzitola, Anjos-Afonso et al. 2014). Another group utilized

CD123 CAR transduced cytokine induced killer (CIK) cells, and demonstrated in vitro efficacy

against CD123+ cell line and primary AML targets.(Tettamanti, Marin et al. 2013).

We also demonstrate the therapeutic efficacy of KHYG-1 in an AML xenograft model, providing

a rationale to develop this as a novel platform for AML therapy. While KHYG-1 cannot mediate

ADCC because it is FcγRIIIA (CD16) negative, it highly expresses the activating receptors

NKp30 and NKp44, which can facilitate reverse ADCC against Fcγ receptor positive targets.

Given that approximately 90% of primary AML samples express at least one Fcγ receptor, there

is a strong rationale to develop reverse-ADCC targeting of AML as a therapeutic strategy. A

humanized anti-NKp30 antibody is in development, and could be used in combination with

KHYG-1 cell-based therapy for MRD positive AML patients who lack a suitable allogeneic

transplant donor.

Page 194: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

174

7 Chapter 7: Conclusions and future directions

Improvements in therapy of AML have been relatively slow, with most new agents being tested

failing at early phase clinical studies. Part of the reason for this failure is that the drug screening

paradigm that has generated most cancer therapeutics has focused on the impact on bulk tumour

cells, rather than cancer stem cells. The most effective therapy for AML remains hematopoietic

stem cell transplantation, the only established cellular immunotherapy, with evidence for a role

in NK cell-mediated benefits in the haplotype transplant setting.

In this thesis, I have focused on the interaction of immune effectors, specifically NK cell lines,

with both bulk leukemia and leukemic stem cells, using established and newly developed

approaches. This has been used in combination with survival analysis in evaluating therapeutic

potential.

In Chapter 3, the classic stem cell markers (CD34+CD38-) established by John Dick’s research

group in 1994, did not hold for the leukemia cell line KG1 or for many other primary AML

samples which have stem cells in the CD34+CD38+ fraction. This indicated the importance of

using a functional readout such as the clonogenic assay to evaluate the impact of immune

effectors on leukemic stem cells. The lower cytotoxic readouts of bulk cytotoxicity assays with

the clonogenic assay suggested that NK-92 was preferentially able to kill leukemic stem cells.

Combining both assays together provided a more complete picture of the cytotoxic impact of

NK-92 against a heterogeneous tumour population.

In Chapter 4, NK-92 was tested against primary AML samples demonstrating moderate

cytotoxicity with the chromium release assay and near complete abolition of colony growth at a

25:1 effector target ratio. Sorted LSCs were also more sensitive to NK-92 by a significant

margin using the chromium release assay. This suggested that the leukemic stem cells were

more sensitive than bulk tumour over a range of samples. We confirmed that non-irradiated NK-

92 can prolong survival in an AML xenograft model, but more importantly demonstrate that the

irradiated cells also have efficacy, though are less effective. Utilization of a high-affinity

CD16+NK-92 cell line, in combination with anti-CD123 mAbs, allowed for ADCC against

Page 195: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

175

leukemic stem cells. This approach was validated in an in vivo experiment showing proof-in-

principle that NK cell line-mediated ADCC against leukemic stem cells can enhance survival in

a primary AML xenograft model. To follow-up our findings, we will use a higher dose of 7G3

(300 µg x 5) in combination with CD16+NK-92 in an attempt to improve survival outcomes, and

when available utilize humanized anti-CD123 mAbs in these types of experiments.

In Chapter 5, NK-92 and KHYG-1, another NK cell line was utilized to treat AML cell lines and

primary AML. Both cell lines had no inhibitory regulation by classical KIR ligand interactions

either because of lack of expression of receptors or lack of functionality. However, NK-92

appeared to be more effective overall at killing leukemic cell lines and primary AML than

KHYG-1. Serrendipitously, we discovered that pretreatment of NK-92 and KHYG-1 with

antibodies directed against NK activating receptors led to very large enhancements in

cytotoxicity against leukemic cell lines and primary AML cell lines, but not esophageal lines.

Targets with high FcγRII (CD32) expression were most amenable to this cytotoxic enhancement,

which was highly correlated for KHYG-1 pretreated with either anti-NKp30 or anti-NKp44

antibodies. The mechanism was by reverse ADCC, only described in the context of CD64

positive targets, making the connection with CD32 novel. This enhancement could be mediated

against clonogenic leukemia cells for KHYG-1 only. We also demonstrated that KHYG-1 can

be effective in vivo against OCI/AML5-induced leukemia, which is the first time this cell line

has shown in vivo efficacy against any cancer. While anti-NKp30 pretreatment did not enhance

the efficacy of the iKHYG-1 in an in vivo OCI/AML5 xenograft model, we postulate it was from

dissociation of antibody. We plan to treat an OCI/AML5 xenograft model with both iKHYG-1

and infused anti-NKp30 antibody (50 µg x 6), which will lead to therapeutic serum levels of this

antibody that should facilitate binding to the circulating iKHGY-1.

These studies lay the framework for combination therapies incorporating cell lines and

monoclonal antibody therapies, whereby leukemic stem cells can be targeted via ADCC or

reverse ADCC. Cell line therapy allows for standardization of the dose and schedule, as well as

anticipated efficacy and toxicity. The ongoing development of novel humanized monoclonal

antibodies makes ADCC capable cell lines an ideal adjunct to the more established antibody

therapies.

Page 196: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

176

8 Chapter 8: References

Aggarwal, B. B. (2003). "Signalling pathways of the TNF superfamily: a double-edged sword."

Nat Rev Immunol 3(9): 745-756.

Agliano, A., I. Martin-Padura, P. Mancuso, P. Marighetti, C. Rabascio, G. Pruneri, L. D. Shultz

and F. Bertolini (2008). "Human acute leukemia cells injected in NOD/LtSz-scid/IL-

2Rgamma null mice generate a faster and more efficient disease compared to other

NOD/scid-related strains." Int J Cancer 123(9): 2222-2227.

Ailles, L. E., B. Gerhard, H. Kawagoe and D. E. Hogge (1999). "Growth characteristics of acute

myelogenous leukemia progenitors that initiate malignant hematopoiesis in nonobese

diabetic/severe combined immunodeficient mice." Blood 94(5): 1761-1772.

Al-Hajj, M., M. S. Wicha, A. Benito-Hernandez, S. J. Morrison and M. F. Clarke (2003).

"Prospective identification of tumorigenic breast cancer cells.[Erratum appears in Proc

Natl Acad Sci U S A. 2003 May 27;100(11):6890]." Proc Natl Acad Sci U S A 100(7):

3983-3988.

Aldemir, H., V. Prod'homme, M. J. Dumaurier, C. Retiere, G. Poupon, J. Cazareth, F. Bihl and

V. M. Braud (2005). "Cutting edge: lectin-like transcript 1 is a ligand for the CD161

receptor." Journal of Immunology 175(12): 7791-7795.

Andersson, L. C., K. Nilsson and C. G. Gahmberg (1979). "K562—A human erythroleukemic

cell line." International Journal of Cancer 23(2): 143-147.

Andrews, R. G., J. W. Singer and I. D. Bernstein (1989). "Precursors of colony-forming cells in

humans can be distinguished from colony-forming cells by expression of the CD33 and

CD34 antigens and light scatter properties." Journal of Experimental Medicine 169(5):

1721-1731.

Anthony, R. S., N. D. McKelvie, A. J. Cunningham, J. I. Craig, S. Y. Rogers and A. C. Parker

(1998). "Flow cytometry using annexin V can detect early apoptosis in peripheral blood

stem cell harvests from patients with leukaemia and lymphoma." Bone Marrow

Transplantation 21(5): 441-446.

Arai, S., R. Meagher, M. Swearingen, H. Myint, E. Rich, J. Martinson and H. Klingemann

(2008). "Infusion of the allogeneic cell line NK-92 in patients with advanced renal cell

cancer or melanoma: a phase I trial." Cytotherapy 10(6): 625-632.

Armas, L. R. d. (2009). Chapter 16: Natural killer cytolytic activity, pg 216.

Arnon, T. I., H. Achdout, N. Lieberman, R. Gazit, T. Gonen-Gross, G. Katz, A. Bar-Ilan, N.

Bloushtain, M. Lev, A. Joseph, E. Kedar, A. Porgador and O. Mandelboim (2004). "The

mechanisms controlling the recognition of tumor- and virus-infected cells by NKp46."

Blood 103(2): 664-672.

Page 197: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

177

Arnon, T. I., M. Lev, G. Katz, Y. Chernobrov, A. Porgador and O. Mandelboim (2001).

"Recognition of viral hemagglutinins by NKp44 but not by NKp30." European Journal of

Immunology 31(9): 2680-2689.

Arora, M., D. J. Weisdorf, S. R. Spellman, M. D. Haagenson, J. P. Klein, C. K. Hurley, G. B.

Selby, J. H. Antin, N. A. Kernan, C. Kollman, A. Nademanee, P. McGlave, M. M.

Horowitz and E. W. Petersdorf (2009). "HLA-identical sibling compared with 8/8

matched and mismatched unrelated donor bone marrow transplant for chronic phase

chronic myeloid leukemia." Journal of Clinical Oncology 27(10): 1644-1652.

Augugliaro, R., S. Parolini, R. Castriconi, E. Marcenaro, C. Cantoni, M. Nanni, L. Moretta, A.

Moretta and C. Bottino (2003). "Selective cross-talk among natural cytotoxicity receptors

in human natural killer cells." European Journal of Immunology 33(5): 1235-1241.

Baer, M. R. (2002). "Detection of minimal residual disease in acute myeloid leukemia." Current

Oncology Reports 4(5): 398-402.

Bari, R., T. Bell, W. H. Leung, Q. P. Vong, W. K. Chan, N. Das Gupta, M. Holladay, B. Rooney

and W. Leung (2009). "Significant functional heterogeneity among KIR2DL1 alleles and

a pivotal role of arginine 245." Blood 114(25): 5182-5190.

Bassan, R., O. Spinelli, E. Oldani, T. Intermesoli, M. Tosi, B. Peruta, G. Rossi, E. Borlenghi, E.

M. Pogliani, E. Terruzzi, P. Fabris, V. Cassibba, G. Lambertenghi-Deliliers, A.

Cortelezzi, A. Bosi, G. Gianfaldoni, F. Ciceri, M. Bernardi, A. Gallamini, D. Mattei, E.

Di Bona, C. Romani, A. M. Scattolin, T. Barbui and A. Rambaldi (2009). "Improved risk

classification for risk-specific therapy based on the molecular study of minimal residual

disease (MRD) in adult acute lymphoblastic leukemia (ALL)." Blood 113(18): 4153-

4162.

Bauer, S., V. Groh, J. Wu, A. Steinle, J. H. Phillips, L. L. Lanier and T. Spies (1999).

"Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA."

Science 285(5428): 727-729.

Baumhueter, S., N. Dybdal, C. Kyle and L. A. Lasky (1994). "Global vascular expression of

murine CD34, a sialomucin-like endothelial ligand for L-selectin." Blood 84(8): 2554-

2565.

Belldegrun, A., L. M. Muul and S. A. Rosenberg (1988). "Interleukin 2 expanded tumor-

infiltrating lymphocytes in human renal cell cancer: isolation, characterization, and

antitumor activity." Cancer Research 48(1): 206-214.

Bene, M. C., G. Castoldi, W. Knapp, W. D. Ludwig, E. Matutes, A. Orfao and M. B. van't Veer

(1995). "Proposals for the immunological classification of acute leukemias. European

Group for the Immunological Characterization of Leukemias (EGIL)." Leukemia 9(10):

1783-1786.

Bennett, J. (1845). "Case of hypertrophy of the spleen and liver in

Page 198: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

178

which death took place from suppuration of the blood." Edinburgh Medical and Surgical

Journal(64): 413-423.

Bennett, J. M., D. Catovsky, M. T. Daniel, G. Flandrin, D. A. Galton, H. R. Gralnick and C.

Sultan (1976). "Proposals for the classification of the acute leukaemias. French-

American-British (FAB) co-operative group." British Journal of Haematology 33(4):

451-458.

Berenson, R. J., R. G. Andrews, W. I. Bensinger, D. Kalamasz, G. Knitter, C. D. Buckner and I.

D. Bernstein (1988). "Antigen CD34+ marrow cells engraft lethally irradiated baboons."

Journal of Clinical Investigation 81(3): 951-955.

Beresford, P. J., Z. Xia, A. H. Greenberg and J. Lieberman (1999). "Granzyme A loading

induces rapid cytolysis and a novel form of DNA damage independently of caspase

activation." Immunity 10(5): 585-594.

Biassoni, R., C. Cantoni, M. Falco, S. Verdiani, C. Bottino, M. Vitale, R. Conte, A. Poggi, A.

Moretta and L. Moretta (1996). "The human leukocyte antigen (HLA)-C-specific

"activatory" or "inhibitory" natural killer cell receptors display highly homologous

extracellular domains but differ in their transmembrane and intracytoplasmic portions."

Journal of Experimental Medicine 183(2): 645-650.

Biffi, A., C. C. Bartolomae, D. Cesana, N. Cartier, P. Aubourg, M. Ranzani, M. Cesani, F.

Benedicenti, T. Plati, E. Rubagotti, S. Merella, A. Capotondo, J. Sgualdino, G. Zanetti, C.

von Kalle, M. Schmidt, L. Naldini and E. Montini (2011). "Lentiviral vector common

integration sites in preclinical models and a clinical trial reflect a benign integration bias

and not oncogenic selection." Blood 117(20): 5332-5339.

Binici, J., J. Hartmann, J. Herrmann, C. Schreiber, S. Beyer, G. Guler, V. Vogel, F. Tumulka, R.

Abele, W. Mantele and J. Koch (2013). "A soluble fragment of the tumor antigen BCL2-

associated athanogene 6 (BAG-6) is essential and sufficient for inhibition of NKp30

receptor-dependent cytotoxicity of natural killer cells." Journal of Biological Chemistry

288(48): 34295-34303.

Binyamin, L., R. K. Alpaugh, T. L. Hughes, C. T. Lutz, K. S. Campbell and L. M. Weiner

(2008). "Blocking NK cell inhibitory self-recognition promotes antibody-dependent

cellular cytotoxicity in a model of anti-lymphoma therapy." Journal of immunology

(Baltimore, Md. : 1950) 180(9): 6392-6401.

Bloushtain, N., U. Qimron, A. Bar-Ilan, O. Hershkovitz, R. Gazit, E. Fima, M. Korc, I.

Vlodavsky, N. V. Bovin and A. Porgador (2004). "Membrane-associated heparan sulfate

proteoglycans are involved in the recognition of cellular targets by NKp30 and NKp46."

Journal of Immunology 173(4): 2392-2401.

Blunt, T., N. J. Finnie, G. E. Taccioli, G. C. Smith, J. Demengeot, T. M. Gottlieb, R. Mizuta, A.

J. Varghese, F. W. Alt, P. A. Jeggo and S. P. Jackson (1995). "Defective DNA-dependent

protein kinase activity is linked to V(D)J recombination and DNA repair defects

associated with the murine scid mutation." Cell 80(5): 813-823.

Page 199: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

179

Blunt, T., D. Gell, M. Fox, G. E. Taccioli, A. R. Lehmann, S. P. Jackson and P. A. Jeggo (1996).

"Identification of a nonsense mutation in the carboxyl-terminal region of DNA-dependent

protein kinase catalytic subunit in the scid mouse." Proceedings of the National Academy

of Sciences of the United States of America 93(19): 10285-10290.

Bonnet, D. and J. E. Dick (1997). "Human acute myeloid leukemia is organized as a hierarchy

that originates from a primitive hematopoietic cell." Nat Med 3(7): 730-737.

Borowitz, M. J., M. Devidas, S. P. Hunger, W. P. Bowman, A. J. Carroll, W. L. Carroll, S.

Linda, P. L. Martin, D. J. Pullen, D. Viswanatha, C. L. Willman, N. Winick and B. M.

Camitta (2008). "Clinical significance of minimal residual disease in childhood acute

lymphoblastic leukemia and its relationship to other prognostic factors: a Children's

Oncology Group study." Blood 111(12): 5477-5485.

Bosma, G. C., R. P. Custer and M. J. Bosma (1983). "A severe combined immunodeficiency

mutation in the mouse." Nature 301(5900): 527-530.

Bosma, M. J. and A. M. Carroll (1991). "The SCID mouse mutant: definition, characterization,

and potential uses." Annual Review of Immunology 9: 323-350.

Bottino, C., R. Castriconi, D. Pende, P. Rivera, M. Nanni, B. Carnemolla, C. Cantoni, J. Grassi,

S. Marcenaro, N. Reymond, M. Vitale, L. Moretta, M. Lopez and A. Moretta (2003).

"Identification of PVR (CD155) and Nectin-2 (CD112) as cell surface ligands for the

human DNAM-1 (CD226) activating molecule." Journal of Experimental Medicine

198(4): 557-567.

Bowles, J. A., S. Y. Wang, B. K. Link, B. Allan, G. Beuerlein, M. A. Campbell, D. Marquis, B.

Ondek, J. E. Wooldridge, B. J. Smith, J. B. Breitmeyer and G. J. Weiner (2006). "Anti-

CD20 monoclonal antibody with enhanced affinity for CD16 activates NK cells at lower

concentrations and more effectively than rituximab." Blood 108(8): 2648-2654.

Bradley, T. R. and D. Metcalf (1966). "The growth of mouse bone marrow cells in vitro."

Australian Journal of Experimental Biology & Medical Science 44(3): 287-299.

Brandt, C. S., M. Baratin, E. C. Yi, J. Kennedy, Z. Gao, B. Fox, B. Haldeman, C. D. Ostrander,

T. Kaifu, C. Chabannon, A. Moretta, R. West, W. Xu, E. Vivier and S. D. Levin (2009).

"The B7 family member B7-H6 is a tumor cell ligand for the activating natural killer cell

receptor NKp30 in humans." Journal of Experimental Medicine 206(7): 1495-1503.

Braud, V. M., D. S. Allan, C. A. O'Callaghan, K. Soderstrom, A. D'Andrea, G. S. Ogg, S.

Lazetic, N. T. Young, J. I. Bell, J. H. Phillips, L. L. Lanier and A. J. McMichael (1998).

"HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C." Nature

391(6669): 795-799.

Bremer, E. (2013). "Targeting of the tumor necrosis factor receptor superfamily for cancer

immunotherapy." ISRN Oncol 2013: 371854.

Brentjens, R. J., M. L. Davila, I. Riviere, J. Park, X. Wang, L. G. Cowell, S. Bartido, J.

Stefanski, C. Taylor, M. Olszewska, O. Borquez-Ojeda, J. Qu, T. Wasielewska, Q. He, Y.

Page 200: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

180

Bernal, I. V. Rijo, C. Hedvat, R. Kobos, K. Curran, P. Steinherz, J. Jurcic, T. Rosenblat,

P. Maslak, M. Frattini and M. Sadelain (2013). "CD19-targeted T cells rapidly induce

molecular remissions in adults with chemotherapy-refractory acute lymphoblastic

leukemia." Science Translational Medicine 5(177): 177ra138.

Brown, M. H., K. Boles, P. A. van der Merwe, V. Kumar, P. A. Mathew and A. N. Barclay

(1998). "2B4, the natural killer and T cell immunoglobulin superfamily surface protein, is

a ligand for CD48." Journal of Experimental Medicine 188(11): 2083-2090.

Brown, T. M. and E. Fee (2006). "Rudolf Carl Virchow: medical scientist, social reformer, role

model." American Journal of Public Health 96(12): 2104-2105.

Bruce, W. R. and C. L. Ash (1963). "Survival of Patients Treated for Cancer of the Breast,

Cervix, Lung, and Upper Respiratory Tract at the Ontario Cancer Institute (Toronto) from

1930 to 1957." Radiology 81: 861-870.

Brunner, K. T., J. Mauel, J. C. Cerottini and B. Chapuis (1968). "Quantitative assay of the lytic

action of immune lymphoid cells on 51-Cr-labelled allogeneic target cells in vitro;

inhibition by isoantibody and by drugs." Immunology 14(2): 181-196.

Bryceson, Y. T., M. E. March, H. G. Ljunggren and E. O. Long (2006). "Activation,

coactivation, and costimulation of resting human natural killer cells." Immunological

Reviews 214: 73-91.

Buccisano, F., L. Maurillo, A. Spagnoli, M. I. Del Principe, E. Ceresoli, F. Lo Coco, W. Arcese,

S. Amadori and A. Venditti (2009). "Monitoring of minimal residual disease in acute

myeloid leukemia." Current Opinion in Oncology 21(6): 582-588.

Buccisano, F., L. Maurillo, A. Spagnoli, M. I. Del Principe, D. Fraboni, P. Panetta, T. Ottone, M.

I. Consalvo, S. Lavorgna, P. Bulian, E. Ammatuna, D. F. Angelini, A. Diamantini, S.

Campagna, L. Ottaviani, C. Sarlo, V. Gattei, G. Del Poeta, W. Arcese, S. Amadori, F. Lo

Coco and A. Venditti (2010). "Cytogenetic and molecular diagnostic characterization

combined to postconsolidation minimal residual disease assessment by flow cytometry

improves risk stratification in adult acute myeloid leukemia." Blood 116(13): 2295-2303.

Buckner, C. D., R. B. Epstein, R. H. Rudolph, R. A. Clift, R. Storb and E. D. Thomas (1970).

"Allogeneic marrow engraftment following whole body irradiation in a patient with

leukemia." Blood 35(6): 741-750.

Burke, M. J., Q. Cao, B. Trotz, B. Weigel, A. Kumar, A. Smith and M. R. Verneris (2009).

"Allogeneic hematopoietic cell transplantation (allogeneic HCT) for treatment of

pediatric Philadelphia chromosome-positive acute lymphoblastic leukemia (ALL)."

Pediatric Blood & Cancer 53(7): 1289-1294.

Burnett, A., M. Wetzler and B. Lowenberg (2011). "Therapeutic advances in acute myeloid

leukemia." Journal of Clinical Oncology 29(5): 487-494.

Page 201: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

181

Burshtyn, D. N., A. M. Scharenberg, N. Wagtmann, S. Rajagopalan, K. Berrada, T. Yi, J. P.

Kinet and E. O. Long (1996). "Recruitment of tyrosine phosphatase HCP by the killer

cell inhibitor receptor." Immunity 4(1): 77-85.

Busfield, S. J., M. Biondo, M. Wong, H. S. Ramshaw, E. M. Lee, S. Ghosh, H. Braley, C.

Panousis, A. W. Roberts, S. Z. He, D. Thomas, L. Fabri, G. Vairo, R. B. Lock, A. F.

Lopez and A. D. Nash (2014). "Targeting of acute myeloid leukemia in vitro and in vivo

with an anti-CD123 mAb engineered for optimal ADCC." Leukemia.

Cantoni, C., C. Bottino, M. Vitale, A. Pessino, R. Augugliaro, A. Malaspina, S. Parolini, L.

Moretta, A. Moretta and R. Biassoni (1999). "NKp44, a triggering receptor involved in

tumor cell lysis by activated human natural killer cells, is a novel member of the

immunoglobulin superfamily." Journal of Experimental Medicine 189(5): 787-796.

Cao, X., E. W. Shores, J. Hu-Li, M. R. Anver, B. L. Kelsall, S. M. Russell, J. Drago, M.

Noguchi, A. Grinberg, E. T. Bloom and et al. (1995). "Defective lymphoid development

in mice lacking expression of the common cytokine receptor gamma chain." Immunity

2(3): 223-238.

Caretto, P., M. Forni, G. d'Orazi, S. Scarpa, P. Feraiorni, C. Jemma, A. Modesti, M. Ferrarini, S.

Roncella, R. Foa and et al. (1989). "Xenotransplantation in immunosuppressed nude mice

of human solid tumors and acute leukemias directly from patients or in vitro cell lines."

Ricerca in Clinica e in Laboratorio 19(3): 231-243.

Cesano, A. and D. Santoli (1992). "Two unique human leukemic T-cell lines endowed with a

stable cytotoxic function and a different spectrum of target reactivity analysis and

modulation of their lytic mechanisms." In Vitro Cellular & Developmental Biology

28A(9-10): 648-656.

Chapman, T. L., A. P. Heikema, A. P. West, Jr. and P. J. Bjorkman (2000). "Crystal structure

and ligand binding properties of the D1D2 region of the inhibitory receptor LIR-1

(ILT2)." Immunity 13(5): 727-736.

Chendamarai, E., P. Balasubramanian, B. George, A. Viswabandya, A. Abraham, R. Ahmed, A.

A. Alex, S. Ganesan, K. M. Lakshmi, U. Sitaram, S. C. Nair, M. Chandy, N. B. Janet, V.

M. Srivastava, A. Srivastava and V. Mathews (2012). "Role of minimal residual disease

monitoring in acute promyelocytic leukemia treated with arsenic trioxide in frontline

therapy." Blood 119(15): 3413-3419.

Cheng, J., S. Baumhueter, G. Cacalano, K. Carver-Moore, H. Thibodeaux, R. Thomas, H. E.

Broxmeyer, S. Cooper, N. Hague, M. Moore and L. A. Lasky (1996). "Hematopoietic

defects in mice lacking the sialomucin CD34." Blood 87(2): 479-490.

Cheng, M., Y. Chen, W. Xiao, R. Sun and Z. Tian (2013). "NK cell-based immunotherapy for

malignant diseases." Cellular & Molecular Immunology 10(3): 230-252.

Cheung, A. M., T. S. Wan, J. C. Leung, L. Y. Chan, H. Huang, Y. L. Kwong, R. Liang and A. Y.

Leung (2007). "Aldehyde dehydrogenase activity in leukemic blasts defines a subgroup

Page 202: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

182

of acute myeloid leukemia with adverse prognosis and superior NOD/SCID engrafting

potential." Leukemia 21(7): 1423-1430.

Christianson, S. W., D. L. Greiner, R. A. Hesselton, J. H. Leif, E. J. Wagar, I. B. Schweitzer, T.

V. Rajan, B. Gott, D. C. Roopenian and L. D. Shultz (1997). "Enhanced human CD4+ T

cell engraftment in beta2-microglobulin-deficient NOD-scid mice." Journal of

Immunology 158(8): 3578-3586.

Cieciura, S. J., P. I. Marcus and T. T. Puck (1956). "Clonal growth in vitro of epithelial cells

from normal human tissues." Journal of Experimental Medicine 104(4): 615-628.

Clarkson, B. D. (1969). "Review of recent studies of cellular proliferation in acute leukemia."

National Cancer Institute Monographs 30: 81-120.

Clift, R. A., J. A. Hansen, E. D. Thomas, C. D. Buckner, J. E. Sanders, E. M. Mickelson, R.

Storb, F. L. Johnson, J. W. Singer and B. W. Goodell (1979). "Marrow transplantation

from donors other than HLA-identical siblings." Transplantation 28(3): 235-242.

Colonna, M., H. Nakajima and M. Cella (1999). "Inhibitory and activating receptors involved in

immune surveillance by human NK and myeloid cells." Journal of Leukocyte Biology

66(5): 718-722.

Colonna, M., F. Navarro, T. Bellon, M. Llano, P. Garcia, J. Samaridis, L. Angman, M. Cella and

M. Lopez-Botet (1997). "A common inhibitory receptor for major histocompatibility

complex class I molecules on human lymphoid and myelomonocytic cells." Journal of

Experimental Medicine 186(11): 1809-1818.

Costello, R. T., F. Mallet, B. Gaugler, D. Sainty, C. Arnoulet, J. A. Gastaut and D. Olive (2000).

"Human acute myeloid leukemia CD34+/CD38- progenitor cells have decreased

sensitivity to chemotherapy and Fas-induced apoptosis, reduced immunogenicity, and

impaired dendritic cell transformation capacities." Cancer Research 60(16): 4403-4411.

Cretney, E., K. Takeda, H. Yagita, M. Glaccum, J. J. Peschon and M. J. Smyth (2002).

"Increased susceptibility to tumor initiation and metastasis in TNF-related apoptosis-

inducing ligand-deficient mice." Journal of Immunology 168(3): 1356-1361.

Curtis, D. J., D. Metcalf, B. Alexander and C. G. Begley (2000). "Leukemic cells from murine

myeloid leukemia display an intrinsic ability for autonomous proliferation." Experimental

Hematology 28(1): 36-45.

Curtis, J. E., M. D. Minden, S. Minkin and E. A. McCulloch (1995). "Sensitivities of AML blast

stem cells to idarubicin and daunorubicin: a comparison with normal hematopoietic

progenitors." Leukemia 9(3): 396-404.

Darro, F., C. Decaestecker, J. F. Gaussin, S. Mortier, R. Van Ginckel and R. Kiss (2005). "Are

syngeneic mouse tumor models still valuable experimental models in the field of anti-

cancer drug discovery?" International Journal of Oncology 27(3): 607-616.

Page 203: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

183

Dennehy, K. M., S. N. Klimosch and A. Steinle (2011). "Cutting edge: NKp80 uses an atypical

hemi-ITAM to trigger NK cytotoxicity." Journal of Immunology 186(2): 657-661.

Dennert, G. and E. R. Podack (1983). "Cytolysis by H-2-specific T killer cells. Assembly of

tubular complexes on target membranes." Journal of Experimental Medicine 157(5):

1483-1495.

Deschler, B. and M. Lubbert (2006). "Acute myeloid leukemia: epidemiology and etiology."

Cancer 107(9): 2099-2107.

Diefenbach, A., A. M. Jamieson, S. D. Liu, N. Shastri and D. H. Raulet (2000). "Ligands for the

murine NKG2D receptor: expression by tumor cells and activation of NK cells and

macrophages." Nat Immunol 1(2): 119-126.

Diermayr, S., H. Himmelreich, B. Durovic, A. Mathys-Schneeberger, U. Siegler, U.

Langenkamp, J. Hofsteenge, A. Gratwohl, A. Tichelli, M. Paluszewska, W. Wiktor-

Jedrzejczak, C. P. Kalberer and A. Wodnar-Filipowicz (2008). "NKG2D ligand

expression in AML increases in response to HDAC inhibitor valproic acid and

contributes to allorecognition by NK-cell lines with single KIR-HLA class I

specificities." Blood 111(3): 1428-1436.

DiSanto, J. P., W. Muller, D. Guy-Grand, A. Fischer and K. Rajewsky (1995). "Lymphoid

development in mice with a targeted deletion of the interleukin 2 receptor gamma chain."

Proceedings of the National Academy of Sciences of the United States of America 92(2):

377-381.

Dohring, C. and M. Colonna (1996). "Human natural killer cell inhibitory receptors bind to HLA

class I molecules." European Journal of Immunology 26(2): 365-369.

Drexler, H. (2010). "Guide to Leukemia-Lymphoma Cell Lines, Braunshweig ".

Drexler, H. G. and Y. Matsuo (2000). "Malignant hematopoietic cell lines: in vitro models for

the study of natural killer cell leukemia-lymphoma." Leukemia 14(5): 777-782.

Dudley, M. E., J. C. Yang, R. Sherry, M. S. Hughes, R. Royal, U. Kammula, P. F. Robbins, J.

Huang, D. E. Citrin, S. F. Leitman, J. Wunderlich, N. P. Restifo, A. Thomasian, S. G.

Downey, F. O. Smith, J. Klapper, K. Morton, C. Laurencot, D. E. White and S. A.

Rosenberg (2008). "Adoptive cell therapy for patients with metastatic melanoma:

evaluation of intensive myeloablative chemoradiation preparative regimens." Journal of

Clinical Oncology 26(32): 5233-5239.

Dworzak, M. N. (2002). "Prognostic significance and modalities of flow cytometric minimal

residual disease detection in childhood acute lymphoblastic leukemia." Blood 99(6):

1952-1958.

Eagle, R. A., J. A. Traherne, J. R. Hair, I. Jafferji and J. Trowsdale (2009). "ULBP6/RAET1L is

an additional human NKG2D ligand." European Journal of Immunology 39(11): 3207-

3216.

Page 204: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

184

Eagle, R. A. and J. Trowsdale (2007). "Promiscuity and the single receptor: NKG2D." Nat Rev

Immunol 7(9): 737-744.

Ema, H., T. Suda, Y. Miura and H. Nakauchi (1990). "Colony formation of clone-sorted human

hematopoietic progenitors." Blood 75(10): 1941-1946.

Eppert, K., K. Takenaka, E. R. Lechman, L. Waldron, B. Nilsson, P. van Galen, K. H. Metzeler,

A. Poeppl, V. Ling, J. Beyene, A. J. Canty, J. S. Danska, S. K. Bohlander, C. Buske, M.

D. Minden, T. R. Golub, I. Jurisica, B. L. Ebert and J. E. Dick (2011). "Stem cell gene

expression programs influence clinical outcome in human leukemia." Nature Medicine

17(9): 1086-1093.

Falini, B., M. P. Martelli, N. Bolli, P. Sportoletti, A. Liso, E. Tiacci and T. Haferlach (2011).

"Acute myeloid leukemia with mutated nucleophosmin (NPM1): is it a distinct entity?"

Blood 117(4): 1109-1120.

Faure, M. and E. O. Long (2002). "KIR2DL4 (CD158d), an NK cell-activating receptor with

inhibitory potential." Journal of Immunology 168(12): 6208-6214.

Fauriat, C., S. Just-Landi, F. Mallet, C. Arnoulet, D. Sainty, D. Olive and R. T. Costello (2007).

"Deficient expression of NCR in NK cells from acute myeloid leukemia: Evolution

during leukemia treatment and impact of leukemia cells in NCRdull phenotype

induction." Blood 109(1): 323-330.

Fefer, A., M. Benyunes, C. Higuchi, A. York, C. Massumoto, C. Lindgren, C. D. Buckner and J.

A. Thompson (1993). "Interleukin-2 +/- lymphocytes as consolidative immunotherapy

after autologous bone marrow transplantation for hematologic malignancies." Acta

Haematologica 89 Suppl 1: 2-7.

Feldmann, J., I. Callebaut, G. Raposo, S. Certain, D. Bacq, C. Dumont, N. Lambert, M.

Ouachee-Chardin, G. Chedeville, H. Tamary, V. Minard-Colin, E. Vilmer, S. Blanche, F.

Le Deist, A. Fischer and G. de Saint Basile (2003). "Munc13-4 is essential for cytolytic

granules fusion and is mutated in a form of familial hemophagocytic lymphohistiocytosis

(FHL3)." Cell 115(4): 461-473.

Fernandez, H. F., Z. Sun, X. Yao, M. R. Litzow, S. M. Luger, E. M. Paietta, J. Racevskis, G. W.

Dewald, R. P. Ketterling, J. M. Bennett, J. M. Rowe, H. M. Lazarus and M. S. Tallman

(2009). "Anthracycline dose intensification in acute myeloid leukemia." New England

Journal of Medicine 361(13): 1249-1259.

Ferrara, F. and C. A. Schiffer (2013). "Acute myeloid leukaemia in adults." Lancet 381(9865):

484-495.

Ferrebee, J. W., H. L. Lochte, Jr., A. Jaretzki, 3rd, O. D. Sahler and E. D. Thomas (1958).

"Successful marrow homograft in the dog after radiation." Surgery 43(3): 516-520.

Fisher, B., B. S. Packard, E. J. Read, J. A. Carrasquillo, C. S. Carter, S. L. Topalian, J. C. Yang,

P. Yolles, S. M. Larson and S. A. Rosenberg (1989). "Tumor localization of adoptively

Page 205: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

185

transferred indium-111 labeled tumor infiltrating lymphocytes in patients with metastatic

melanoma." Journal of Clinical Oncology 7(2): 250-261.

Fisher, G. H., F. J. Rosenberg, S. E. Straus, J. K. Dale, L. A. Middleton, A. Y. Lin, W. Strober,

M. J. Lenardo and J. M. Puck (1995). "Dominant interfering Fas gene mutations impair

apoptosis in a human autoimmune lymphoproliferative syndrome." Cell 81(6): 935-946.

Flanagan, S. P. (1966). "'Nude', a new hairless gene with pleiotropic effects in the mouse."

Genetical Research 8(3): 295-309.

Foster, C. E., M. Colonna and P. D. Sun (2003). "Crystal structure of the human natural killer

(NK) cell activating receptor NKp46 reveals structural relationship to other leukocyte

receptor complex immunoreceptors." Journal of Biological Chemistry 278(46): 46081-

46086.

Franks, C. R., D. Bishop, F. R. Balkwill, R. T. Oliver and W. G. Spector (1977). "Growth of

acute myeloid leukaemia as discrete subcutaneous tumours in immune-deprived mice."

British Journal of Cancer 35(5): 697-700.

Fuchs, A., M. Cella, T. Kondo and M. Colonna (2005). "Paradoxic inhibition of human natural

interferon-producing cells by the activating receptor NKp44." Blood 106(6): 2076-2082.

Furth, J. (1935). "TRANSMISSION OF MYELOID LEUKEMIA OF MICE : ITS RELATION

TO MYELOMA." Journal of Experimental Medicine 61(3): 423-446.

Furth, J., M. C. Kahn and C. Breedis (1937). "The Transmission of Leukemia of Mice with a

Single Cell." The American Journal of Cancer 31(2): 276-282.

Gasser, S., S. Orsulic, E. J. Brown and D. H. Raulet (2005). "The DNA damage pathway

regulates innate immune system ligands of the NKG2D receptor." Nature 436(7054):

1186-1190.

Gau, J. P., J. H. Young, T. H. Lin and Y. S. Yang (2000). "Diagnosis and follow-up of acute

promyelocytic leukemia by detection of PML-RAR alpha gene rearrangement."

Zhonghua Yi Xue Za Zhi (Taipei) 63(3): 175-181.

Geller, M. A., S. Cooley, P. L. Judson, R. Ghebre, L. F. Carson, P. A. Argenta, A. L. Jonson, A.

Panoskaltsis-Mortari, J. Curtsinger, D. McKenna, K. Dusenbery, R. Bliss, L. S. Downs

and J. S. Miller (2011). "A phase II study of allogeneic natural killer cell therapy to treat

patients with recurrent ovarian and breast cancer." Cytotherapy 13(1): 98-107.

Gill, S., S. K. Tasian, M. Ruella, O. Shestova, Y. Li, D. L. Porter, M. Carroll, G. Danet-

Desnoyers, J. Scholler, S. A. Grupp, C. H. June and M. Kalos (2014). "Preclinical

targeting of human acute myeloid leukemia and myeloablation using chimeric antigen

receptor-modified T cells." Blood 123(15): 2343-2354.

Goardon, N., E. Marchi, A. Atzberger, L. Quek, A. Schuh, S. Soneji, P. Woll, A. Mead, K. A.

Alford, R. Rout, S. Chaudhury, A. Gilkes, S. Knapper, K. Beldjord, S. Begum, S. Rose,

N. Geddes, M. Griffiths, G. Standen, A. Sternberg, J. Cavenagh, H. Hunter, D. Bowen, S.

Page 206: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

186

Killick, L. Robinson, A. Price, E. Macintyre, P. Virgo, A. Burnett, C. Craddock, T.

Enver, S. E. Jacobsen, C. Porcher and P. Vyas (2011). "Coexistence of LMPP-like and

GMP-like leukemia stem cells in acute myeloid leukemia." Cancer Cell 19(1): 138-152.

Gong, J. H., G. Maki and H. G. Klingemann (1994). "Characterization of a human cell line (NK-

92) with phenotypical and functional characteristics of activated natural killer cells."

Leukemia 8(4): 652.

Grimwade, D., R. K. Hills, A. V. Moorman, H. Walker, S. Chatters, A. H. Goldstone, K.

Wheatley, C. J. Harrison and A. K. Burnett (2010). "Refinement of cytogenetic

classification in acute myeloid leukemia: determination of prognostic significance of rare

recurring chromosomal abnormalities among 5876 younger adult patients treated in the

United Kingdom Medical Research Council trials." Blood 116(3): 354-365.

Groh, V., R. Rhinehart, H. Secrist, S. Bauer, K. H. Grabstein and T. Spies (1999). "Broad tumor-

associated expression and recognition by tumor-derived gamma delta T cells of MICA

and MICB." Proceedings of the National Academy of Sciences of the United States of

America 96(12): 6879-6884.

Gross, G., G. Gorochov, T. Waks and Z. Eshhar (1989). "Generation of effector T cells

expressing chimeric T cell receptor with antibody type-specificity." Transplantation

Proceedings 21(1 Pt 1): 127-130.

Gruen, J. R. and S. M. Weissman (2001). "Human MHC class III and IV genes and disease

associations." Frontiers in Bioscience 6: D960-972.

Grupp, S. A., M. Kalos, D. Barrett, R. Aplenc, D. L. Porter, S. R. Rheingold, D. T. Teachey, A.

Chew, B. Hauck, J. F. Wright, M. C. Milone, B. L. Levine and C. H. June (2013).

"Chimeric antigen receptor-modified T cells for acute lymphoid leukemia." New England

Journal of Medicine 368(16): 1509-1518.

Guan, Y., B. Gerhard and D. E. Hogge (2003). "Detection, isolation, and stimulation of quiescent

primitive leukemic progenitor cells from patients with acute myeloid leukemia (AML)."

Blood 101(8): 3142-3149.

Gurney, J. G., S. Davis, R. K. Severson, J. Y. Fang, J. A. Ross and L. L. Robison (1996).

"Trends in cancer incidence among children in the U.S." Cancer 78(3): 532-541.

Hamann, J., K. T. Montgomery, S. Lau, R. Kucherlapati and R. A. van Lier (1997). "AICL: a

new activation-induced antigen encoded by the human NK gene complex."

Immunogenetics 45(5): 295-300.

Hameed, A., D. M. Lowrey, M. Lichtenheld and E. R. Podack (1988). "Characterization of three

serine esterases isolated from human IL-2 activated killer cells." Journal of Immunology

141(9): 3142-3147.

He, S. Z., S. Busfield, D. S. Ritchie, M. S. Hertzberg, S. Durrant, I. D. Lewis, P. Marlton, A. J.

McLachlan, I. Kerridge, K. F. Bradstock, G. Kennedy, A. W. Boyd, T. M. Yeadon, A. F.

Lopez, H. S. Ramshaw, H. Iland, S. Bamford, M. Barnden, M. DeWitte, R. Basser and A.

Page 207: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

187

W. Roberts (2014). "A Phase 1 study of the Safety, Pharmacokinetics, and Anti-leukemic

Activity of the anti-CD123 monoclonal antibody, CSL360, in Relapsed, Refractory or

High-Risk Acute Myeloid Leukemia (AML)." Leukemia and Lymphoma: 1-23.

Henkart, P. A. (1985). "Mechanism of lymphocyte-mediated cytotoxicity." Annual Review of

Immunology 3: 31-58.

Herberman, R. B., M. E. Nunn, H. T. Holden and D. H. Lavrin (1975). "Natural cytotoxic

reactivity of mouse lymphoid cells against syngeneic and allogeneic tumors. II.

Characterization of effector cells." International Journal of Cancer 16(2): 230-239.

Herberman, R. B., M. E. Nunn and D. H. Lavrin (1975). "Natural cytotoxic reactivity of mouse

lymphoid cells against syngeneic acid allogeneic tumors. I. Distribution of reactivity and

specificity." International Journal of Cancer 16(2): 216-229.

Ho, A. D., B. Brado, R. Haas and W. Hunstein (1991). "Etoposide in acute leukemia. Past

experience and future perspectives." Cancer 67(1 Suppl): 281-284.

Hoffman, R. M. (1991). "In vitro sensitivity assays in cancer: a review, analysis, and prognosis."

Journal of Clinical Laboratory Analysis 5(2): 133-143.

Horowitz, M. M., R. P. Gale, P. M. Sondel, J. M. Goldman, J. Kersey, H. J. Kolb, A. A. Rimm,

O. Ringden, C. Rozman, B. Speck and et al. (1990). "Graft-versus-leukemia reactions

after bone marrow transplantation." Blood 75(3): 555-562.

Hosen, N., C. Y. Park, N. Tatsumi, Y. Oji, H. Sugiyama, M. Gramatzki, A. M. Krensky and I. L.

Weissman (2007). "CD96 is a leukemic stem cell-specific marker in human acute

myeloid leukemia." Proc Natl Acad Sci U S A 104(26): 11008-11013.

Houchins, J. P., T. Yabe, C. McSherry and F. H. Bach (1991). "DNA sequence analysis of

NKG2, a family of related cDNA clones encoding type II integral membrane proteins on

human natural killer cells." Journal of Experimental Medicine 173(4): 1017-1020.

Howlader N, N. A., Krapcho M, Garshell J, Miller D, Altekruse SF, Kosary CL, Yu M, Ruhl J,

Tatalovich Z,Mariotto A, Lewis DR, Chen HS, Feuer EJ, Cronin KA (eds). SEER Cancer

Statistics Review, 1975-2011, National Cancer Institute. Bethesda, MD,

http://seer.cancer.gov/csr/1975_2011/, based on November 2013 SEER data submission,

posted to the SEER web site, April 2014.

Huang, S., Z. Chen, J. F. Yu, D. Young, A. Bashey, A. D. Ho and P. Law (1999). "Correlation

between IL-3 receptor expression and growth potential of human CD34+ hematopoietic

cells from different tissues." Stem Cells 17(5): 265-272.

Hurwitz, C. A., K. G. Mounce and H. E. Grier (1995). "Treatment of patients with acute

myelogenous leukemia: review of clinical trials of the past decade." Journal of Pediatric

Hematology/Oncology 17(3): 185-197.

Iliopoulou, E. G., P. Kountourakis, M. V. Karamouzis, D. Doufexis, A. Ardavanis, C. N.

Baxevanis, G. Rigatos, M. Papamichail and S. A. Perez (2010). "A phase I trial of

Page 208: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

188

adoptive transfer of allogeneic natural killer cells in patients with advanced non-small

cell lung cancer." Cancer Immunology, Immunotherapy 59(12): 1781-1789.

Iriyama, N., Y. Hatta, J. Takeuchi, Y. Ogawa, S. Ohtake, T. Sakura, K. Mitani, F. Ishida, M.

Takahashi, T. Maeda, T. Izumi, H. Sakamaki, S. Miyawaki, S. Honda, Y. Miyazaki, T.

Taki, M. Taniwaki and T. Naoe (2013). "CD56 expression is an independent prognostic

factor for relapse in acute myeloid leukemia with t(8;21)." Leukemia Research 37(9):

1021-1026.

Ishikawa, F., M. Yasukawa, B. Lyons, S. Yoshida, T. Miyamoto, G. Yoshimoto, T. Watanabe,

K. Akashi, L. D. Shultz and M. Harada (2005). "Development of functional human blood

and immune systems in NOD/SCID/IL2 receptor {gamma} chain(null) mice." Blood

106(5): 1565-1573.

Ito, M., H. Hiramatsu, K. Kobayashi, K. Suzue, M. Kawahata, K. Hioki, Y. Ueyama, Y.

Koyanagi, K. Sugamura, K. Tsuji, T. Heike and T. Nakahata (2002).

"NOD/SCID/gamma(c)(null) mouse: an excellent recipient mouse model for engraftment

of human cells." Blood 100(9): 3175-3182.

Jacobs, P. and L. Wood (2005). "Clonogenic growth patterns correlate with chemotherapy

response in acute myeloid leukaemia." Hematology 10(4): 321-326.

Jan, M., M. P. Chao, A. C. Cha, A. A. Alizadeh, A. J. Gentles, I. L. Weissman and R. Majeti

(2011). "Prospective separation of normal and leukemic stem cells based on differential

expression of TIM3, a human acute myeloid leukemia stem cell marker." Proc Natl Acad

Sci U S A 108(12): 5009-5014.

Jemal, A., A. Thomas, T. Murray and M. Thun (2002). "Cancer statistics, 2002." CA: A Cancer

Journal for Clinicians 52(1): 23-47.

Jin, L., E. M. Lee, H. S. Ramshaw, S. J. Busfield, A. G. Peoppl, L. Wilkinson, M. A. Guthridge,

D. Thomas, E. F. Barry, A. Boyd, D. P. Gearing, G. Vairo, A. F. Lopez, J. E. Dick and R.

B. Lock (2009). "Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha

chain, eliminates human acute myeloid leukemic stem cells." Cell Stem Cell 5(1): 31-42.

Jondal, M. and H. Pross (1975). "Surface markers on human b and t lymphocytes. VI.

Cytotoxicity against cell lines as a functional marker for lymphocyte subpopulations."

International Journal of Cancer 15(4): 596-605.

Jones, K. H. and J. A. Senft (1985). "An improved method to determine cell viability by

simultaneous staining with fluorescein diacetate-propidium iodide." Journal of

Histochemistry and Cytochemistry 33(1): 77-79.

Jordan, C. T., D. Upchurch, S. J. Szilvassy, M. L. Guzman, D. S. Howard, A. L. Pettigrew, T.

Meyerrose, R. Rossi, B. Grimes, D. A. Rizzieri, S. M. Luger and G. L. Phillips (2000).

"The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous

leukemia stem cells." Leukemia 14(10): 1777-1784.

Page 209: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

189

Kagami, Y., S. Nakamura, R. Suzuki, S. Iida, Y. Yatabe, Y. Okada, T. Kobayashi, T. Tsurumi,

M. Seto, M. Ogura, O. Taguchi and Y. Morishima (1998). "Establishment of an IL-2-

dependent cell line derived from 'nasal-type' NK/T-cell lymphoma of CD2+, sCD3-,

CD3epsilon+, CD56+ phenotype and associated with the Epstein-Barr virus." British

Journal of Haematology 103(3): 669-677.

Kagi, D., B. Ledermann, K. Burki, P. Seiler, B. Odermatt, K. J. Olsen, E. R. Podack, R. M.

Zinkernagel and H. Hengartner (1994). "Cytotoxicity mediated by T cells and natural

killer cells is greatly impaired in perforin-deficient mice." Nature 369(6475): 31-37.

Kalos, M., B. L. Levine, D. L. Porter, S. Katz, S. A. Grupp, A. Bagg and C. H. June (2011). "T

cells with chimeric antigen receptors have potent antitumor effects and can establish

memory in patients with advanced leukemia." Science Translational Medicine 3(95):

95ra73.

Kam, C. M., D. Hudig and J. C. Powers (2000). "Granzymes (lymphocyte serine proteases):

characterization with natural and synthetic substrates and inhibitors." Biochimica et

Biophysica Acta 1477(1-2): 307-323.

Kamel-Reid, S., M. Letarte, C. Sirard, M. Doedens, T. Grunberger, G. Fulop, M. H. Freedman,

R. A. Phillips and J. E. Dick (1989). "A model of human acute lymphoblastic leukemia in

immune-deficient SCID mice." Science 246(4937): 1597-1600.

Karimi, M., T. M. Cao, J. A. Baker, M. R. Verneris, L. Soares and R. S. Negrin (2005).

"Silencing human NKG2D, DAP10, and DAP12 reduces cytotoxicity of activated CD8+

T cells and NK cells." Journal of Immunology 175(12): 7819-7828.

Karlhofer, F. M., R. K. Ribaudo and W. M. Yokoyama (1992). "MHC class I alloantigen

specificity of Ly-49+ IL-2-activated natural killer cells." Nature 358(6381): 66-70.

Karre, K., H. G. Ljunggren, G. Piontek and R. Kiessling (1986). "Selective rejection of H-2-

deficient lymphoma variants suggests alternative immune defence strategy." Nature

319(6055): 675-678.

Katz, G., R. Gazit, T. I. Arnon, T. Gonen-Gross, G. Tarcic, G. Markel, R. Gruda, H. Achdout, O.

Drize, S. Merims and O. Mandelboim (2004). "MHC class I-independent recognition of

NK-activating receptor KIR2DS4." Journal of Immunology 173(3): 1819-1825.

Khalidi, H. S., L. J. Medeiros, K. L. Chang, R. K. Brynes, M. L. Slovak and D. A. Arber (1998).

"The immunophenotype of adult acute myeloid leukemia: high frequency of lymphoid

antigen expression and comparison of immunophenotype, French-American-British

classification, and karyotypic abnormalities." American Journal of Clinical Pathology

109(2): 211-220.

Kiessling, R., E. Klein, H. Pross and H. Wigzell (1975). ""Natural" killer cells in the mouse. II.

Cytotoxic cells with specificity for mouse Moloney leukemia cells. Characteristics of the

killer cell." European Journal of Immunology 5(2): 117-121.

Page 210: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

190

Kiessling, R., E. Klein and H. Wigzell (1975). ""Natural" killer cells in the mouse. I. Cytotoxic

cells with specificity for mouse Moloney leukemia cells. Specificity and distribution

according to genotype." European Journal of Immunology 5(2): 112-117.

Kirchgessner, C. U., C. K. Patil, J. W. Evans, C. A. Cuomo, L. M. Fried, T. Carter, M. A.

Oettinger and J. M. Brown (1995). "DNA-dependent kinase (p350) as a candidate gene

for the murine SCID defect." Science 267(5201): 1178-1183.

Klein, E., H. Ben-Bassat, H. Neumann, P. Ralph, J. Zeuthen, A. Polliack and F. Vanky (1976).

"Properties of the K562 cell line, derived from a patient with chronic myeloid leukemia."

International Journal of Cancer 18(4): 421-431.

Klingemann, H., E. Wong and G. Maki (1996). "A cytotoxic NK-cell line (NK-92) for ex vivo

purging of leukemia from blood." Blood and Marrow Transplantation 2(2): 68-75.

Klingemann, H. G., E. Wong and G. Maki (1996). "A cytotoxic NK-cell line (NK-92) for ex

vivo purging of leukemia from blood." Biology of Blood & Marrow Transplantation 2(2):

68-75.

Kobayashi, E., S. Motoi, M. Sugiura, M. Kajikawa, S. Kojima, J. Kohroki and Y. Masuho

(2014). "Antibody-dependent cellular cytotoxicity and cytokine/chemokine secretion by

KHYG-1 cells stably expressing FcgammaRIIIA." Immunology Letters 161(1): 59-64.

Kochenderfer, J. N., M. E. Dudley, R. O. Carpenter, S. H. Kassim, J. J. Rose, W. G. Telford, F.

T. Hakim, D. C. Halverson, D. H. Fowler, N. M. Hardy, A. R. Mato, D. D. Hickstein, J.

C. Gea-Banacloche, S. Z. Pavletic, C. Sportes, I. Maric, S. A. Feldman, B. G. Hansen, J.

S. Wilder, B. Blacklock-Schuver, B. Jena, M. R. Bishop, R. E. Gress and S. A.

Rosenberg (2013). "Donor-derived CD19-targeted T cells cause regression of malignancy

persisting after allogeneic hematopoietic stem cell transplantation." Blood 122(25): 4129-

4139.

Koeffler, H. P., R. Billing, A. J. Lusis, R. Sparkes and D. W. Golde (1980). "An undifferentiated

variant derived from the human acute myelogenous leukemia cell line (KG-1)." Blood

56(2): 265-273.

Koeffler, H. P. and D. W. Golde (1978). "Acute myelogenous leukemia: a human cell line

responsive to colony-stimulating activity." Science 200(4346): 1153-1154.

Kohler, G. and C. Milstein (1975). "Continuous cultures of fused cells secreting antibody of

predefined specificity." Nature 256(5517): 495-497.

Kojima, Y., A. Kawasaki-Koyanagi, N. Sueyoshi, A. Kanai, H. Yagita and K. Okumura (2002).

"Localization of Fas ligand in cytoplasmic granules of CD8+ cytotoxic T lymphocytes

and natural killer cells: participation of Fas ligand in granule exocytosis model of

cytotoxicity." Biochemical and Biophysical Research Communications 296(2): 328-336.

Kondo, T., T. Setoguchi and T. Taga (2004). "Persistence of a small subpopulation of cancer

stem-like cells in the C6 glioma cell line." Proc Natl Acad Sci U S A 101(3): 781-786.

Page 211: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

191

Kottaridis, P. D., R. E. Gale, M. E. Frew, G. Harrison, S. E. Langabeer, A. A. Belton, H. Walker,

K. Wheatley, D. T. Bowen, A. K. Burnett, A. H. Goldstone and D. C. Linch (2001). "The

presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia

(AML) adds important prognostic information to cytogenetic risk group and response to

the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom

Medical Research Council AML 10 and 12 trials." Blood 98(6): 1752-1759.

Krause, D. S., M. J. Fackler, C. I. Civin and W. S. May (1996). "CD34: structure, biology, and

clinical utility." Blood 87(1): 1-13.

Krause, S. W., R. Gastpar, R. Andreesen, C. Gross, H. Ullrich, G. Thonigs, K. Pfister and G.

Multhoff (2004). "Treatment of colon and lung cancer patients with ex vivo heat shock

protein 70-peptide-activated, autologous natural killer cells: a clinical phase i trial."

Clinical Cancer Research 10(11): 3699-3707.

Kuang, A. A., G. E. Diehl, J. Zhang and A. Winoto (2000). "FADD is required for DR4- and

DR5-mediated apoptosis: lack of trail-induced apoptosis in FADD-deficient mouse

embryonic fibroblasts." Journal of Biological Chemistry 275(33): 25065-25068.

Langenkamp, U., U. Siegler, S. Jorger, S. Diermayr, A. Gratwohl, C. P. Kalberer and A.

Wodnar-Filipowicz (2009). "Human acute myeloid leukemia CD34+CD38- stem cells

are susceptible to allorecognition and lysis by single KIR-expressing natural killer cells."

Haematologica 94(11): 1590-1594.

Lanier, L. L. (2003). "Natural killer cell receptor signaling." Current Opinion in Immunology

15(3): 308-314.

Lanier, L. L. (2005). "NK cell recognition." Annual Review of Immunology 23: 225-274.

Lanier, L. L., C. Chang and J. H. Phillips (1994). "Human NKR-P1A. A disulfide-linked

homodimer of the C-type lectin superfamily expressed by a subset of NK and T

lymphocytes." Journal of Immunology 153(6): 2417-2428.

Lanier, L. L., B. Corliss, J. Wu and J. H. Phillips (1998). "Association of DAP12 with activating

CD94/NKG2C NK cell receptors." Immunity 8(6): 693-701.

Lapidot, T., C. Sirard, J. Vormoor, B. Murdoch, T. Hoang, J. Caceres-Cortes, M. Minden, B.

Paterson, M. A. Caligiuri and J. E. Dick (1994). "A cell initiating human acute myeloid

leukaemia after transplantation into SCID mice." Nature 367(6464): 645-648.

Laubach, J. and A. V. Rao (2008). "Current and emerging strategies for the management of acute

myeloid leukemia in the elderly." Oncologist 13(10): 1097-1108.

Lazetic, S., C. Chang, J. P. Houchins, L. L. Lanier and J. H. Phillips (1996). "Human natural

killer cell receptors involved in MHC class I recognition are disulfide-linked

heterodimers of CD94 and NKG2 subunits." Journal of Immunology 157(11): 4741-

4745.

Page 212: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

192

Lee, N., M. Llano, M. Carretero, A. Ishitani, F. Navarro, M. Lopez-Botet and D. E. Geraghty

(1998). "HLA-E is a major ligand for the natural killer inhibitory receptor

CD94/NKG2A." Proceedings of the National Academy of Sciences of the United States

of America 95(9): 5199-5204.

Legrand, O., J. Y. Perrot, M. Baudard, A. Cordier, R. Lautier, G. Simonin, R. Zittoun, N.

Casadevall and J. P. Marie (2000). "The immunophenotype of 177 adults with acute

myeloid leukemia: proposal of a prognostic score." Blood 96(3): 870-877.

Leyton, J. V., M. Hu, C. Gao, P. V. Turner, J. E. Dick, M. Minden and R. M. Reilly (2011).

"Auger electron radioimmunotherapeutic agent specific for the CD123+/CD131-

phenotype of the leukemia stem cell population." Journal of Nuclear Medicine 52(9):

1465-1473.

Li, H. Y., D. H. Deng, Y. Huang, F. H. Ye, L. L. Huang, Q. Xiao, B. Zhang, B. B. Ye, Y. R. Lai,

Z. N. Mo and Z. F. Liu (2014). "Favorable Prognosis of Biallelic CEBPA Gene

Mutations in Acute Myeloid Leukemia Patients: a meta-analysis." European Journal of

Haematology.

Lichtenheld, M. G., K. J. Olsen, P. Lu, D. M. Lowrey, A. Hameed, H. Hengartner and E. R.

Podack (1988). "Structure and function of human perforin." Nature 335(6189): 448-451.

Lieber, M. R., J. E. Hesse, S. Lewis, G. C. Bosma, N. Rosenberg, K. Mizuuchi, M. J. Bosma and

M. Gellert (1988). "The defect in murine severe combined immune deficiency: joining of

signal sequences but not coding segments in V(D)J recombination." Cell 55(1): 7-16.

Lieberman, J. (2010). "Granzyme A activates another way to die." Immunological Reviews

235(1): 93-104.

Litzow, M. R., S. Tarima, W. S. Perez, B. J. Bolwell, M. S. Cairo, B. M. Camitta, C. S. Cutler,

M. de Lima, J. F. Dipersio, R. P. Gale, A. Keating, H. M. Lazarus, S. Luger, D. I. Marks,

R. T. Maziarz, P. L. McCarthy, M. C. Pasquini, G. L. Phillips, J. D. Rizzo, J. Sierra, M.

S. Tallman and D. J. Weisdorf (2010). "Allogeneic transplantation for therapy-related

myelodysplastic syndrome and acute myeloid leukemia." Blood 115(9): 1850-1857.

Locafaro, G., G. Amodio, D. Tomasoni, C. Tresoldi, F. Ciceri and S. Gregori (2014). "HLA-G

expression on blasts and tolerogenic cells in patients affected by acute myeloid

leukemia." J Immunol Res 2014: 636292.

Lowenberg, B., J. R. Downing and A. Burnett (1999). "Acute myeloid leukemia." New England

Journal of Medicine 341(14): 1051-1062.

Lozzio, C. B. and B. B. Lozzio (1975). "Human chronic myelogenous leukemia cell-line with

positive Philadelphia chromosome." Blood 45(3): 321-334.

Machado, E. A., D. A. Gerard, C. B. Lozzio, B. B. Lozzio, J. R. Mitchell and D. W. Golde

(1984). "Proliferation and differentiation of human myeloid leukemic cells in

immunodeficient mice: electron microscopy and cytochemistry." Blood 63(5): 1015-

1022.

Page 213: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

193

Majumder, D., D. Bandyopadhyay, S. Chandra, N. Mukherjee and S. Banerjee (2006). "Lack of

HLA-E surface expression is due to deficiency of HLA-E transcripts in the malignant

hematopoietic cells of leukemic patients." Leukemia Research 30(2): 242-245.

Maki, G., H. G. Klingemann, J. A. Martinson and Y. K. Tam (2001). "Factors regulating the

cytotoxic activity of the human natural killer cell line, NK-92." Journal of hematotherapy

& stem cell research 10(3): 369-383.

Malarkannan, S. (2006). "The balancing act: inhibitory Ly49 regulate NKG2D-mediated NK cell

functions." Seminars in Immunology 18(3): 186-192.

Malavasi, F., S. Deaglio, A. Funaro, E. Ferrero, A. L. Horenstein, E. Ortolan, T. Vaisitti and S.

Aydin (2008). "Evolution and function of the ADP ribosyl cyclase/CD38 gene family in

physiology and pathology." Physiological Reviews 88(3): 841-886.

Malynn, B. A., T. K. Blackwell, G. M. Fulop, G. A. Rathbun, A. J. Furley, P. Ferrier, L. B.

Heinke, R. A. Phillips, G. D. Yancopoulos and F. W. Alt (1988). "The scid defect affects

the final step of the immunoglobulin VDJ recombinase mechanism." Cell 54(4): 453-460.

Manaster, I., S. Mizrahi, D. Goldman-Wohl, H. Y. Sela, N. Stern-Ginossar, D. Lankry, R. Gruda,

A. Hurwitz, Y. Bdolah, R. Haimov-Kochman, S. Yagel and O. Mandelboim (2008).

"Endometrial NK cells are special immature cells that await pregnancy." Journal of

Immunology 181(3): 1869-1876.

Mandelboim, O., N. Lieberman, M. Lev, L. Paul, T. I. Arnon, Y. Bushkin, D. M. Davis, J. L.

Strominger, J. W. Yewdell and A. Porgador (2001). "Recognition of haemagglutinins on

virus-infected cells by NKp46 activates lysis by human NK cells." Nature 409(6823):

1055-1060.

Mandelboim, O., P. Malik, D. M. Davis, C. H. Jo, J. E. Boyson and J. L. Strominger (1999).

"Human CD16 as a lysis receptor mediating direct natural killer cell cytotoxicity."

Proceedings of the National Academy of Sciences of the United States of America

96(10): 5640-5644.

Mandelboim, O. and A. Porgador (2001). "NKp46." International Journal of Biochemistry and

Cell Biology 33(12): 1147-1150.

Mandelboim, O., H. T. Reyburn, M. Vales-Gomez, L. Pazmany, M. Colonna, G. Borsellino and

J. L. Strominger (1996). "Protection from lysis by natural killer cells of group 1 and 2

specificity is mediated by residue 80 in human histocompatibility leukocyte antigen C

alleles and also occurs with empty major histocompatibility complex molecules." Journal

of Experimental Medicine 184(3): 913-922.

Mardiros, A., C. Dos Santos, T. McDonald, C. E. Brown, X. Wang, L. E. Budde, L. Hoffman, B.

Aguilar, W. C. Chang, W. Bretzlaff, B. Chang, M. Jonnalagadda, R. Starr, J. R. Ostberg,

M. C. Jensen, R. Bhatia and S. J. Forman (2013). "T cells expressing CD123-specific

chimeric antigen receptors exhibit specific cytolytic effector functions and antitumor

effects against human acute myeloid leukemia." Blood 122(18): 3138-3148.

Page 214: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

194

Marin, R., F. Ruiz-Cabello, S. Pedrinaci, R. Mendez, P. Jimenez, D. E. Geraghty and F. Garrido

(2003). "Analysis of HLA-E expression in human tumors." Immunogenetics 54(11): 767-

775.

Markel, G., R. Seidman, M. J. Besser, N. Zabari, R. Ortenberg, R. Shapira, A. J. Treves, R.

Loewenthal, A. Orenstein, A. Nagler and J. Schachter (2009). "Natural killer lysis

receptor (NKLR)/NKLR-ligand matching as a novel approach for enhancing anti-tumor

activity of allogeneic NK cells." PLoS ONE 4(5): e5597.

Martinvalet, D., D. M. Dykxhoorn, R. Ferrini and J. Lieberman (2008). "Granzyme A cleaves a

mitochondrial complex I protein to initiate caspase-independent cell death." Cell 133(4):

681-692.

Masson, D. and J. Tschopp (1987). "A family of serine esterases in lytic granules of cytolytic T

lymphocytes." Cell 49(5): 679-685.

Masson, D., M. Zamai and J. Tschopp (1986). "Identification of granzyme A isolated from

cytotoxic T-lymphocyte-granules as one of the proteases encoded by CTL-specific

genes." FEBS Letters 208(1): 84-88.

Matsui, W., C. A. Huff, Q. Wang, M. T. Malehorn, J. Barber, Y. Tanhehco, B. D. Smith, C. I.

Civin and R. J. Jones (2004). "Characterization of clonogenic multiple myeloma cells."

Blood 103(6): 2332-2336.

Matsuo, Y. (2003). "Immunoprofiling of cell lines derived from natural killer-cell and natural

killer-like T-cell leukemia–lymphoma." Leukemia Research 27(10): 935-945.

Maul-Pavicic, A., S. C. Chiang, A. Rensing-Ehl, B. Jessen, C. Fauriat, S. M. Wood, S. Sjoqvist,

M. Hufnagel, I. Schulze, T. Bass, W. W. Schamel, S. Fuchs, H. Pircher, C. A. McCarl, K.

Mikoshiba, K. Schwarz, S. Feske, Y. T. Bryceson and S. Ehl (2011). "ORAI1-mediated

calcium influx is required for human cytotoxic lymphocyte degranulation and target cell

lysis." Proceedings of the National Academy of Sciences of the United States of America

108(8): 3324-3329.

McCann, F. E., B. Vanherberghen, K. Eleme, L. M. Carlin, R. J. Newsam, D. Goulding and D.

M. Davis (2003). "The size of the synaptic cleft and distinct distributions of filamentous

actin, ezrin, CD43, and CD45 at activating and inhibitory human NK cell immune

synapses." Journal of Immunology 170(6): 2862-2870.

McCulloch, E. A. and J. E. Till (1960). "The radiation sensitivity of normal mouse bone marrow

cells, determined by quantitative marrow transplantation into irradiated mice." Radiation

Research 13: 115-125.

McCune, J. M., R. Namikawa, H. Kaneshima, L. D. Shultz, M. Lieberman and I. L. Weissman

(1988). "The SCID-hu mouse: murine model for the analysis of human hematolymphoid

differentiation and function." Science 241(4873): 1632-1639.

McLaughlin, P., A. J. Grillo-Lopez, B. K. Link, R. Levy, M. S. Czuczman, M. E. Williams, M.

R. Heyman, I. Bence-Bruckler, C. A. White, F. Cabanillas, V. Jain, A. D. Ho, J. Lister,

Page 215: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

195

K. Wey, D. Shen and B. K. Dallaire (1998). "Rituximab chimeric anti-CD20 monoclonal

antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose

treatment program." Journal of Clinical Oncology 16(8): 2825-2833.

Meshinchi, S., W. G. Woods, D. L. Stirewalt, D. A. Sweetser, J. D. Buckley, T. K. Tjoa, I. D.

Bernstein and J. P. Radich (2001). "Prevalence and prognostic significance of Flt3

internal tandem duplication in pediatric acute myeloid leukemia." Blood 97(1): 89-94.

Miller, J. S., Y. Soignier, A. Panoskaltsis-Mortari, S. A. McNearney, G. H. Yun, S. K. Fautsch,

D. McKenna, C. Le, T. E. Defor, L. J. Burns, P. J. Orchard, B. R. Blazar, J. E. Wagner,

A. Slungaard, D. J. Weisdorf, I. J. Okazaki and P. B. McGlave (2005). "Successful

adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with

cancer." Blood 105(8): 3051-3057.

Miller, R. D., J. Hogg, J. H. Ozaki, D. Gell, S. P. Jackson and R. Riblet (1995). "Gene for the

catalytic subunit of mouse DNA-dependent protein kinase maps to the scid locus."

Proceedings of the National Academy of Sciences of the United States of America

92(23): 10792-10795.

Miller, W. H., Jr., K. Levine, A. DeBlasio, S. R. Frankel, E. Dmitrovsky and R. P. Warrell, Jr.

(1993). "Detection of minimal residual disease in acute promyelocytic leukemia by a

reverse transcription polymerase chain reaction assay for the PML/RAR-alpha fusion

mRNA." Blood 82(6): 1689-1694.

Moretta, A., C. Bottino, D. Pende, G. Tripodi, G. Tambussi, O. Viale, A. Orengo, M. Barbaresi,

A. Merli, E. Ciccone and et al. (1990). "Identification of four subsets of human CD3-

CD16+ natural killer (NK) cells by the expression of clonally distributed functional

surface molecules: correlation between subset assignment of NK clones and ability to

mediate specific alloantigen recognition." Journal of Experimental Medicine 172(6):

1589-1598.

Moretta, A., C. Bottino, M. Vitale, D. Pende, R. Biassoni, M. C. Mingari and L. Moretta (1996).

"Receptors for HLA class-I molecules in human natural killer cells." Annual Review of

Immunology 14: 619-648.

Moretta, A., S. Sivori, M. Vitale, D. Pende, L. Morelli, R. Augugliaro, C. Bottino and L. Moretta

(1995). "Existence of both inhibitory (p58) and activatory (p50) receptors for HLA-C

molecules in human natural killer cells." Journal of Experimental Medicine 182(3): 875-

884.

Moretta, A., G. Tambussi, C. Bottino, G. Tripodi, A. Merli, E. Ciccone, G. Pantaleo and L.

Moretta (1990). "A novel surface antigen expressed by a subset of human CD3- CD16+

natural killer cells. Role in cell activation and regulation of cytolytic function." Journal of

Experimental Medicine 171(3): 695-714.

Moretti, S., F. Lanza, M. Dabusti, A. Tieghi, D. Campioni, M. Dominici and G. L. Castoldi

(2001). "CD123 (interleukin 3 receptor alpha chain)." Journal of Biological Regulators

and Homeostatic Agents 15(1): 98-100.

Page 216: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

196

Morgan, R. A., J. C. Yang, M. Kitano, M. E. Dudley, C. M. Laurencot and S. A. Rosenberg

(2010). "Case report of a serious adverse event following the administration of T cells

transduced with a chimeric antigen receptor recognizing ERBB2." Mol Ther 18(4): 843-

851.

Mosier, D. E., R. J. Gulizia, S. M. Baird and D. B. Wilson (1988). "Transfer of a functional

human immune system to mice with severe combined immunodeficiency." Nature

335(6187): 256-259.

Nagata, H., A. Konno, N. Kimura, Y. Zhang, M. Kimura, A. Demachi, T. Sekine, K. Yamamoto

and N. Shimizu (2001). "Characterization of novel natural killer (NK)-cell and

gammadelta T-cell lines established from primary lesions of nasal T/NK-cell lymphomas

associated with the Epstein-Barr virus." Blood 97(3): 708-713.

Nakajima, H., M. Cella, A. Bouchon, H. L. Grierson, J. Lewis, C. S. Duckett, J. I. Cohen and M.

Colonna (2000). "Patients with X-linked lymphoproliferative disease have a defect in

2B4 receptor-mediated NK cell cytotoxicity." European Journal of Immunology 30(11):

3309-3318.

Nakajima, H., M. Cella, H. Langen, A. Friedlein and M. Colonna (1999). "Activating

interactions in human NK cell recognition: the role of 2B4-CD48." European Journal of

Immunology 29(5): 1676-1683.

Nakajima, H. and M. Colonna (2000). "2B4: an NK cell activating receptor with unique

specificity and signal transduction mechanism." Human Immunology 61(1): 39-43.

Nara, N. and T. Miyamoto (1982). "Direct and serial transplantation of human acute myeloid

leukaemia into nude mice." British Journal of Cancer 45(5): 778-782.

Neri, S., E. Mariani, A. Meneghetti, L. Cattini and A. Facchini (2001). "Calcein-acetyoxymethyl

cytotoxicity assay: standardization of a method allowing additional analyses on recovered

effector cells and supernatants." Clinical and Diagnostic Laboratory Immunology 8(6):

1131-1135.

Nguyen, S., V. Beziat, N. Dhedin, M. Kuentz, J. P. Vernant, P. Debre and V. Vieillard (2009).

"HLA-E upregulation on IFN-gamma-activated AML blasts impairs CD94/NKG2A-

dependent NK cytolysis after haplo-mismatched hematopoietic SCT." Bone Marrow

Transplantation 43(9): 693-699.

Nielsen, J. S. and K. M. McNagny (2008). "Novel functions of the CD34 family." Journal of Cell

Science 121(Pt 22): 3683-3692.

Nilsson, K., B. C. Giovanella, J. S. Stehlin and G. Klein (1977). "Tumorigenicity of human

hematopoietic cell lines in athymic nude mice." International Journal of Cancer 19(3):

337-344.

Nonoyama, S., F. O. Smith, I. D. Bernstein and H. D. Ochs (1993). "Strain-dependent leakiness

of mice with severe combined immune deficiency." Journal of Immunology 150(9):

3817-3824.

Page 217: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

197

Noonan, K., W. Matsui, P. Serafini, R. Carbley, G. Tan, J. Khalili, M. Bonyhadi, H. Levitsky, K.

Whartenby and I. Borrello (2005). "Activated marrow-infiltrating lymphocytes

effectively target plasma cells and their clonogenic precursors." Cancer research 65(5):

2026-2034.

Notter, M., W. D. Ludwig, S. Bremer and E. Thiel (1993). "Selective targeting of human

lymphokine-activated killer cells by CD3 monoclonal antibody against the interferon-

inducible high-affinity Fc gamma RI receptor (CD64) on autologous acute myeloid

leukemic blast cells." Blood 82(10): 3113-3124.

O'Brien, C. A., A. Kreso and J. E. Dick (2009). "Cancer stem cells in solid tumors: an overview."

Seminars in radiation oncology 19(2): 71-77.

O'Brien, C. A., A. Pollett, S. Gallinger and J. E. Dick (2007). "A human colon cancer cell

capable of initiating tumour growth in immunodeficient mice." Nature 445(7123): 106-

110.

Ogawa, M., D. E. Bergsagel and E. A. McCulloch (1973). "Sensitivity of human and murine

hemopoietic precursor cells to chemotherapeutic agents assessed in cell culture." Blood

42(6): 851-856.

Ohbo, K., T. Suda, M. Hashiyama, A. Mantani, M. Ikebe, K. Miyakawa, M. Moriyama, M.

Nakamura, M. Katsuki, K. Takahashi, K. Yamamura and K. Sugamura (1996).

"Modulation of hematopoiesis in mice with a truncated mutant of the interleukin-2

receptor gamma chain." Blood 87(3): 956-967.

Olcese, L., P. Lang, F. Vely, A. Cambiaggi, D. Marguet, M. Blery, K. L. Hippen, R. Biassoni, A.

Moretta, L. Moretta, J. C. Cambier and E. Vivier (1996). "Human and mouse killer-cell

inhibitory receptors recruit PTP1C and PTP1D protein tyrosine phosphatases." Journal of

Immunology 156(12): 4531-4534.

Olesen, L. H., N. Clausen, A. Dimitrijevic, G. Kerndrup, E. Kjeldsen and P. Hokland (2004).

"Prospective application of a multiplex reverse transcription-polymerase chain reaction

assay for the detection of balanced translocations in leukaemia: a single-laboratory study

of 390 paediatric and adult patients." British Journal of Haematology 127(1): 59-66.

Orange, J. S. (2006). "Human natural killer cell deficiencies." Curr Opin Allergy Clin Immunol

6(6): 399-409.

Ostergaard, H. L., K. P. Kane, M. F. Mescher and W. R. Clark (1987). "Cytotoxic T lymphocyte

mediated lysis without release of serine esterase." Nature 330(6143): 71-72.

Ozdemir, O., Y. Ravindranath and S. Savasan (2003). "Cell-mediated cytotoxicity evaluation

using monoclonal antibody staining for target or effector cells with annexinV/propidium

iodide colabeling by fluorosphere-adjusted counts on three-color flow cytometry."

Cytometry A 56(1): 53-60.

Ozdemir, O., Y. Ravindranath and S. Savasan (2003). "Cell-mediated cytotoxicity evaluation

using monoclonal antibody staining for target or effector cells with annexinV/propidium

Page 218: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

198

iodide colabeling by fluorosphere-adjusted counts on three-color flow cytometry."

Cytometry Part A: The Journal of the International Society for Analytical Cytology

56(1): 53-60.

Palu, G., P. Selby, R. Powles and P. Alexander (1979). "Spontaneous regression of human acute

myeloid leukaemia xenografts and phenotypic evidence for maturation." British Journal

of Cancer 40(5): 731-735.

Park, C. H., M. Amare, M. A. Savin, J. W. Goodwin, M. M. Newcomb and B. Hoogstraten

(1980). "Prediction of chemotherapy response in human leukemia using an in vitro

chemotherapy sensitivity test on the leukemic colony-forming cells." Blood 55(4): 595-

601.

Park, C. H., D. E. Bergsagel and E. A. McCulloch (1971). "Mouse myeloma tumor stem cells: a

primary cell culture assay." Journal of the National Cancer Institute 46(2): 411-422.

Parkhurst, M. R., J. P. Riley, M. E. Dudley and S. A. Rosenberg (2011). "Adoptive transfer of

autologous natural killer cells leads to high levels of circulating natural killer cells but

does not mediate tumor regression." Clinical Cancer Research 17(19): 6287-6297.

Parolini, S., C. Bottino, M. Falco, R. Augugliaro, S. Giliani, R. Franceschini, H. D. Ochs, H.

Wolf, J. Y. Bonnefoy, R. Biassoni, L. Moretta, L. D. Notarangelo and A. Moretta (2000).

"X-linked lymphoproliferative disease. 2B4 molecules displaying inhibitory rather than

activating function are responsible for the inability of natural killer cells to kill Epstein-

Barr virus-infected cells." Journal of Experimental Medicine 192(3): 337-346.

Pearce, D. J., D. Taussig, K. Zibara, L.-L. Smith, C. M. Ridler, C. Preudhomme, B. D. Young,

A. Z. Rohatiner, T. A. Lister and D. Bonnet (2006). "AML engraftment in the

NOD/SCID assay reflects the outcome of AML: implications for our understanding of the

heterogeneity of AML." Blood 107(3): 1166-1173.

Pende, D., C. Cantoni, P. Rivera, M. Vitale, R. Castriconi, S. Marcenaro, M. Nanni, R. Biassoni,

C. Bottino, A. Moretta and L. Moretta (2001). "Role of NKG2D in tumor cell lysis

mediated by human NK cells: cooperation with natural cytotoxicity receptors and

capability of recognizing tumors of nonepithelial origin." European Journal of

Immunology 31(4): 1076-1086.

Pende, D., S. Parolini, A. Pessino, S. Sivori, R. Augugliaro, L. Morelli, E. Marcenaro, L.

Accame, A. Malaspina, R. Biassoni, C. Bottino, L. Moretta and A. Moretta (1999).

"Identification and molecular characterization of NKp30, a novel triggering receptor

involved in natural cytotoxicity mediated by human natural killer cells." Journal of

Experimental Medicine 190(10): 1505-1516.

Perez-Quintero, L. A., R. Roncagalli, H. Guo, S. Latour, D. Davidson and A. Veillette (2014).

"EAT-2, a SAP-like adaptor, controls NK cell activation through phospholipase

Cgamma, Ca++, and Erk, leading to granule polarization." Journal of Experimental

Medicine 211(4): 727-742.

Page 219: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

199

Pessino, A., S. Sivori, C. Bottino, A. Malaspina, L. Morelli, L. Moretta, R. Biassoni and A.

Moretta (1998). "Molecular cloning of NKp46: a novel member of the immunoglobulin

superfamily involved in triggering of natural cytotoxicity." Journal of Experimental

Medicine 188(5): 953-960.

Pierce, R. A., E. D. Field, T. Mutis, T. N. Golovina, C. Von Kap-Herr, M. Wilke, J. Pool, J.

Shabanowitz, M. J. Pettenati, L. C. Eisenlohr, D. F. Hunt, E. Goulmy and V. H.

Engelhard (2001). "The HA-2 minor histocompatibility antigen is derived from a diallelic

gene encoding a novel human class I myosin protein." Journal of Immunology 167(6):

3223-3230.

Pizzitola, I., F. Anjos-Afonso, K. Rouault-Pierre, F. Lassailly, S. Tettamanti, O. Spinelli, A.

Biondi, E. Biagi and D. Bonnet (2014). "Chimeric antigen receptors against

CD33/CD123 antigens efficiently target primary acute myeloid leukemia cells in vivo."

Leukemia 28(8): 1596-1605.

Podack, E. R. and G. Dennert (1983). "Assembly of two types of tubules with putative cytolytic

function by cloned natural killer cells." Nature 302(5907): 442-445.

Podack, E. R. and P. J. Konigsberg (1984). "Cytolytic T cell granules. Isolation, structural,

biochemical, and functional characterization." Journal of Experimental Medicine 160(3):

695-710.

Pogge von Strandmann, E., V. R. Simhadri, B. von Tresckow, S. Sasse, K. S. Reiners, H. P.

Hansen, A. Rothe, B. Boll, V. L. Simhadri, P. Borchmann, P. J. McKinnon, M. Hallek

and A. Engert (2007). "Human leukocyte antigen-B-associated transcript 3 is released

from tumor cells and engages the NKp30 receptor on natural killer cells." Immunity

27(6): 965-974.

Polakova, K., M. Krcova, D. Kuba and G. Russ (2003). "Analysis of HLA-G expression in

malignant hematopoetic cells from leukemia patients." Leukemia Research 27(7): 643-

648.

Porter, D. L., B. L. Levine, M. Kalos, A. Bagg and C. H. June (2011). "Chimeric antigen

receptor-modified T cells in chronic lymphoid leukemia." New England Journal of

Medicine 365(8): 725-733.

Potter, G. K., R. N. Shen and J. W. Chiao (1984). "Nude mice as models for human leukemia

studies." American Journal of Pathology 114(3): 360-366.

Pross, H. F. and M. Jondal (1975). "Cytotoxic lymphocytes from normal donors. A functional

marker of human non-T lymphocytes." Clinical and Experimental Immunology 21(2):

226-235.

Puck, T. T., P. I. Marcus and S. J. Cieciura (1956). "Clonal growth of mammalian cells in vitro;

growth characteristics of colonies from single HeLa cells with and without a feeder

layer." Journal of Experimental Medicine 103(2): 273-283.

Page 220: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

200

Reiffers, J., M. H. Gaspard, D. Maraninchi, M. Michallet, G. Marit, A. M. Stoppa, B. Corront, B.

David, J. A. Gastaut, J. J. Scotto and et al. (1989). "Comparison of allogeneic or

autologous bone marrow transplantation and chemotherapy in patients with acute

myeloid leukaemia in first remission: a prospective controlled trial." British Journal of

Haematology 72(1): 57-63.

Ribeiro, R. C., B. I. Razzouk, S. Pounds, N. Hijiya, C. H. Pui and J. E. Rubnitz (2005).

"Successive clinical trials for childhood acute myeloid leukemia at St Jude Children's

Research Hospital, from 1980 to 2000." Leukemia 19(12): 2125-2129.

Ricci-Vitiani, L., D. G. Lombardi, E. Pilozzi, M. Biffoni, M. Todaro, C. Peschle and R. De

Maria (2007). "Identification and expansion of human colon-cancer-initiating cells."

Nature 445(7123): 111-115.

Ries LAG, M. D., Krapcho M, Stinchcomb DG, Howlader N, Horner MJ, Mariotto A, Miller

BA, Feuer EJ, Altekruse SF, Lewis DR, Clegg L, Eisner MP, Reichman M, Edwards BK

(eds). (2008). SEER Cancer Statistics Review, 1975-2005, National Cancer Institute.

Bethesda, MD.

Rieux-Laucat, F., F. Le Deist, C. Hivroz, I. A. Roberts, K. M. Debatin, A. Fischer and J. P. de

Villartay (1995). "Mutations in Fas associated with human lymphoproliferative syndrome

and autoimmunity." Science 268(5215): 1347-1349.

Robertson, M. J., M. A. Caligiuri, T. J. Manley, H. Levine and J. Ritz (1990). "Human natural

killer cell adhesion molecules. Differential expression after activation and participation in

cytolysis." Journal of Immunology 145(10): 3194-3201.

Robertson, M. J., K. J. Cochran, C. Cameron, J. M. Le, R. Tantravahi and J. Ritz (1996).

"Characterization of a cell line, NKL, derived from an aggressive human natural killer

cell leukemia." Experimental Hematology 24(3): 406-415.

Rosen, D. B., M. Araki, J. A. Hamerman, T. Chen, T. Yamamura and L. L. Lanier (2004). "A

Structural basis for the association of DAP12 with mouse, but not human, NKG2D."

Journal of Immunology 173(4): 2470-2478.

Rosen, D. B., J. Bettadapura, M. Alsharifi, P. A. Mathew, H. S. Warren and L. L. Lanier (2005).

"Cutting edge: lectin-like transcript-1 is a ligand for the inhibitory human NKR-P1A

receptor." Journal of Immunology 175(12): 7796-7799.

Rosen, D. B., W. Cao, D. T. Avery, S. G. Tangye, Y. J. Liu, J. P. Houchins and L. L. Lanier

(2008). "Functional consequences of interactions between human NKR-P1A and its

ligand LLT1 expressed on activated dendritic cells and B cells." Journal of Immunology

180(10): 6508-6517.

Rosenberg, S. A., M. T. Lotze, L. M. Muul, S. Leitman, A. E. Chang, S. E. Ettinghausen, Y. L.

Matory, J. M. Skibber, E. Shiloni, J. T. Vetto and et al. (1985). "Observations on the

systemic administration of autologous lymphokine-activated killer cells and recombinant

interleukin-2 to patients with metastatic cancer." New England Journal of Medicine

313(23): 1485-1492.

Page 221: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

201

Rosenberg, S. A., M. T. Lotze, J. C. Yang, P. M. Aebersold, W. M. Linehan, C. A. Seipp and D.

E. White (1989). "Experience with the use of high-dose interleukin-2 in the treatment of

652 cancer patients." Annals of Surgery 210(4): 474-484; discussion 484-475.

Rosenberg, S. A., M. T. Lotze, J. C. Yang, S. L. Topalian, A. E. Chang, D. J. Schwartzentruber,

P. Aebersold, S. Leitman, W. M. Linehan, C. A. Seipp and et al. (1993). "Prospective

randomized trial of high-dose interleukin-2 alone or in conjunction with lymphokine-

activated killer cells for the treatment of patients with advanced cancer." Journal of the

National Cancer Institute 85(8): 622-632.

Rosenberg, S. A., P. Spiess and R. Lafreniere (1986). "A new approach to the adoptive

immunotherapy of cancer with tumor-infiltrating lymphocytes." Science 233(4770):

1318-1321.

Rouvier, E., M. F. Luciani and P. Golstein (1993). "Fas involvement in Ca(2+)-independent T

cell-mediated cytotoxicity." Journal of Experimental Medicine 177(1): 195-200.

Rowe, J. M. and M. S. Tallman (2010). "How I treat acute myeloid leukemia." Blood 116(17):

3147-3156.

Rubnitz, J. E. (2012). "How I treat pediatric acute myeloid leukemia." Blood 119(25): 5980-

5988.

Rubnitz, J. E., H. Inaba, G. Dahl, R. C. Ribeiro, W. P. Bowman, J. Taub, S. Pounds, B. I.

Razzouk, N. J. Lacayo, X. Cao, S. Meshinchi, B. Degar, G. Airewele, S. C. Raimondi, M.

Onciu, E. Coustan-Smith, J. R. Downing, W. Leung, C.-H. Pui and D. Campana (2010).

"Minimal residual disease-directed therapy for childhood acute myeloid leukaemia:

results of the AML02 multicentre trial." Lancet Oncology 11(6): 543-552.

Rubnitz, J. E., H. Inaba, R. C. Ribeiro, S. Pounds, B. Rooney, T. Bell, C. H. Pui and W. Leung

(2010). "NKAML: a pilot study to determine the safety and feasibility of haploidentical

natural killer cell transplantation in childhood acute myeloid leukemia." J Clin Oncol

28(6): 955-959.

Ruggeri, L., M. Capanni, A. Mancusi, M. F. Martelli and A. Velardi (2005). "The impact of

donor natural killer cell alloreactivity on allogeneic hematopoietic transplantation."

Transplant Immunology 14(3-4): 203-206.

Ruggeri, L., M. Capanni, E. Urbani, K. Perruccio, W. D. Shlomchik, A. Tosti, S. Posati, D.

Rogaia, F. Frassoni, F. Aversa, M. F. Martelli and A. Velardi (2002). "Effectiveness of

donor natural killer cell alloreactivity in mismatched hematopoietic transplants." Science

295(5562): 2097-2100.

Ruggeri, L., A. Mancusi, K. Perruccio, E. Burchielli, M. F. Martelli and A. Velardi (2005).

"Natural killer cell alloreactivity for leukemia therapy." Journal of Immunotherapy 28(3):

175-182.

Page 222: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

202

Ryan, J. C., E. C. Niemi, M. C. Nakamura and W. E. Seaman (1995). "NKR-P1A is a target-

specific receptor that activates natural killer cell cytotoxicity." Journal of Experimental

Medicine 181(5): 1911-1915.

San Miguel, J. F., M. B. Vidriales, C. Lopez-Berges, J. Diaz-Mediavilla, N. Gutierrez, C.

Canizo, F. Ramos, M. J. Calmuntia, J. J. Perez, M. Gonzalez and A. Orfao (2001). "Early

immunophenotypical evaluation of minimal residual disease in acute myeloid leukemia

identifies different patient risk groups and may contribute to postinduction treatment

stratification." Blood 98(6): 1746-1751.

Savani, B. N., S. Mielke, S. Adams, M. Uribe, K. Rezvani, A. S. Yong, J. Zeilah, R. Kurlander,

R. Srinivasan, R. Childs, N. Hensel and A. J. Barrett (2007). "Rapid natural killer cell

recovery determines outcome after T-cell-depleted HLA-identical stem cell

transplantation in patients with myeloid leukemias but not with acute lymphoblastic

leukemia." Leukemia 21(10): 2145-2152.

Sawyers, C. L., M. L. Gishizky, S. Quan, D. W. Golde and O. N. Witte (1992). "Propagation of

human blastic myeloid leukemias in the SCID mouse." Blood 79(8): 2089-2098.

Saxena, R. K., Q. B. Saxena and W. H. Adler (1982). "Identity of effector cells participating in

the reverse antibody-dependent cell-mediated cytotoxicity." Immunology 46(2): 459-464.

Schlecker, E., N. Fiegler, A. Arnold, P. Altevogt, S. Rose-John, G. Moldenhauer, A. Sucker, A.

Paschen, E. P. von Strandmann, S. Textor and A. Cerwenka (2014). "Metalloprotease-

mediated tumor cell shedding of B7-H6, the ligand of the natural killer cell-activating

receptor NKp30." Cancer Research 74(13): 3429-3440.

Schmaltz, C., O. Alpdogan, B. J. Kappel, S. J. Muriglan, J. A. Rotolo, J. Ongchin, L. M. Willis,

A. S. Greenberg, J. M. Eng, J. M. Crawford, G. F. Murphy, H. Yagita, H. Walczak, J. J.

Peschon and M. R. M. van den Brink (2002). "T cells require TRAIL for optimal graft-

versus-tumor activity." Nat Med 8(12): 1433-1437.

Scholler, J., T. L. Brady, G. Binder-Scholl, W. T. Hwang, G. Plesa, K. M. Hege, A. N. Vogel, M.

Kalos, J. L. Riley, S. G. Deeks, R. T. Mitsuyasu, W. B. Bernstein, N. E. Aronson, B. L.

Levine, F. D. Bushman and C. H. June (2012). "Decade-long safety and function of

retroviral-modified chimeric antigen receptor T cells." Science Translational Medicine

4(132): 132ra153.

Scott, A. M., J. P. Allison and J. D. Wolchok (2012). "Monoclonal antibodies in cancer therapy."

Cancer Immun 12: 14.

Segre, J. A., J. L. Nemhauser, B. A. Taylor, J. H. Nadeau and E. S. Lander (1995). "Positional

cloning of the nude locus: genetic, physical, and transcription maps of the region and

mutations in the mouse and rat." Genomics 28(3): 549-559.

Shlush, L. I., S. Zandi, A. Mitchell, W. C. Chen, J. M. Brandwein, V. Gupta, J. A. Kennedy, A.

D. Schimmer, A. C. Schuh, K. W. Yee, J. L. McLeod, M. Doedens, J. J. Medeiros, R.

Marke, H. J. Kim, K. Lee, J. D. McPherson, T. J. Hudson, A. M. Brown, F. Yousif, Q. M.

Trinh, L. D. Stein, M. D. Minden, J. C. Wang and J. E. Dick (2014). "Identification of

Page 223: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

203

pre-leukaemic haematopoietic stem cells in acute leukaemia." Nature 506(7488): 328-

333.

Shultz, L. D., S. J. Banuelos, J. Leif, M. C. Appel, M. Cunningham, K. Ballen, L. Burzenski and

D. L. Greiner (2003). "Regulation of human short-term repopulating cell (STRC)

engraftment in NOD/SCID mice by host CD122+ cells." Experimental hematology 31(6):

551-558.

Shultz, L. D., F. Ishikawa and D. L. Greiner (2007). "Humanized mice in translational

biomedical research." Nature Reviews.Immunology. 7(2): 118-130.

Shultz, L. D., F. Ishikawa and D. L. Greiner (2007). "Humanized mice in translational

biomedical research." Nat Rev Immunol 7(2): 118-130.

Shultz, L. D., B. L. Lyons, L. M. Burzenski, B. Gott, X. Chen, S. Chaleff, M. Kotb, S. D. Gillies,

M. King, J. Mangada, D. L. Greiner and R. Handgretinger (2005). "Human lymphoid and

myeloid cell development in NOD/LtSz-scid IL2R gamma null mice engrafted with

mobilized human hemopoietic stem cells." Journal of Immunology 174(10): 6477-6489.

Shultz, L. D., P. A. Schweitzer, S. W. Christianson, B. Gott, I. B. Schweitzer, B. Tennent, S.

McKenna, L. Mobraaten, T. V. Rajan, D. L. Greiner and et al. (1995). "Multiple defects

in innate and adaptive immunologic function in NOD/LtSz-scid mice." Journal of

Immunology 154(1): 180-191.

Silla, L. M., J. Chen, R. K. Zhong, T. L. Whiteside and E. D. Ball (1995). "Potentiation of lysis

of leukaemia cells by a bispecific antibody to CD33 and CD16 (Fc gamma RIII)

expressed by human natural killer (NK) cells." British Journal of Haematology 89(4):

712-718.

Singh, S. K., I. D. Clarke, T. Hide and P. B. Dirks (2004). "Cancer stem cells in nervous system

tumors." Oncogene 23(43): 7267-7273.

Singh, S. K., I. D. Clarke, M. Terasaki, V. E. Bonn, C. Hawkins, J. Squire and P. B. Dirks

(2003). "Identification of a cancer stem cell in human brain tumors." Cancer Research

63(18): 5821-5828.

Singh, S. K., C. Hawkins, I. D. Clarke, J. A. Squire, J. Bayani, T. Hide, R. M. Henkelman, M. D.

Cusimano and P. B. Dirks (2004). "Identification of human brain tumour initiating cells."

Nature 432(7015): 396-401.

Sivori, S., S. Parolini, E. Marcenaro, R. Castriconi, D. Pende, R. Millo and A. Moretta (2000).

"Involvement of natural cytotoxicity receptors in human natural killer cell-mediated lysis

of neuroblastoma and glioblastoma cell lines." Journal of Neuroimmunology 107(2): 220-

225.

Sivori, S., M. Vitale, L. Morelli, L. Sanseverino, R. Augugliaro, C. Bottino, L. Moretta and A.

Moretta (1997). "p46, a novel natural killer cell-specific surface molecule that mediates

cell activation." Journal of Experimental Medicine 186(7): 1129-1136.

Page 224: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

204

Skrtic, M., S. Sriskanthadevan, B. Jhas, M. Gebbia, X. Wang, Z. Wang, R. Hurren, Y. Jitkova,

M. Gronda, N. Maclean, C. K. Lai, Y. Eberhard, J. Bartoszko, P. Spagnuolo, A. C.

Rutledge, A. Datti, T. Ketela, J. Moffat, B. H. Robinson, J. H. Cameron, J. Wrana, C. J.

Eaves, M. D. Minden, J. C. Y. Wang, J. E. Dick, K. Humphries, C. Nislow, G. Giaever

and A. D. Schimmer (2011). "Inhibition of mitochondrial translation as a therapeutic

strategy for human acute myeloid leukemia." Cancer Cell 20(5): 674-688.

Smyth, M. J., E. Cretney, J. M. Kelly, J. A. Westwood, S. E. Street, H. Yagita, K. Takeda, S. L.

van Dommelen, M. A. Degli-Esposti and Y. Hayakawa (2005). "Activation of NK cell

cytotoxicity." Molecular Immunology 42(4): 501-510.

Smyth, M. J., E. Cretney, K. Takeda, R. H. Wiltrout, L. M. Sedger, N. Kayagaki, H. Yagita and

K. Okumura (2001). "Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)

contributes to interferon gamma-dependent natural killer cell protection from tumor

metastasis." Journal of Experimental Medicine 193(6): 661-670.

Smyth, M. J., J. Swann, E. Cretney, N. Zerafa, W. M. Yokoyama and Y. Hayakawa (2005).

"NKG2D function protects the host from tumor initiation." Journal of Experimental

Medicine 202(5): 583-588.

Stepp, S. E., R. Dufourcq-Lagelouse, F. Le Deist, S. Bhawan, S. Certain, P. A. Mathew, J. I.

Henter, M. Bennett, A. Fischer, G. de Saint Basile and V. Kumar (1999). "Perforin gene

defects in familial hemophagocytic lymphohistiocytosis." Science 286(5446): 1957-1959.

Stirewalt, D. L., C. L. Willman and J. P. Radich (2001). "Quantitative, real-time polymerase

chain reactions for FLT3 internal tandem duplications are highly sensitive and specific."

Leukemia Research 25(12): 1085-1088.

Strehl, S., M. Konig, G. Mann and O. A. Haas (2001). "Multiplex reverse transcriptase-

polymerase chain reaction screening in childhood acute myeloblastic leukemia." Blood

97(3): 805-808.

Suck, G. (2006). "Novel approaches using natural killer cells in cancer therapy." Seminars in

Cancer Biology 16(5): 412-418.

Suck, G., D. R. Branch, P. Aravena, M. Mathieson, S. Helke and A. Keating (2006).

"Constitutively polarized granules prime KHYG-1 NK cells." International Immunology

18(9): 1347-1354.

Suck, G., D. R. Branch and A. Keating (2006). "Irradiated KHYG-1 retains cytotoxicity:

potential for adoptive immunotherapy with a natural killer cell line." International Journal

of Radiation Biology 82(5): 355-361.

Suck, G., D. R. Branch and A. Keating (2006). "Irradiated KHYG-1 retains cytotoxicity:

potential for adoptive immunotherapy with a natural killer cell line." Int J Radiat Biol

82(5): 355-361.

Page 225: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

205

Suck, G., D. R. Branch, M. J. Smyth, R. G. Miller, J. Vergidis, S. Fahim and A. Keating (2005).

"KHYG-1, a model for the study of enhanced natural killer cell cytotoxicity."

Experimental Hematology 33(10): 1160-1171.

Sugamura, K., H. Asao, M. Kondo, N. Tanaka, N. Ishii, K. Ohbo, M. Nakamura and T.

Takeshita (1996). "The interleukin-2 receptor gamma chain: its role in the multiple

cytokine receptor complexes and T cell development in XSCID." Annual Review of

Immunology 14: 179-205.

Sutlu, T. and E. Alici (2009). "Natural killer cell-based immunotherapy in cancer: current

insights and future prospects." J Intern Med 266(2): 154-181.

Sutton, V. R., M. E. Wowk, M. Cancilla and J. A. Trapani (2003). "Caspase activation by

granzyme B is indirect, and caspase autoprocessing requires the release of proapoptotic

mitochondrial factors." Immunity 18(3): 319-329.

Swann, J. B. and M. J. Smyth (2007). "Immune surveillance of tumors." Journal of Clinical

Investigation 117(5): 1137-1146.

Swift, B. E., B. A. Williams, Y. Kosaka, X. H. Wang, J. A. Medin, S. Viswanathan, J. Martinez-

Lopez and A. Keating (2012). "Natural killer cell lines preferentially kill clonogenic

multiple myelomacells and decrease myeloma engraftment in a bioluminescentxenograft

mouse model." Haematologica.

Tahara-Hanaoka, S., K. Shibuya, Y. Onoda, H. Zhang, S. Yamazaki, A. Miyamoto, S. Honda, L.

L. Lanier and A. Shibuya (2004). "Functional characterization of DNAM-1 (CD226)

interaction with its ligands PVR (CD155) and nectin-2 (PRR-2/CD112)." International

Immunology 16(4): 533-538.

Takahashi, T., M. Tanaka, C. I. Brannan, N. A. Jenkins, N. G. Copeland, T. Suda and S. Nagata

(1994). "Generalized lymphoproliferative disease in mice, caused by a point mutation in

the Fas ligand." Cell 76(6): 969-976.

Takeda, K., Y. Hayakawa, M. J. Smyth, N. Kayagaki, N. Yamaguchi, S. Kakuta, Y. Iwakura, H.

Yagita and K. Okumura (2001). "Involvement of tumor necrosis factor-related apoptosis-

inducing ligand in surveillance of tumor metastasis by liver natural killer cells." Nature

Medicine 7(1): 94-100.

Tallerico, R., M. Todaro, S. Di Franco, C. Maccalli, C. Garofalo, R. Sottile, C. Palmieri, L.

Tirinato, P. N. Pangigadde, R. La Rocca, O. Mandelboim, G. Stassi, E. Di Fabrizio, G.

Parmiani, A. Moretta, F. Dieli, K. Karre and E. Carbone (2013). "Human NK cells

selective targeting of colon cancer-initiating cells: a role for natural cytotoxicity receptors

and MHC class I molecules." Journal of Immunology 190(5): 2381-2390.

Tam, Y. K., B. Miyagawa, V. C. Ho and H. G. Klingemann (1999). "Immunotherapy of

malignant melanoma in a SCID mouse model using the highly cytotoxic natural killer

cell line NK-92." Journal of hematotherapy 8(3): 281-290.

Page 226: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

206

Tangye, S. G., H. Cherwinski, L. L. Lanier and J. H. Phillips (2000). "2B4-mediated activation

of human natural killer cells." Molecular Immunology 37(9): 493-501.

Taussig, D. C., F. Miraki-Moud, F. Anjos-Afonso, D. J. Pearce, K. Allen, C. Ridler, D.

Lillington, H. Oakervee, J. Cavenagh, S. G. Agrawal, T. A. Lister, J. G. Gribben and D.

Bonnet (2008). "Anti-CD38 antibody-mediated clearance of human repopulating cells

masks the heterogeneity of leukemia-initiating cells." Blood 112(3): 568-575.

Tettamanti, S., V. Marin, I. Pizzitola, C. F. Magnani, G. M. Giordano Attianese, E. Cribioli, F.

Maltese, S. Galimberti, A. F. Lopez, A. Biondi, D. Bonnet and E. Biagi (2013).

"Targeting of acute myeloid leukaemia by cytokine-induced killer cells redirected with a

novel CD123-specific chimeric antigen receptor." British Journal of Haematology

161(3): 389-401.

Thacker, J. D. and D. E. Hogge (1994). "Cytokine-dependent engraftment of human myeloid

leukemic cell lines in immunosuppressed nude mice." Leukemia 8(5): 871-877.

Thomas, E. D., H. L. Lochte, Jr., W. C. Lu and J. W. Ferrebee (1957). "Intravenous infusion of

bone marrow in patients receiving radiation and chemotherapy." New England Journal of

Medicine 257(11): 491-496.

Till, J. and E. McCulloch (1961). "A direct measurement of the radiation sensitivity of normal

mouse bone marrow cells." Radiation Research 14: 213-222.

Till, J. E., E. A. McCulloch and L. Siminovitch (1964). "A STOCHASTIC MODEL OF STEM

CELL PROLIFERATION, BASED ON THE GROWTH OF SPLEEN COLONY-

FORMING CELLS." Proceedings of the National Academy of Sciences of the United

States of America 51: 29-36.

Toba, K., K. Kishi, T. Koike, E. F. Winton, H. Takahashi, K. Nagai, S. Maruyama, T. Furukawa,

S. Hashimoto, M. Masuko, Y. Uesugi, T. Kuroha, N. Tsukada and A. Shibata (1996).

"Profile of cell cycle in hematopoietic malignancy by DNA/RNA quantitation using

7AAD/PY." Experimental Hematology 24(8): 894-901.

Tonn, T., S. Becker, R. Esser, D. Schwabe and E. Seifried (2001). "Cellular immunotherapy of

malignancies using the clonal natural killer cell line NK-92." Journal of Hematotherapy

& Stem Cell Research 10(4): 535-544.

Tonn, T., D. Schwabe, H. G. Klingemann, S. Becker, R. Esser, U. Koehl, M. Suttorp, E. Seifried,

O. G. Ottmann and G. Bug (2013). "Treatment of patients with advanced cancer with the

natural killer cell line NK-92." Cytotherapy 15(12): 1563-1570.

Traggiai, E., L. Chicha, L. Mazzucchelli, L. Bronz, J. C. Piffaretti, A. Lanzavecchia and M. G.

Manz (2004). "Development of a human adaptive immune system in cord blood cell-

transplanted mice." Science 304(5667): 104-107.

Trenn, G., H. Takayama and M. V. Sitkovsky (1987). "Exocytosis of cytolytic granules may not

be required for target cell lysis by cytotoxic T-lymphocytes." Nature 330(6143): 72-74.

Page 227: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

207

Trinchieri, G. (1989). "Biology of natural killer cells." Advances in Immunology 47: 187-376.

Trinchieri, G. and N. Valiante (1993). "Receptors for the Fc fragment of IgG on natural killer

cells." Natural Immunity 12(4-5): 218-234.

Tsuchiyama, J., T. Yoshino, M. Mori, E. Kondoh, T. Oka, T. Akagi, A. Hiraki, H. Nakayama, A.

Shibuya, Y. Ma, T. Kawabata, S. Okada and M. Harada (1998). "Characterization of a

novel human natural killer-cell line (NK-YS) established from natural killer cell

lymphoma/leukemia associated with Epstein-Barr virus infection." Blood 92(4): 1374-

1383.

van den Broek, M. E., D. Kagi, F. Ossendorp, R. Toes, S. Vamvakas, W. K. Lutz, C. J. Melief,

R. M. Zinkernagel and H. Hengartner (1996). "Decreased tumor surveillance in perforin-

deficient mice." Journal of Experimental Medicine 184(5): 1781-1790.

van Rhenen, A., N. Feller, A. Kelder, A. H. Westra, E. Rombouts, S. Zweegman, M. A. van der

Pol, Q. Waisfisz, G. J. Ossenkoppele and G. J. Schuurhuis (2005). "High stem cell

frequency in acute myeloid leukemia at diagnosis predicts high minimal residual disease

and poor survival." Clin Cancer Res 11(18): 6520-6527.

van Rhenen, A., B. Moshaver, A. Kelder, N. Feller, A. W. Nieuwint, S. Zweegman, G. J.

Ossenkoppele and G. J. Schuurhuis (2007). "Aberrant marker expression patterns on the

CD34+CD38- stem cell compartment in acute myeloid leukemia allows to distinguish the

malignant from the normal stem cell compartment both at diagnosis and in remission."

Leukemia 21(8): 1700-1707.

van Rhenen, A., G. A. M. S. van Dongen, A. Kelder, E. J. Rombouts, N. Feller, B. Moshaver, M.

Stigter-van Walsum, S. Zweegman, G. J. Ossenkoppele and G. Jan Schuurhuis (2007).

"The novel AML stem cell associated antigen CLL-1 aids in discrimination between

normal and leukemic stem cells." Blood 110(7): 2659-2666.

Vardiman, J. W., J. Thiele, D. A. Arber, R. D. Brunning, M. J. Borowitz, A. Porwit, N. L. Harris,

M. M. Le Beau, E. Hellstrom-Lindberg, A. Tefferi and C. D. Bloomfield (2009). "The

2008 revision of the World Health Organization (WHO) classification of myeloid

neoplasms and acute leukemia: rationale and important changes." Blood 114(5): 937-951.

Veillette, A. (2006). "NK cell regulation by SLAM family receptors and SAP-related adapters."

Immunological Reviews 214: 22-34.

Velardi, A. (2008). "Role of KIRs and KIR ligands in hematopoietic transplantation." Curr Opin

Immunol 20(5): 581-587.

Virchow, R. (1856). "Leukamie, Gesammelte Abhandlungen zur wissenschaftlichen medicin."

Frankfurt: Meidinger Sohn & comp: 190–212.

Visonneau, S., A. Cesano, D. L. Porter, S. L. Luger, L. Schuchter, M. Kamoun, M. H. Torosian,

K. Duffy, C. Sickles, E. A. Stadtmauer and D. Santoli (2000). "Phase I trial of TALL-104

cells in patients with refractory metastatic breast cancer." Clinical Cancer Research 6(5):

1744-1754.

Page 228: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

208

Vitale, M., C. Bottino, S. Sivori, L. Sanseverino, R. Castriconi, E. Marcenaro, R. Augugliaro, L.

Moretta and A. Moretta (1998). "NKp44, a novel triggering surface molecule specifically

expressed by activated natural killer cells, is involved in non-major histocompatibility

complex-restricted tumor cell lysis." Journal of Experimental Medicine 187(12): 2065-

2072.

Vitale, M., M. Falco, R. Castriconi, S. Parolini, R. Zambello, G. Semenzato, R. Biassoni, C.

Bottino, L. Moretta and A. Moretta (2001). "Identification of NKp80, a novel triggering

molecule expressed by human NK cells." European Journal of Immunology 31(1): 233-

242.

Vivier, E., N. Rochet, J. P. Kochan, D. H. Presky, S. F. Schlossman and P. Anderson (1991).

"Structural similarity between Fc receptors and T cell receptors. Expression of the

gamma-subunit of Fc epsilon RI in human T cells, natural killer cells and thymocytes."

Journal of Immunology 147(12): 4263-4270.

Voskoglou-Nomikos, T., J. L. Pater and L. Seymour (2003). "Clinical predictive value of the in

vitro cell line, human xenograft, and mouse allograft preclinical cancer models." Clinical

Cancer Research 9(11): 4227-4239.

Walter, R. B., B. Gyurkocza, B. E. Storer, C. D. Godwin, J. M. Pagel, S. A. Buckley, M. L.

Sorror, B. L. Wood, R. Storb, F. R. Appelbaum and B. M. Sandmaier (2014).

"Comparison of minimal residual disease as outcome predictor for AML patients in first

complete remission undergoing myeloablative or nonmyeloablative allogeneic

hematopoietic cell transplantation." Leukemia.

Wang, C., J. E. Curtis, M. D. Minden and E. A. McCulloch (1989). "Expression of a retinoic

acid receptor gene in myeloid leukemia cells." Leukemia 3(4): 264-269.

Wang, C., P. Koistinen, G. S. Yang, D. E. Williams, S. D. Lyman, M. D. Minden and E. A.

McCulloch (1991). "Mast cell growth factor, a ligand for the receptor encoded by c-kit,

affects the growth in culture of the blast cells of acute myeloblastic leukemia." Leukemia

5(6): 493-499.

Watanabe-Fukunaga, R., C. I. Brannan, N. G. Copeland, N. A. Jenkins and S. Nagata (1992).

"Lymphoproliferation disorder in mice explained by defects in Fas antigen that mediates

apoptosis." Nature 356(6367): 314-317.

Williams, B. A., B. E. Swift, R. Cheng and A. Keating (2013). NK-92 cytotoxicity against

cancer stem cells in hematologic malignancies. Stem Cells and Cancer Stem Cells:

Therapeutic Applications in Disease and Injury. M. A. Hayat, Springer. 9 (24): 249-257.

Williams, B. A., X. H. Wang and A. Keating (2010). "Clonogenic assays measure leukemia stem

cell killing not detectable by chromium release and flow cytometric cytotoxicity assays."

Cytotherapy 12(7): 951-960.

Williams, S. A., W. C. Anderson, M. T. Santaguida and S. J. Dylla (2013). "Patient-derived

xenografts, the cancer stem cell paradigm, and cancer pathobiology in the 21st century."

Laboratory Investigation 93(9): 970-982.

Page 229: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

209

Witte, K. E., J. Ahlers, I. Schafer, M. Andre, G. Kerst, H. G. Scheel-Walter, C. P. Schwarze, M.

Pfeiffer, P. Lang, R. Handgretinger and M. Ebinger (2011). "High proportion of leukemic

stem cells at diagnosis is correlated with unfavorable prognosis in childhood acute

myeloid leukemia." Pediatr Hematol Oncol 28(2): 91-99.

Wooden, S. L., S. R. Kalb, R. J. Cotter and M. J. Soloski (2005). "Cutting edge: HLA-E binds a

peptide derived from the ATP-binding cassette transporter multidrug resistance-

associated protein 7 and inhibits NK cell-mediated lysis." Journal of Immunology 175(3):

1383-1387.

Woods, W. G. (2001). "A comparison of allogeneic bone marrow transplantation, autologous

bone marrow transplantation, and aggressive chemotherapy in children with acute

myeloid leukemia in remission: a report from the Children's Cancer Group." Blood 97(1):

56-62.

Wu, J., J. C. Edberg, P. B. Redecha, V. Bansal, P. M. Guyre, K. Coleman, J. E. Salmon and R. P.

Kimberly (1997). "A novel polymorphism of FcgammaRIIIa (CD16) alters receptor

function and predisposes to autoimmune disease." Journal of Clinical Investigation

100(5): 1059-1070.

Wu, Z., B. Markovic, C. N. Chesterman and B. H. Chong (1996). "Characterization of IgG Fc

receptors on CD34 antigen-expressing cell lines (KG-1 and KG-1a)." Immunology and

Cell Biology 74(1): 57-64.

Wulf, G. G., R. Y. Wang, I. Kuehnle, D. Weidner, F. Marini, M. K. Brenner, M. Andreeff and

M. A. Goodell (2001). "A leukemic stem cell with intrinsic drug efflux capacity in acute

myeloid leukemia." Blood 98(4): 1166-1173.

Yagita, M., C. L. Huang, H. Umehara, Y. Matsuo, R. Tabata, M. Miyake, Y. Konaka and K.

Takatsuki (2000). "A novel natural killer cell line (KHYG-1) from a patient with

aggressive natural killer cell leukemia carrying a p53 point mutation." Leukemia 14(5):

922-930.

Yamada, M., M. Mori and T. Sugimura (1983). "Myeloperoxidases of human myeloid leukemia

cells HL-60 grown in culture and in nude mice." J Biochem 93(6): 1661-1668.

Yan, Y., P. Steinherz, H. G. Klingemann, D. Dennig, B. H. Childs, J. McGuirk and R. J. O'Reilly

(1998). "Antileukemia activity of a natural killer cell line against human leukemias."

Clinical Cancer Research 4(11): 2859-2868.

Yang, W., J. H. Williams, P. F. Hogan, S. S. Bruinooge, G. I. Rodriguez, M. P. Kosty, D. F.

Bajorin, A. Hanley, A. Muchow, N. McMillan and M. Goldstein (2014). "Projected

supply of and demand for oncologists and radiation oncologists through 2025: an aging,

better-insured population will result in shortage." J Oncol Pract 10(1): 39-45.

Yodoi, J., K. Teshigawara, T. Nikaido, K. Fukui, T. Noma, T. Honjo, M. Takigawa, M. Sasaki,

N. Minato, M. Tsudo and et al. (1985). "TCGF (IL 2)-receptor inducing factor(s). I.

Regulation of IL 2 receptor on a natural killer-like cell line (YT cells)." Journal of

Immunology 134(3): 1623-1630.

Page 230: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

210

Yu, M. C., L. L. Su, L. Zou, Y. Liu, N. Wu, L. Kong, Z. H. Zhuang, L. Sun, H. P. Liu, J. H. Hu,

D. Li, J. L. Strominger, J. W. Zang, G. Pei and B. X. Ge (2008). "An essential function

for beta-arrestin 2 in the inhibitory signaling of natural killer cells." Nat Immunol 9(8):

898-907.

Page 231: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

211

Appendices

Page 232: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

212

Appendix I: NK-92 HTS flow cytometry percent positivity and MFI of antigens

# Antigen % + MFI Antigen name(s)

1 BLTR-1 1.83 133 Leukotriene B4 receptor

2 B2m 99.8 13283 Beta-2 microglobulin

3 CA9 5.39 135 Carbonic anhydrase IX

4 CDH3 3.06 133 Cadherin 3

5 CDH6 5.42 207 Cadherin 6

6 CDH11 1.52 561 Cadherin 11

7 CDw93 4.31 506 C1q-binding protein

8 CDw198 2.02 129 CCR8

9 CDw199 0.0702 188 CCR9

10 CDw210 8.48 172 IL-10 receptor

11 CDw218a 11.9 147 IL-18 receptor

12 CDw329 1.03 524 SIGLEC-9

13 CD1a 0.266 120 T6

14 CD1b 1.19 534 T6

15 CD1c 0.964 123 T6

16 CD1d 0.282 128 CD1d

17 CD2 100 1521 T11, LFA-2, SRBC-R

18 CD3 0.842 133 T3

19 CD3e 0.0639 182 T3

20 CD4 0.345 119 T4

21 CD5 0.324 111 T1, Tp67

22 CD6 56.5 187 T12

23 CD7 99.9 2512 LEU-9

24 CD8 0.198 130 T8, Leu-2, CD8alpha

25 CD8b 1.56 135 CD8beta

26 CD9 0.268 129 p24, MRP-1

27 CD10 0.406 118 CALLA, NEP, gp100

28 CD11a 100 10258 LFA-1, integrin alpha L

29 CD11b 5.67 136 Mac-1, integrin alphaM

30 CD11c 18.2 153 p150, CR4, integrin alphaX

31 CD13 4.09 137 Aminopeptidase N, APN

32 CD14 4.36 134 LPS-R

33 CD15 0.212 128 Lewis-x, Lex

34 CD16 1.78 127 Fcgamma RIIIA

35 CD16b 0.355 122 FcgammaRIIIB

Page 233: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

213

36 CD17 3.06 537 Lactosylceramide

37 CD18 100 642 Integrin beta2

38 CD19 0.251 130 B4

39 CD20 0.28 113 B1, Bp35

40 CD21 0.525 133 C3DR, CR2, EBV-R

41 CD22 0.566 131 BL-CAM, Siglec-2

42 CD23 0.345 121 FcepsilonRII

43 CD24 0.617 125 BA-1

44 CD25 8.52 141 IL-2Ralpha, Tac, p55

45 CD26 100 4223 DPP IV

46 CD27 0.401 127 TNFRSF7, T14

47 CD28 41.3 201 Tp44, T44

48 CD29 100 1034 Integrin beta1

49 CD30 88.4 398 Ki-1

50 CD31 0.175 122 PECAM-1

51 CD32 0.759 128 IgSF

52 CD33 1.46 133 p67, Siglec-3

53 CD34 12.1 138 HPCA1

54 CD35 0.0932 139 CR1

55 CD36 5.14 203 GPIV

56 CD37 1.33 533 N/A

57 CD38 99.8 890 T10

58 CD39 1.94 225 ENTPD1

59 CD40 0.522 166 TNFRSF5

60 CD41a 0.225 127 gpIIb

61 CD41b 1.28 536 HPA-3

62 CD42a 0.248 129 GPIX

63 CD42b 0.445 127 GPIba

64 CD43 100 773 Leukosialin, sialophorin

65 CD44 99 1510 Hyalunorate receptor

66 CD45 99.9 5891 LCA

67 CD45RA 89.8 353 LCA

68 CD45RB 99.6 487 LCA, T200, B220

69 CD45RO 100 2149 LCA, UCHL-1

70 CD46 57.8 548 Membrane cofactor protein

71 CD47 98.8 900 IAP

72 CD48 99.6 1195 Blast-1

73 CD49a 97.2 363 VLA-1

74 CD49b 0.614 127 VLA-2

75 CD49c 0.882 126 VLA-3

76 CD49d 100 3227 VLA-4

77 CD49e 2.36 141 VLA-5

Page 234: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

214

78 CD49f 100 863 VLA-6

79 CD50 1.11 543 ICAM-3

80 CD51/CD61 2.2 1436 Vitronectin receptor

81 CD52 7.53 137 CAMPATH-1 antigen

82 CD53 92.7 579 MRC OX44

83 CD54 100 4549 ICAM-1

84 CD55 4.2 135 DAF

85 CD56 100 2032 NCAM

86 CD57 1.01 551 HNK-1, Leu-7

87 CD58 99.9 1359 LFA-3

88 CD59 13.7 144 Protectin, MAC- inhibitor

89 CD60b 78.1 2451 9-O-sialyl GD3

90 CD61 6.85 136 GPIIIa

91 CD62E 1.27 130 E-selectin, ELAM-1

92 CD62L 3.29 140 L-selectin, LECAM-1

93 CD62P 3.35 139 P-selectin, PADGEM

94 CD63 99.9 1120 LIMP, LAMP-3

95 CD64 0.827 128 FcgammaRI

96 CD65 1.24 548 VIM2

97 CD65s 1.34 546 VIM2

98 CD66 99.9 998 BGP-1, NCA-160

99 CD66b 1.11 572 CD67, CGM6

100 CD66c 9.48 141 NCA

101 CD66d 98.7 427 CGM1; CEACAM-3

102 CD66e 72.6 249 CEACAM-5

103 CD69 0.946 132 AIM

104 CD70 0.467 118 Ki-24

105 CD71 99.6 1372 T9

106 CD72 1.39 552 Lyb-2

107 CD73 1.48 128 L-VAP-2

108 CD74 1.5 543 Invariant chain

109 CD75 1.36 518 LN-1

110 CD77 1.86 530 Gb3, Pk blood group

111 CD79a 4.46 133 Iga

112 CD79b 2.45 140 Igb

113 CD80 16.9 172 B7, B7-1, BB1

114 CD81 100 2072 TAPA-1

115 CD82 42 193 R2

116 CD83 0.881 130 HB15

117 CD84 2.58 136 SLAMF5

118 CD85a 1.94 127 LILRB3

119 CD85d 12.2 141 LIRB2, ILT-4, LIR-2

Page 235: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

215

120 CD85g 27.5 156 ILT-7

121 CD85h 4.29 133 LILRA-2, ILT-1, LIR-7

122 CD85j 95.7 370 LIRRB-1, ILT-2

123 CD86 100 1359 B70, B7-2

124 CD87 0.334 124 UPA-R

125 CD88 1.12 135 C5aR

126 CD89 1.37 129 FcalphaR

127 CD90 28.3 190 Thy-1

128 CD91 4.86 135 TGFBR5

129 CD92 22.9 151 CTL1B

130 CD94 100 1876 NKG2C, KP43

131 CD95 99 410 Apo-1, Fas

132 CD96 99.5 650 TACTILE

133 CD97 65.9 172 AURA51

134 CD98 100 4383 4F2

135 CD99 35.1 150 MIC2, E2

136 CD100 89.4 865 SEMA4D

137 CD101 0.478 129 V7, p126

138 CD102 29 161 ICAM-2

139 CD103 0.21 109 HML-1, alpha6, integrin alphaE

140 CD104 0.462 131 Beta4 integrin

141 CD105 2.94 135 Endoglin

142 CD106 0.608 129 VCAM-1

143 CD107a 34 149 LAMP-1

144 CD107b 1.98 575 LAMP-2

145 CD108 95.4 468 SEMA7A

146 CD109 0.812 132 7D1, 8A3

147 CD110 46.1 199 MPL, TPO-R

148 CD111 37.6 168 PRR1, Nectin-1

149 CD112 21.8 178 PRR2, Nectin-2

150 CD114 6.19 216 G-CSFR

151 CD115 1.61 141 M-CSFR, c-fms

152 CD116 18.8 217 GM-CSFR alpha

153 CD117 8.48 250 c-kit, SCFR

154 CD118 2.55 131 LIFR, gp190

155 CD119 42.3 185 IFNgammaR

156 CD120a 1.99 131 TNFR-I

157 CD120b 98.7 773 TNFR-II

158 CD121b 20 147 IL-1R, type II

159 CD122 96 328 IL-2Rbeta

160 CD123 10.5 145 IL-3Ralpha

161 CD124 1.72 129 IL-4Ralpha

Page 236: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

216

162 CD125 1.8 141 IL-5Ralpha

163 CD126 1.6 131 IL-6Ralpha

164 CD127 27.9 1015 IL-7Ralpha

165 CD129 0.33 112 IL-9R

166 CD130 6.51 133 IL-6Rbeta, gp130

167 CD131 0.821 132 IL-3Rbeta

168 CD132 85.7 280 Common gamma

169 CD133 0.189 184 AC133, prominin-like 1

170 CD134 3.56 134 OX-40

171 CD135 1.2 130 Flt3/Flk2

172 CD136 0.377 124 MSP-R, RON

173 CD137 0.471 119 4-1BB, TNRFSF9

174 CD137L 29.5 159 4-1BB L

175 CD138 0.434 131 Syndecan-1

176 CD140a 0.22 113 PDGFRalpha

177 CD140b 82.1 215 PDGFRbeta

178 CD141 0.265 119 Thrombomodulin

179 CD142 0.852 129 Tissue Factor

180 CD143 2.18 249 ACE

181 CD144 0.306 127 VE-Cadherin, Cadherin-5

182 CD146 0.307 126 MUC18, S-endo

183 CD147 100 3806 Neurothelin, basoglin

184 CD148 99.9 630 HPTP-eta

185 CD150 0.992 130 SLAM

186 CD151 92 381 PETA-3

187 CD152 17.3 141 CTLA-4

188 CD153 4.88 158 CD30L

189 CD154 0.428 125 CD40L, gp39, TRAP

190 CD155 33.9 160 PVR

191 CD156b 1.48 131 ADAM8

192 CD157 0.365 130 BST-1

193 CD158a 0.243 112 KIR2DL1, p58.1

194 CD158b 0.263 130 KIR2DL2, p58.2

195 CD158b2 0.395 130 KIR2DL3

196 CD158d 37.3 156 KIR2DL4

197 CD158e2 0.225 131 KIR3DL1

198 CD158f 6.73 138 KIR2DL5

199 CD158i 6.01 135 KIR2DS4

200 CD159a 1.82 131 NKG2A

201 CD159c 0.971 128 NKG2C

202 CD160 73.2 427 BY55

203 CD161 8.76 144 NKR-P1

Page 237: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

217

204 CD162 99.9 2457 PSGL-1

205 CD163 0.453 106 Ber-MAC3, M130

206 CD164 4.53 138 MGC-24

207 CD165 0.203 128 AD2, gp37

208 CD166 13.6 142 ALCAM

209 CD167 12.1 138 DDR1

210 CD169 1.74 131 Sialoadhesin, Siglec-1

211 CD170 2.18 127 Siglec-5, CD33-like2

212 CD171 0.905 129 L1

213 CD172a 2.55 134 SIRPgamma

214 CD172b 0.208 124 SIRPbeta, SIRB1

215 CD172g 84.6 259 SIRPgamma, SIRPB2

216 CD175s 87.5 807 Sialyl-Tn

217 CD177 1.08 544 NB1

218 CD178 30.4 152 FasL, CD95L

219 CD179a 25 146 VpreB

220 CD180 1.18 131 RP-105

221 CD181 11.5 146 CXCR1, IL-8RA

222 CD182 5.38 32217 CXCR2, IL-8RB

223 CD183 36.5 156 CXCR3

224 CD184 12.4 144 CXCR4, fusin

225 CD185 2.32 473 CXCR5, BLR1

226 CD186 0.97 130 CXCR6, BONZO

227 CD191 6.22 142 CCR1, MIP-1alphaR, RANTES-R

228 CD192 0.789 192 CCR2, MCP-1-R

229 CD193 30.4 164 CCR3, CKR3

230 CD194 5.34 209 CCR4

231 CD195 0.969 133 CCR5

232 CD196 20.1 159 CCR6, LARC receptor, DRY6

233 CD197 0.658 128 CCR7

234 CD200 0.68 127 OX-2

235 CD201 0.341 129 EPC-R

236 CD202b 17.3 148 Tie2, Tek

237 CD203c 9.65 141 NPP3 / PDNP3, ENpp1, PD-1b

238 CD204 6.17 311 Macrophage scavenger-R

239 CD205 93.2 238 DEC-205

240 CD206 0.3 108 Macrophage mannose-R

241 CD207 58.9 301 Langerin

242 CD208 1.02 128 DC-LAMP, LAMP-3

243 CD209 0.0858 137 DC-SIGN

244 CD212 24 146 IL-12-R beta1

245 CD213a2 0.189 110 IL-13-R alpha2

Page 238: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

218

246 CD215 6.78 141 IL-15R alpha

247 CD217 16.9 208 IL-17-R

248 CD218b 3.31 140 IL-18Rbeta, IL18RAP

249 CD220 11.9 215 Insulin-R

250 CD221 0.544 128 IGF-1 R

251 CD222 2.99 525 IGF-II R

252 CD223 10.4 140 Lag3

253 CD226 75.1 207 DNAM-1, PTA-1, TLiSA1

254 CD227 2.08 542 MUC1, EMA

255 CD229 4.48 139 Ly-9

256 CD230 98.3 517 PRNP

257 CD231 30.8 161 TALLA-1, A15

258 CD234 6.3 144 Duffy, DARC

259 CD235a 24.7 154 Glycophorin A

260 CD243 (BC) 24.8 147 MDR-1, p170, P-gp

261 CD243 (BD) 0.313 136 MDR-1, p170, P-gp

262 CD244 6.79 134 2B4

263 CD245 99.1 756 p220/240

264 CD249 40.6 156 Aminopeptidase A

265 CD252 15.8 1046 OX-40Ligand, gp34

266 CD253 36.3 182 TRAIL, TNFSF10

267 CD254 2.59 130 TRANCE, RANKL, OPGL

268 CD255 8.64 139 TWEAK

269 CD256 25.3 187 APRIL, TALL-2

270 CD257 49.5 198 BLyS, BAFF, TALL-1

271 CD258 4.51 137 LIGHT, HVEM-L

272 CD261 7.58 137 TRAIL-R1, DR4

273 CD262 99.3 386 TRAIL-R2, DR5

274 CD263 0.79 127 TRAIL-R3, DcR1, LIT

275 CD264 1.84 130 TNFRSF10D, TRAILR4

276 CD267 70.6 323 TACI, TNFR SF13B

277 CD268 26.2 362 BAFFR, TR13C

278 CD269 1.55 136 BCMA, TNFRSF13B

279 CD270 30.4 152 TNFSF14

280 CD271 0.746 133 NGFR, p75 (NTR)

281 CD272 4 7770 BTLA

282 CD273 0.301 113 B7DC, PD-L2, PDCD1L2

283 CD274 0.988 131 B7-H1, PD-L1

284 CD275 10.4 140 B7-H2, ICOSL, B7-RP1

285 CD276 22 149 B7-H3

286 CD277 0.832 129 BT3.1, butyrophilin SF3 A1

287 CD278 1.41 127 ICOS, AILIM

Page 239: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

219

288 CD279 2.25 133 PD1, SLEB2

289 CD281 37 166 TLR1

290 CD282 1.04 557 TLR2

291 CD283 37.9 170 TLR3

292 CD284 3.91 135 TLR4

293 CD286 32.7 164 TLR6

294 CD288 34.4 166 TLR8

295 CD289 24.6 151 TLR9

296 CD290 49.5 187 TLR-10

297 CD292 1.56 562 BMPR1A, ALK3

298 CD294 10.3 207 CRTH2. GPR44

299 CD295 21.7 147 LeptinR, LEPR

300 CD298 100 1588 Na/K ATPase beta3 subunit

301 CD299 28.4 164 DC-SIGN-related, LSIGN, DC-SIGN2

302 CD300a 25.3 174 CMRF35H, IRC1, IRp60

303 CD300c 42.9 174 CMRF35A, LIR

304 CD300e 33.3 160 CMRF35L

305 CD301 0.186 141 MGL, HML

306 CD303 51.6 184 BDCA2, HECL

307 CD304 8.14 137 BDCA4, neuropilin 1

308 CD305 32 177 LAIR1

309 CD307 7.07 141 IRTA2

310 CD309 34.2 167 VEGFR2, KDR

311 CD312 30.7 181 EMR2

312 CD314 78.9 298 NKG2D, KLR

313 CD317 99.9 2764 BST2, HM1.24

314 CD318 1.86 131 CDCP1, SIMA135

315 CD319 92.8 316 CRACC, SLAMF7

316 CD321 94.1 346 JAM1, F11 receptor

317 CD322 2.14 557 JAM2, VE-JAM

318 CD324 7.62 140 E-Cadherin, Uvomorulin

319 CD325 2.22 130 N-Cadherin, NCAD

320 CD326 0.25 129 Ep-CAM, Ly74

321 CD328 3.46 133 SIGLEC7, AIRM-1

322 CD332 13 204 FGFR2, BEK, KGFR

323 CD333 2.18 138 FGFR3, ACH, CEK2

324 CD334 0.65 123 FGFR4, JTK2, TKF

325 CD335 36.9 155 NKp46, Ly-94 homolog

326 CD336 4.55 145 NKp44, Ly-95 homolog

327 CD337 41.6 556 NKp30, Ly117

328 CD338 25.5 156 ABCG2, BCRP, Bcrp1, MXR

Page 240: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

220

329 CD339 1.61 518 Jagged-1, JAG1, JAGL1, hJ1

330 CD340 0.366 192 erbB2, HER-2, EGFR-2

331 CD344 51.8 199 EVR1

332 CD351 73.2 493 FCAMR

333 CD352 100 1094 SLAMF6, NTB-A

334 CD354 19.5 150 TREM-1

335 CD355 10.1 139 CRTAM

336 CD357 16.1 219 TNFSF18, GITR

337 CD358 11.9 145 TNFSF21, DR6

338 CD360 (BD) 21.5 152 IL-21RA

339 CD360 (BL) 18.9 147 IL-21RA

340 CD362 1.15 233 SDC2, HSPG-1

341 CD363 6.95 139 S1PR1

342 CLA 22.6 502 CLA

343 CLIP 1.33 534 CLIP

344 DCIR 2.02 192 DCIR

345 EGF-R 0.533 123 EGF-R

346 FMC7 1.16 552 FMC7

347 HLA-ABC 100 4234 HLA-ABC

348 HLA-A2 0.947 129 HLA-A2

349 HLA-DM 0.337 124 HLA-DM

350 HLA-DR 0.304 128 HLA-DR

351 HPC 3.54 132 HPC

352 ITGB7 99.9 1153 ITGB7

353 LTBR 20.3 168 LTBR, TNFRSF3

354 Lgr-5 1.54 133 Lgr-5

355 MIC A/B 0.506 143 MIC A/B

356 Notch1 81.5 334 Notch1

357 Notch2 91.3 489 Notch2

358 Notch3 15.1 215 Notch3

359 Notch4 1.09 129 Notch4

360 PAC-1 1.12 549 PAC-1

361 Podoplanin 6.11 136 PDPN

362 SSEA-3 1.01 133 SSEA-3

363 SSEA-4 0.232 110 SSEA-4

364 Stro-1 34 257 Stro-1

365 TCR alpha beta 0.279 127 TCR alpha beta

366 TCR gamma delta 18.1 158 TCR gamma delta

367 TPBG 0.378 214 TPBG

368 VB8 TCR 3.93 319 VB8 TCR

369 VD2 TCR 14.4 163 VD2 TCR

370 fMLP-R 10.8 142 fMLP-R

Page 241: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

221

Appendix II: KHYG-1 HTS flow cytometry percent positivity and MFI of antigens

# Antigen % + MFI Antigen name(s)

1 BLTR-1 39.8 320 Leukotriene B4 receptor

2 B2m 100 14037 Beta-2 microglobulin

3 CA9 28.3 355 Carbonic anhydrase IX

4 CDH3 6.38 261 Cadherin 3

5 CDH6 0.0527 252 Cadherin 6

6 CDH11 0.1 367 Cadherin 11

7 CDw93 55.6 425 C1q-binding protein

8 CDw198 62.4 453 CCR8

9 CDw199 0.508 394 CCR9

10 CDw210 34 322 IL-10 receptor

11 CDw218a 94.4 468 IL-18 receptor

12 CDw329 0.445 374 SIGLEC-9

13 CD1a 0.0827 263 T6

14 CD1b 1.41 371 T6

15 CD1c 0.959 243 T6

16 CD1d 0.29 233 CD1d

17 CD2 100 6723 T11, LFA-2, SRBC-R

18 CD3 0.265 289 T3

19 CD3e 0.033 452 T3

20 CD4 0.224 237 T4

21 CD5 0.126 239 T1, Tp67

22 CD6 26.2 469 T12

23 CD7 100 8271 LEU-9

24 CD8 99.9 3641 T8, Leu-2, CD8alpha

25 CD8b 14.4 278 CD8beta

26 CD9 6.38 280 p24, MRP-1

27 CD10 11.8 274 CALLA, NEP, gp100

28 CD11a 100 52431 LFA-1, integrin alpha L

29 CD11b 52.7 346 Mac-1, integrin alphaM

30 CD11c 100 2357 p150, CR4, integrin alphaX

31 CD13 38.7 312 Aminopeptidase N, APN

32 CD14 67.7 353 LPS-R

33 CD15 0.0246 303 Lewis-x, Lex

34 CD16 16.5 279 Fcgamma RIIIA

35 CD16b 0.23 274 FcgammaRIIIB

Page 242: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

222

36 CD17 12 380 Lactosylceramide

37 CD18 100 2422 Integrin beta2

38 CD19 0.0926 223 B4

39 CD20 0.851 250 B1, Bp35

40 CD21 16.6 274 C3DR, CR2, EBV-R

41 CD22 1.27 251 BL-CAM, Siglec-2

42 CD23 0.183 258 FcepsilonRII

43 CD24 7.28 274 BA-1

44 CD25 35.4 307 IL-2Ralpha, Tac, p55

45 CD26 100 1974 DPP IV

46 CD27 1.65 257 TNFRSF7, T14

47 CD28 84.7 2340 Tp44, T44

48 CD29 99.9 1903 Integrin beta1

49 CD30 86.2 551 Ki-1

50 CD31 0.0854 253 PECAM-1

51 CD32 24.8 289 IgSF

52 CD33 61.2 759 p67, Siglec-3

53 CD34 0.0692 241 HPCA1

54 CD35 0.0919 228 CR1

55 CD36 0.268 235 GPIV

56 CD37 0.22 345 N/A

57 CD38 22.8 375 T10

58 CD39 0.0554 281 ENTPD1

59 CD40 0.0725 208 TNFRSF5

60 CD41a 0.012 352 gpIIb

61 CD41b 0.23 350 HPA-3

62 CD42a 0.524 258 GPIX

63 CD42b 1.21 257 GPIba

64 CD43 100 3931 Leukosialin, sialophorin

65 CD44 100 6773 Hyalunorate receptor

66 CD45 100 2087 LCA

67 CD45RA 13.6 290 LCA

68 CD45RB 99.9 5508 LCA, T200, B220

69 CD45RO 99.9 917 LCA, UCHL-1

70 CD46 99.9 979 Membrane cofactor protein

71 CD47 99.7 779 IAP

72 CD48 100 4416 Blast-1

73 CD49a 97.5 1757 VLA-1

74 CD49b 65.9 406 VLA-2

75 CD49c 67.4 417 VLA-3

76 CD49d 100 6316 VLA-4

77 CD49e 1.88 240 VLA-5

Page 243: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

223

78 CD49f 97.8 1488 VLA-6

79 CD50 100 2690 ICAM-3

80 CD51/CD61 2.15 244 Vitronectin receptor

81 CD52 99.8 966 CAMPATH-1 antigen

82 CD53 97.3 707 MRC OX44

83 CD54 100 10759 ICAM-1

84 CD55 100 6623 DAF

85 CD56 88.7 7373 NCAM

86 CD57 0.128 390 HNK-1, Leu-7

87 CD58 100 2710 LFA-3

88 CD59 85.2 393 Protectin, MAC- inhibitor

89 CD60b 17.2 386 9-O-sialyl GD3

90 CD61 1.23 243 GPIIIa

91 CD62E 72.7 515 E-selectin, ELAM-1

92 CD62L 3.53 284 L-selectin, LECAM-1

93 CD62P 8.12 274 P-selectin, PADGEM

94 CD63 98.7 849 LIMP, LAMP-3

95 CD64 0.583 243 FcgammaRI

96 CD65 1.41 396 VIM2

97 CD65s 8.88 378 VIM2

98 CD66 80.7 1314 BGP-1, NCA-160

99 CD66b 0.128 366 CD67, CGM6

100 CD66c 49.3 337 NCA

101 CD66d 61.8 384 CGM1; CEACAM-3

102 CD66e 91 839 CEACAM-5

103 CD68 6.46 259

104 CD69 93.7 498 AIM

105 CD70 100 1241 Ki-24

106 CD71 99.5 1757 T9

107 CD72 0.278 356 Lyb-2

108 CD73 1.89 258 L-VAP-2

109 CD74 10.1 434 Invariant chain

110 CD75 0.183 324 LN-1

111 CD77 11.6 375 Gb3, Pk blood group

112 CD79a 0.254 224 Iga

113 CD79b 59.6 402 Igb

114 CD80 31.2 321 B7, B7-1, BB1

115 CD81 100 2518 TAPA-1

116 CD82 99.8 3500 R2

117 CD83 50.9 324 HB15

118 CD84 29.2 317 SLAMF5

119 CD85a 0.894 259 LILRB3

Page 244: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

224

120 CD85d 38.3 323 LIRB2, ILT-4, LIR-2

121 CD85g 26.1 295 ILT-7

122 CD85h 14.6 279 LILRA-2, ILT-1, LIR-7

123 CD85j 81.8 519 LIRRB-1, ILT-2

124 CD86 56.4 345 B70, B7-2

125 CD87 1.23 251 UPA-R

126 CD88 2.79 256 C5aR

127 CD89 11.1 271 FcalphaR

128 CD90 71.3 740 Thy-1

129 CD91 3.25 257 TGFBR5

130 CD92 41.4 321 CTL1B

131 CD94 98.8 1212 NKG2C, KP43

132 CD95 98 541 Apo-1, Fas

133 CD96 98.6 1069 TACTILE

134 CD97 94.8 533 AURA51

135 CD98 100 8916 4F2

136 CD99 23.3 279 MIC2, E2

137 CD100 99.8 1017 SEMA4D

138 CD101 3.27 272 V7, p126

139 CD102 100 2213 ICAM-2

140 CD103 0.0205 279 HML-1, alpha6, integrin alphaE

141 CD104 9.83 274 Beta4 integrin

142 CD105 22.1 296 Endoglin

143 CD106 16.3 272 VCAM-1

144 CD107a 19 299 LAMP-1

145 CD107b 2.09 375 LAMP-2

146 CD108 65.1 376 SEMA7A

147 CD109 0.276 271 7D1, 8A3

148 CD110 73.1 589 MPL, TPO-R

149 CD111 11.6 281 PRR1, Nectin-1

150 CD112 37.1 325 PRR2, Nectin-2

151 CD114 71.5 723 G-CSFR

152 CD115 9.08 272 M-CSFR, c-fms

153 CD116 74.1 1049 GM-CSFR alpha

154 CD117 8.04 25938 c-kit, SCFR

155 CD118 36.9 327 LIFR, gp190

156 CD119 13.5 71350 IFNgammaR

157 CD120a 19.2 279 TNFR-I

158 CD120b 99.4 2052 TNFR-II

159 CD121b 39.2 329 IL-1R, type II

160 CD122 95.8 794 IL-2Rbeta

161 CD123 58.5 430 IL-3Ralpha

Page 245: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

225

162 CD124 12.8 282 IL-4Ralpha

163 CD125 25.4 311 IL-5Ralpha

164 CD126 34 319 IL-6Ralpha

165 CD127 53.6 543 IL-7Ralpha

166 CD129 0.0833 217 IL-9R

167 CD130 6.12 263 IL-6Rbeta, gp130

168 CD131 1.68 268 IL-3Rbeta

169 CD132 92.6 1049 Common gamma

170 CD133 0.0177 326 AC133, prominin-like 1

171 CD134 45.5 340 OX-40

172 CD135 24.7 302 Flt3/Flk2

173 CD136 20 296 MSP-R, RON

174 CD137 13.5 285 4-1BB, TNRFSF9

175 CD137L 76.6 596 4-1BB L

176 CD138 0.192 284 Syndecan-1

177 CD140a 4.29 263 PDGFRalpha

178 CD140b 14.4 280 PDGFRbeta

179 CD141 1.97 245 Thrombomodulin

180 CD142 0.68 270 Tissue Factor

181 CD143 15.8 289 ACE

182 CD144 0.558 277 VE-Cadherin, Cadherin-5

183 CD146 0.0478 305 MUC18, S-endo

184 CD147 100 9563 Neurothelin, basoglin

185 CD148 99.8 1719 HPTP-eta

186 CD150 0.635 260 SLAM

187 CD151 100 6606 PETA-3

188 CD152 50.9 364 CTLA-4

189 CD153 29.3 305 CD30L

190 CD154 4.4 276 CD40L, gp39, TRAP

191 CD155 83.9 873 PVR

192 CD156b 56.5 346 ADAM8

193 CD157 0.232 231 BST-1

194 CD158a 0.175 280 KIR2DL1, p58.1

195 CD158b 0.193 285 KIR2DL2, p58.2

196 CD158b2 14.5 277 KIR2DL3

197 CD158d 73.5 381 KIR2DL4

198 CD158e2 0.162 275 KIR3DL1

199 CD158f 13.5 275 KIR2DL5

200 CD158i 18.8 298 KIR2DS4

201 CD159a 17 290 NKG2A

202 CD159c 47.9 359 NKG2C

203 CD160 28.6 462 BY55

Page 246: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

226

204 CD161 30.5 305 NKR-P1

205 CD162 100 7888 PSGL-1

206 CD163 12.7 284 Ber-MAC3, M130

207 CD164 16.4 315 MGC-24

208 CD165 0.0872 238 AD2, gp37

209 CD166 3.48 261 ALCAM

210 CD167 19 286 DDR1

211 CD169 26.3 289 Sialoadhesin, Siglec-1

212 CD170 34 327 Siglec-5, CD33-like2

213 CD171 1.4 245 L1

214 CD172a 17.3 289 SIRPgamma

215 CD172b 0.706 269 SIRPbeta, SIRB1

216 CD172g 39.2 332 SIRPgamma, SIRPB2

217 CD175s 53 412 Sialyl-Tn

218 CD177 1.47 263 NB1

219 CD178 31.9 311 FasL, CD95L

220 CD179a 57.2 382 VpreB

221 CD180 1.32 266 RP-105

222 CD181 64.8 471 CXCR1, IL-8RA

223 CD182 50.8 409 CXCR2, IL-8RB

224 CD183 94.2 507 CXCR3

225 CD184 17.8 309 CXCR4, fusin

226 CD185 1.8 370 CXCR5, BLR1

227 CD186 5.9 401 CXCR6, BONZO

228 CD191 11.8 281 CCR1, MIP-1alphaR, RANTES-R

229 CD192 0.172 304 CCR2, MCP-1-R

230 CD193 80.8 1139 CCR3, CKR3

231 CD194 3.49 421 CCR4

232 CD195 11.5 276 CCR5

233 CD196 92.1 911 CCR6, LARC receptor, DRY6

234 CD197 0.0369 269 CCR7

235 CD200 0.359 252 OX-2

236 CD201 0.0709 292 EPC-R

237 CD202b 73 595 Tie2, Tek

238 CD203c 6.82 266 NPP3 / PDNP3, ENpp1, PD-1b

239 CD204 22.5 286 Macrophage scavenger-R

240 CD205 80.6 493 DEC-205

241 CD206 0.837 257 Macrophage mannose-R

242 CD207 3.12 398 Langerin

243 CD208 23.9 295 DC-LAMP, LAMP-3

244 CD209 0.0316 238 DC-SIGN

245 CD212 40.1 313 IL-12-R beta1

Page 247: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

227

246 CD213a2 0.314 303 IL-13-R alpha2

247 CD215 50.8 359 IL-15R alpha

248 CD217 29.9 434 IL-17-R

249 CD218b 59 417 IL-18Rbeta, IL18RAP

250 CD220 5.88 401 Insulin-R

251 CD221 3.16 277 IGF-1 R

252 CD222 4.87 369 IGF-II R

253 CD223 99 6858 Lag3

254 CD226 82.6 398 DNAM-1, PTA-1, TLiSA1

255 CD227 95.7 1046 MUC1, EMA

256 CD229 99.3 758 Ly-9

257 CD230 99.9 1172 PRNP

258 CD231 73.8 602 TALLA-1, A15

259 CD234 16.9 279 Duffy, DARC

260 CD235a 56.3 411 Glycophorin A

261 CD243 (BC) 19.7 282 MDR-1, p170, P-gp

262 CD243 (BD) 0.094 268 MDR-1, p170, P-gp

263 CD244 3.64 261 2B4

264 CD245 99.2 1105 p220/240

265 CD247 19.4 291

266 CD249 18.3 320 Aminopeptidase A

267 CD252 17.2 496 OX-40Ligand, gp34

268 CD253 59.9 514 TRAIL, TNFSF10

269 CD254 51.4 370 TRANCE, RANKL, OPGL

270 CD255 30.6 325 TWEAK

271 CD256 18 287 APRIL, TALL-2

272 CD257 73.4 476 BLyS, BAFF, TALL-1

273 CD258 36 302 LIGHT, HVEM-L

274 CD261 46.4 313 TRAIL-R1, DR4

275 CD262 86.7 460 TRAIL-R2, DR5

276 CD263 9.81 268 TRAIL-R3, DcR1, LIT

277 CD264 10.5 269 TNFRSF10D, TRAILR4

278 CD267 17.6 479 TACI, TNFR SF13B

279 CD268 10.9 11864 BAFFR, TR13C

280 CD269 31.1 316 BCMA, TNFRSF13B

281 CD270 72.3 486 TNFSF14

282 CD271 1.08 279 NGFR, p75 (NTR)

283 CD272 4.53 68697 BTLA

284 CD273 0.316 231 B7DC, PD-L2, PDCD1L2

285 CD274 2.03 277 B7-H1, PD-L1

286 CD275 38.4 337 B7-H2, ICOSL, B7-RP1

287 CD276 76.3 863 B7-H3

Page 248: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

228

288 CD277 11.8 277 BT3.1, butyrophilin SF3 A1

289 CD278 15.3 289 ICOS, AILIM

290 CD279 27.2 303 PD1, SLEB2

291 CD281 47 350 TLR1

292 CD282 0.231 373 TLR2

293 CD283 76.5 1102 TLR3

294 CD284 42.4 334 TLR4

295 CD286 63.4 419 TLR6

296 CD288 72.9 623 TLR8

297 CD289 35.9 323 TLR9

298 CD290 70.1 549 TLR-10

299 CD292 1.46 357 BMPR1A, ALK3

300 CD294 11 412 CRTH2. GPR44

301 CD295 40.8 340 LeptinR, LEPR

302 CD298 100 7447 Na/K ATPase beta3 subunit

303 CD299 57.2 405 DC-SIGN-related, LSIGN, DC-SIGN2

304 CD300a 100 8067 CMRF35H, IRC1, IRp60

305 CD300c 72.3 481 CMRF35A, LIR

306 CD300e 12.4 274 CMRF35L

307 CD301 0.812 397 MGL, HML

308 CD303 2.99 4223 BDCA2, HECL

309 CD304 21.2 289 BDCA4, neuropilin 1

310 CD305 92.1 634 LAIR1

311 CD307 28.5 308 IRTA2

312 CD309 42.7 339 VEGFR2, KDR

313 CD312 48 400 EMR2

314 CD314 94.7 2723 NKG2D, KLR

315 CD317 100 2482 BST2, HM1.24

316 CD318 75 355 CDCP1, SIMA135

317 CD319 82.2 469 CRACC, SLAMF7

318 CD321 90.7 1311 JAM1, F11 receptor

319 CD322 0.535 367 JAM2, VE-JAM

320 CD324 1.1 370 E-Cadherin, Uvomorulin

321 CD325 38.5 321 N-Cadherin, NCAD

322 CD326 1.05 404 Ep-CAM, Ly74

323 CD328 61.8 436 SIGLEC7, AIRM-1

324 CD332 0.416 354 FGFR2, BEK, KGFR

325 CD333 36.3 307 FGFR3, ACH, CEK2

326 CD334 6.89 266 FGFR4, JTK2, TKF

327 CD335 50.8 327 NKp46, Ly-94 homolog

328 CD336 99.5 1039 NKp44, Ly-95 homolog

Page 249: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

229

329 CD337 99.3 4130 NKp30, Ly117

330 CD338 80.5 756 ABCG2, BCRP, Bcrp1, MXR

331 CD339 0.539 362 Jagged-1, JAG1, JAGL1, hJ1

332 CD340 0.38 403 erbB2, HER-2, EGFR-2

333 CD344 69.1 546 EVR1

334 CD349 7.02 409

335 CD351 81 2159 FCAMR

336 CD352 100 3164 SLAMF6, NTB-A

337 CD354 66.3 481 TREM-1

338 CD355 47.3 337 CRTAM

339 CD357 4.37 399 TNFSF18, GITR

340 CD358/DR6 69.9 589 TNFSF21, DR6

341 CD360 (BD) 74.4 461 IL-21RA

342 CD360 (BL) 54.7 367 IL-21RA

343 CD362 0.281 485 SDC2, HSPG-1

344 CD363 58.9 389 S1PR1

345 CLA 81.2 871 CLA

346 CLIP 0.601 368 CLIP

347 DCIR 1.24 394 DCIR

348 EGF-R 1.39 264 EGF-R

349 FMC7 0.119 374 FMC7

350 FOXP3 76.8 446

351 Galectin-3 0.549 248 352 HLA-ABC 100 3164 HLA-ABC

353 HLA-A2 1.01 248 HLA-A2

354 HLA-DM 1.16 272 HLA-DM

355 HLA-DR 0.383 260 HLA-DR

356 HPC 0.288 229 HPC

357 ITGB7 98.2 1395 ITGB7

358 LTBR 57 655 LTBR, TNFRSF3

359 MIC A/B 4.6 266 MIC A/B

360 NPM-ALK 0.143 272 361 Notch1 0.0578 401 Notch1

362 Notch2 21 283 Notch2

363 Notch3 37.5 464 Notch3

364 PAC-1 0.169 387 PAC-1

365 Podoplanin 7.72 262 PDPN

366 SSEA-3 18 279 SSEA-3

367 SSEA-4 0.368 230 SSEA-4

368 Stro-1 4.45 402 Stro-1

369 TCR alpha beta 0.26 242 TCR alpha beta

370 TCR gamma delta 9.29 17690 TCR gamma delta

Page 250: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

230

371 TPBG 0.0174 265 TPBG

372 VB8 TCR 41.4 362 VB8 TCR

373 VD2 TCR 66.7 552 VD2 TCR

374 fMLP-R 29 302 fMLP-R

Page 251: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

231

Appendix III: OCI/AML3 HTS flow cytometry percent positivity and MFI of antigens

# Antigen % + MFI Antigen name(s)

1 BLTR-1 1.65 1286 Leukotriene B4 receptor

2 B2m 98.9 3133 Beta-2 microglobulin

3 CA9 0.89 604 Carbonic anhydrase IX

4 CDH3 0.198 490 Cadherin 3

5 CDH6 1.57 427 Cadherin 6

6 CDH11 8.12 707 Cadherin 11

7 CDw93 15 816 C1q-binding protein

8 CDw198 74.4 5413 CCR8

9 CDw199 56.2 438 CCR9

10 CDw210 0.186 547 IL-10 receptor

11 CDw218a 0.128 1277 IL-18 receptor

12 CDw329 0.28 847 SIGLEC-9

13 CD1a 0.0733 1206 T6

14 CD1b 0.212 945 T6

15 CD1c 0.695 620 T6

16 CD1d 0.152 376 CD1d

17 CD2 0.132 1429 T11, LFA-2, SRBC-R

18 CD3 0.0687 1147 T3

19 CD3e 0.0163 839 T3

20 CD4 69.3 1346 T4

21 CD5 0.0462 1280 T1, Tp67

22 CD6 1.29 1180 T12

23 CD7 0.0707 898 LEU-9

24 CD8 0.0649 849 T8, Leu-2, CD8alpha

25 CD8b 0.174 1024 CD8beta

26 CD9 0.186 596 p24, MRP-1

27 CD10 0.0898 1336 CALLA, NEP, gp100

28 CD11a 98.4 8983 LFA-1, integrin alpha L

29 CD11b 0.613 602 Mac-1, integrin alphaM

30 CD11c 0.208 730 p150, CR4, integrin alphaX

31 CD13 68.6 2149 Aminopeptidase N, APN

32 CD14 0.164 1150 LPS-R

33 CD15 0.27 408 Lewis-x, Lex

34 CD16 0.214 598 Fcgamma RIIIA

35 CD16b 0.128 1126 FcgammaRIIIB

Page 252: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

232

36 CD17 77.1 12014 Lactosylceramide

37 CD18 12.9 738 Integrin beta2

38 CD19 0.0783 1250 B4

39 CD20 0.0769 998 B1, Bp35

40 CD21 0.205 898 C3DR, CR2, EBV-R

41 CD22 0.211 1596 BL-CAM, Siglec-2

42 CD23 0.147 839 FcepsilonRII

43 CD24 0.176 1262 BA-1

44 CD25 0.125 588 IL-2Ralpha, Tac, p55

45 CD26 0.202 768 DPP IV

46 CD27 0.0803 1339 TNFRSF7, T14

47 CD28 0.219 839 Tp44, T44

48 CD29 78.3 2476 Integrin beta1

49 CD30 0.0464 1342 Ki-1

50 CD31 31.5 1840 PECAM-1

51 CD32 64.5 1183 IgSF

52 CD33 19.7 827 p67, Siglec-3

53 CD34 0.0794 998 HPCA1

54 CD35 2.63 1800 CR1

55 CD36 42.2 1554 GPIV

56 CD37 0.0675 1005 N/A

57 CD38 29.5 913 T10

58 CD39 0.284 291 ENTPD1

59 CD40 0.0697 348 TNFRSF5

60 CD41a 0.119 596 gpIIb

61 CD41b 0.0506 621 HPA-3

62 CD42a 0.131 833 GPIX

63 CD42b 0.13 1197 GPIba

64 CD43 99.9 3605 Leukosialin, sialophorin

65 CD44 99.8 6039 Hyalunorate receptor

66 CD45 99.8 4828 LCA

67 CD45RA 82.2 2003 LCA

68 CD45RB 37.3 885 LCA, T200, B220

69 CD45RO 1.71 948 LCA, UCHL-1

70 CD46 98.7 1238 Membrane cofactor protein

71 CD47 15.4 752 IAP

72 CD48 0.0854 991 Blast-1

73 CD49a 0.213 656 VLA-1

74 CD49b 4.01 959 VLA-2

75 CD49c 0.133 1463 VLA-3

76 CD49d 99.7 6428 VLA-4

77 CD49e 98.3 24850 VLA-5

Page 253: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

233

78 CD49f 8.34 1029 VLA-6

79 CD50 0.127 766 ICAM-3

80 CD51/CD61 0 ¥ Vitronectin receptor

81 CD52 0.0884 808 CAMPATH-1 antigen

82 CD53 1.74 610 MRC OX44

83 CD54 42.9 1699 ICAM-1

84 CD55 99.9 4757 DAF

85 CD56 0.944 575 NCAM

86 CD57 0.0926 673 HNK-1, Leu-7

87 CD58 91.7 2218 LFA-3

88 CD59 0.909 1950 Protectin, MAC- inhibitor

89 CD60b 0.472 728 9-O-sialyl GD3

90 CD61 0 ¥ GPIIIa

91 CD62E 0 ¥ E-selectin, ELAM-1

92 CD62L 0.129 1241 L-selectin, LECAM-1

93 CD62P 0.0783 705 P-selectin, PADGEM

94 CD63 0.808 471 LIMP, LAMP-3

95 CD64 0.757 522 FcgammaRI

96 CD65 99.7 9119 VIM2

97 CD65s 90 2295 VIM2

98 CD66 0.155 805 BGP-1, NCA-160

99 CD66b 0.0543 928 CD67, CGM6

100 CD66c 4.42 1081 NCA

101 CD66d 0.725 728 CGM1; CEACAM-3

102 CD66e 69.6 1876 CEACAM-5

103 CD69 0.134 447 AIM

104 CD70 30.2 1034 Ki-24

105 CD71 70.9 1298 T9

106 CD72 0.0881 970 Lyb-2

107 CD73 58 1715 L-VAP-2

108 CD74 0.784 592 Invariant chain

109 CD75 0.0637 959 LN-1

110 CD77 0.125 943 Gb3, Pk blood group

111 CD79a 0.349 473 Iga

112 CD79b 0.46 968 Igb

113 CD80 0.174 1139 B7, B7-1, BB1

114 CD81 79.9 5074 TAPA-1

115 CD82 87.3 5399 R2

116 CD83 0.121 1031 HB15

117 CD84 5.47 699 SLAMF5

118 CD85a 0.267 954 LILRB3

119 CD85d 0.951 493 LIRB2, ILT-4, LIR-2

Page 254: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

234

120 CD85g 1.83 579 ILT-7

121 CD85h 2.19 937 LILRA-2, ILT-1, LIR-7

122 CD85j 5.14 1139 LIRRB-1, ILT-2

123 CD86 7.4 1311 B70, B7-2

124 CD87 16.3 945 UPA-R

125 CD88 0.28 1271 C5aR

126 CD89 0.644 1474 FcalphaR

127 CD90 12.3 966 Thy-1

128 CD91 1.44 939 TGFBR5

129 CD92 2.4 745 CTL1B

130 CD94 0.0415 1203 NKG2C, KP43

131 CD95 5.79 829 Apo-1, Fas

132 CD96 0.186 675 TACTILE

133 CD97 39.2 902 AURA51

134 CD98 99.9 8271 4F2

135 CD99 1.6 472 MIC2, E2

136 CD100 0.879 609 SEMA4D

137 CD101 28.5 1569 V7, p126

138 CD102 9.74 697 ICAM-2

139 CD103 0.0848 1015 HML-1, alpha6, integrin alphaE

140 CD104 0.154 937 Beta4 integrin

141 CD105 31.7 900 Endoglin

142 CD106 0.0799 952 VCAM-1

143 CD107a 0.184 837 LAMP-1

144 CD107b 0.111 952 LAMP-2

145 CD108 1.46 558 SEMA7A

146 CD109 4.28 807 7D1, 8A3

147 CD110 2.19 568 MPL, TPO-R

148 CD111 70.8 1333 PRR1, Nectin-1

149 CD112 0.241 522 PRR2, Nectin-2

150 CD114 2.56 155000 G-CSFR

151 CD115 0.728 663 M-CSFR, c-fms

152 CD116 16.3 1022 GM-CSFR alpha

153 CD117 3.27 2229 c-kit, SCFR

154 CD118 0.321 708 LIFR, gp190

155 CD119 27.4 934 IFNgammaR

156 CD120a 9.38 732 TNFR-I

157 CD120b 8.15 792 TNFR-II

158 CD121b 1.69 584 IL-1R, type II

159 CD122 1.45 532 IL-2Rbeta

160 CD123 16.1 754 IL-3Ralpha

161 CD124 0.165 812 IL-4Ralpha

Page 255: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

235

162 CD125 0.0869 1691 IL-5Ralpha

163 CD126 22.1 790 IL-6Ralpha

164 CD127 8.74 1180 IL-7Ralpha

165 CD129 0.0382 532 IL-9R

166 CD130 0.116 458 IL-6Rbeta, gp130

167 CD131 0.112 510 IL-3Rbeta

168 CD132 2.87 1292 Common gamma

169 CD133 0.0558 436 AC133, prominin-like 1

170 CD134 3.62 600 OX-40

171 CD135 11.9 847 Flt3/Flk2

172 CD136 0.129 661 MSP-R, RON

173 CD137 0.102 961 4-1BB, TNRFSF9

174 CD137L 3.85 661 4-1BB L

175 CD138 0.0771 1311 Syndecan-1

176 CD140a 0.103 1346 PDGFRalpha

177 CD140b 0.167 742 PDGFRbeta

178 CD141 81.6 7263 Thrombomodulin

179 CD142 0.131 1034 Tissue Factor

180 CD143 0.906 503 ACE

181 CD144 0.0641 672 VE-Cadherin, Cadherin-5

182 CD146 0.163 799 MUC18, S-endo

183 CD147 100 9563 Neurothelin, basoglin

184 CD148 91.4 1619 HPTP-eta

185 CD150 0.16 1120 SLAM

186 CD151 82.4 6069 PETA-3

187 CD152 0.145 765 CTLA-4

188 CD153 0.523 1107 CD30L

189 CD154 0.0938 1336 CD40L, gp39, TRAP

190 CD155 93.7 3457 PVR

191 CD156b 32 975 ADAM8

192 CD157 0.222 1158 BST-1

193 CD158a 0.0807 1178 KIR2DL1, p58.1

194 CD158b 0.0645 623 KIR2DL2, p58.2

195 CD158b2 0.189 562 KIR2DL3

196 CD158d 4.8 754 KIR2DL4

197 CD158e2 0.165 937 KIR3DL1

198 CD158f 0.492 558 KIR2DL5

199 CD158i 0.436 705 KIR2DS4

200 CD159a 0.291 687 NKG2A

201 CD159c 0.299 522 NKG2C

202 CD160 1.68 515 BY55

203 CD161 0.174 930 NKR-P1

Page 256: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

236

204 CD162 95.8 2318 PSGL-1

205 CD163 0.0548 1241 Ber-MAC3, M130

206 CD164 0.244 667 MGC-24

207 CD165 0.121 777 AD2, gp37

208 CD166 2.67 623 ALCAM

209 CD167 0.355 1346 DDR1

210 CD169 1.59 1235 Sialoadhesin, Siglec-1

211 CD170 0.0975 1092 Siglec-5, CD33-like2

212 CD171 0.186 737 L1

213 CD172a 98.3 4029 SIRPgamma

214 CD172b 0.0731 564 SIRPbeta, SIRB1

215 CD172g 0.349 524 SIRPgamma, SIRPB2

216 CD175s 65 934 Sialyl-Tn

217 CD177 0.0385 957 NB1

218 CD178 3.22 572 FasL, CD95L

219 CD179a 0.219 575 VpreB

220 CD180 0.213 648 RP-105

221 CD181 0.923 683 CXCR1, IL-8RA

222 CD182 2.96 184000 CXCR2, IL-8RB

223 CD183 0.0995 667 CXCR3

224 CD184 2.05 697 CXCR4, fusin

225 CD185 26.2 810 CXCR5, BLR1

226 CD186 0.243 618 CXCR6, BONZO

227 CD191 9.37 725 CCR1, MIP-1alphaR, RANTES-R

228 CD192 0.205 301 CCR2, MCP-1-R

229 CD193 1.81 610 CCR3, CKR3

230 CD194 34.2 389 CCR4

231 CD195 0.114 1195 CCR5

232 CD196 2.17 552 CCR6, LARC receptor, DRY6

233 CD197 0.089 335 CCR7

234 CD200 0.114 984 OX-2

235 CD201 0.0791 1164 EPC-R

236 CD202b 1.76 596 Tie2, Tek

237 CD203c 4.61 1359 NPP3 / PDNP3, ENpp1, PD-1b

238 CD204 0.293 1139 Macrophage scavenger-R

239 CD205 64.1 1175 DEC-205

240 CD206 0.074 678 Macrophage mannose-R

241 CD207 0.149 610 Langerin

242 CD208 0.135 1372 DC-LAMP, LAMP-3

243 CD209 0.275 1153 DC-SIGN

244 CD212 0.0685 369 IL-12-R beta1

245 CD213a2 0.132 689 IL-13-R alpha2

Page 257: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

237

246 CD215 1.2 634 IL-15R alpha

247 CD217 98.5 1102 IL-17-R

248 CD218b 0.61 826 IL-18Rbeta, IL18RAP

249 CD220 30.2 361 Insulin-R

250 CD221 9.18 599 IGF-1 R

251 CD222 1.49 556 IGF-II R

252 CD223 4.84 1153 Lag3

253 CD226 0.12 474 DNAM-1, PTA-1, TLiSA1

254 CD227 20 740 MUC1, EMA

255 CD229 3.02 592 Ly-9

256 CD230 77.4 3614 PRNP

257 CD231 7.34 744 TALLA-1, A15

258 CD234 4.57 885 Duffy, DARC

259 CD235a 11 934 Glycophorin A

260 CD243 (BC) 0.116 1280 MDR-1, p170, P-gp

261 CD243 (BD) 0.123 2482 MDR-1, p170, P-gp

262 CD244 0.407 427 2B4

263 CD245 68.9 1456 p220/240

264 CD249 0.309 613 Aminopeptidase A

265 CD252 18.8 1305 OX-40Ligand, gp34

266 CD253 12.8 801 TRAIL, TNFSF10

267 CD254 0.32 541 TRANCE, RANKL, OPGL

268 CD255 1.1 497 TWEAK

269 CD256 14.8 1298 APRIL, TALL-2

270 CD257 16.1 941 BLyS, BAFF, TALL-1

271 CD258 1.11 571 LIGHT, HVEM-L

272 CD261 0.61 478 TRAIL-R1, DR4

273 CD262 12.5 989 TRAIL-R2, DR5

274 CD263 1.63 681 TRAIL-R3, DcR1, LIT

275 CD264 1.77 636 TNFRSF10D, TRAILR4

276 CD267 45.4 1142 TACI, TNFR SF13B

277 CD268 25.9 1546 BAFFR, TR13C

278 CD269 0.167 855 BCMA, TNFRSF13B

279 CD270 12.8 1217 TNFSF14

280 CD271 0.419 636 NGFR, p75 (NTR)

281 CD272 20.4 1120 BTLA

282 CD273 0.269 661 B7DC, PD-L2, PDCD1L2

283 CD274 0.102 1150 B7-H1, PD-L1

284 CD275 2.21 564 B7-H2, ICOSL, B7-RP1

285 CD276 81.5 4223 B7-H3

286 CD277 0.514 539 BT3.1, butyrophilin SF3 A1

287 CD278 0.13 892 ICOS, AILIM

Page 258: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

238

288 CD279 0.459 761 PD1, SLEB2

289 CD281 4.33 607 TLR1

290 CD282 21 699 TLR2

291 CD283 5.05 717 TLR3

292 CD284 27.5 1320 TLR4

293 CD286 4.04 712 TLR6

294 CD288 7.11 859 TLR8

295 CD289 0.475 514 TLR9

296 CD290 1.9 533 TLR-10

297 CD292 0.334 1017 BMPR1A, ALK3

298 CD294 1.24 310 CRTH2. GPR44

299 CD295 1.59 512 LeptinR, LEPR

300 CD298 99.9 6207 Na/K ATPase beta3 subunit

301 CD299 2.86 810 DC-SIGN-related, LSIGN, DC-SIGN2

302 CD300a 1.04 1049 CMRF35H, IRC1, IRp60

303 CD300c 1.47 599 CMRF35A, LIR

304 CD300e 3.58 643 CMRF35L

305 CD301 0.158 1453 MGL, HML

306 CD303 7.64 807 BDCA2, HECL

307 CD304 1.3 1333 BDCA4, neuropilin 1

308 CD305 99.6 9684 LAIR1

309 CD307 4.12 1051 IRTA2

310 CD309 10.6 717 VEGFR2, KDR

311 CD312 62.3 1382 EMR2

312 CD314 9.95 808 NKG2D, KLR

313 CD317 83.8 5346 BST2, HM1.24

314 CD318 0.224 831 CDCP1, SIMA135

315 CD319 5.13 689 CRACC, SLAMF7

316 CD321 29 1059 JAM1, F11 receptor

317 CD322 2.9 33040 JAM2, VE-JAM

318 CD324 2.64 869 E-Cadherin, Uvomorulin

319 CD325 0.225 623 N-Cadherin, NCAD

320 CD326 0.148 865 Ep-CAM, Ly74

321 CD328 0.0697 1346 SIGLEC7, AIRM-1

322 CD332 0.372 277 FGFR2, BEK, KGFR

323 CD333 9.75 890 FGFR3, ACH, CEK2

324 CD334 0.151 488 FGFR4, JTK2, TKF

325 CD335 0.15 968 NKp46, Ly-94 homolog

326 CD336 0.14 1097 NKp44, Ly-95 homolog

327 CD337 8.92 869 NKp30, Ly117

328 CD338 1.32 1051 ABCG2, BCRP, Bcrp1, MXR

Page 259: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

239

329 CD339 0.0956 839 Jagged-1, JAG1, JAGL1, hJ1

330 CD340 0.468 1481 erbB2, HER-2, EGFR-2

331 CD344 12.3 779 EVR1

332 CD351 45.9 1283 FCAMR

333 CD352 0.168 1134 SLAMF6, NTB-A

334 CD354 12 984 TREM-1

335 CD355 0.771 520 CRTAM

336 CD357 43 415 TNFSF18, GITR

337 CD358 5.71 673 TNFSF21, DR6

338 CD360 (BD) 1.49 521 IL-21RA

339 CD360 (BL) 1.88 607 IL-21RA

340 CD362 93.8 2352 SDC2, HSPG-1

341 CD363 3.51 1172 S1PR1

342 CLA 1.26 735 CLA

343 CLIP 0.0982 518 CLIP

344 DCIR 0.176 344 DCIR

345 EGF-R 0.0654 950 EGF-R

346 FMC7 0.115 979 FMC7

347 HLA-ABC 79.4 1372 HLA-ABC

348 HLA-A2 94.2 2154 HLA-A2

349 HLA-DM 0.0945 539 HLA-DM

350 HLA-DR 0.128 717 HLA-DR

351 HPC 0.132 1268 HPC

352 ITGB7 10.5 1118 ITGB7

353 LTBR 86.7 1978 LTBR, TNFRSF3

354 Lgr-5 0.643 740 Lgr-5

355 MIC A/B 0.201 737 MIC A/B

356 Notch1 86.4 694 Notch1

357 Notch2 36.4 1097 Notch2

358 Notch3 7.33 320 Notch3

359 Notch4 1.04 1333 Notch4

360 PAC-1 0.034 1094 PAC-1

361 Podoplanin 0.105 759 PDPN

362 SSEA-3 0.164 730 SSEA-3

363 SSEA-4 0.0964 1183 SSEA-4

364 Stro-1 13.6 342 Stro-1

365 TCR alpha beta 0.0803 1268 TCR alpha beta

366 TCR gamma delta 6.17 833 TCR gamma delta

367 TPBG 0.0689 346 TPBG

368 VB8 TCR 4.75 119000 VB8 TCR

369 VD2 TCR 8.77 1425 VD2 TCR

370 fMLP-R 0.105 712 fMLP-R

Page 260: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

240

Appendix IV: OCI/AML5 HTS flow cytometry percent positivity and MFI of antigens

# Antigen % + MFI Antigen name(s)

1 BLTR-1 18.6 368 Leukotriene B4 receptor

2 B2m 99.8 14251 Beta-2 microglobulin

3 CA9 16.4 362 Carbonic anhydrase IX

4 CDH3 20.7 376 Cadherin 3

5 CDH6 0.311 250 Cadherin 6

6 CDH11 6.36 422 Cadherin 11

7 CDw93 46 456 C1q-binding protein

8 CDw198 66.2 533 CCR8

9 CDw199 0.0214 280 CCR9

10 CDw210 19.2 377 IL-10 receptor

11 CDw218a 24.9 388 IL-18 receptor

12 CDw329 2.89 467 SIGLEC-9

13 CD1a 1.34 552 T6

14 CD1b 2.54 411 T6

15 CD1c 16 497 T6

16 CD1d 26.7 410 CD1d

17 CD2 0.237 318 T11, LFA-2, SRBC-R

18 CD3 0.13 323 T3

19 CD3e 0.0249 264 T3

20 CD4 0.243 326 T4

21 CD5 0.13 314 T1, Tp67

22 CD6 0.815 298 T12

23 CD7 0.846 326 LEU-9

24 CD8 0.695 312 T8, Leu-2, CD8alpha

25 CD8b 0.541 325 CD8beta

26 CD9 0.476 318 p24, MRP-1

27 CD10 0.288 326 CALLA, NEP, gp100

28 CD11a 100 41148 LFA-1, integrin alpha L

29 CD11b 52.5 620 Mac-1, integrin alphaM

30 CD11c 25.4 440 p150, CR4, integrin alphaX

31 CD13 15.9 391 Aminopeptidase N, APN

32 CD14 0.376 297 LPS-R

33 CD15 6.47 512 Lewis-x, Lex

34 CD16 15.3 448 Fcgamma RIIIA

35 CD16b 0.232 325 FcgammaRIIIB

Page 261: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

241

36 CD17 52.8 717 Lactosylceramide

37 CD18 99.9 1654 Integrin beta2

38 CD19 0.136 323 B4

39 CD20 0.157 334 B1, Bp35

40 CD21 3.76 340 C3DR, CR2, EBV-R

41 CD22 1.14 343 BL-CAM, Siglec-2

42 CD23 2.88 346 FcepsilonRII

43 CD24 1.19 305 BA-1

44 CD25 17.3 453 IL-2Ralpha, Tac, p55

45 CD26 0.834 315 DPP IV

46 CD27 0.618 323 TNFRSF7, T14

47 CD28 2.98 328 Tp44, T44

48 CD29 100 4151 Integrin beta1

49 CD30 82 827 Ki-1

50 CD31 58.2 712 PECAM-1

51 CD32 98.9 4029 IgSF

52 CD33 100 3483 p67, Siglec-3

53 CD34 0.273 316 HPCA1

54 CD35 3.39 438 CR1

55 CD36 4.91 643 GPIV

56 CD37 0.354 412 N/A

57 CD38 99.8 1889 T10

58 CD39 3.36 442 ENTPD1

59 CD40 0.138 309 TNFRSF5

60 CD41a 0.177 313 gpIIb

61 CD41b 0.289 426 HPA-3

62 CD42a 4.05 328 GPIX

63 CD42b 6.1 334 GPIba

64 CD43 99.8 2658 Leukosialin, sialophorin

65 CD44 100 12728 Hyalunorate receptor

66 CD45 99.9 5413 LCA

67 CD45RA 100 27005 LCA

68 CD45RB 98.6 797 LCA, T200, B220

69 CD45RO 1.5 318 LCA, UCHL-1

70 CD46 98.1 835 Membrane cofactor protein

71 CD47 99.8 1912 IAP

72 CD48 91.4 952 Blast-1

73 CD49a 1.58 365 VLA-1

74 CD49b 67.5 461 VLA-2

75 CD49c 0.527 322 VLA-3

76 CD49d 100 16612 VLA-4

77 CD49e 100 7084 VLA-5

Page 262: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

242

78 CD49f 0.819 325 VLA-6

79 CD50 0.518 416 ICAM-3

80 CD51/CD61 1.56 303 Vitronectin receptor

81 CD52 3.09 356 CAMPATH-1 antigen

82 CD53 85 623 MRC OX44

83 CD54 100 9468 ICAM-1

84 CD55 99.8 1849 DAF

85 CD56 72.8 742 NCAM

86 CD57 0.384 410 HNK-1, Leu-7

87 CD58 100 4351 LFA-3

88 CD59 99.4 890 Protectin, MAC- inhibitor

89 CD60b 9.2 422 9-O-sialyl GD3

90 CD61 0.359 320 GPIIIa

91 CD62E 1.78 317 E-selectin, ELAM-1

92 CD62L 2.64 329 L-selectin, LECAM-1

93 CD62P 1.33 332 P-selectin, PADGEM

94 CD63 62.4 421 LIMP, LAMP-3

95 CD64 0.415 309 FcgammaRI

96 CD65 99.7 5891 VIM2

97 CD65s 95.3 1215 VIM2

98 CD66 4.57 323 BGP-1, NCA-160

99 CD66b 0.36 424 CD67, CGM6

100 CD66c 27.7 371 NCA

101 CD66d 3.89 338 CGM1; CEACAM-3

102 CD66e 98.4 2128 CEACAM-5

103 CD69 3.46 335 AIM

104 CD70 99.3 1912 Ki-24

105 CD71 95.1 1178 T9

106 CD72 0.369 400 Lyb-2

107 CD73 0.35 317 L-VAP-2

108 CD74 15.3 431 Invariant chain

109 CD75 0.225 393 LN-1

110 CD77 55.3 452 Gb3, Pk blood group

111 CD79a 72.2 607 Iga

112 CD79b 5.2 339 Igb

113 CD80 8.84 461 B7, B7-1, BB1

114 CD81 99.9 6192 TAPA-1

115 CD82 37.4 468 R2

116 CD83 1.23 331 HB15

117 CD84 87.5 1686 SLAMF5

118 CD85a 4.82 318 LILRB3

119 CD85d 34.8 420 LIRB2, ILT-4, LIR-2

Page 263: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

243

120 CD85g 45.1 441 ILT-7

121 CD85h 14.2 357 LILRA-2, ILT-1, LIR-7

122 CD85j 43.7 432 LIRRB-1, ILT-2

123 CD86 39.4 424 B70, B7-2

124 CD87 0.559 334 UPA-R

125 CD88 2.34 334 C5aR

126 CD89 4.8 328 FcalphaR

127 CD90 36.4 561 Thy-1

128 CD91 14.8 338 TGFBR5

129 CD92 92 1049 CTL1B

130 CD94 0.124 323 NKG2C, KP43

131 CD95 93.2 742 Apo-1, Fas

132 CD96 30 372 TACTILE

133 CD97 90.1 547 AURA51

134 CD98 100 37957 4F2

135 CD99 78.1 475 MIC2, E2

136 CD100 59.1 511 SEMA4D

137 CD101 40.4 559 V7, p126

138 CD102 96.3 1180 ICAM-2

139 CD103 0.156 279 HML-1, alpha6, integrin alphaE

140 CD104 3.07 325 Beta4 integrin

141 CD105 9.3 336 Endoglin

142 CD106 2.12 316 VCAM-1

143 CD107a 1.76 346 LAMP-1

144 CD107b 0.765 409 LAMP-2

145 CD108 4.91 374 SEMA7A

146 CD109 14.7 399 7D1, 8A3

147 CD110 50.7 495 MPL, TPO-R

148 CD111 97.3 3605 PRR1, Nectin-1

149 CD112 16.8 357 PRR2, Nectin-2

150 CD114 5.78 426 G-CSFR

151 CD115 55.6 574 M-CSFR, c-fms

152 CD116 51.3 566 GM-CSFR alpha

153 CD117 15.3 624 c-kit, SCFR

154 CD118 12.7 367 LIFR, gp190

155 CD119 94.2 950 IFNgammaR

156 CD120a 86.4 869 TNFR-I

157 CD120b 25 372 TNFR-II

158 CD121b 70.4 604 IL-1R, type II

159 CD122 38.1 449 IL-2Rbeta

160 CD123 96.4 1099 IL-3Ralpha

161 CD124 3.69 367 IL-4Ralpha

Page 264: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

244

162 CD125 36.9 451 IL-5Ralpha

163 CD126 86.4 723 IL-6Ralpha

164 CD127 23.3 3989 IL-7Ralpha

165 CD129 0.277 318 IL-9R

166 CD130 3.43 316 IL-6Rbeta, gp130

167 CD131 1.9 315 IL-3Rbeta

168 CD132 44.8 518 Common gamma

169 CD133 0.0134 206 AC133, prominin-like 1

170 CD134 20.8 350 OX-40

171 CD135 84.1 824 Flt3/Flk2

172 CD136 2.4 333 MSP-R, RON

173 CD137 1.5 340 4-1BB, TNRFSF9

174 CD137L 70.9 609 4-1BB L

175 CD138 0.359 337 Syndecan-1

176 CD140a 0.376 322 PDGFRalpha

177 CD140b 9.12 348 PDGFRbeta

178 CD141 59.5 466 Thrombomodulin

179 CD142 0.987 315 Tissue Factor

180 CD143 45.5 496 ACE

181 CD144 0.444 311 VE-Cadherin, Cadherin-5

182 CD146 0.182 328 MUC18, S-endo

183 CD147 100 18791 Neurothelin, basoglin

184 CD148 98.9 1686 HPTP-eta

185 CD150 14.3 376 SLAM

186 CD151 99.8 1964 PETA-3

187 CD152 10.1 366 CTLA-4

188 CD153 8.31 341 CD30L

189 CD154 0.965 332 CD40L, gp39, TRAP

190 CD155 98.6 1740 PVR

191 CD156b 92 1110 ADAM8

192 CD157 7.88 340 BST-1

193 CD158a 0.186 333 KIR2DL1, p58.1

194 CD158b 0.182 302 KIR2DL2, p58.2

195 CD158b2 4.68 337 KIR2DL3

196 CD158d 22.1 380 KIR2DL4

197 CD158e2 0.193 322 KIR3DL1

198 CD158f 14.9 352 KIR2DL5

199 CD158i 24 380 KIR2DS4

200 CD159a 4.09 316 NKG2A

201 CD159c 33.4 403 NKG2C

202 CD160 2.66 282 BY55

203 CD161 4.96 328 NKR-P1

Page 265: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

245

204 CD162 99.8 8460 PSGL-1

205 CD163 3.2 310 Ber-MAC3, M130

206 CD164 21.7 370 MGC-24

207 CD165 0.612 307 AD2, gp37

208 CD166 99.7 1561 ALCAM

209 CD167 20.3 353 DDR1

210 CD169 4.8 328 Sialoadhesin, Siglec-1

211 CD170 93 831 Siglec-5, CD33-like2

212 CD171 2.24 331 L1

213 CD172a 99.8 9468 SIRPgamma

214 CD172b 0.458 340 SIRPbeta, SIRB1

215 CD172g 11.6 344 SIRPgamma, SIRPB2

216 CD175s 96.8 1089 Sialyl-Tn

217 CD177 0.154 406 NB1

218 CD178 42.6 465 FasL, CD95L

219 CD179a 24.8 361 VpreB

220 CD180 39.6 393 RP-105

221 CD181 26.4 402 CXCR1, IL-8RA

222 CD182 13.7 130000 CXCR2, IL-8RB

223 CD183 13.1 350 CXCR3

224 CD184 1.66 337 CXCR4, fusin

225 CD185 8.59 445 CXCR5, BLR1

226 CD186 3.17 320 CXCR6, BONZO

227 CD191 37.8 438 CCR1, MIP-1alphaR, RANTES-R

228 CD192 0.406 284 CCR2, MCP-1-R

229 CD193 41.8 455 CCR3, CKR3

230 CD194 10.6 334 CCR4

231 CD195 0.812 331 CCR5

232 CD196 38.7 489 CCR6, LARC receptor, DRY6

233 CD197 0.231 320 CCR7

234 CD200 0.738 298 OX-2

235 CD201 0.437 361 EPC-R

236 CD202b 27.5 411 Tie2, Tek

237 CD203c 23.2 375 NPP3 / PDNP3, ENpp1, PD-1b

238 CD204 6.05 455 Macrophage scavenger-R

239 CD205 99.9 3440 DEC-205

240 CD206 0.631 398 Macrophage mannose-R

241 CD207 68.3 413 Langerin

242 CD208 3.65 328 DC-LAMP, LAMP-3

243 CD209 0.194 329 DC-SIGN

244 CD212 14.3 331 IL-12-R beta1

245 CD213a2 85 466 IL-13-R alpha2

Page 266: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

246

246 CD215 29.2 402 IL-15R alpha

247 CD217 87.7 616 IL-17-R

248 CD218b 14.1 375 IL-18Rbeta, IL18RAP

249 CD220 59.2 378 Insulin-R

250 CD221 97.5 799 IGF-1 R

251 CD222 10.8 408 IGF-II R

252 CD223 44.9 424 Lag3

253 CD226 2.01 393 DNAM-1, PTA-1, TLiSA1

254 CD227 48.4 522 MUC1, EMA

255 CD229 50 427 Ly-9

256 CD230 98.6 1940 PRNP

257 CD231 41.2 473 TALLA-1, A15

258 CD234 18.5 373 Duffy, DARC

259 CD235a 52.5 471 Glycophorin A

260 CD243 (BC) 34.8 423 MDR-1, p170, P-gp

261 CD243 (BD) 0.251 342 MDR-1, p170, P-gp

262 CD244 22.3 345 2B4

263 CD245 96 1192 p220/240

264 CD249 34 371 Aminopeptidase A

265 CD252 9.23 5522 OX-40Ligand, gp34

266 CD253 36.9 456 TRAIL, TNFSF10

267 CD254 7.84 353 TRANCE, RANKL, OPGL

268 CD255 17 353 TWEAK

269 CD256 43.8 497 APRIL, TALL-2

270 CD257 55.4 581 BLyS, BAFF, TALL-1

271 CD258 72.8 712 LIGHT, HVEM-L

272 CD261 35 372 TRAIL-R1, DR4

273 CD262 98.7 1398 TRAIL-R2, DR5

274 CD263 31.7 403 TRAIL-R3, DcR1, LIT

275 CD264 48.1 536 TNFRSF10D, TRAILR4

276 CD267 65.3 896 TACI, TNFR SF13B

277 CD268 50.7 720 BAFFR, TR13C

278 CD269 10.8 394 BCMA, TNFRSF13B

279 CD270 45.3 448 TNFSF14

280 CD271 4.98 425 NGFR, p75 (NTR)

281 CD272 2.4 25873 BTLA

282 CD273 2.02 327 B7DC, PD-L2, PDCD1L2

283 CD274 0.65 326 B7-H1, PD-L1

284 CD275 20.9 369 B7-H2, ICOSL, B7-RP1

285 CD276 99.6 3027 B7-H3

286 CD277 4.99 356 BT3.1, butyrophilin SF3 A1

287 CD278 0.604 320 ICOS, AILIM

Page 267: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

247

288 CD279 6.26 346 PD1, SLEB2

289 CD281 79.4 758 TLR1

290 CD282 0.368 402 TLR2

291 CD283 57.7 524 TLR3

292 CD284 22.1 353 TLR4

293 CD286 80.5 666 TLR6

294 CD288 58.4 596 TLR8

295 CD289 15.9 356 TLR9

296 CD290 30.5 397 TLR-10

297 CD292 2.38 405 BMPR1A, ALK3

298 CD294 2.52 284 CRTH2. GPR44

299 CD295 43.2 389 LeptinR, LEPR

300 CD298 100 8169 Na/K ATPase beta3 subunit

301 CD299 29.5 393 DC-SIGN-related, LSIGN, DC-SIGN2

302 CD300a 99.7 3911 CMRF35H, IRC1, IRp60

303 CD300c 42.2 433 CMRF35A, LIR

304 CD300e 48.6 467 CMRF35L

305 CD301 0.111 355 MGL, HML

306 CD303 54 519 BDCA2, HECL

307 CD304 91.1 725 BDCA4, neuropilin 1

308 CD305 99.9 5228 LAIR1

309 CD307 5.91 329 IRTA2

310 CD309 70.4 607 VEGFR2, KDR

311 CD312 92 1449 EMR2

312 CD314 33.6 442 NKG2D, KLR

313 CD317 99.1 1770 BST2, HM1.24

314 CD318 7.57 330 CDCP1, SIMA135

315 CD319 14.9 356 CRACC, SLAMF7

316 CD321 100 5563 JAM1, F11 receptor

317 CD322 5.83 403 JAM2, VE-JAM

318 CD324 19.7 373 E-Cadherin, Uvomorulin

319 CD325 5.15 325 N-Cadherin, NCAD

320 CD326 0.585 330 Ep-CAM, Ly74

321 CD328 11.2 340 SIGLEC7, AIRM-1

322 CD332 2.16 285 FGFR2, BEK, KGFR

323 CD333 76.5 740 FGFR3, ACH, CEK2

324 CD334 2 320 FGFR4, JTK2, TKF

325 CD335 0.759 300 NKp46, Ly-94 homolog

326 CD336 0.261 316 NKp44, Ly-95 homolog

327 CD337 4.88 19861 NKp30, Ly117

328 CD338 46.2 448 ABCG2, BCRP, Bcrp1, MXR

Page 268: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

248

329 CD339 2.47 397 Jagged-1, JAG1, JAGL1, hJ1

330 CD340 0.655 444 erbB2, HER-2, EGFR-2

331 CD344 35.3 411 EVR1

332 CD351 59.2 633 FCAMR

333 CD352 0.838 318 SLAMF6, NTB-A

334 CD354 25.2 398 TREM-1

335 CD355 54.3 1283 CRTAM

336 CD357 77.1 566 TNFSF18, GITR

337 CD358 49.6 512 TNFSF21, DR6

338 CD360 (BD) 42.2 414 IL-21RA

339 CD360 (BL) 23.9 384 IL-21RA

340 CD362 0.345 359 SDC2, HSPG-1

341 CD363 22.6 380 S1PR1

342 CLA 99.2 1056 CLA

343 CLIP 79 669 CLIP

344 DCIR 0.638 290 DCIR

345 EGF-R 0.324 285 EGF-R

346 FMC7 0.251 425 FMC7

347 HLA-ABC 99.8 4471 HLA-ABC

348 HLA-A2 0.415 321 HLA-A2

349 HLA-DM 0.628 318 HLA-DM

350 HLA-DR 68 477 HLA-DR

351 HPC 0.748 316 HPC

352 ITGB7 93.9 1298 ITGB7

353 LTBR 23.8 420 LTBR, TNFRSF3

354 Lgr-5 4.56 331 Lgr-5

355 MIC A/B 1.65 313 MIC A/B

356 Notch1 84 526 Notch1

357 Notch2 73.3 875 Notch2

358 Notch3 11.2 298 Notch3

359 Notch4 21.5 368 Notch4

360 PAC-1 0.916 470 PAC-1

361 Podoplanin 8.51 342 PDPN

362 SSEA-3 26.3 349 SSEA-3

363 SSEA-4 0.507 350 SSEA-4

364 Stro-1 12.3 305 Stro-1

365 TCR alpha beta 0.521 326 TCR alpha beta

366 TCR gamma delta 52.1 686 TCR gamma delta

367 TPBG 0.141 251 TPBG

368 VB8 TCR 7.86 343 VB8 TCR

369 VD2 TCR 12.5 401 VD2 TCR

370 fMLP-R 14.8 370 fMLP-R

Page 269: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

249

Copyright Acknowledgements

Page 270: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

250

Chapter 3: Reprinted with minor modifications from: B.A. Williams, X.-H. Wang and A.

Keating. Clonogenic assays measure leukemia stem cell killing not detectable by chromium

release and flow cytometric cytotoxicity assays. Cytotherapy 2010: 12(7);951-60.

SPRINGER LICENSE

TERMS AND CONDITIONS

Feb 09, 2015

This is a License Agreement between Brent Williams ("You") and Springer ("Springer")

provided by Copyright Clearance Center ("CCC"). The license consists of your order

details, the terms and conditions provided by Springer, and the payment terms and

conditions.

All payments must be made in full to CCC. For payment instructions, please see information listed at the bottom of this

form.

License Number 3552210425609

License date Jan 18, 2015

Licensed content publisher Springer

Licensed content publication Springer eBook

Licensed content title NK-92 Cytotoxicity Against Cancer Stem Cells in Hematologic Malignancies

Licensed content author Brent A. Williams

Licensed content date Jan 1, 2013

Type of Use Thesis/Dissertation

Portion Full text

Number of copies 10

Author of this Springer article Yes and you are the sole author of the new work

Order reference number None

Title of your thesis / dissertation Immunotherapy of Leukemic Stem Cells

Expected completion date Jan 2015

Estimated size(pages) 260

Total 0.00 CAD

Page 271: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

251

Section 1.3.3: Reprinted with minor modifications from: B. A. Williams, B. E. Swift, R. Cheng

and A. Keating. 2013. NK-92 cytotoxicity against cancer stem cells in hematologic

malignancies. (Springer) Stem Cells and Cancer Stem Cells: Therapeutic Applications in Disease

and Injury, Ed. M.A. Hayat, 9 (24); 249-257.

ELSEVIER LICENSE

TERMS AND CONDITIONS

Feb 09, 2015

This is a License Agreement between Brent Williams ("You") and Elsevier ("Elsevier")

provided by Copyright Clearance Center ("CCC"). The license consists of your order

details, the terms and conditions provided by Elsevier, and the payment terms and

conditions.

All payments must be made in full to CCC. For payment instructions, please see information listed at the bottom of this

form.

Supplier Elsevier Limited

The Boulevard,Langford Lane

Kidlington,Oxford,OX5 1GB,UK

Registered Company Number 1982084

Customer name Brent Williams

Customer address 5 Colty Drive

Markham, ON L6C 2W1

License number 3564810548348

License date Feb 09, 2015

Licensed content publisher Elsevier

Licensed content publication Cytotherapy

Licensed content title Clonogenic assays measure leukemia stem cell killing not detectable by chromium release

and flow cytometric cytotoxicity assays

Licensed content author None

Licensed content date November 2010

Licensed content volume number 12

Licensed content issue number 7

Number of pages 10

Start Page 951

End Page 960

Type of Use reuse in a thesis/dissertation

Portion full article

Format both print and electronic

Are you the author of this Elsevier

article?

Yes

Will you be translating? No

Title of your thesis/dissertation Immunotherapy of Leukemic Stem Cells

Expected completion date Jan 2015

Estimated size (number of pages)

Elsevier VAT number GB 494 6272 12

Page 272: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

252

Permissions price 0.00 CAD

Page 273: Immunotherapy of Leukemic Stem Cells using Natural Killer ... · Immunotherapy of Leukemic Stem Cell using Natural Killer Cell lines Brent A. Williams Doctor of Philosphy Institute

253