+ All Categories
Home > Documents > the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1...

the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1...

Date post: 25-Jan-2019
Category:
Upload: phunghanh
View: 214 times
Download: 0 times
Share this document with a friend
178
Studies of the Immune System of HOX11 Transgenic Mice for Lymphoma Immunotherapy Suzana Rosic-Kablar, M.D. A thesis subrnitted in conformation with the requiremenb for the degree of Master of Science Graduate department of Laboratory Medicine and Pathobiology University of Toronto @ Copyright by Suuna Rosic-Kablar, 1999
Transcript
Page 1: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Studies of the Immune System of HOX11 Transgenic Mice for Lymphoma Immunotherapy

Suzana Rosic-Kablar, M.D.

A thesis subrnitted in conformation with the requiremenb for the degree of Master of Science

Graduate department of Laboratory Medicine and Pathobiology University of Toronto

@ Copyright by Suuna Rosic-Kablar, 1999

Page 2: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

National Library Bibliothèque nationale du Canada

Acquisitions and Acquisitions et Bibliographii Services senrices bibliographiques 395 Wellington Street 395. rue Weilingtm OnawaûN KlAûN4 OüawaON U l A W Canada Canada

The author has granted a non- exclusive licence allowing the National Library of Canada to reproduce, loan, distriiute or sell copies of this thesis in microforni, paper or electronic formats.

The author retains ownership of the copyright in this thesis. Neither the thesis nor substantial extracts fiom it may 'De printed or otherwise reproduced without the author's permission.

L'auteur a accordé une licence non exclusive permettant à la Bibliothèque nationale du Canada de reproduire, prêter, distribuer OU

vendre des copies de cette thèse sous la fome de microfiche/film, de reproduction sur papier ou sur format électronique.

L'auteur conserve la propriété du droit d'auteur qui protège cette thèse. Ni la thèse ni des extraits substantiels de celle-ci ne doivent être imprimés ou autrement reproduits sans son autorisation.

Page 3: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Studies of the Immune System of HOX11 Transgenic Mice for Lymphorna

lrnmunotherapy

A thesis submitted in conformation with the requirernents for the degree of Master of Science

Graduate department of Laboratory Medicine and Pathobiology University of Toronto

1999

Suzana Rosic-Kablar, M.D.

Abstract

We assessed the applicability of dendntic (DC) cell based

immunotherapeutic protocols for the treatment of B cell lymphoma, using the

HOXI1 transgenic mouse model. These mice develop large B cell lyrnphomas in

the spleen. Towards this goal, we analyzed dendritic and T cells from HOX1 1

mice. Results indicate that HOX11 DCs express normal levels of ceil surface

molecules and have normal ability to present foreign antigens, while T cells show

normal proliferative response to mitogenic stimulation, foreign MHC and OVA

protein. In addition, HOXI1 derived peptides or lymphoma cell lysates are

sufficiently immunogenic to induce specific immune responses in naïve mice.

However, HOXl1 derived T cells do not show induction of immune responses.

Our results suggest that lymphoma development in HOXll rnice is associated

with the toleration of lymphoma antigen specific T cells. making HOXll

transgenic mice a good model ta develop DC-based immunotherapeutic

protocols for the treatment of nonfollicular lymphomas.

Page 4: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

ACKNOWLEDGMENTS

I would Iike to thank my supervison, Drs. lan Dube and Margaret Hough for

being patient, supportive and encouraging throughout my studies.

Dr. Dubé believed in me and gave me the opportunity to be a member of his

research team. He provided a great environment in which I could leam and I

considered myself lucky to be a part of it.

I had the opportunity to know Dr. Hough as a supewisor and a friend.

Margaret has continually challenged me throughout this project with intriguing

questions and has provided me with valuable advice and direction in both work

and life.

My appreciation also goes to Dr. Serge Jothy who provided me with important

feedback at my cornmittee meetings and always reminded me how wonderful

science is, even when baniers are faœd.

Thanks to my committee members Drs. Charles Catzavelos, Corrinne Lobé

and David Spaner for providing adviœ and guidance du ring my graduate studies.

My gratitude also goes to al1 rnembers of the Dubé research team who were

always ready to provide moral and technical support. In this respect I would

especially like to thank Dr. Carolyn Lutzko, Kin Chan and Renita Yap.

Thanks to Drs. Marciano Reis and Megan Lim for sharing their knowledge in

hernopoiesis and for the hours spent in reviewing the histopathology slides.

Thanks to the members of Sunnybrook Animal Facility for technical support

and assistance with animal husbandry.

A special thanks goes to Dr. Barbara Guinn who introduced me to many of the

challenging techniques that I used in my project and turned our relationship into a

great friendship.

My special gratitude goes to my mother for her love and support. She has

always encouraged hard work and stood behind any of my decisions.

Finally, but most importantly, I would like to thank rny husband, Nijaz and my

son, Adi who believed in me and gave me the support and tirne I needed to

complete this degree. Thanks is such a small word considering how much you

deserve.

Page 5: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

4

Table of Contents

CHAPTER 1 1

LNTRODUCTION

1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN

1.2 MALIGNANT HEMATOPOIESIS

1.3 NEOPLA~MS OF m L~LMUNE SYSTEM

1.4 NON-HODGKM L'YWPHOMA

1.4. I Developmenz and Classtjkation

1-42 Incidence and Etiology

1.4 3 Trearment and Prognosii of NhZ

1.5 TUMOR IMMUNOLOGY

I . 5 I Tumor anrigens

1-52 me Immune System 's Rote in Tumorigeneiris

15.2.1 B Cells and Antibody-Dependent Killing

1 -5.2.2 T Ce11 Mediated Cytotoxicity

1.5.3 Mechankm by Which Tumor Ce Ils Escape an Immune Response

1.6 ~ ~ U N O T H E R A P Y FOR NON-HODGKM LYMPKOMA (NHL)

1.6.1 Eady Approaches of Lymphoma Immun0 fherapy

1.6.2 Crarenr Concepts of Lymphoma Zinmunotherapy

1.7 DENDRITIC CELLS AS PROFESSIONAL ANTIGEN P R E S ~ G CELLS

1.7.1 *gin and Generation of Cuitwed De&-tic Cells

1.7.2. Rule of Denilritic Celk in T cell Activation

1.8 D m m c CELLS AND OR hm-mrw

1.8.1 Denchitic Cell Vaccination in Murine ModeIs: Preclinical Sludies

1.8.2 Dendriric Cell Vaccination for Human Cancer: Clinical Trrals

1.8.2.1 Non-Hodgkin Lymphoma

1.8.2.2 Malignant Melanoma B.

1.8.2.3 Multiple *elorna

'1.8.2.4 Ofher Cancers

1.8.3 F u m e Prospects for DC Based Cancer Vaccines

1 -9 HOX 1 1 TRANSGENK MICE

Page 6: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

1.9.1 Homeobox Genes in Hematopoiesis and Hematologic Malignancies

1.9.2 Production of HOX1 1 Trmgenic Mice

1.9.3 Lymphoma Developrnenr In HOXl 2 Mce

1.10 SWY RATIONALE

1.1 1 HYPoTHESIS

1.12 OBJEC~VES

CHAPTER 2 48

~ ~ T E R U L S AND METHODS

2.1 ANIMALS

2.2 REAGENTS USED FOR TKE ~ O L A T I O N AND C U L T U ~ G OF DCS AND T CELLS

2.2.1 Growth Factors

2.2.2 Collagenase D

2.2.3 Bovine Serum Albumine (BSA)

2.2.4 Preparation of Steriked Nylon Wool Columrts

2.3 bfATERIALS USED FOR MIXED LYMPHOCYE W ~ O N W R ) AND CYTOTOXIC T

LYMPHOCYTE (Cil) ASSAY

2.3.1 Cell Lines Used ai^ Targets in CTL Assqv

2.3.2 Production of h vitro Transcribed (m RNA

2.3.3 Preparation of Tumor Lysate

2.4 GENERATION OF DC CULTURES

2-41 Dendritic Cell I.solationfiom Spleen

2.4.1.1 Density gradient centrifugation using Bovine Senim Albumin @SA)

2.4.1.2 Separation using MACS CD1 l c MicroBeads

2.4.2 Dendritic Cell Isolation fiom Bone Marrow

2.5 DENDRlIK CELL SURFACE PHENoTYPE CHARACIFRIZA~ON

2 .3 1 Flow Cytomeiric Anabsis of Spleen and Bone M m o w Dendritic CelLs

2.6 T CELL ISOLATION AND ENRICHMENT

2.6.2 Isolation of T celkfi.orn Peripheral Blood

2.6.2 Isolation of T Cells fiom Spleen and Thymus

2.6.2.1 Separation with MACS System

2.6.2.2 Isolation with Nylon WooI Columns

2.7 OPT[MIZATION OF MIXED LYMPHOCYTE REACTION (MLR)

Page 7: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

2.7.1 Optirniration of T Cell Prolijeraiive Respowes

2.8 FUNCTIONAL STUDiES USMG HOXI1 DWED DCS

3 8.1 Presenfarion of Foreip A2HC by HOXl l Derived DCs

2-82 Presenfarion of O VA Protein by HUXl l Derived DCs

2.9 FUNCTIONAL STUDIES USMG HOX11 DERIVED T CELLS

2.9.1 T Cell Prolferution Assay

2.9.2 T Cell Aesponses tu Foreign MHC Anrigens

2.9.3 T Cefl Responses ro O VA Prorein

2.10 ~WUREMENT OF CYTOTOXIC T LYMPHOCYTE (CTL) ACTIV~TY

2.1 O. 1 In Vitro CTL

3 10.2 In Vivo CTL

2.1 1 ASSESSMENT OF I M M U N O G E N E I C ~ OF HOXI 1 DENVED Amx~ms 2.1 1. 1 Induction of CTL Response ro in Vitro Tramcribed (rm HOXI 1 RNA

2.11.2 Induction of CTL Response to HOXI1 Tumor Lysutes

RESULTS

3.1 LDENTTFICATION OF HOXl 1 TP.SGEN1C MCE

3.2 O P ~ Z A T I O N OF MIXED LYMPHOCYTE REAC~ON (MLR)

3.3 CHARA~RIZATION OF HOX11 TRANSGENIC M m DERIVED D E N D ~ C CELLS

3.3.1 ESTABLISHMENT OF DENDRITIC CEU CULWRES

3.3.2 Phenotype Characterïzation

3- 3.3 HOXI i Derived DCs: Presenraiion of Foreign MHC

3.3.4 HOXI i Derived DCs: Presentaiion of OVA Prolein

3.4 CHARACTERIZATION OF HOXI I TRANSGEMC MICE DEWED T CELLS

3 - 4 1 Prolijeration Assay (PNA Stimulation)

3.42 T Cell Prol~erative Responses to Foreign MHC

3.43 T Ceil Proliferaiïve Responses to OVA Protein

3 -5 MEASUREMENT OF CWOTOXIC T LYMPHOCYTE (Cm) A c w m

3.5.1 In Viîro CTL Assay

3.22 in vivo CTL Assay

3 -6 AsSESSMENT OF IMMtMoGENEICITY OF HOX 1 1

3.6.1 T CeZi Prof$erative Responses upon the Stimulaion with IVT RNA

Page 8: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

3.6.2 Inducrion of Primary CTL Respome zo XVTNOX'l RNA in Nafie Mice 114

3.6.3 Inducfion of CTL Response u p n St i~~dat ion with Timor Lysares 120

CHAPTER 4 124

0

D~scussxo~ AND FUTURE EXPWUMENTS

vii

Page 9: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

List of Figures

Figure 1 : PCR analysis of HOX11 transgenic and nontransgenic mice .............. ... ... 74

Figure 2: Optimization of MLR ........................................................................ ïï

Figure 3: Flow cytometric analysis of HOXl1 and nontransgenic splenic DCs ........... 81

Figure 4: Flow cytometric analysis of HOXI 1 and nontransgenic bone marrow

Derived DCs .................................................................................. 82

Figure 5: Presentation of foreign MHC by HOXI1 derived DCs ............................. 86

Figure 6: HOXI1 derived DCs presenting OVA protein ........................................ 88

Figure 7: HOXi 1 derived T cell proliferation assay (PHA) ..................................... 91

Figure 8: HOX11 derived T cell proliferative responses to foreign MHC ................... 94

Figure 9: HOX11 derived T cell proliferative responses to DOTAP-OVA pulsed

DCs ............................................................................................. 96

Figure 10: ln vitro CTL assay using OVA pulsed DCs ........................................... 99

Figure 11 : In vivo CTL assay using OVA protein pulsed DCs ............................... 102

Figure 12: MLR stimulated with IVT HOXI 1 RNA pulsed DCs used for immunization . . ....................................................................................... ln VIVO. 105

........... Figure 1 3: CTL assay after immunization with IVT HOX11 RNA pulsed DCs 108

............ Figure 14: In vitro CTL assay using DCs pulsed with lymphoma cell lysates 113

Page 10: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

List of Illustrations

...... Illustration 1 : Tissue sections from HOX11 transgenic mice showing lymphorna -42

Illustration 2: HOX11 derived DCs in culture .................................................... -81

Illustration 3: In vitro CTL assay using HOX11 transgenic mice dedved DCs pulsed

....................................................................... with OVA protein 102

Illustration 4: In vitro restimulation of splenocytes isolated from HOX11 transgenic.

nontransgenic and C57BU6 after irnmunization ............................... 116

Page 11: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

List of Tables

Table 1 : Nurnber of DCs obtained from spleens of HOX11 transgenic mice ............... 80

Table 2: Nurnber of HOXl 1 bone manow derived DCs ......................................... 81

Table 3: Flow cytometric analysis of HOXl1 and nontransgenic spleen derived DCs.83

Table 4: Flow cytometric analysis of HOX11 and nontransgenic bone marrow

denved DCs .................................................................................... -87

Table 5: In vitro CTL assay using OVA protein pulsed DCs .................................. 105

Table 6: In vivo CTL using DC pulsed with OVA ................................................. 109

..... .............. Table 7: In vivo CTL assay using DCs pulsed with IVT HOX11 RNA ... 115

Table 8: In vjtm CTL assay using DCs pulsed with lymphoma cell lysates .............. .121

Page 12: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Chapter 1

ln traduction

1.1 Hematopoietic Cell Origin, Growth and Differentiation

Hematopoiesis is a process through which blood cells grow, divide and

differentiatel. All ceIl types are derived from a pool of hematopoietic stem œlls

(HSCs), which reside in the bone manow and have the unique ability, under

the appropriate conditions, to give rise to al1 of the different mature blood cells.

As the cells differentiate, their capacity for replication and self-renewal

declines. Thus, the less differentiated cells in a given pathway are rare but

replicate actively whereas the mature cells are more numerous but generally

are mitotically inert. Some daughter cells remain as HSCs (self-renewing cells),

while othen commit to any one of several alternative differentiation pathways

(myelo-erythroid progenitors and lymphoid progenitor cells) that lead to the

production of three general classes of cells: (1) erythrocytes, responsible for

oxygen transportation; and (2) myeloid and (3) lyrnphoid cells, both critical to

host defense.

The mature cells of the myeloid lineage include neutrophils, monocytes,

mast cells, eosinophils, basophils and megakaryocytes. Mature cells of the

lymphocyte lineage include B lymphocytes, T lymphocytes and natural killer

(NU) cells. B cells express immunoglobulin (lg) receptors, which recognize

soluble antigens whereas T cells express T cell receptor FCR) molecules that

recognize antigenic peptides in the context of major histocompatibility

molecules on the surface of antigen presenting cells. Consequer!tly, B and T

Page 13: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

lymphocytes. with other blood cells. play important roles in host immune

responses and in the elimination of foreign pathogens.

1.2 Malignant Hernatopoiesis

Vast numbers of mature blood cells are produced daily in the

hematopoietic system. Hematopoiesis is precisely controlled at several levels:

(1) the maintenance of a lifelang pool of HSCs; (2) regulation of cornmhent,

proliferation and differentiation of cells at al1 stages of each hematopoietic

pathway; and (3) modulation of the activities of each pathway in response to

physiological demands. Normal hematopoietic cell activities are coordinated so

that the rate of loss due to natural death of mature differentiated cells closely

approxirnates the rate of appearance of new cells from the less mature

proliferating cell pool*. The transformation of a normal hematopoietic progenitor

ce11 to one with malignant potential can result from a variety of causes. It may

be induced by chemical. physical and viral carcinogens or may occur

spontaneously by random mutation or gene rearrangements. For example, the

mechanisms by which chemical carcinogens induce neoplastic transformation

predominantly reflect the mutagenic adivity of the carcinogens? Physical

carcinogens, such as radioactivity or ultraviolet radiation. often induce damage

to cellular DNA resulting in a variety of cancers of epithelial cell origin'. Viral

oncogenesis is of particular interest because several human DNA viruses have

been found to contain potential onwgenes that have been associated with the

development of malignancies. These include links between Epstein-Ban virus

Page 14: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

(EBV) and Burkitt's lymphoma and some forms of Hodgkin's disease, and

human papillomavirus and skin cancer. In addition to DNA viruses, a class of

human retroviruses. the human T cell Ieukemia viruses (HTLV) are responsible

for the development of a subset of T cell leukemias. Another important

oncogenic retrovirus is human immunodeficiency virus (HIV). which is

responsible for the development of acquired irnmunodeficiency syndrome

(AIDS), a disease that among other symptoms. makes its vidims susceptible to

nume rous cancers inciuding non-Hodgkin's lymp homa (NHL) and Kaposi's

sarcoma (KS).

Advances in molecular biology have provided the tools to better

understand the essential role of many cellular oncogenes in normal growth and

development, and in the transformation of normal cells into malignant cells.

Proteins expressed by advated oncogenes are involved in the process of

transformation. Oncogene activation may occur by a variety of mechanisms

including gene amplification, mutation. and chrornosomal translocation. Gene

translocations are frequently observed in the malignant cells of patients with

leukemia and lymphoma. The most commonly obseived translocation in 6 cell

tumon places the c-MYC oncogene next to the immunoglobulin gene locisI6.

Aberrant MYC expression can lead to the development of malignant tumors,

presumably by interfering with the normal control of cell g r m . These

translocations are seen in over 90% of endemic and sporadic Burkitt's

lymphomas. T lymphocyte derived tumors frequently contain translocations

involving the T cell receptor gene loci. In pathologic conditions resuiting from

Page 15: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

oncogene activation, an individual cell may acquire the potential to produce

daughter cells that proliferate independently of extemal growth signals and this

rnay represent the basis of a malignant proœss? Such transformed cells rnay

be less sensitive to normal cell death pathways and may also fait to respond to

the regulatory signals responsible for normal growth and tissue repair. The

additional accumulation of mutations within the expanding monoclonal

populations often results in genotypic and phenotypic heterogeneity. Genomic

abnormalities may also eventually result in invasive growth through normal

tissue boundaries and metastasis to distant organs. following entry into blood

and lymph channels.

The immunologic responses of the host to the growth of malignant cells

are based on the host's immune system and the way in which the host defense

is triggered to promote tumor eradication. Immune rejection of malignant cells

is often difficult to accomplish because tumor cells have many similarities to

normal cells, despite often exhibiting an abnormal ability to proliferate, an ability

to spread throughout the host, and capacities to interfere with normal organ

functions.

1.3 Neoplasms of the lmmune System

Neoplasms of the hematopoietic system are cancers that directly affect

the host immune defense. They are similar to other neoplasms in many

respects, but considering the crucial role of the host immune system, some

Page 16: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

notable differenœs occur. First. sinœ neoplastic lymphocytes may circulate in

the penpheral blood and lymphatic systems. like their non-neoplastic

counterparts, many lymp hoid neoplasms are widely disseminated at diagnosis.

A second feature of neoplasms of the immune system is that they often retain

some functional characteristics of normal cells. For example. neoplastic T cells

may secrete cytokines and neoplastic plasma cells usually secrete

immunoglobulins. but generally the functional propertïes of such transformecl

cells do not occur in response to physiological stimuli. These cellular functions

become autonomous. often contributing to further tumor development and

clonal expansion.

Neoplasms of the hematopoietic system are classified into leukemias

and lymphomas. The leukemias are hernatologic neoplasms in which rnalignant

cells are present in the bone marrow and the blood, whereas lymphomas are

localized proliferations of lymphoid cells forrning a solid tissue mas. However,

many hematologic neoplasms show both patterns, to varying degrees, making

distinctions between leukemia and lymphoma sometimes rather difficult in

clinical practice. Neoplastic cell classification and the establishment of a correct

diagnosis is important for the selection of the appropriate therapy.

1.4 Non-Hodgkin Lymphoma

7.4.1 Development and Classfication

Lymphomas are divided into Hodgkin's disease and non-Hodgkin

lymphoma. NHLs make up about 5% of al1 cancers and 85% of al1 lymphomas.

Page 17: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

In 1832. Thomas Hodgkin first described the neoplastic transformations of cells

that reside predominantly within lymphoid tissues. The f o m he described is

now called Hodgkin's disease. Despite their diversity, al1 of the other malignant

lymphomas are refened to as non-Hodgkin's lymphomas (NHLs). Primary sites

of NHL growth are the organs of the immune system such as lymph nodes,

spleen. tonsils and thymus. The stomach, small intestine and skin may also be

affected. The development of lyrnphomas reflects in large part the relatively

unrestrained growth of lyrnphoid cells that locally invade and disrupt normal

tissue as well as metastasize and growth in distant organs. Lymphoid

neoplasrns can be divided into 6 or T cell in origin. Furthemore. since normal

lymphocytes go through several discrete stages of development, lymphoid

malignances may arise from cells arrested at a particular stage of

differentiation? Consequently a cell population that has undergone malignant

proliferation often shows a combination of surface markers that are known to

arise during discrete stage of T or 6 lymphopoiesis1o.

Appropriate classification is of significant importance because of the

multiple subgroups that are associated with distinct clinical outcornes. A variety

of systems have been used to classify NHL subtypes according to their

morp holog ical appearance, p henotype and clinical behavior of the disease. The

Rappaport classification is based on assessrnent of the overall pattern of lymph

node architecture as well as the cytological classification of the neoplastic cell

subdividing NHL into diffuse and follicular lyrnphomasll. In follicular

lymphomas the neoplastic cells appear to recapitulate normal lymphoid

Page 18: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

germinal centers while in difise types normal cortical and paracortical lymph

node architecture is largely affeded. The International Working Formulation,

developed in 1982. defined 10 major subtypes of lyrnphoma, categorized as

low, intemediate or high grade. More recently, in 1994, the International

Lyrnphoma Study Group developed the Revised European-American

Lyrnphoma (REAL) classificationl2. It describes several lymphoma types with

distinct clinical and morphological features and defines the most important

immunophenotypic, cytogenetic, and molecular features of each.

Currently, lymp homas are graded as aggressive (intemediate and hig h

grade) and indolent tumors (low-grade lymphomas). Aggressive lymphomas

are generally large cell lymphomas with rapid cell division and rapid

progression of d isease. Indolent lymphomas are small cell lymphomas and,

although patients may be asymptomatic for prolonged periods, these diseases

usually progress to an acute phase-

1.4.2 Incidence and Etiology

The number of new cases of NHL in Canada in 1999 is estimated to be

5,800. Based on the American Cancer Society statistics, NHLs are the second

leading cause of cancer deaths in patients aged 15 to 34 and the third leading

cause of cancer death in patients aged 35 to 54. Although childhood cancer is

rare, NHL is the third most common pediatnc cancer and accounts for 6

percent of childhood cancers. An increase in the incidence of NHL between

1973 and 1987 was larger than the increase during that period for any other

Page 19: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

major cancer except rnelanoma and lung cancer? Eight to 27 percent of the

increase was associated with the development of NHL in association with

AIDS. For children with AIDS. the rate of developing NHL is about ten times

higher than that of adults with AIDS.

The etiology of most types of NHLs is unknown. However, several

factors suggest possible causal relationships. In addition to chernical and viral

etiologies, NHL has been obsewed as a late complication of prior

chemotherapy and radiation therapy for unrelated disorders. Patients with

Hodgkin's disease who were treated with conventional therapy exhibit an

increased risk of developing secondary large-cell lymphornas'4. The incidence

of NHL is increased nearly 100-fold for patients undergoing organ

transplantation necessitating chronic imrnunosuppression, and it is greatest

during the first year posttransplantl? Diseases of inherited and acquired

irnmunodeficiency as well as autoimmune diseases are associated with an

increased incidence of lymphoma, indicating that immune deregulation may

significantly contribute to the pathogenesis of lymphoma16.

7.4.3 Treatmenf and Prognosk of NHL

The treatrnent of lymphoma is detenined according to the grade or

claçsificatic?n of the disease at the time of diagnossq? Non-Hodgkin

lymphomas often demand multidisciplinary treatment regimes to achieve

optimal cure rates's. Aggressive lymphomas are rapidly growing tumors thus

are generally treated using aggressive chemotherapyl? This results in a high

Page 20: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

percentage of durable, long-temi disease-free survival and with approximately

45 percent of patients being cured using a variety of chemotherapeutic

regimens. Unforhinately, a significant nurnber of patients with aggressive

lymphoma either fail to respond or eventually have disease relapse. Prognosis

then is poor and almost al1 patients in this category can be expected to die of

progressive lymphoma.

lndolent lymphomas are slow growing tumors and patients diagnosed

with this type of cancer can have median survivals of 7.5 to 9 years. For

patients with early stage disease. especially those under 40 years of age, local

radiation is the standard treatrnent? A standard treatment for advanced

indolent lymphomas has not yet k e n determined. Chemotherapy with one

drug or combinations of dnigs is effective in treating recurrences. Although

generally regarded as an indolent disease, slow growing tumors are

progressive and ultimately fatal. lndolent lymphomas that progress into more

aggressive types are generally more difficult to treat with standard therapies

than lymphomas that are aggressive at diagnosis.

Relapse following initial therapy confers a poor outlook with overall

survival of less than I O percent? The principal curative approach involves

aggressive chemotherapy in combination with radiation therapy. Autologous

bone marrow transplantation has also been considered as a form of primary

treatment. Such treatrnent requires further validation before it is acœpted as

standard practice21. In addition, allogeneic bone manow transplantation has

been performed on a small number of patients with resistant disease.

Page 21: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Unfortunately, this procedure is restricted due to the need for a matched donor

and hig h treatrnent related mortalip.

1.5 Tumor lmmunology

The discussion of tumor immunology will be categorized as follows: (1)

antigenic properties of transformed cells, (2) consequences of the growth of

malignant cells, (3) host immune responses to tumor cells, (4) tumor

immunosurveillance and (5) modulation of the immune system to recognize

and promote tumor eradication.

7 - 5 7 Tumor antigens

Malignant cells may express tumor antigens that can be recagnized by T

cells or antibodies. These fall into two major categories, which are tumor

specific antigens and turnor associated antigens. Tumor specific antigens

(TSA) are found only on tumor cells and therefore represent ideal targets for an

irnmunologic attack. Although the existence of TSA has been refened through

studies in animal models, their identification in human tumors has been ditficult

and thus the existence of these antigens remains a debated issue. In one of

the early studies, Prehn et al. induœd sarcomas in inbred strains of mice. by

painting mouse skin with the chernical carcinogen. methylcholanthren*.

Arising tumon were transplanted inducing growth and eventually killing the

new host, demonstrating the existence of antigens expressed exclusively by

tumor cells. In contrast, the injection of tumor cells into the original host

Page 22: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

resulted in a specific immunologic rejection of the tumor, indicating the

presence of activated tumor antigen specific T cells. Altematively, irndiated

tumor cells used to immunize secondary syngeneic mice induced immune

responses that protected hosts from additional tumor transplants. One potential

example of a TSA found in human malignancy may be the specific lg idiotype

expressed by 6 cell lymphoma (see below pg. 22)

Tumor associated antigens (TAA) are found on both tumor cells and

normal cells. and rnost TAA identified to date represent differentiation antigens.

An example of the diversity of TAAs is seen in the phenotype of human

melanoma24. Melanome cells express highly immunogenic surface proteins

that can stimulate immune responses ri, vivo. Several of the melanoma

antigens isolated to date are also expressed within normal tissues. MAGE-1 is

expressed in testis, whereas tyrosinase, MART-1 and gplOO are present in

rnelanocytes and retniel pigment epithelium. When cancers from different sites

are studied, the expression of tyrosinase, MART-1 and gp100 appear lirnited to

melanoma, in contrast to MAGE-1. -2, -3 that are found in a variety of tumors24.

The best-characterized human TAAs are the on cofetal antigens 25.26.

These antigens are expressed during embryogenesis but are absent or very

difFicult to detect in normal adult tissue. The prototype antigen is

carcinoernbryonic antigen (CEA), which is greatly increased on human colon

cancers, breast cancers and non-small-cell lung can~ers2~. Assays for senim

CEA are used to monitor the spread of carcinoma or its recurrence after

Page 23: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

primary treatment28-30. Moreover, human studies have suggested that antî-

turnor T cell immunity can be elicited to tumors expressing CE@.

Other types of TAAs are lymphoid-specific or differentiation antigens that

are expressed in normal cells reflecting distinct stages of differentiation. These

lymphoma-associated antigens may be aberrantly expressed on some tumor

cells. For example, malignant lymphomas ansing during a specific stage of 6

cell development may be diagnosed as derived from the 6 cell lineage by the

detection of surface markers characteristic for nomal pre-B cells. such as

CD10. Tumors arising from more mature B cells are characterired by the

presence of surface immunoglobulin3~.

1.5.2 The Immune System's Role In Tumongenesis

The concept of host immune surveillance, in which the immune system

provides the framework for recognition and eradication of developing

immunogenic tumor cells was first proposed by F. M. Bumet32. The immune

surveillance hypothesis has since stimulated rnany experimental studies al1

aimed at detennining whether the immune system has a general role in

controlling tumor growth. Under this model. al1 of the effector components of

the immune system potentially contribute to the eradication of tumor cells.

However. specific immune responses are classified into two types: humoral

and cellular irnmunity, rnediated by B and T cells. respecüvely.

Page 24: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

1 - 5.2. ? B Cells and Antibody-Dependent Killriig

The development of monoclonal antibody technology by Kohler and

Milstein in 1975 resulted in the generation of reagents pemitting precise

characterization of ceIl surface antigens associated with B cell

differentiation33.34. The initial stages of B cell differentiation are independent of

antigens, but subsequent difFerentiation requires antigens and proœeds

optimally in the presence of T cellderived factors. Mature 6 cells eventually

arise that synthesize and express immunoglobulins on their surface. After

interacting with antigens and T cells, B cells clones differentiate into plasma

cells that produce multiple copies of a single antibody with the ability to bind a

particular antigen35-36.

There are two major mechanisms by which antibodies may mediate

tumor cell lysis. The first involves complernent-dependent IgG and IgM

antibodies37. These antibodies bind to antigenic sites on target cells and

promote attachment of complement components that create pores in the

target's cell membrane, resulting eventually in cell death due to loss of osmotic

and structural integrity. An alternative mechanism is antibodydependent

cellular cytotoxicity (ADCC) in which antibodies, usually of the IgG class, act as

bridges between tumor targets and effector cells that express Fc receptors,

particularly natural killer (NK) cells, macrophages and granulocytes. ADCC is a

more efficient lytic mechanism than cornplement-mediated cytotoxicity,

requiring fewer antibody molecules per cell for a cytotoxic response37.

Page 25: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Antibodies that bind to human TAA have been found in sera from

patients with various cancers, such as melanorna and renal ceIl cancer?

Approximately 10 percent of patients with melanorna have antibodies that bind

to gp75, which is a melanosorne-rnembraneassociated protein found in both

melanomas and normal rnelanocytes38. Antibodies may also be fomied against

antigens induced by EBV. herpes sirnplex virus 2 (HSV-2) and papillomavirus.

In addition to their role as antibody producing cells. B cells also play a

role in the immune system as antigen presenting cells (APC) in a secondary

immune response. 6 cells can take up extracellular antigens via lg molecules

expressed on the cell surface. Sinœ B cells express both MHC class I and

class II molecules, these antigens can be processed and presented for the

activation of CD4 or CD8 T cells. However, they do not express costirnulatory

molecules required for the activation of naïve T cells, thus B cells can not

function as APC in a prirnary immune response.

7.5.2.2 T Cell Mediated Cytotoxicity

T lymphocyte progenitors arise in bone marrow, migrate to the thymus

where they proliferate and differentiate into mature CD4 and CD8 thymocytes.

Part of the maturation process involves selection of thymocytes expressing

functional T cell receptor ( K R ) that can interact with either MHC class I or

class II molecule but do not recognize self peptides. After selection, mature

thymocytes expressing productive TCR and either CD4 or CD8 CO-receptor

migrate from the thymus and circulate in the penphery as mature T cells. Over

Page 26: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

90 percent of thymocytes generated in the thymus are eliminated during the

maturation process. Mature T cells mediate cellular immune responses with the

specifïcity of interaction against different antigens being mediated by the T cell

receptor. In contrast to B cells, which can recognize antigens in the fluid phase,

T cells recognize antigenic peptides cornplexed to major histocornpatibility

complex (MHC) class I or class II molecules presented on the surface of

antigen presenting cellsJ?

T cells play important roles in host defenses against tumor and foreign

antigens (see illustration on pg.16). There are two T cell subsets: CD4 helper T

cells mediate their effect by the secretion of cytokines to acüvate other effector

cells, while CD8 cytotoxic T cells also secrete cytokines but primarily mediate

their effect by direct lysis of tumor cells4Oe41. Extracellular derived peptide

antigens are presented to CD4 T cells by antigen presenting cells in the context

of peptide-MHC dass II complexes. CD4 T cells differentiate into (wo types of

effector cells. TH1 cells produœ INF-y and 11-2 cytokines, which contribute to

the activation of macrophages and induction of IgG antibodies. TH2 c e k

secrete 11-4, 11-5 and IL-IO, which initiate humoral immune responses by

activating naïve antigen-specific B cells to produce IgM antibodies. CD8 T cells

recognize peptides derived from proteins that are processed within the

cytoplasm of the cells and are presented by MHC class I molecules. These T

cells differentiate into cytotoxic T cells that kill foreign antigen presenting cells.

The precise mechanism of T cell mediated cytotoxicity is not known, but

Page 27: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

cytotoxii

TNF recepto

Fas band

r

:as

virus-infected cell bacteria

macrophage

antigen-specific B cell --

O " 1 I F - i l F I L I l I L ( IL-. 1 Gmnzyrnes TNF-P TNF-a TN F-P IL-5 GM-CSF Fas ligand TNF-a CD40 ligand (IL-2) 11-1 0 CD40 ligand TGF-B

Fas ligand

CD4 and CD8 T cells in host defense: The three main types of effector T cells pro- duce distinct sets of molecules

Source: Imrnunobiology, Janeway CA and Travers P;Garland Publishing Inc, 1997

Page 28: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

perforins, lym p hotoxins, direct membrane interactions and induction of

apoptosis by Fas-Fas ligand pathway have been proposed4*+.

The role of CTLs in rejecüng tumor cells has been demonstrated in

numerous studies. For example, in the case of melanoma patients, antitumor

CTLs have been shown to recognize and lyse cells expressing the MEGA-1

TAA presented by HLA-Al'? Furthemore. in vitro studies have shown that

human sarcomas, renal cell cancers, breast cancers, ovarian cancers, head

and neck cancers as well as HTLV-I induced leukemias are lysed by T cells

that recognize TAA in the context of MHC antigens.

Other cell types, such as natural killer (NK) cells and macrophages, also

play significant roles in tumor immunology. NU cells represent a firçt Iine of host

defense against the proliferation of transforrned cells at both primary and

metastatic sites. They provide an efiector rnechanism recruited by T cells to

supplement specific antitumor responses. These cells are large granular

lymphocytes which have been shown by in vitro studies to possess the ability

to lyse certain tumor cell lines in vit#. Cytolysis by NK cells is mediated by

the release of cytotoxic factors and the use of perforins that puncture holes in

the target cells membraned? Like NK cells, macrophages are also important in

tumor immunology not only as potential effector cells to mediate tumor lysis but

also as antigen presenting cells to initiate antitumor immune responses. The

rnechanisms by which macrophages recognize tumor cells and mediate their

lysis are not defined, but activated macrophages bind to and lyse transfomied

Page 29: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

cells in marked preference to normal cells. Intercellular transfer of lysosomal

products, release of neutral proteinases, TNFu and 11-2 al1 rnay contnbute to

macrophage mediated cytotoxicity, depending on the macrophage-activating

factors (MAK) responsible for activation48.

1.5.3 Mechanisms by Which lurnor Celk Escape an Immune Response

Many potential mechanisms that rnay allow tumor cells to escape

immune rejection by the host have been identified49eso. One of the hypotheses

is the lack of antigen expression by tumor cells, because antigens rnay not

appear at the cell surface or rnay not be adequately processed or presented in

the context of appropriate MHC determinants. Although most cells within a

tumor express potentially immunogenic antigens, a subpopuIation of tumor

cells rnay lose expression of these antigensS1. This loss rnay result in a

reduction or elimination of tumor antigens that can be recognized by the

immune system resulting in the outgrowth of a variant subpopulation of tumor

cells that become the dominant population.

For T cells to recognize an antigen, it must be degraded into small

peptides that are transported to the endoplasmic reticulum and presented by

MHC class I molecules on the cell surface or via endosomes and presented by

class II molecules. Studies have shown that some tumor cells have defective

antigen processing rnachinery, with the result that class I MHC molecubs are

not loaded with peptides and transported to the cell surface5? In addition, MHC

expression rnay be down regulated and in such circumstances even potentially

Page 30: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

highly immunogenic tumor antigens may not be presented to the immune

s ysterns?

In addition to the first signal received via the TCR through its interaction

with MHC-peptide cornplex. T cells require a second signal. which is provided

by costimulatory molecules 87-1 and 87-2 (CD80 and CD86)54. These

molecules interact with CD28 molecules expressed on T cells inducing its full

activity. TCR ligation by peptides in the absence of the second signal results in

T cell anergy or a state of non-responsiveness. Since most tumor cells do not

express costimulatory molecules required for the activation of naïve T cells, it

has been hypothesized that tumor antigen specific T cells are rendered non-

responsive upon encountering TAA expressed on malignant ceIls5557.

There are many other mechanisms by which tumor cells may non-

specifically interfere with the induction of immunity in the host. Some tumors

can produce a vaMy of soluble factors such as transforrning growth factor4

(TGF-B) that can affect immune effectors. TGF-B has antiproliferative effeds

on a wide variety of cell types, including T and B lymphocytes. It also

suppresses the production of rnost lymphokines and reduces the cellular

expression of MHC class II mole cul es^. In addition to TGF-B. 11-10 may

prevent the difFerentiation of monocytes into dendntic cells or induce

suppression of MHC class II expression and consequent loss of the accessory

functions of macrophages and dendritic cells5? In fact, both TGF-13 and 11-10

are expressed by lymphoma c ~ I I s ~ O - ~ ~ .

Page 31: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

1.6 lmmunotherapy for Non-Hodgkin Lymphorna (NHL)

The majority of NHL patients are not cured with conventional therapy,

although signifiant advances have been made in the treatrnent of NHL62. Only

20-50 percent of patients with intermediate- and high-grade lyrnphoma

experience prolonged diseasefree survival and nearly al1 patients with low-

grade NHL relapse63. Based on the overall treatment results, rising incidence

rates that contribute to a slight increase in mortality rate and the reality that

chemotherapy cannot be further increased due to unacceptable toxicity , other

strategies for treatrnent of NHLs, including immunotherapy, have been

increasingly investigated. Cancer immunotherapy is a mode of cancer

treatment that stimulates the natural host immune system to destroy tumor

cells thereby mediating cancer regression, in contrast with conventional

therapies, which act by directly attacking, or killing tumor cells. A better

understanding of the factors involved in the induction of an immune response

and the biology of tumor progression have created new interest in

immunotherapy for the treatment of many malignancies including lymphoma. In

addition, technological advances that permit the isolation of lymphocyte

subpopulations, identification and purification of tumor antigens, growth of

selected antigen specific T cells, and amplification of immune responses with

cytokines have created new approaches for the development of

immunotherapeutic treatment of cancer.

Page 32: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

1.6.1 Early Approeches of Lymphoma lmmunotherapy

Early cancer immunotherapeutic approaches used recombinant

cytokines and immune celis, in particular lAK cells and TIL, to increase

immune responses. lAK cells are a phenotypically heterogeneous population

of cells with the major effector cells being NK cells. Tumor-infiltrating

lymphocytes (TILs) can be isolated directly from tumors and expanded in vitm

with 11-2. Murine studies showed that treatment of mice with high doses of 11-2

alone or in conjunction with LAK cells could mediate the regression of

established liver and lung metastasis, using a variety of tumor rnodels. These

studies fostered the development of human clinical protocols. In clinical trials

based on this approach, Ki vitro treatment of peripheral blood cells with 11-2

induces large numben of LAK cells but did not significantly increase specific

killing of autologous lymphorna cells when used as a vaccine in vivo64. ln

clinical trials involving TlLs, TlLs from more than 50 patients with rnalignant

melanoma were expanded in culture with IL-2. These in vitro expanded cells

appeared to have specific cytotoxic reactivity against autologous tumor cells

with tumor regression being reported in about thirty-eight percent of patients65.

In addition to LAK and TIL, treatment with monoclonal antibodies (mAbs)

directed against turnor surface antigens provided a rational therapeutic

approach for some types of cancers. For example, antibodies specific for CD20

rnolecule, which is expressed on B cells have been shown to produce clinical

responses when administered to B ceIl lymphorna patients who have previously

Page 33: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

failed chemotherapya6. Conjugation of cytotoxic drugs, toxins, or radioisotopes

to the antibody are other approaches used to deliver a lethal hit directly to

tumor cells without requiring the participation of host effector ce l ls~ . Studies

utilizing radiolabeled forrns of anti-CD20 have demonstrated potentially curative

effects and have also been tested for clinical uses? Evidence fmm other

cancers such as colon or breast cancer have also shown reduction in mortality

and support a rote of monoclonal antibodies in cancer treatment.

7.6.2 Current Concepts of Lymphoma lmmunotherapy

Current strategies for irnmunotherapy of 6 cell NHL are focused on

approaches to induce effective antilymphoma specific T ceIl mediated immune

responses. Two main strategies in B cell lymphoma immunotherapy are being

explored. First, since most B cell lymphomas are characterized by the clonal

expansion of a single B cell with a unique rearrangernent of both the heavy and

light immunoglobulin genes, their specific B cell receptors (idiotype, Id) are not

only clonal markers but also tumor specific antigens to which an immune

response might be directed68. Vaccination with either Id bound to keyhole

limpet hemacyanin (KLH) alone or in combination with GM-CSF has shown

efficacy against established tumors in murine models and to some extent. in

humans69-70. However, there are several disadvantages to this approach. For

example, it has been demonstrated that clonal expansion in low grade NHL

could potentially lead to loss of recognition by the effector cells introduced by

Id-vaccines if the antigen components are altered. In addition, production of Id-

Page 34: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

vaccines is highly labor intensive and cosUy, and thus would not be practical as

a general treatrnent for NHL6gS70.

A second strategy for B cell lymphoma immunotherapy is based on

knowledge that normal 6 cells do not express costirnulatory molecules and

therefore can not activate naïve T cells. However, 8 cells can function as APC

for the activation of experienced T cells. Consequently malignant B cells could

potentially function as APC, although clinically significant immune responses in

patients with B cell lymphoma are not readily detected. A second

immunotherapeutic strategy for the treatment of B cell NHL has attempted ta

enhance the ability of B lymphoma cells to function as APC by transfecting

malig nant cell Iines with 87 wstimulatory molecules required for the activation

of primary T cell responses71. In addition to transfection of primary B cell

lymphoma cells, it is possible to upregulate the expression of 67 molecules.

One approach for the induction of 87-1 and 8-2 molecules in normal and

malignant B cells is by cross-linking of CD40 molecules necessary for B cell

proliferation and activation72. This approach has the advantage that co-

stimulation and adhesion as well as antigen presentation is enhanced. CD40-

activated lymphoma celfs can be used to induce T cell response either as a

vaccine or for in vitro stimulation of T cells. These in vitro activated T cells are

capable of mediating effedor responses against the CWO-activated follicular

lymphoma. Once primed, these T cells should rewgnize the wild-type tumor

celI~73~74.

Page 35: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Several animal tumor model systems support the relevance of exploring

87 CO-stimulatory pathway in the generation of tumor specific immune

responses. In some experimental models an effective antitumor immune

response was initiated in animals irnmunized with tumor cells genetically

altered to express 87 molecules~=75. When 87 is transfected into a highly

malignant melanoma ceIl Iine and then implanted into an immunocompetent

mouse, tumon initially grew but then completely regressed over three to four

weeks. Further, these animals were protected against a subseqwnt

rechallenge with the parental cell line. However, expression of these molecules

has not proven to be sufficient in al1 tumor models76. In clinical trials, a breast

cancer cell line was transduced with B7 to vaccinate patients in combination

with GM-CSF as an adjuvantT

In addition to the above described approaches, transfection of various

cytokine genes such as GM-CSF or 11-2 that assist in host immune cell

recruitment and activation, into tumor cells has also been assessed with the

idea that providing cytokines to cytotoxic T cells would bypass the need for

CD4 helper T cells78.79.

1.7 Denditic Cells as Professional Antigen Presenting Cells

Studies carried out more than 20 year ago led to the identification of

dendritic cells (DC). Since DCs have been shown to be particularly effective

and potent APCs, their use in cancer immunotherapy protocols is emerging as

a viable alternative to radiation or chemotherapy. This new strategy of

Page 36: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

enhancing the recognition of tumor cells by the immune system is based on the

presentation of tumor antigens by dendritic cells80. Theoretically, autologous

dendritic cells presenting tumor antigens may be used as vaccines to trigger an

antitumor specific T cell response.

As professional antigen presenting celk (APC), dendntic cells (DC) are

critical for the initiation of ceIl-mediated immune responses. DCs express high

levels of MHC class I and class II molecuies. and therefore have the

exceptional ability to present class I and class II restricted peptides and

activate either CD8 or CD4 T cellsatla. They also express the additional

accessory molecules required for T cell activation, including CD40,

costirnulatory molecules B7- 1 /CD80 and B7-2/CD86, intercellular ad hesion

molecules I CAM4 /CDS, ICAM-3/CD50, Iyrnp hocyte function-associated

proteins LFA-IdCD1 1 a, LFA03/CD58 and Mac-VCDl 1 b, CD1 1 c and HSA

molecules56~83. DCs are specialized to take up, process and present antigen.

and have the capacity to stimulate naïve T cells as part of a primary immune

responseV

1.7.1 Ongin and Generation of Cultured Dendntic Cells

DCs were first recognized by Steinman and Cohn in 1973. They were

characterized on the basis of their morphology. low density and migratory

capacity. DCs circulate from pen'pheral tissues to lymphoid organs, where they

corne into contact with mature T cefls85. Mernbers of the DC family include

interstitial DCs which are found in mainly al1 tissues but are primarily

Page 37: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

concentrated along epithelial and body cavity surface, the Langerhan's cefk

(LCs) of the skin, and the interdigitating cells of the thymusm.

DCs differentiate from bone marrow derived CD34+ granulocyte-

macrophage precursor ce l l~8~. Immature DC precursors exit the bone manow

and circulate via the bloodstream to reach peripheral tissues. Immature DCs

are highly efficient at antigen uptake and proœssing but do not present these

processed peptides at the cell surface. Thus, at this early stage of

development, they are unable to activate T ~ells88~". Mature DCs migrate

through lymphatic vessels to T cell areas of lymph nodes, where they forrn

clusters with T cells, to maximize encounters with rare antigen reactive

clonesg0. DCs maturation results in the loss of their ability to uptake antigens,

but their capacity to present antigens and stimulate T cells is greatly increased.

Maturation also coincides with the upregulated expression of adhesion and CO-

stimulatory molecules. Ligation of CD40 on the surface of activated DCs by

CD40 ligand expressed by T cells induces further elevated expression of 87-1

and B7-2 molecules. In addition, DCs initiate the secretion of high levels of IL-

12, which supports the development of Tnl CD4 T cells and the maturation of

CTLs91. DCs also enhance the proliferation and maturation of B cells either

directly or by stimulating CD4 T cells to produce cytokinesgz.

In addition to DCs derived from CD34' myeloid precursors, lymphoid

derived DC subpopulations have also been identifiedg? Lymphoid DC

precursors have little or no capacity to fonn myeloid cells, indicating that they

are of lymphoid lineage rather than a myeloid-related lineage. The

Page 38: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

developrnent of thymic DCs and thymocytes is Iinked via a common precursor

at an early stage of thymocyte developmentw. Thymic DCs induce the death of

the developing, serreacting T cells, rather than presenting foreign antigens.

These DCs, as well as a subgroup of DCs in spleen and lymph nodes, express

some markers normally expressed on lymphoid cells.

DC cultures rnay be established using BM. peripheral blood and spleen

cells or early thymic precursor ceIl. DCs may be expanded in vitro by

supplementing spleen or bone marrow derived cell cultures with GM-CSF and

IL4, although lymphoid-related DCs do not require GM-CSFS3.95w% The

development of DCs in culture is rapid, with expanded populations reaching

DCs purity of more than 85%. Cultured DCs are similar to freshly isolated DCs

in their expression of surface antigens. their ability to stimulate allogeneic T

cells in vitro and to proœsses and present antigens.

7.7.2. Role of Dendntic Cells in T ce// Activation

Antigens that are captured by antigen presenting cells (APC) such as

dendritic cells (DC), become enclosed within membrane-lined vesicles,

undergo a series of alterations, which includes cleavage of antigens into

peptides by denaturation and proteolityc digestiong7(see illistration on pg.28)

The resulting peptides then associate with major histocompatibility complex

(MHC) proteins and are transported to the APC cell surface. There are two

different classes of MHC proteins, each of which is recognized by one of the

two major subpopulations of T lymphocytes. Class I MHC molecules are

expressed by

Page 39: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

for Ag "Or (TCR) / & ~

l MHC

C

Activation of the naive T cells in the proces of immune response: Interaction between APC and T cells

Adapted from Medical Immunology, Stites DP, Ten Al and Parslow TG, 1997

Page 40: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

virtually al1 somatic cell types and are used to present antigen to CD8 T cells.

Thus, almost any cell can present antigen to cytotoxic T cells and serve as the

target of a cytotoxic response. Class II MHC molecules are expressed by DCs,

macrophages and B cells and are necessary for antigen presentation to CD4-

helper T cellsg*. Since helper cell activation is important for immune responses,

class Il-bearing APCs play a crucial role in mediating such responses. T cells

express a number of adhesion molecules, such as leukocyte fundional

antigen-1 (LFA-1) and CD2 that bind ligands on the surface of DCs. and

stabilize T cells and DCs interactions.

Despite their complexity, the signals delivered by TCRs are not sufficient

to fully activate T cells. T cell activation requires a second signal delivered by

costimulatory molecules8J. CD28 is a costimulatory molecule expressed on al1

CD4 T cells and a number of CD8 T cells. CD28 binds the cell surface

molecule B7 expressed on DC, and together fully activate T cells to proliferate,

differentiate and perfonn their effector functionsg? Activation of helper T cells

leads to the production of cytokines that promote cellular and humoral immune

responses, whereas activation of tumor antigen specific T cells results in the

lysis of the tumor antigen-beanng ~ e l l s ~ ~ ~ 0 2 .

1.8 Dendritic Cells and Antitumor lmmunity

The rote of DCs in T cell activation and the demonstrated ability to

manipulate DCs to present tumor antigens has generated great interest in the

use of DCs as cancer vaccines and to their recent evaluation in pilot clinical

Page 41: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

trials. One of the most promising areas of DCs research has been the

development of OC-based strategies for enhancing immune responses against

tumors.

7.8.7 Dendritic Cell Vaccination in Murine Models: Preclinical Studies

The induction of anti-tumor immune responses by the injection of

antigen-loaded DCs has been studied extensively in animals. Antigen-loading

of DCs can be achieved through pulsing of DCs in vitro with protein or peptide,

tumor cell lysates, tumor-derived RNA ennched for tumor specific mRNA, or

transfection of DCs with cDNA encoding tumor antigensf03-107.

Protein loading of DCs is accomplished by CO-culturing of DC and protein

preparations in the presence of a lipid carrier such as DOTAPW The protein is

taken into the DC, processed intracellulary, and class I and class II restncted

peptide fragments presented at the DC surface, for the activation of CD4 and

CD8 T cells. DCs rnay also be pulsed with MHC class 1 or class II restricted

peptides to enhance either CD8 or CD4 T cell responseslo? Numerous studies

have clearly established the effectiveness of tumor antigen or peptide pulsed

DCs to induce potent antitumor immune responses and to provide protective

tumor immunity from a subsequent lethal tumor challenge104~108-110.

The approach of using tumor lysate-pulsed DCs offers the potential

advantage of increasing T cell immune response to tumor-associated antigens

that may not be obtained by pulsing DCs with single tumor peptideslo? Pulsing

DCs with tumor cell lysates avoids the problem of defining TAAs and facilitates

Page 42: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

the presentation of a large anay of antigens derived from a heterogeneous

population of tumor cells thus reducing the risk of %scape mutationsn. In a

recent study, Fields et al. demonstrated that immunization of healthy mice

subcutaneously (SC. ) with tumor cell lysate-pulsed DCs provided strong

protection against a subsequent challenge with a lethal dose of the viable

parental tumor cells and rnediated reduction in the number of experimentally

established pulmonary metastasesW

The amount and purity of tumor tissue used for the isolation of proteins,

peptides or turnor-lysates is often insufficient for vaccination. Recently

investigators have focused on discovering more effective methods of delivering

tumor antigens to DCs, not requiring knowledge of the relevant tumor peptide

sequences. A potential advantage of using RNA rather then proteins or

peptides as a source of tumor antigens is that sufficient amounts of antigen can

be generated from very small amounts of tumor tissue using PCR amplification

techniques. Boczkowski et al. demonstrated that rnunne DCs could be

efficientiy transfected with RNA encoding the chicken ovalbumin (OVA) antigen

and used as a potent APC for the induction of T cells mediated responses'o3.

In addition, the ability to transfect human DCs with CEA RNA and the induction

of CTL response with these DCs in vitro has been also rep0rtedlll1~~2. cDNAs

cloned in bacterial plasmids can also be used for the production of RNA. by in

vitro transcription, allowing the applicability of RNA-based vaccines to patients

with very small tumors. Another promising strategy for DC loading wlh tumor

antigens is direct transfection with cDNA libraries derived from tumor cellsfl3.

Page 43: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Although DCs pulsed with cDNA have been shown to be effective in reversing

tumor progression. the generation of cDNA expression libraries is labor-

intensive and therefore pulsing DCs with mRNA will likely prove to be more

advantageousl14. In addlion to the above strategies. the fusion of tumor celfs

to DCs has also proven to be an efficient method for presenting tumor antigens

and inducing an anti tumor immune respon~e115*'~6.

Thus. various shidies h mouse tumor rnodels suggest that DCs pulsed

with tumor antigens in vitm and administrated to murine rnodels may initiate an

effective antitumor immune response. Such immunity protects animais against

inoculation with tumor cells and often causes suppression of tumor cell growth

and even regression of established tumor~105~117*118.

7.8.2 Dendntic Ce// Vaccination for Human Cancer Clinical Trials

Based on the results of animal studies, several investigators have

speculated that the administration of manipulated DCs may also be effective in

generating cellular immunity to tumors in humansllg. The development of

methods for obtaining large numbers of human DCs has made testing this

hypothesis feasible. Clinical trials of OC vaccination are based on two general

methods including purification of immature DCs precunon from peripheral

blood, and in vitm differentiation of DCs from peripheral blood monocytes or

CD34+ hematopoietic progenitor cells120- The potential of isolated human DCs

to prime tumor antigen specific T cells in vitm has been reported repeatedfy.

Page 44: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

7.8.2.1 Non-Hodgkin Lymphoma

Hsu et al. first reported the intravenous administration of autologous DCs

pulsed rii vitro with idiotypic proîeins into patients for the treatrnent of NHL121.

In that study, peripheral bloodderived DCs from four patients with low-grade

lymphoma were pulsed with KLH or tumorderived idiotype protein and infused

intravenously. Each patient received several DC infusions with a final given five

to six months later. The treatment approach resutted in cellular protiferative

responses specific to the patient's own idiotypic protein, as welf as humoral and

cellular responses to the control KLH protein. In addition, clinical responses

were also found- Complete remission of pericardial and periaortic masses were

seen in one patient, who remained in remission for 42 months. In the case of

another patient, polymerase chain reaction (PCR) analysis of blood and bone

marrow for tumor specific DNA became negative and complete remission was

found for more than 36 months after vaccination. The two other patients

exhibited stabilization of disease. Wih respect to safety of idiotype-pulsed DC

vaccinations in patients with NHL, no delay or long-terni toxicity was identified

in this study.

A similar approach being evaluated in clinical studies. demonstrated that

vaccination with DCs pulsed with idiotype protein directly coupled to KLH may

improve immunogenicity and tumor protection in a murine lymphorna model

when compared with vaccination using idiotype-pulsed DCsl*.

Page 45: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

1.8.2.2 Malignant Melanoma

The characterizaüon of melanoma antigens recognized by T cells lead to

the initiation of a variety of melanoma vaccine trials, using antigen pulsed APC.

One of the first studies, done by Mukherji and colleagues, involved the

treatment of patients with advanced melanoma with GM-CSF expanded and

MAGE-1 MHC class 1-restricted peptide pulsed monocytes. Following

intradermal injection, induction of melanoma-reactive CTL responses was

detected in patients with advanced melanoma. although no significant

therapeutic responses were seen123.

In a similar study by Nestle and colleagues, DCs were pulsed with either

a cocktail of HLA class I restricted peptides including gp100, MART-1,

tyrosinase, MAGE-1 and MAGE-3 peptides or with tumor lysates, if patient's

HLA haplotype was not HLA class I specific and was inappropriate for peptide

loadingl24. To induce CD4 T cell response and to improve immunogenicity KLH

was included during antigen pulsing. Patients received 6-10 intralymphatic

injection, with only minor toxicity noted, which was limited at the injection site.

Regression of tumors was seen in five of the sixteen patients, including two

patients that received only tumor lysate pulsed DCs, applicable to cancers

lacking defined tumor antigensl*?

1.8.2.3 Mulftiple Myeloma

Multiple myeloma is a late B cell disorder involving malignant

transformations of a mature, antibody secreting B dl. The lg idiotype

Page 46: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

expressed by the transfonned cells (M protein) represents a tumor specific

antigen, which may be used as a target antigen by host T cells. Preclinical

studies have dernonstrated that host T cells could recognize the idiotypic

deteminants of myeloma proteins (M protein)W Additional studies assessed

the therapeutic benefits of injecting rnyeloma patients with penpheral blood

derived DCs pulsed with M protein after the patients had received high dose

chernotherapy, total body irradiation and autologous penpheral blood stem cell

transplantation. DCs injection was followed by five subcutaneous booster

injections of idiotype coupled to KLH. ldiotype specific T cell proliferation and

cytolytic responses were induced but it was observed only in a group of

patients who achieved complete remission prior to vaccination. ln a similar

study carried out by Wen et al, results demonstrated the induction of anti-

idiotypic T cell response but no significant clinical responses, respectively'27.

1.8.2.4 Other Cancers

Prostate cancer is another type of malignancy that may be amenable to

immunotherapy, since severai prostate tissue associated antigens are now

being characterized. These include prostatic alkaline phosphatase (PAP),

prostate specific membrane antigen (PSMA) and prostate-specific antigen

(PSA). Valone et al. used PAP protein to pulse penpheral blood derived DCs.

These cells were injected into twelve patients, which induœd T cell proliferative

responses to PAP protein128. However. when monocyte derived DCs were

Page 47: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

pulsed with PSMA and then administrated to eighty-two patients. only two of

the patients mounted T cell responses against PSMA peptides129JJo.

DC based immunotherapy has also been assessed for the treatrnent of

renal carcinoma. In one study monocyte derived DCs were pulsed with KLH

and autologous tumor cell l ysa te~~3~ . Only two patients completed the

treatment with one exhibiting reduction of tumor rnass and proliferative

responses of peripheral blood monocytes against tumor ceIl lysates-

Although vanous studies in murine model systerns and human clinical

trials have demonstrated the in vivo activity of antigen-pulsed DCs to function

as vaccines against tumor challenges, the animal model systems used are

artificial. In rnost experiments, genes encading foreign proteins have been

introduced into tumor cells to serve as model tumors antigens, making them

highly irnmunogenic. In contrast, most spontaneously arising human cancers

are nonimmunogenic. Moreover, in most transplantable tumor models, animals

with healthy immune systems are useds? However. rnalignancies usually

develop spontaneously as a result of the expression of a specific genetic

abrnormality. Clona1 expression and the accumulation of genetic abnormalities

leads to the generation of a heterogenous population of tumor cells, some of

which have the ability to escape host immune rejection.

7.8.3 Future Prospects for DC Based Cancer Vaccines

Vaccination of cancer patients with antigen pulsed DCs illustrates the

powerful antigen-presenting capacity of DCs for induction of an antitumor-

Page 48: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

specific immune response. The general safety of DC vaccination in patients

and therapeutic potential of this approach have been amply demonstrated.

Several variables should be considered in the future studies:

1) Since both peripheral blood and monocyte derived DCs have shown

eficiency but have never been compared the optimal source of DCs for

clinical use must be determined;

2) It remains to be determined whether DCs pulsed with autologous tumor cell

antigens induce the maximum antitumor response or whether more effort

should be directed at the identification of tumor specific antigens that would

serve as a general antigen to induce an anti tumor response in al1 patients

with a particular malignancy;

3) The sufficient amount of DCs and the most efficient methods for antigen

loading of DCs that will permit the presentation of large array of tumor

antigens, and

4)- The optimal route and frequency of administration that is sufficient for

induction of tumor specific immune responses.

Immune responses may be further enhanced by the development of

techniques that may improve antigen presenting properties of tumor cells and

to inactivate the immunosuppressive factors produced by tumor cells-

Page 49: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

1.9 HOX11 Transgenic Mice

1.9.1 Homeobox Genes in Hematopoiesis and Hematologic Malignancies

Horneobox genes are a family of genes containing the conserved

domain of 61-amino acid132. Homeobox genes are classifed into class I HOX

genes and divergent homeobox genes. Thirty-eight known class I mammalian

HOX genes are arranged in four clusters designated HOX A, BI C and Dl while

more than 200 divergent horneobox genes are located outside the HOX loci

often without forming clusters133*134.

The products of homeodomain containing genes, homeoproteins,

function as transcription factors and play an important role in forming the body

structure during fetal developmentr3? In addition. many of these genes are

expressed in hematopoietic cells and in leukemia cel ls136~~~~. Genetic alteration

and aberrant expression of homeobox genes have been reported in human

leukemias. In early studies hurnan leukemia cell lines were used to determine

the expression pattern of HOX genes138. Subsequent studies used normal

peripheral blood and bone marrow cells to determine which HOX genes are

expressed during normal hematopoiesisl3? Results from these studies showed

general preference of the HOX genes from each cluster in their expression

pattern. The genes of HOX A, 6 and C clusters are predominantly expressed in

rnyelornonocytic, erythroid and lymphoid lineage, respectively140. However,

HOX genes of each cluster play a role in not only a single lineage but also

Page 50: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

hernatopoietic cells. For example. Care et al. showed that HOX 6 cluster genes

are expressed in PHA activated T cells, while Petrini et al. demonstrated the

expression of several HOX B locus genes in lymphoid cell lines with a natural

killer phenotype141-142.

As with class I HOX genes. divergent homeobox genes are also

expressed in normal and malignant hematopoietic cells143. One of the genes

that belong to this family is the HOX11 gene. HOXI 1 was originally isolated

from the breakpoint of the nonrandom t(l O;l4)(qZ4;q 1 1) chromosome

translocation, which is found in the malignant cells of 5% of patients with T cell

acute lymphoblastic leukemias (T-ALL)lun14? Another group reported that

thirty-three percent of pediatnc T-ALL patients showed overexpression of the

HOXll gene in their leukemic blast cells. although normal T cells did not

express HOXI1 as detennined by RT-PCRW Sirnilar HOX11 rearrangements

have been also seen in some cases of B Iineage malignancy.

HOX11 is nomally expressed during murine ernbryonic development in

the surface ectodem, certain brachial arches, as well as within cranial nerves

innervating these structures147-149. Human Iiver and CD34' bone marrow cells

show HOXlI expression, but it does not appear to be expressed in normal T

cells, although one report suggests that mature T cells may express HOX11150-

152. Expression of HOX11 is also found in the splanchnic mesoderm. which

ultimately gives rise to the spleen. It is interesting to note that in HOX11

deficient mica, spleen formation is inliated during early embryogenesis, which

is followed by atrophy of the organ due to apoptosis. These miœ are norrnal at

Page 51: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

birth but show no spleen thus indicating that HOX11 may contribute to the

survival of the primordial cells, which develop into the spleen147.

1.9.2 Production of HOXl1 Ti-ansgenic Mice

Transgenic technology offers possibillies for generating precise animal

models for human genetic disease. Rapid generation tirne and the ease of

maintaining inbred strains make mouse models useful mammalian genetic

systems. One of the beneficial applications of this technology has been the

study of the behavior of overexpressed or ectopically expressed oncogenes.

While transgenic animals bear the introduced oncogenes in every tissue.

expression of that gene may either be widespread or directed to a particular

cell Iineage, depending upon the regulatory sequences chosen.

Much work has been done in Our laboratory to generate transgenic mice

for characterizing the oncogenic potential of HOX11. We have developed a

HOXll transgenic mouse modal for human lymphoma that may be more

consistent with spontaneous lymphoma growth found in patients. The

oncogenic potential of HOX11 was assessed by placing the human HOX11

cDNA under the transcriptional control of munne immunoglobulin heavy chain

(IgH) prornoter and enhancer sequences with expression directed in B

lymphocytes. The transgene (IgHp -HOXI 1 ) was injected into pronuclei of a

fertilized ovum and three lines, C2, C5 and D l 1 were obtained. Each line

contained approximately 2 4 copies of the transgene integrated in head-to-tail

orientation. Histopathologic analyses showed that low nurnbers of HOX11

Page 52: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

transgenic mice develop lymphoid hyperplasia dun'ng their first year of life and

85 percent of mice developed lymphorna in the second year of life 1%.

7.9.3 Lymphoma Development in HOXl1 Mice

Detailed analyses confimed that HOXl 1 transgenic mice developed

mature B cell lymphomas153. Assessment of the effects of HOXl l transgene

expression on lymphopoiesis indicated that low level expression of HOX11 in

B-cells resulted in no significant effects on normal lymphopoiesis. However. al1

HOXl1 transgenic rnice died during their second year of life with the average

age of death being 14 to 15 month. The lymphomas were predominantly

detected in spleen with progression to an aggressive phenotype characterized

by metastasis to the lymph nodes. thymus. liver, lungs. kidney and pancreas.

The B cell origin of the lymphomas was confirmed by analyzing thymuses

infiltrated with lymphoma cells. Since the lymphoma cells expressed both IgM

and IgD. it is most likely they are mature B cells. Furthemore. the injection of

SClD mice with spleen cells obtained from HOXI 1 transgenic mice confimed

the malignant phenotype of these cells- This study provided the first direct

evidence that expression of HOX11 leads to rnalignant transformation- The

long latency period suggests that other genetic abnonnalities in conjunction

with ectopic expression of HOX11 in lymphoma cells are required for the

development of lymphomas. The course of lyrnphomagenesis in tiûX11

transgenic mice is similar to a subtype of human, indolent nonfollicular

lymphoma temed splenic marginal zone lymphoma.

Page 53: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Illustration 1. Tissue sections from HOXlt transgenic mice stained with

hematoxylin and eosin showing lyrnphoma.

(A) HOX11 transgenic spleen (B) Lymphoma cells x 200 in spleen (C)

Thymus (D) Lung (E) Liver (F) Kidney

Page 54: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS
Page 55: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

For example, the lymphocytes in HOX11 hyperplastic spleens are small and

show maturation to plasma cells. Progression to lymphoma is associated with

enlarged marginal zones, early marginal zone lymphorna and the appearance

of large lymphoma cells. Therefore. HOX11 transgenic mice provide a clinically

relevant model in which to assess novel approaches for the treatrnent of

indolent nonfollicular 8 cell lymphoma-

1 -1 0 Study Rationale

Making up about 5% of al1 cancers and 85% of al1 lymphomas. non-

Hodgkin lymphoma is a significant cause of morbidity and mortality'w. It is

primarily seen in older individuals and dMded into B and T celk lymphoma by

origin. 8 cell tumors are most common and have better prognosis than T cell

tumors. Lyrnphomas may be classified into indolent and aggressive tumors.

Conventional treatments, such as radiation or chemotherapy are most effective

against aggressive lymphoma. In cases where patients are not cured. they

generally have a short life span. Patients with tumors that grow slowly may

have a much better prognosis. However, some indolent lymphomas may

transform into aggressive types, which are generally more difficult to treat with

standard therapies.

In addition to killing tumor cells, conventional treatments cause damage

in normal cells and produce side effeds, and often do not result in cures.

Based on these data, several different approaches to improve cure rate and

survival are being investigated. One of the most promising is cancer

Page 56: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

immunotherapy involving a new strategy based on enhancing the recognition of

turnor cells by the host immune system through the presentation of tumor

antigens by dendritic cellss7Jos.1ss1s7- Animal expenments as well as clinical

experience indicates that the immune system can recognize and kill tumor

cells. However the presence of tumor antigens by themselves is often

inadequate to stimulate the host immune system to control tumor growth. The

cumulative cunent knowledge suggests an approach to cancer

immunotherapy. in which autologous DCs from tumor-bearing hosts are

expanded ex vivo, pulsed with potential tumor antigens and reinfused into

patients to induce tumor specific T cells including CTLs. Data discussed above

suggest that DC may be used as a physiological adjuvant in vivo to activate

antigen-specific T cells and wnsequently induce T cell dependent humoral

responsesll2.114.158.159.

Mode1 systems cunently used to assess DC based therapeutic

modalities often involve treating miœ after subcutaneous or intravenous

injection of tumor cells into healthy mi~eio3~105. Although tnese experiments

show very promising results including prevention of tumor developrnent and

reduction of metastasis to distant organs, results of human clinical trials are not

conclusive. One possible reason is that immunosurveillance of tumor cells by

the cellular immune systern is often ineffective in vivo, which is reflected in the

outgrowth of tumors that are apparently unable to evoke an immune

response". One explanation for this is that the tumor cells have downregulated

expression of the cell surface molecules such as costirnulatory molecules or

Page 57: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

MHC. In addition. it is also possible that T cells with receptors specific for tumor

antigens have been rendered anergic.

Our laboratory recently developed a transgenic mouse model for 6

lineage lymphomas. Sinœ lymphoma developrnent in these mica is

spontaneous with a long latency period, we believe that this transgenic model

is a unique system in which to assess the therapeutic potential of dendritic

cells, Before these studies c m be initiated, however, it is essential to show that

HOXI1 transgenic mice have a competent immune system capable of initiating

an immune response against foreign antigens. Furthemore it is important to

determine whether the process of lymphomagenesis results in the generation

of TAA to which an immune response can be induced.

1 -1 1 Hypothesis

HOXl 1 transgenic mice spontaneously develop lymphomas, which

ultirnately evade the host immune system and therefore may be used as a

model system to develop dendritic cell based cancer vaccines. We hypothesize

that young HOXI 1 mice have functionally competent dendritic and T cells and

that HOX11 lymphomas express TAA that can be recognized by naïve T cells.

We also hypothesize that lymphomagenesis in HOXll transgenic mice is

associated with the development of a comprornised immune system andior the

development of the host T cell tolerance to lymphoma associated antigens.

Page 58: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

1 -1 2 Objectives

The objectives of the experiments undertaken in this thesis were to:

assess the functionality of DCs from HOXll transgenic mice including

number, phenotype and capacity for antigen presentation,

asssss the proliferative response of T cells from HOX11 transgenic mice to

mitogenic stimulators and foreign proteins presented by DCs,

assess the cytolytic activity of antigen specifïc cytotoxic T cells from HOXl1

transgenic rnice, and

assess the immunogenicity of HOX11 denved peptides or HOX11

lymphomas using either in vitm transcribed HOXI 1 RNA or HOXl1

lymphoma cell lysates as a source of tumor antigens

In future studies, tumor antigen pulsed DCs may be used as cell

vaccines to abrogate or prevent lymphoma development in this unique murine

mode1 system. Eventually, a better understanding of DC-based immunotherapy

may lead to human clinical trials.

Page 59: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Chapter 2

Materials and Methods

2.1 Animals

HOXll transgenic mice were originally produced in Our laboratory and

we have ample access to them in our breeding colony153. Transgenic mice

were identified by amplification of HOX11 DNA (20 ng) using the Polymerase

Chain Reaction (PCR), by using the forward primer, 5'-

AACCGCAGATACACAAAGGA-3' and the reverse primer, 5'-

TGGGCCAGGCTCTTCTGGAA-3'. Samples were first incubated at 95°C for 5

min and then subjected to 35 cycles of PCR amplification. PCR condition were

as followed: denaturation at 94°C for 1 min, prÏmer annealing at 62°C for 1 min,

extension at 72°C for 2 min and a final incubation at 72°C for 10 min- The

product was detected in 2.5% agarose gels. In addition, CDI, C57BU6 (H-2b)

and C3H (H-2*) mice were purchased from Charles River (Quebec, Canada)

and bred and maintained in microisolators under pathogen-free conditions in

the animal facility of the Sunnybrook Hospital Research Institute. All animal

manipulations were in accordance with Sunnybrook Health Science Center

Animal Care Committee guidelines.

Page 60: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

2.2 Reagents Used for the Isolation and Culturing of DCs and T cells

2.2.1 Growth Factors

The interleukin-4 (114) producing cell Iine was cultured in RPMl 1640

(GIBCO BRL) supplemented with 10% heat inactivated FCS (Gibco), 2 mM L-

glutamine (Gibco), 100 Ufml penicillin (Gibco) and 100 mg/ml streptomycin

(Gibco)W Cultures were left without feeding untir viability dropped below 80%

(as determined by trypan blue staining), at which tirne. cells were harvested.

centrifuged for 10 min at 200xg (Beckman) and supernatant filter sterilized and

stored at -20°C.

Granulocyte macrophage-colony stirnulator factor (GM-CSF) was

produced from COS-7 cells transduced with the rnurine GM-CSF cDNA (a gift

of Dr. Nicholas Gough, Victoria, Australia). COS-7 cells were cultured in 100

mm plates in DMEM (Sigma) supplemented with t O % FCS. Before

transfection, cells were washed with TS and then TD. TD contained 140 rnM

NaCI, 5 mM KCI, 25 mM Tris and 0.5 rnM NaH2P04, pH 7.5. TS contained TD

supplemented with 1 mM MgClz and 1 mM CaCI2.

Transfection method

10pg of GM-CSF cDNA was diluted in 2 ml of TS and mked with 2 ml of TS

containing 1 mg/ml DEAE-Dextran, making a final volume of 4 ml of DNA

solution. The DNA solution was added ta the cells and incubated at 37°C. After

50 min, the DNA solution was replaced wRh new TS containing 20% glycerol,

for 2 min. TS was removed and cells washed with TS and then with DMEM

containing 5% FCS. Cells were placed in fresh DMEM containing 5% FCS and

Page 61: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

200 pM Cloroquine and incubated at 37OC for 2 hours. at which time, fresh

DMEM (S%FCS) was added and cells incubated for 48 hours at 37OC, 5%

C02. After incubation, the culture supernatant was filter sterilized and stored at

-20°C.

2.2.2 Collagenase D

Collagenase D was purchased from Boehringer Mannheim and since the

activity is fisted in a specific units called Wunsch units and the protocol required

Mandl units, the entire Collagenase D via1 was dissolved to 4000 Mandl Ulml in

HBSS to a final volume determined by the following equation:

mllvial = (x Wunsch Ufml) x (750 Mandl UMlunsch U) x 1000 mghial / 4000

Mandllml

x = specific activity listed in the package insert

1 Wunsch unit = 750 Mandl units

Calcium containing HBSS is essential to preserve Collagenase D activity. since

it is a Ca" dependent enzyme.

2.2.3 Bovine Serum Albumin (BSA)

DCs from the spleen were isolated by BSA density gradient centrifugation. To

prepare the 35% BSA solution, 186 ml PBS. 29ml of 1 N NaOH. and 65ml

water was combined in a 1-liter beaker. 106 g BSA (Cohn fraction V, Integren)

was placed ont0 the surface of the solution without stirring and refrigerated

ovemight to allow the albumin to dissolve. After 24 hours. a small sample was

removed and the refractive index measured. The correct density of BSA was

Page 62: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

1 -080 g/mI which conesponded to a refractive index between 1 -384 and 1 -385

at 25°C. The solution was filter sterilized (Nalgene. 500 ml) and stored for 3

months at 4°C-

2.2.4 P reparation of Sterilized Nylon Wool Columns

Nylon Wool (Fisher Scientific) was placed in a Cliter beaker and

saturated with an excess volume of 1 % HCI, The contents were boiled for 5 to

10 min to remove contaminants and then washed at least 10x with water.

Nylon wool was completely dried at 25°C and wool combed to remove knots.

Columns were made by placing 0.5-0.8 g or 1.0-1.5 g of nylon wool inside 5

and 10 ml disposable syringes (Becton Dickinson). respectively. Columns were

autoclaved at 1 1 O°C for 15 min at slow exhaust (no dry cycle) and stored for

months in a dry place, at 25OC (room temperature). A total of -10' cells were

added to 5 ml columns and 2-3 xlo8 cells were added to 10 ml columns..

2.3 Materials Used for Mixed Lymphocyte Reaction (MLR) and Cytotoxic T

Lymphocyte (CTL) Assay

a OVA protein (grade VI. Sigma Chemical Co., 1 g) was dissolved in RPMl

1640 to a final concentration of 200mglml and stored at -20°C.

a Phytohemagglutinin (PHA) (Sigma Chemical Co., 1 mg/ml) was dissolved in

RPMA 1640 and stored at -20°C.

Page 63: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

[ 3 ~ ] thymidine (Amersham) was received as a stock of 1mCi. 1 ml of th#

stock was dissolved in 19 ml of RPMl 1640 and stored at 4°C. The working

concentration was 20uCi per well (200 pl).

2.3.1 Cell Lines Used as Targets in CTL Assay

The EL4 ceii Iine (American Type Culture Coilection, Rockville, MD, TI&

39) was established from a T cell thymoma derived from a C57BU6 (H-zb)

rnouse'61. The EG7-OVA cell line (ATCC. CRL-2113) was derived by

transfection of EL4 cells with a plasmid carrying chicken ovalbumin (OVA) and

the neomycin (G418) resistanœ genel62. E.G7-OVA cells contain a single copy

of the inserted plasmid and translate and secrete OVA protein. Cell lines were

maintained in DMEM medium (EL-4) and RPMl 1640 medium (E.G7-OVA)

supplemented with 10% heat inactivated FCS, 2 mM L-glutamine, 100 Ulml

penicillin and 100 rnglml streptomycin. In addition, EG7-OVA culture media

was adjusted to contain 1.5 gA sodium bicarbonate. 4.5 glL glucose. 1 O mM

HEPES. 1 mM sodium pyruvate, 0.05 mM 2-mercaptoethanol and 0.4 mgh l

G418.

In addition to the EL4 and E.G7-OVA cell lines described above, the K3P

cell line was also used as a target in CTL assays. The K3P was cell line was

derived from cells obtained from a T-ALL patient with a t( l O;l4) chromosome

translocation and established according to published methodst63. Cytogenetic

analysis indicated that the cell line had lost both the normal and derivative

chromosomes 10 but retained the derivative chromosome 14 bearing the short

Page 64: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

and proximal long amis of 14 and the translocated chromosome 10 material

distal to the breakpoint in 10q24. The cell Iine expressed the 2.lkb HOXll

transcript and was used to isolate the HOXI1 cDNA1W. K3P cells were

maintained in complete RPMI-1640 medium supplemented with 10% heat

inactivated FCS, 2 mM L-glutamine, 100 U/ml penicillin and 100 mg/ml

streptomycin.

2.3.2 Production of In vitro Tiiinscribed (IVT) RNA

The pGEMHOXl1 vector was generated by cloning nucleotides 165-2058

of the human HOXll cDNA into the Smal site of the multiple cloning site of

pGEM7Zf (Promega. Madison, WI) thereby placing the HOX11 cONA

downstrearn of the T7 prornoter. The vector was Iinearized with BamHl or

Hindlll to minimize plasmid sequences from the in vitro transcribed mRNA. In

vitro transcription of HOXl1 was carried out at 37OC for one hour using the

Promega RiboprobeO in vitro Transcription System according to the

manufacturer's instructions. Template DNA was digested with RNase free

DNase 1 and RNA was recovered by triasoVchlorofon extraction followed by

centrifugation and the pellet was washed with 70% ethanol. The pellet was air-

dried and resuspended in sterile water. RNA was quantitated by measunng the

optical density (OD) at 260-280 nm and stored at -80°C.

Page 65: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

2.3.3 Preparation of Tumor Lysate

Tumor biopsies from HOXll mice that had developed lymphoma were

removed under the sterile conditions using forceps and scissors and placed in

PBS. Nonmalignant tissue was removed and tumor cells disperseci to create

single cell suspensions. Cells were transferred into 2 ml cryopreservation tubes

(Falcon) and lysed using the following procedure. First, tubes were placed in

iiquid nitrogen for 5 min and then lef€ ai 25°C to thaw completely. This freezel

thaw procedure was repeated 6x. After the last thaw cycle, cells were

centrifuged at 14Oxg for 10 min to remove large particles, and supernatant

passed through a sterile MiIlex@-HV 0.22 prn filter (Millipcre) and aliquots

stored at -80%.

2.4 Generation of DC Cultures

The main objective of this initial expen'ment was to detemine the most

efficient technique for the isolation of DCs from spleen and bone marrow of

HOXI 1 mice. Further, we tested culture conditions under which we would

routinely obtain sufficient number of DCs to be used in different assays (see

below).

2.4.1 Dendn'tic Cell Isolation from Spleen

2.4.1.1 Density gradient cenHfugation using Bovine Semm Albumin (BSA)

Collagenase digested splenocyte suspensions were centrifuged in dense

bovine serum albumin to obtain a fraction with a low buoyant density enriched

for DCs.

Page 66: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Spleens were removed and plaœd in 60 mm tissue culture dishes

containing 5 ml of Collagenase D solution. Each spleen was injected with 100

pl of Collagenase D using a 22-G needle attached to a 5 ml syringe. Forceps

were used to tease the spleen into srnall fragments. Dishes were placed in a

37°C incubator for 45-60 minutes to release DCs. At the end of the incubation

period, the fragments were placed into a 70 pn nybn cell strainer (Falcon),

pressed through the strainer using a plunger from 5 ml syringe, and celi

suspensions collected into 50ml polypropylene tubes (Falcon). Tubes were

centrifuged in a refrigerated centrifuge (Beckman) held at 4OC for 10 min at

280x9. After centrifugation. supematants were removed and pellets

resuspended in 35% BSA (1 mwspleen). Cell suspensions were transferred to

the 6 ml polypropylene tubes (Falcon), overlaid with 1.5 ml of 4OC RPMl 1640

to fomi a sharp interface, and low density cells obtained after a 15 min

centrifugation (Beckman, L8-70 Ultracentrifuge) (on a 35% BSA gradient) at

9500xg and 4°C. The centrifuge was set up for slow acceleration and with the

break turned off. Using a pipette, low density cells were carefully collected from

the interface region, taking the full volume of RPMl 1640 and the top 1 ml of

BSA. Cells were resuspended in wld (4°C) RPMl 1640 and centrifuged for 10

min (280xg, 4OC), after which supematants were removed. and cells

resuspended in 5-10 ml of DCs medium (RPMI 1640 medium supplemented

with 10% FCS (Gibco). 50 mM 2-mercaptoethanol (Sigma), 10 mM HEPES

(Sigma). 2 mM glutamine (Gibco). 100 Ulml penicillin (Gibco), 50 pglml

streptomycin (Gibco), and 20 nglml GM-CSF). Cell viability was detemined by

Page 67: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

trypan blue exclusion and 4 ml of the DC suspension (- 1 4 x lo5 cells/ml) was

placed in 60 mm tissue culture dishes. After 90 minutes, medium containing the

non-adherent cells was removed and adherent cells cultured ovemight. All

tissue culture procedures were done under sterile conditions in a level 2

containment facility.

2.4.1.2 Separation using MA CS CD f fc MicroSeads

For MACS systern separation. colloidal super-paramagnetic MicroBeads

conjugated to monoclonal hamster anti-mouse CDl lc antibodies were

purchased from Milteny Biotech. The CD1 1c antibody clone N418 is specïfic for

the leukocyte integrin expressed primarily but not exclusively on mouse DCs.

Using this technique DCs can be efficiently enriched.

lsolated spleens (4-6) were placed in a 60 mm tissue culture dishes with

Collagenase D solution (5ml) and the procedure for the spleen preparation

followed as described above. After incubation, cell suspensions were prepared

and cells washed in 15 ml of buffer followed by centrifugation at 200x0 for 10

min (4°C). The bmer solution consisted of phosphate buffered saline (PBS) pH

7.2, supplemented with 0.5% BSA. Cell pellets were resuspended in 400 pl of

buffer per 1 o8 total cells. 100 pl of CD1 1c MicroBeads per 10' cells were added

and cells were incubated at 6°-120C. After 15 min cells were washed by adding

4-8 ml of buffer and centrifuged at 200x9 for 10 min. Cell pellets were

resuspended in 500 pl of buffer per 108 cells and passed through a separation

column (column type MS+/RS+ for up to 2x10' total cells), which was held in

Page 68: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

the magnetic field of the MACS separator. The magnetically labeled CD1 lc'

DCs were retained in the column while the unlabeled CD116 cells passed

through. Columns were washed with 500 pl of buffer and removed from the

magnetic field. Retained CD1 1c' DCs were eluted as the positively selected

cell fraction using 1 ml of OC medium and cultured under the same conditions

as BSA isolated DCs (above). To obtain a highly pure population of DCs for

some experiments, the magnetic selection step was repeated.

2.4.2 Dendn'tic Ce11 Isolation from Bone Manow

After removing al1 muscle tissues from the femun using forceps and

scissors, the bones were plaœd into 35 mm culture dish with 4°C RPMl 1640

medium. Both ends of the bones were cut with scissors, and the marrow was

flushed out using 3 ml of RPMl 1640 and a 5 ml syringe with a 25-gauge

needle. Red blood cells were lysed using 3 ml of lysis bMer (0.165 M

ammonium chlonde and Tris, pH 7.4) at 25OC for 10 min. Bone marrow cells

were washed in RPMl 1640 supplemented with 2% FCS followed by

centrifugation for 10 min at 200x9. Cells were plaœd in 60 mm tissue culture

dishes in RPMl supplemented with 10% FCS, 50 mM 2-mercaptoethanol, 10

mM HEPES, 2 mM glutamine. 100 Ufml penicillin, 50 pglml streptomycin, 20

nglml GM-CSF and 20 nglml 114. The cell concentration was - 1-5 x 10'

cellslml. The cultures were incubated at 37*C, 5% CO2 and fed every 2 days by

removing 75% of the medium and returnhg a similar volume of fresh, warm

Page 69: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

(37°C) DC medium. On day 7, cells were harvested by gentle pipetting. Further

purification of DCs was done using CD1 1 c MicroBeads (described above).

2.5 Dendritic Cell Surface Phenotype Charactenzation

We used flow cytometric analysis to determine the phenotype of HOXI 1

spleen and bone mamw denved DCs, since the expression of specific

molecules on the surface of DCs is essential for their function as APCs-

2.5.1 Flow Cytometric Analysis of Spleen and Bone Manow Dendritic Cells

Two groups of six mice (HOX11 x C57BU6. F4) were used to detennine the

phenotype of spleen derived DCs. One group consisted of three HOXI1

transgenic and three non-transgenic mice (three months old) while another

included nine month old mice. To evaluate bone marrow derived DCs, HOXI 1

(HOX11 x C57BU6, F4) and non-transgenic mice were used. Each group

consisted of three mice (three months old). Spleen derived DCs were isolated

by BSA density gradient centrifugation, while bone marrow derived DCs were

isolated using CDl lc MicrBeads (MACS system). Both techniques were

described above. DCs (-1o6) were stained with primary antibodies in HANK'S

medium supplemented with 2% FCS (HF) on ice for 30 min. The cells were

subsequently washed twice with HF and resuspended in HF containing 1 pl/rnl

propidium iodide. Antibodies used in this study were: Fluoresœin

isothiocyanate (FITC)-labeled monoclonal antibodies to CD1 lc , CD1 1 b (Mac-

1). Ml169 (HSA), CD80 (87-1). CD86 (87-2). CD40. CD3 and 8220, al1

purchased from PharMingen (Mississauga, Canada); FITC-labeled H ~ K ~ (MHC

Page 70: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

class 1) and I A ~ (MHC class II) purchased from Cedarlane (Homby, Canada).

FITC-labeled polyclonal anti-rat IgG (PharMingen) was used as negative

controls. Flow cytometric analysis was performed using a Coulter Elite

(Beckman Coulter) flow cytorneter equipped with EXP02 software (Applied

Cytometry System).

2.6 T Ceii Isolation and Enrichment

The aim of this experiment was to determine the best method for the

isolation of sufficient numbers of an enriched T cell population from HOX11 and

control mice. These T cells were used for the optimization of the mixed

lymphocyte reactions (described below) followed by dfierent functional studies

to characterize HOXl 1 derived T cells.

2.6.1 Isolation of T cells f m Peripheral Blood

Blood was obtained by cardiac puncture using a 19-gauge needle and 5 ml

syringe filled with RPMl 1640 with 100 U/ml heparin (-2 ml/mouse). Blood was

transferred into 14 ml polypropylene tubes (Corning). diluted 1:2 in medium and

carefully layered onto the density gradient, FicolCPaque, using a pipette to

produce a clean interface between the tw layers (the proportion of blood ta

Ficoll was 1:3). Tubes were centrifuged at 400x9 at 20°C. After 25 min. the

white opaque mononuclear fraction from the interface between the diluent

(medium) and Ficoll-Paque was removed using a disposable pipette and the

cells washed at least two times with 5-10 ml of medium to remove Ficoll-Paque

(300x9, for 10 min).

Page 71: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

2.6.2 Isolation of T Cells from Spleen and Thymus

2.6.2.7 Separa tion with MA CS System

T cells were isolated from spleens using either the MACS separation

system or nylon wool columns. For MACS separation. a single cell suspension

was prepared from spleens (described above) or thymuses (same procedure

as with spleen cells) and red cells lysed with 0.165 M NHdCI-Tris pH 7.4. The

protocol for the MicroBeads separation was similar to the method for the DCs

isolation from spleen, previously described, with the only difference being the

type of MicroBeads used for T cell isolation. CD90 (Thy 1.2) MicroBeads were

used since CD90 is expressed on thymocytes. peripheral T cells and some

interepithelial T cells. Thymus or spleen cell suspensions depleted of red cells

were washed. and labeled with CD90 MicroBeads (1 Op1 of beads/l o7 total cells)

and passed through separation columns. The rnagnetically retained CD90' T

cells were eluted as a positively selected cell fraction. Eluted cells were -90%

CD3+ as deterrnined by flow cytometric analysis.

2.6.2.2 Isolation with Nylon Wool Columns

T cell enrichment by nylon wool is based on the adherent properties of B

cells and accessory cells to nylon wool at 37°C in the presence of FCS. In

contrast T cells do not adhere but pass through the columns. Sterilization of the

nylon wool and assembly of the columns was described above. Columns were

washed with warm (37OC) RPMl 1640 supplemented with 5% FCS, sealed with

Page 72: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

parafilm and incubated at 37°C. 5% CO2 (with 1 ml of medium remaining in the

columns). After 1 hour, columns were rinsed with 5 ml of warm medium and 1

ml of the spleen cell suspension (prepared as described above) was added to

columns and incubated at 37°C for 45 min. After incubation, columns were

washed with 15 to 20 ml of warm medium, T cells collected into 50 ml tubes

(Falcon) and concentrated by centrifugation (200x9, 10 min).

2.7 Optimization of Mixed Lymphocyte Reaction (MLR)

The MLR assay was used to measure T cell proliferation ability after

stimulation. Suspensions of responder T cells were cultured with stimulator

cells and based on the activating stimuli, an allogeneic or syngeneic MLR was

established. The activating stimuli in an allogeneic MLR were the mismatched

MHC class I and class II antigens processed and presented by allogeneic

stimulator cells (DCs). while in syngeneic MLR OVA protein pulsed stimulator

cells (DCs) were used to present foreign antigen and induœ proliferation of

responder cells (T cells). Parameters affecting the magnitude of T cell

proliferative responses include cell concentration, type of medium, type and

concentration of activation agent, type of responding T cells and culture time.

We optimized experimental conditions with respect to these variables.

Page 73: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

2.7.1 Optimization of T CeIl Pmliferative Responses

To optimize MLR. a dose-response assay was performed using different

sources and nurnbers of responder T cells. T cells were isolated from spleen,

peripheral blood and thymus using the previously described techniques.

T h e replicate wells were set up containing each of the following:

T cell proliferation was induced by phytohemagglutinin (PHA, Sigma) at a

concentration of 3 4 pgfml. Cells were plated at different concentration (as

presented in the table above), cultured in 96-well U-bottom plates (Corning)

and I~H] thymidine (1 pCilwell, Amersham) added on days 3. 4. and 5 to

determine the time of the maximum T celI proliferative response. Cells were

harvested 1848 hou= later, using a Titertek-cell harvester. I~H] thymidine

incorporation was measured by liquid scintillator(Wallac-1205 Betaplate).

Spleen

Peripheral blood

Thymus

2.8 Çunctional Studies Using HOX11 Derived DCs

2.8.1 Presentation of Foreign MHC by HOX7 7 Derived DCs

The prirnary aim in this experiment was to test the ability of HOX11

spleen derived DCs to process and present their own MHC class I and class II

0.5

1 .O

1 .O

3.0

2.0

5.0

8.0 4.0

5.0

6.0

Page 74: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

molecules and to induce proliferation of allogeneic T cell isolated from C3H

mice (H-23.

Four month old HOX11 transgenic (bred in the CD4 genetic

background), C3H (H-2*) and C578U6 (H-2b) female mice were used in this

experiment. 1 x Io4 irradiated (>ZOO0 rad) spleen derived DCs from HOX11

transgenic mice were cultureci for 5 days in 96-well plates with 1 x 10' splenic T

cefls isolated from C3H and C57BU6 mice. Thirty six hours before assay

completion, i 3 ~ ] thymidine (1 pCi1well) was added to each well. At completion,

plates were harvested and 1 3 ~ ] thymidine uptake measured in a liquid

scintillation counter. Responses were reported as mean cpm from triplicate

samples. As a positive control, C3H DCs were plated with C57BU6 T cells,

while as a negative control, HOXI1 derived DCs were mixed with autologous

HOX11 derived T cells.

2.8.2 Presentation of OVA Protein by HOXI 1 Derived DCs

Samples from three month old HOX11 transgenic and normal female mice

were used in this assay. OVA protein (grade VI-Sigma) was dissolved in OC

culture medium at 40, 60, 100 and 120 pg/ml to detemine the best

concentration of OVA protein required for maximum proliferation of autologous

T cells. 1 x 105 spleen DCslml were plated in 6-well dishes and incubated

(37OC, 5% COz) in 1 ml of culture medium containing OVA protein and 2%

FBS. After 30 min, 4 ml of OC medium supplemented with 6% FBS was added

and cells were incubated overnight. After 18 hours, DCs were washed,

Page 75: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

irradiated at > 2000 rad and added (1 x lo4 per well) to naïve autologous T

cells (1 x l o5 per well) in 96-well plates. After 5 days. PH] thymidine was

added, and cells harvested 36 hr later and thymidine uptake measured in a

liquid scintillation counter. Proliferative responses were reported as mean cpm

frorn triplicate samples. As a negative control. T cells were either cultured alone

or with unpulsed DCs.

2.9 Functional Studies Using HOXI 1 Derived T Cells

2.9.1 T Cell Proliferation Assay

Mice ranging in ages from 3.5 to 18 month of age were used. MicroBead

purified T cells from spleen were plated in 96-well plates at concentrations of 3

x 1 o5 per well. PHA was added at a concentration of 5 pgiml. The final volume

was 200 pl per well. Cells were cultured for 3 days at which tirne VH] thymidine

was added and plates incubated at 37OC in 5% COz for 38 hr. Cells were then

harvested and ['HI thymidine incorporation measured. Data are presented as

the difference in cpm between stimulated and non-stimulated T cells.

2.9.2 T Ce11 Responses to Foreign MHC Antigens

Four rnonth old HOX11 transgenic, C57BU6 and C3H female mice were

used in this experiment. T cells were isolated from the spleens of HOXll

transgenic mice using the MACS systern and plated in 96-well plates (1 x

105/well) with C3H irradiated spleen DCs (1 x 104 per well). On day 5, PH]

Page 76: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

thymidine was added and cells were harvested. Thymidine uptake was

measured using a liquid scintillation counter (Wallac-1205 Betaplate).

Proliferative responses were reported as mean cpm from triplicate samples. As

a positive control. C57BU6 cells were cuitured with C3H DCs. As a negative

control, C3H T cells and HOX11 derived T cells were mixed with autologous

C3H DCs and HOXl 1 transgenic DCs, respectively-

2.9.3 T Ce11 Responses to O VA Pmtein

Three month old HOX11 transgenic, normal female mice were used in

this experiment. The protocol used was similar to that previously described to

assess the ability of HOXll derived DCs to present OVA protein. The only

modification was that OVA protein was incubated with N-(1-(2,3-

dioleoyloxy)propyl]-N, N. N-trimethylammonium methylsulfate (DOTAP)

(Boehringer Mannheim), which is a lipid component that increase antigen

uptake by DCs. 1 x 1 o4 DCs were pulsed overnight with DOTAP-OVA protein

(D0TAP:OVA was 100pg:40pg and 100pg:lOOpg). After a 18 hour incubation,

cells were irradiated and added to naïve HOXll transgenic mice derived T

cells in 96-well plates at the concentration of 1 x 104 cells/well DCs and 1 x lo5

cells/well T cells. After 5 days in culture, [ 3 ~ ] thymidine was added and uptake

measured. As negative controk, T cells were elher cultured alone or with

unpulsed DCs.

Page 77: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

2.1 0 Measurement of Cytotoxic T Lymphocyte (CTL) Activity

This method was used to examine ability of HOX11 transgenic mouse

derived DCs to present antigen and induce primary CTL responses as well as

to test the capacity of HOX11 derived CTL to lyse different target cells. The

CTL killing assay is frequently used to measure the activity of CTL. generated

b y precursor T lymphocytes following stimulation by specific antigens

presented by DCs. The most sensitive method to assay CTL adivity is JAM

test @ust Another Method). This assay is based on the evidence that target

cells that are killed by CTL effector cells undergo apoptosis, which results in

the degradation of their ONA into srnall fragments. Target cells are labeled with

[ 3 ~ ] thymidine. washed and then mixed with cytotoxic effector cells. If target

cells are killed their DNA fragments will be washed through a filter, whereas

target cells, which are not killed uptake the PH] thymidine that will be captured

by the filter and measured. The percent lysis can be calculated by comparing

the amount of PH] thymidine bound to the filters in the presence and absence

of effector cells.

CTL activitv determination

%Spontaneous lysis = [(T-S)m x 100

T - total counts wells

S - spontaneous release wells

Page 78: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

This value is not used to calculate % cytotoxicity, but is important because high

levels of spontaneous lysis can seriously distort the expenmental results.

Specific cytotoxicity for each expenmental point was measured by the formula:

% Cytotoxicity = ((S-€11 x 100

S - spontaneous release

E - the average values of the replicates from expenments

T and S value were measured, for each CTL assay, using the following

procedure:

100p1 of target cells were added to control wells containing 100p1 of

medium (to measure spontaneous target cell lysis during assay) as well as on

a separate plate to measure total counts. All plates were centrifuged for 2 min

at 400xg at room temperature. A separate plate containing cells used to

measure total munts was harvested immediately while other plates were

incubated for 5 hrs at 37°C. After incubation, plates were harvested and

radioactivity counted on al1 filters (from test plate and total count plate) by a

liquid scintillation counter. Percentage of cytotoxicity was measured as the total

counts as well as spontaneous release for each experiment.

Page 79: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

2.1 O. 7 In Vitro CTL

This assay was used to determine the ability of OVA protein pulsed

HOXl1 derived DCs to induce CTL responses in vitm.

Six month old HOXI1 (HOXI1 x C57BU6, F5). non-transgenic and

C57BU6 female mice were used in this assay. Bone marrow derived DCs (1 x

I o 5 cells/ml) were pulsed with OVA protein in the presence of DOTAP as

descri bed a bave. After ovemight incubation, cells were washed , irradiated with

>2000 rads and added to naïve responder syngeneic T cells (1 x 106 cellshi)

in 24-well plate at responderlstimulator ratio of 20:l. The cultured medium was

complete RPMl 1640 supplemented with 1mM sodium pyruvate and l x

nonessential amino acids. After 6 days, the T cells were hawested, adjusted to

1 x 10~1rnl and restimulated with freshly pulsed DCs. Twelve to fourteen days

after the first stimulation, T cells were harvested and tested for cytolytic activity

with either OVA protein pulsed autologous BM DCs, EG7-OVA, EL4 or

unpulsed DCs serving as target cells. Based on preliminary studies to optimize

the time for the best PH] thymidine uptake, target cells were labeled with [=HI

thymidine for 5-12 hours and washed and centrifuged for two times for 7 min at

1200 rpm. In the blockinglinhibition experiment using anti-MHC class I mAb,

OVA protein pulsed target cells were incubated at 37OC for 30 min with anti-H-

2~~ mAb before use in cytotoxicity assays. The final rnAb concentration was

50pg/ml. Cytotoxic T cells and target cells were plated in 96-well plate dishes at

effectorltarget ratio of 50:l. 20:1 and 10:l using RPMl 1640 medium with 10%

FBS. In addition, plates to measure spontaneous target lysis and total counts

Page 80: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

were prepared as described above. All plates were centrifuged. further

incu bated, harvested and radioactivity counted as described above. Each

assay was perforrned in triplicates with spontaneous release < 15% (see

a bove).

2.1 O. 2 ln Vivo C TL

Based on the results of the in vitro study, we next determined whether

HOXl1 derived DCs were able to induce primary CTL immune responses to

OVA protein in vivo.

Three month old HOX11 transgenic (HOXI 1 x C57BU6-F5). non-

transgenic littermates and C57BU6 female mice were used in this assay. Bone

marrow cells were placed in culture medium supplemented with GM-CSF and

11-4 to expand DCs, while DCs isolated from spleens using CD1 l c Microbeads

were pulsed with OVA protein in presenœ of DOTAP as descnbed above. After

18 hours, pulsed cells were irradiated with >2000 rads, washed three times in

PBS and injected intraperitonealy (i-p.) into recipient mice at a concentration of

5 x 105 cellslml in 2004 PBS. Seven days post immunization mice received a

second round of immunization using expanded, CD? 1c Microbeads purified BM

derived DCs that were freshly pulsed with OVA protein in the presence of

DOTAP and irradiated. Seven to eight days after the second injection with OVA

puised DCs, spleen cells from primed mice were removed and red cells lysed.

Splenocytes (2 x I o 6 cells/ml) were cultured with either 1 x Io5 celldml OVA

protein pulsed DCs irradiated at 2000 rads or E.G7-OVA cells inadiated at

Page 81: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

20,000 rads, as a stimulators. The same culture medium describeâ for in vitro

CTL was used and plates were incubated for 5 days at 37OC and 5% CO. In

addition, bone marrow cells isolated from prirned mice were cultured in GM-

CSF and I L 4 to expand DCs and serve as autologous target cells. Effector T

cells were harvested on day 5 and assayed for antigen specific CTL activity.

Targets used to asses CTL adivity were autologous DCs pulsed with OVA

protein, E.G7-OVA, EL4 and unpulsed autologous DCs. Target cells were

labeled with ('HI thymidine for 5-12 hous and washed two times and

centrifuged for 7 min at 1200 rpm. The killing assay was set up at

effectodtarget ratios of 50:l to 5 3 in 200~1 of RPMl 1640 with 10% FBS in 96-

well plates. In addition, plates to measure spontaneous target lysis and total

counts were prepared as described above. All plates were centrifuged, further

incu bated , harvested and radioactivity coun ted as described above. Each

assay was perfomed in triplicate with spontaneous release being c 20%.

2.1 1 Assessrnent of Immunogeneicity of HOXl1 Derived Antigens

2.1 7.1 Induction of CTL Response fo In Vitro lmnscribed (IV? HOXl1 RNA

Pulsing of DCs with IVT HOXI 7 RNA

Two to three month old HOX11 transgenic (HOX11 x C57BU6-F5),

normal and C57BU6 female mice were used in this assay. CD1 lc' DCs were

isolated from spleens by MACS systern. Negatively labeled cells were washed

and served as a source of 1 cells, which were purified by nylon wool columns.

Bone marrow cells were cultured in medium supplemented with GM-CSF and

Page 82: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

11-4 to expand BM-DCs (to be useâ later as APC in a second immunization).

DOTAP (in 250-500 pl RPMI-1640) was incubated with IVT RNA (2-5 pgl250-

500 pl RPM 1-1 640) (described above) at room temperature for 20 minutes after

which the complex was added to the 5-10 x 105 cells/ml spleen derived DCs

and incubated at 37OC for 4 hours. In addition. sorne DCs were pulsed with

DOTAP-OVA protein (as descnbed above) and used as controls. DCs were

washed with PBS, centrifuged at 200x0 for 1 O min. irradiated at ,2000 rads

and used as stimulators in an MLR as well as to prime mice in an in vivo CTL

experiment as descnbed below.

MLR Assay Using DCs Pulsed with HOXl1 I M RNA

The main objective of this experiment was to determine whether DCs

pulsed with IVT HOXll RNA could activate T cells isolated from HOX11

transgenic and naïve C57BU6 mice. DCs pulsed with IVT HOXll RNA were

used as stimulator cells, while nylon wool purified T cells were used as

responder cells. MLR assay was set up in 96-well plates at the

responder/stimulator ratio 1O:l. On day 5. thymidine was added and cells

were harvested. Thymidine uptake was measured using a liquid scintillation

counter. Proliferative responses were reported as mean cpm from triplicate

sarnples. PHA and unpulsed DCs were used as controls.

In Vivo Induction of CTL Response to IVT HOX11 RNA

Page 83: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Our primary aim was to determine whether HOXll derived peptides.

presented by DCs could be used to induce antigen specific CTL response in

vivo in HOXl1 transgenic and naïve mice.

DCs were pulsed with IVT HOXll RNA. inadiated, washed in PBS

(three times) and 3 x 10' cells/ml in 2004 PBS were injected intraperitonealy

@p.) into recipient mice. Seven days post immunization mice received a

second round of immunization using expanded, CDllc* irradiated bone

manow DCs that were freshly pulsed with IM HOXI 1 RNA or OVA protein.

Seven to eight days after the previous injection, spleen cells from primed mice

were removed, red cells lysed and in v i t . CTL assays were set up. For in vifro

CTL, splenocytes (2 x 1 o6 cells/ml) were cultured with inadiated IVT HOXII

RNA pulsed DCs (1 x 10' cellslml), which sewed as a stimulator'57. The same

culture medium described for in vitro CTL was used and plates were incubated

for 5 days. In addition. bone manow cells from primed mice were cultured in

media supplemented with GM-CSF and 11-4 to expand DCs, which served as

autologous target cells. Effector T cells were harvested on day 5 and assayed

for anti-HOX11 peptide specific CTL activity. Targets used to asses CTL

activity were autologous DCs pulsed with HOX11 IVT RNA, OVA protein, K3P.

EG7-OVA and EL4 cells. All target cells were labeled with [ 3 ~ ] thymidine for 5-

12 hours and washed two times and centrifuged for 7 min at 1200 rpm. Killing

assays were set up at effectwnarget ratios of 50:l to 12.5:1 in 200pl of RPMl

1640 with 10% FBS in 96-well plates. In addition, plates to measure

spontaneous target lysis and total counts were prepared as described above.

Page 84: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

All plates were œntrifuged . further incu bated. harvested and radioactivity

counted as described above. Each assay was perfonned in tripkate with

spontaneous release being < 20%.

2.1 1.2 Induction of CTL Resporise to HOX11 Tumor Lysates

Pulshg of DCs with Tumor Ce11 Lysates and In Vitro CTL Assay

HOXll transgenic and C57BU6 bone marrow cells were cultured in

medium supplemented with GM-CSF and 114 to expand DCs and expanded

DCs purÎfied using CD1 1c Microbeads. Spleens were removed, red cells iysed

and spleen cells cryopreserved as a source of T cells. Bone mamw derived

DCs were incubated with freezelthawed tumor lysates ovemight in DCs

cultured medium. After 18 hours DCs were harvested, irradiated with 2000 rads

washed and used as stimulators in CTL assays. T cells to serve as responder

cells were isolated from spleen ceils suspensions using nylon wool columns.

CTL assays were set up in 24-well plates (Corning), in RPMl supplemented

medium (described above), at responder/stimulator ratio 20:l. The same day

the CTL assay was set up, additional T cells were isolated from another

C57BU6 mouse and used as additional responder cells. After five days in

culture, T cells were harvested, adjusted to 1 x 1 o6 cellslml and then stimuiated

with DCs pulsed with tumor cell lysates. After a total of 12 days in culture, T

cells were harvested and tested for cytolytic ability using different target cells,

including DCs pulsed with tumor lysates, DCs pulsed with IVT HOXll RNA or

unpulsed DCs. All target cells were labeled with thymidine for 5 hou= and

Page 85: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

washed two times for 7 min at 1200 rpm. Killing assays were set up at effector

to target ratio of 80:l to 1O:l in 200~1 of RPMl 1640 with 10% FBS in 96-well

plates. In addition. plates were prepared to measure both spontaneous target

cell lysis and total counts as described above. All plates were centrifuged.

further incubated, harvested and radioactivity counted as described above.

Each assay was perforrned in triplicate with spontaneous release c 20%.

Page 86: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Chapter 3

RESULTS

3.1 Identification of HOX11 Transgenic Mice

PCR analysis was used to distinguish between control (non-transgenic)

and HOX11 transgenic mice as described in the Materials and Methods. As

shown in Figure 1, the primers amplify both endogenous murine (mHOX11)

and HOXI 1 transgenic (HOXI 1 Tg) fragment.

3.2 Optimization of Mixed Lymphocyte Reaction (MLR)

The MLR is one of the most widely used methods to evaluate overall T

cell proliferative responses as well as the ability of DCs to present antigens.

However, to utilize this technique several parameters must be deterrnined

including the source and concentration of T cells, the concentration of

activation agent, and cuiture times. In the following study, T celk were isolated

from spleens, peripheral blood and thymuses from three C57BU6 mice as

described in the Methods. To deterrnine the optimum cell number required for

maximum proliferation of T cells induced by the mitogenic stimulator, PHA,

different combinations of cell concentrations were plated and T cell proliferative

responses assayed by TH] thymidine incorporation. The data presented in Fig.

2 indicated that T cell proliferation was dependent on the number of T cells

plated. The best proliferation of spleen derived T cells was obtained using 2 x

10' cells while 3 x 10"as optimal for PB derived T cells.

Page 87: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Figure 1. PCR analysis used to dîstinguish HOXII transgenic (HOX11

Tg) and non-transgenic mice. Amplification of both endogenous and

HOXI 1 transgenic fragment was observed. Lane 1 contained a negative

control, and lines 2-1 5 contained amplified DNA from transgenic and non-

transgenic miœ followed by CD-1 non-transgenic DNA (lane 16), positive

transgenic DNA (lane 17). and a plasmid containing the enüre HOXI1

cDNA (lane 18).

Page 88: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Figure 2. Optimization of MLR. Spleen, peripheral blood (PB) and thymus denved

T cells from C57BU6 mice were stimulated with PHA and T cell proliferation

measured by PH] thymidine uptake, after 3 days in culture for spleen and 5 days

for peripheral blood and thymic T cells.

Page 89: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Dose-response for Spleen T cells

OTcell alone PHA

O

Dose-response for PB T cells

Dose-reaponse for Thymus T cells

Page 90: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

When these concentrations of cells were used, 10-1 2 fold higher proliferation

was observed, when compared wÏth unstimulated T cells. Although high

concentrations of thymocytes were used (8 x 10' cellolml), the maximum

proliferation was only 3 foM above unstimulated T cells. One possible

explanation for this result couM be the low frequency of responding T cells in

the thymus. The results also suggested that the highest proliferation was

obtained on day 3 (for spleen T celts) or day 5 (for PB-T cells) after PHA

stimulation (Fig. 2).

3.3 Characterization of HOXI1 Transgenic Mice Derived Dendritic Cells

3.3.1 Establishment of Dendn'ic Ce11 Cultures

Spleen derived DCs were purified by either BSA density gradient

centrifugation or MACS CD1 1c magnetic separation and DC cell nurnben

expanded in culture supplemented with GM-CSF. Initially, DCs adhered to the

tissue culture dishes but became nonadherent after ovemight culture.

Nonadherent cells were then transferred to new T25 flasks and fed with GM-

CSF supplemented medium every 3-4 days. After 7-10 days the surface of the

fiasks generally became covered with tightly adherent strornal-like cells and

loosely detached clusters of DCs were apparent. The clusters expanded rapidly

in number and sire. After 10 days, high numbers of spontaneously released

cells were observed. These cells were subcultured and upon transfer most of

the cells adhered within 1-3 days, fonning new clusters (Illustration 2). The final

Page 91: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

yield of DCs isolated from HOX11 transgenic mice ranged from 2 x 1 O' to 5 x

10' DCS per spleen (Table 1).

1 #DC 1 HOXll transgenic 1

Table.? Number of DCs obtained from spleen of HOXI 1 transgenic rnice

(spleen) #1

Bone marrow derived DCs were cultured in the presence of GM-CSF and 11-4.

mice 2x105

Since GM-CSF also induces proliferation of macrophages, 11-4 was added to

cultures to suppress the proliferation of macrophages. Cells were washed to

remove nonadherent granulocytes without dislodging clusters of developing

DCs that were loosely attached to the marrow stroma (Picture 2). Optimal

yields (5 x I o5 DCs per mouse) and purity of DCs were obtained when the

aggregates were harvested on day 7 followed by CD1 1 c magnetic separation

(Table 2). If the clusters were allowed to overgrow without subculturing or

without GM-CSF, the aggregates dissociated and did not refom, indicating that

these factors are critical for normal DC development and proliferation.

#DC (mouse-

Table.2 Number of HOXl 1 bone marrow derived DCs

HOX11 transgenic ' mice

BM) #I #2

4 x 1 0 5 3 x 105

Page 92: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Illustration 2. Dendritic cells isolated frorn HOXll transgenic mice. A and

B represent spleen DCs 18 hr after isolation and culture in medium

supplemented with GM-CSF, while C represents bone mamw derived

DC-cluster after 6 days in the culture supplemented with GM-CSF and IL-

4.

Page 93: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS
Page 94: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

3.3.2 Phenotype Characteniation

DCs isolated from the spleens of non-transgenic and HOX11 transgenic

mice and cultured ovemight were analyzed for the expression of cell surface

antigens associated with DC function by fl ow cytometric analysis (Fig.3 and 4).

The nonadherent cells analyzed were 80.90% DCs. Staining of cells with MHC

class I and class II antibodies indicated that both control and HOX11 derived

DCs express high levels (measured in percentage) of MHC molecubs.

Furthemore, both control and HOXI 1 DCs expressed moderate levels of the

macrophage/granulocyte marker CD1 1 b (Mac-1). normally expressed at levels

ranging from low to high on difterent DC subpopulations. They also expressed

moderate levels of costimulatory molecules CD80 (87-1). CD86 (87-2), HSA

and CD40. The cells did not express or expressed low levels of the T ceII

specific antigen, CD3 and B lineage specific marker 8220. (Fig. 3)

Table 3. Flow cytometric analysis of HOX11 and non-transgenic spleen derived DCs. Results are presented in percentage of cells that stained with specific antibody

Cells isolated from the bone marrow of HOX11 transgenic and

nontransgenic littermate mice and cultured in medium supplemented with GM-

CSF and 11-4 were also phenotypically characterized by flow cytometry

analysis. After 6 days, floating and easily detached cels from aggregates were

harvested, purified using the MACS system and stained with antibodies specific

for surface antigens reflecting DCs ability to function as APC.

Page 95: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Figures 3 and 4 Flow cytometric analysis of HOX11 transgenic and non-

transgenic DCs isolated from either spleen (Fig.3) or bone marrow

(FigA). Solid histogram represents unstained cells. while open and

shaded histograms represent HOXI1 transgenic and non-transgenic

derived DCs , respectively.

Page 96: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

HOX11 Spleen Derived Dendritic Cells

MHC I MHC II

HSA Mac-1 CD3 8220

Figure 3. Surface Phenotype Characterization

Page 97: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

HOX11 Bone Marrow Derived Dendritic Cells

MHC I MHC II HSA

Figure 4. Surface Phenotype Characterization

Page 98: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

HOX11 transgenic and control DCs stained strongly with rnAb to MHC class II

and HSA. Moderate levels of MHC class 1, CD80, CD86 and CD40 staining of

al1 DCs sam ples was noted. Staining with monocyte/macrop hage specific

antigen mAb, CDllb, was weak or undetectable. In addition, cultures

contained only rare B cells (8220) (Fig. 4). To further confirm these finding, a

larger group of mice should be used for isolation of DCs from bone marrow.

Table. 4 Flow cytometric analysis of HOX11 and non-transgenic bone marrow dedved DCs. Results are presented in percentage of cells that stained with specific antibody.

Oh cells

In summary, these studies indicated that we were able to isolate and

expand normal numbers of DCs from the spleen and bone marrow of HOX11

transgenic mice. DCs responded normally to stimulation by growth factors such

as GM-CSF. In addition, HOX11 derived DCs expressed normal levels of MHC

class I and class II molecules required for antigen presentation, costirnulatory

molecules such as B7-1, 87-2 and HSA required for the activation of naïve T

cells and the CD40 activation antigen.

3.3.3 HOXl 1 Derived DCs: Presentation of FoMgn MHC

The capacity of HOX11 derived DCs to prime naïve alfogeneic T cells in

vitro was assessed by measuring T cell proliferative responses to specific

foreign MHC presented by DCs. For this experirnent, HOX11 derived DCs were

used as stimulators (presenting their own MHC as an antigen), white C3H and

MHC I [ MHC II CDl lc , HOXI1 44% , Non-Tg ) 39%

15% 67% 56%

87-1 32%

12% 1 35%

87-2 9% 6%

CD40 31% 30%

HSA 92% 86%

Mac-1 8220 1% 5% 2% 3%

Page 99: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

C57BU6 derived T cells were used as allogeneic responder cells. As shown in

Fig. 5 stimulation of naïve C3H and C57BU6 derived T cells with HOX11

derived DCs (bred in the CD-1 genetic background) resulted in an 8-10 fold

increase in proliferation relative to the unstimulated control T cens. As a

positive control, C3H derived DCs (H-2*) were used to stimulate C57BU6 (H-

2 9 derived T cells. This stimulation resulted in a 9-12 fold increase in

proliferation relative to unstimulated T celk thereby suggesting that HOXll

derived DCs were similar to C3H derived DCs in their ability to present self-

peptides and activate allogeneic T cells. Negative controls in this experiment

included MLR in which HOX11 or C3H derived DCs were used to stimulate

autologous T cells. Under these conditions, no T cell proliferation was detected.

Similar results were obtained in three additional experiments (Fig . 5).

3.3.4 HOXI1 Derived DCs: Presentation of OVA Protein

Prior to initiating these experiments. we detemined that the optimum

conditions required to induce maximum proliferation of T cells were obtained

when 1 x I o5 DCsfml were pulsed with 60 pg/ml of OVA protein. To detennine

whether HOXI 1 derived DCs were able to present a foreign protein and induœ

a specific T cell proliferative response, OVA pulsed DCs were prepared and

used as stimulators in a MLR. Autologous T cells were isolated from two

HOXl1 transgenic and non-transgenic rniœ used as responder cells. The

results of the expenments showed that HOXI1 derived splenic DCs pulsed with

Page 100: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Figure.5 HOXll miœ (bred in the CD-1 genetic background) derived DCs

presenting their own MHC antigens. Spleen derived DCs were used as

stimulator cells and cultured with C3H (H-2k) and C57BU6 (H-2b) derived T

cells that served as responder cells. As a positive control, C3H derived DCs

were cultured with C57BU6 derived T cells, while as a negative control HOX11

C3H derived DCs were cultured with autologous T cells. Proliferation was

measured by the incorporation of PH] thymidine and reported as mean counts

per min with standard deviation. The same experiment was repeated three

times with similar results.

Page 101: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

PRESENTATION OF FOREIGN MHC BY HOX11 DERIVED DCs

Page 102: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Fig u re.6 HOXI 1 transgenic mice denved DCs presenting OVA protein. Spleen

derived DCs from HOXI 1 transgenic and non-transgenic (control) mice were

pulsed with OVA, irradiated and used as stimulator cells in MLR. T cells were

isolated from spleen and served as responder cells. Cultures were incubated for

5 days and T cells proliferative responses to OVA protein measured by

incorporation of [ 3 ~ ] thymidine. The same experiment was repeated three times

and error bars present the standard deviations among the experiments.

Page 103: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

CONTROL AND HOX11 DERIVEO DCs PRESENTING OVA PROTEIN FOR THE ACTIVATION OF AUTOLOGOUS T CELLS

Page 104: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

OVA protein induced 5-6 fold higher proliferation of T cells, relative to unpulsed

control DCs. In addition, no difference in the stimulatory responses between

HOX11 transgenic (5-6 fold) and non-transgenic mice derived DCs (6-7 fold)

was noted. This experiment was repeated three times with similar results. (Fig.

6)

3.4 Characterization of HOX1I Transgenic Mice Derived T Cells

3.4.1 Proliferation Assay (PHA Stimulation)

These experiments were initiated to compare the ability of normal and

HOXl l derived T cells to proliferate in response to mitogenic stimulation. A

number of agents can activate T cells resulting in cytokine production, cytokine

receptor expression and T cell proliferation. The mitogen used in these

experiments was phytohemagglutinin (PHA) which activates T cells by cross-

linking the TCR. TCR-negative cells will not respond to PHA. Ta test the ability

of HOXI 1 derived T cells to proliferate upon stimulation with PHA, T cells were

isolated from spleens from a total of 5 HOX11 transgenic and 5 non-transgenic

mice ranging in age from 3 to 18 months. T cell proliferation was assessed

using a standard PH] thymidine incorporation assay.

Data shown in Fig. 7 demonstrated that PHA stimulation induced an 80-

100 fold increase in proliferation of stimulated HOXI 1 derived T cells as

compared to unstimulated T cells. Furthemore, the proliferative ability of T

cells isolated from HOXI 1 transgenic mice was similar when compared to age

and sex matched non-transgenic mice (75-90 fold) (Fig. 7).

Page 105: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Figure 7. HOXI 1 transgenic mice denved T cells proliferative responses upon

the stimulation with PHA. Spleen derived T cells isolated from HOXI1 transgenic

and non-transgenic (control) mice were stimulated with PHA for 3 days and T cell

proliferation measu red by PH J thymidine incorporation. The same experiment

was repeated five times using miœ ranging in aga from 3 to 18 rnonth. Enor ban

present the standard deviations among the experiments.

Page 106: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

HOXl1 DERIVED T CELL PROLIFERATION ASSAY

( P W

Page 107: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

3.4.2 T Ceii Proliferative Responses to Foreign MHC

To assess the ability of HOX11 derived T cells to proliferate after

stimulation by foreign MHC antigens. T cells were isolated from spleens of

HOX11 transgenic and non-transgenic female mice and used as a responder

cell population in an allogeneic MLR. Each group consisted of three mice. C3H

derived, irradiated DCs (H-zk) were used as stimulator cells presenting foreign

MHC antigen. As shown in Fig. 8, HOXl l derived T celfs proriferated in

responds to allogeneic MHC antigens, presented by C3H derived DCs. Under

these conditions, a 12-15 fold increase in proliferation of T cells was detected

when compared with unstimulated T cells. As a positive control, C3H derived

DCs (H-2k) were used to stimulate C57BU6 derived T cells (H-2b). This

stimulation resulted in 1 3-1 7 fold increase in proliferation as compared with

unstimulated T cells and suggested that HOX11 derived T cells did not difFer in

their proliferative responses when compared with C57BU6 derived T cells. In

additional control experiments neither HOXl l transgenic nor C3H derived T

cells proliferated upon stimulation with autologous DCs (Fig. 8).

3.4.3 T Ce11 Proliferative Responses to OVA Protein

To address the question of whether HOXl1 transgenic mice derived T

cells were functionally capable of responding to specific foreign antigens. we

used DCs pulsed with OVA protein ta stimulate autologous T cells. The

proliferative responses of T cells isolated from two HOX11 transgenic and non-

transgenic mice were compared in MLR. AS shown in figure 9, HOX11 derived

Page 108: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Figure 8 HOX11 mice (bred in the CD-1 genetic background) derived T cells

responding to foreign MHC antigen. Spleen derived T cells were used as

responder cells. C3H (H-2k) derived DCs were used as stimulator cells

presenting their own MHC antigens. As a positive control, C3H derived DCs

were cultured with C57BU6 (H-2b) derived T cells, while as a negative control

HOX11 and C3H derived DCs were cultured with autologous T cells.

Proliferation was measured by the incorporation of [)HI thymidine and reported

as mean counts per min with standard deviation. The same experiment was

repeated three timas with similar results.

Page 109: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

HOX11 DERIVED T CELL PROLIFERATIVE RESPONSES TO

FOREIGN MHC

Page 110: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Figure 9 Proliferative responses of HOXI 1 transgenic mice derived T cells

to OVA protein in MLR. T cells were isolated from HOX11 transgenic and

non-transgenic (non-Tg) mice and cultured with autologous DCs pulsed

with OVA protein in the presence of DOTAP using different D0TAP:OVA

ratio. After 5 days proliferation of T cells was measured by the

incorporation of [ 3 ~ ] thymidine. The same expenment was repeated three

times with similar results. Error bars present the standard deviations

among the mice in each group.

Page 111: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

HOX11 DERNED T CELL PROLIFERATIVE RESPONSES TO DOTAP-

OVA PULSED DCs (pglml)

10 - 9 - 8 - 7 - 6 - 5 - 4 - 3 -

I HOX11 T cells O Non-Tg T cells

2 - 1 - O - m I

Page 112: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

DCs presenting OVA protein induœd 10 to 14 fold increase in proliferation of T

cells when compared to unstimulated T cells. MLRs were set up with cells

isolated from HOXl1 and non-transgenic mice. Furthemore, T cells isolated

from non-transgenic mice showed a 8-13 fold increase in proliferation upon

stimulation by OVA presenting DCs as compared to unstimulated T cells.

These results indicated that HOXI 1 derived T cells showed normal levels of

proliferation upon activation by foreign antigens. We also analyzed whether

HOXl 1 det-ived DCs could uptake OVA protein more efficiently in the presence

of lipid (DOTAP). When these data were compared to those from Fig. 6 the

results suggested that the lipid component was not essential to deliver protein

into DCs. This experiment was repeated three times with similar results, using

total of six mice in each group. (Fig. 9)

3.5 Measurement of Cytotoxic T Lymphocyte (CTL) Activity

3.5.7 ln Vitro CTL Assay

These studies were initiated to compare the ability of HOX transgenic and

non-transgenic (control) derived DCs to present OVA protein and generate

OVA-specific CTL responses in vitro. OVA protein is taken up by DCs and

degraded into the peptides. These are further processed and presented as a

class I or class II restricted peptides by MHC molecules for the activation of

CD8 and CD4 T cells. In our assays, no additional cytokines were added and

thus the CTL assay measured not only the ability of antCOVA specific CD8 T

cells, but also indirectly assayed the ability of OVA specific CD4 T cells to

Page 113: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

!Ilustration 3. In vitro CTL essay using HOXll transgenic mice derÏved

DCs pulsed with OVA protein. Picture 4A represents target cells (DCs

pulsed with OVA protein), 48 represents effector cells (cytotoxic T cells),

while picture 4C represents mixed effectorharget cell in attempt to induce

specific killing of target cells.

Page 114: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS
Page 115: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Figure 10 Induction of CTL response in vitro using DCs pulsed with OVA

protein in the presence of DOTAP. Spleen derived DCs from HOXll

transgenic, non-transgenic and C57BU6 mice were pulsed with OVA protein

for 18 hr, irradiated and used as stimulator cells. T cells were isolated from

spleen of 3 transgenic and non-transgenic mice and used as responder cells.

CTL assay was done after two rounds of stimulation and percentage of specific

lysis measured using different target cells (EG 7-OVA. DCs pulsed with OVA

protein and irrelevant EL4 cells) The same experiment was repeated three

tirnes with similar results. Error bars present standard deviation among the

experiments.

Page 116: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

IN WTRO CTL ASSAY USlNG OVA PULSE0 DCs

5 0 : l 20:1 1 O : l u n p u l s e d D C s

D C - O V A

50:1 20:1 1 O : u n p u l s e d ant i -H- D C s 2 K b

,*rH 0 x 1 1

N o r m a t

+ C 5 7 B L / 6

Page 117: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Target E.G.7-OVA I OC pulsed with OVA I EL4

T cells #1 + Table. 5 In vitro CTL assay using OVA protein pulsed DCs. Table

present ability of cytotoxic T cells to lyse specific target cells. Data in

percentage are presented for each mouse with average percentage for each

group of mice including HOX11, non-transgenic and C57BU6 rnice.

Page 118: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

produce cytokines required by cytatoxic T cells. T cells obtained from three

HOX11 transgenic and three non-transgenic mice were activateci in vitro with

OVA protein pulsed DCs and the resulting T cells evaluated for antigen specific

target cells lysates using the JAM test. Data presented in figure 10 showed that

both control and HOXI 1 transgenic cytotoxic T cells were similar in their ability

to lyse OVA protein pulsed autologous DC targets or E-G7-OVA target cells. In

contrast, there was no or littte response when CTL were rnixed with EL4 or

unpulsed autologous DC targets. Of importance, the anti-OVA cytolytic activity

of the effector T cells was blocked by the addition of a n t i - ~ - 2 ~ ~ mAb, indicating

that the CTL activity detected in these assays was MHC class I restricted (Fig.

1 O).

3.5.2 ln vivo CTL Assay

To test the in vivo ability of HOX11 transgenic DCs presenting foreign

peptides to induce CTL responses, HOXI 1 transgenic, non-transgenic and

C57BU6 mice (two mice in each group) were immunized on two separate

occasions with DCs pulsed with OVA protein. Mice were immunized by

intraperitoneal (1.P) injection of 5 x 10' irradiated OVA pulsed DCslml. with

seven day interval between injections. Sevan days after the second

immunization, mice were sacrificed and isolated splenocytes restimulated in

vitro with either OVA pulsed DCs or E.G7-OVA cells. Cytotoxic activity of

splenocytes was tested using autologous OVA pulsed DCs, E.07-OVA, EL4

Page 119: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Figure 11A and 11 B. Priming of OVA-specific CTLs in mice immunized with DCs

pulsed with OVA protein. Spleen and bone rnarrow DCs were isolated from

HOX11 transgenic. C578U6 and non-transgenic mice, pulsed with OVA protein,

irradiated and injected I.P. into the mice on two separate occasions. Seven days

after the second injections, splenocytes were restimulated in vitro either with DCs

pulsed with OVA or with EG7-OVA tumor cells for 5 days. Cytotoxicity was

measured using a JAM test and presented as percentage. This experiment was

done once using two mice in each group. Error bars represent standard deviation

within the each group.

Page 120: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

IN VlVO CTL ASSAY USlNG OVA PROTEIN PULSED DCs

(CTL RESTIMULATED IN WTRO WlTH OVA PULSED DCsL

Target OC pulseci with OVA

+C57BM

+ Normal

,+ HOX11

Target EL4 œlls

Figure 11A.

Page 121: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

IN VIVO CTL ASSAY USING OVA PROTEIN PULSED DCs

(CTL RESTIMULATED IN VITRO WlTH E.G7-OVA)

Target E.G7-OVA cells

2 5 3 12.5:1

Target: EL4 cells

Figure 11 B.

Page 122: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Non-Tg T L T

1 DCs pulsed with OVA

r r C

C L

- - -

-

-

-

Table. 6a In vivo CTL using DC pulsed with OVA (restimulated in vitm with DC-OVA)

1 Target

Table. 6b In vivo CTL using DC pulsd with OVA (mtimulatmî in vttn> with E.G.7-OVA)

Page 123: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

and autologous unpulsed DCs as targets. As shown in figure 11, HOXl1

derived DCs pulsed with OVA protein were able to induce a strong and specific

in vivo CTL response as rneasured by the percentage of lysed target cells (DC-

OVA pulsed and E-G7-OVA cells). A small percentage of target cells were

lysed in control samples. Since OVA protein is an H-2b restricted protein.

C57BU6 derived cells were used as positive controls in this assay. A

cornparison of the stimulation potential of HOX11 transgenic DCs with C57BU6

and non-transgenic mice derived DCs revealed sirnilar percentages of

cytotoxicity thus demonstrating similar functional ability of DCs and CTL among

different groups of mice (Fig.11).

3.6 Assessrnent of Immunogeneicity of HOXi 1

The ability of HOX11 protein derived peptides and HOXI1 tumor cell

lysates to induce an immune response in naïve mice was assessed using MRL

and CTL assays. To assess the immunogenicity of HOXl1 derived peptides

human HOX11 cDNA was used for the production of in vitro transcnbed (IVT)

HOXlf RNA, while tumor cells, isolated from a seventeen month old HOX11

mouse, served as source of the tumor lysates. It was demonstrated by

histopathofogical analysis that this mouse had splenic lymphoma with

dissemination of lymphoma cells to the lungs, kidney. liver and thymus.

Page 124: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

3.6.1 T Cell Proliferative Responses upon the Stimulation with /VT RNA

The ability of DCs pulsed with IV1 HOX11 RNA to stimulate C57BiJ6,

non-transgenic and HOXI 1 transgenic T cells was assessed in MLR assays.

Results using T cells from naïve. C57BU6 and non-transgenic miœ

demonstrated a 4 to 7 fold increase of T cell proliferation after stimulation with

DCs pulsed with IVT HOX11 RNA as compared to unstimulated T cells or T cell

stirnulated with unpuised DCs (Fig. 12)- This indicated that DCs were efficîently

pulsed with IVT HOXll RNA using previously described technique and

strongly suggested that the T cell proliferation was the result of the presence of

HOXl 1 peptide. It also suggested that HOXI1 derived peptides are suffciently

immunogenic to activate T cells in naTve mice. However. HOXl l derived T

ceils showed less than a 1 fold increase in proliferation as compared to 4-7 fold

increase in proliferation of C57BU6 or non-transgenic T cells. These results

suggested that T cells from HOXll transgenic mice were nonresponsive to

HOXI 1 protein, which is constitutively expressed by B cells in HOX11

transgenic mice.

Page 125: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Figure 12 T cell proliferative responses upon the stimulation with HOXl1 RNA

as tested by MLR. DCs isolated from spleen of HOXI 1 transgenic, C57BU6 and

non-transgenic rnice were pulsed with IVT HOX11 RNA, irradiated and cultured

with spleen derived T cells. After 5 days proliferative responses of T cells were

measured by the [ 3 ~ ] thymidine incorporation. The experiment was done once.

Page 126: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

MLR STIMULATED WlTH IVT HOX11 .. RNA PULSED DCs USED FOR

IMMUNIZATION IN VIVO

U T cells alone DC-HOX1 1 RNA

I D C unpulsed

T cell

Page 127: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

3.6.2 Induction of Primary CTL Response to IVT HOXI 1 RNA in Naïve Mice

Based on the findings from the in vitro experiments, we used an

experirnental system in which primary CTL responses to HOX11 derived

peptide were induœd and evaluated in vivo- Spleen and bone marrow DCs

were isolated from HOX11 transgenic. non-transgenic and C57BU6 mice and

pulsed with HOX11 RNA synthesized in vitro (IVT HOX11 RNA) from a plasmid

encoding the human HOXI1 cDNA. DCs pulsed with OVA protein served as a

positive control. Three groups of mice (five HOX11. six non-transgenic and six

C57BU6 mice) were immunized on two separate occasions with IVT HOXl l

RNA or OVA protein pulsed DCs to stimulate an in vivo HOX11 or OVA specific

CTL response. CTL activity was assayed using difTerent targets such as DCs

pulsed with IVT HOX11 RNA or K3P cells. As shown in figure 13, IVT HOX11

RNA pulsed DCs were capable of ïnducing HOX11 specific responses in naïve

C57BU6 and non-transgenic mice, although HOX11 transgenic DCs pulsed

with IVT HOX11 RNA did not induce a notable CTL responses. Our data

suggest that T cells from HOXll transgenic mice have either been deleted

during thymic selection or have been rendered non-responsive to HOX11-

derived peptides as a result of constitutive expression of the human HOXl l

transgene in B lymphocytes. (Fig. 13).

Page 128: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Table 7 In vivo CTL assay using OC8 pulsed with IVf HOX11 RNA. Cytotoxic T celi

ability of HOXl1 transgenic, C57BU6 and non-transgenic mice to lyse specific target

cells is measured in percentage and presented for each mouse used in this assay with

average for HOX11, C57BU6 and non-transgenic group of mice.

Target

E:T

C57BU6 T cells #1

#2

I #3

#4

Average % lysis

Non-Tg T cells #I

#2

#3

#4

Average % lysis

HOXIi T celIs #1

#2

#3

Average % lysis

DC pulsed with IVT -

K3P

12.5:1

13%

14Oh

18Oh

13Oh

15% '

13%

15%

19%

24Oh

18%

6Oh

0%

0%

2%

HOX11 RNA EL4

50:1

27%

38%

24%

29%

' 2511

14Oh

25%

21%

15%

30%

40%

27%

38%

33%

35%

4%

9%

9%

9%

19%

19%

18%

20°h

25%

21%

10%

1%

0%

4 O h

Page 129: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Illustration 4. Restimulation of splenocytes isolated from HOX11 transgenic,

non-transgenic and C57BU6 mice after immunization with DCs pulsed with IVT

HOX11 RNA. Splenocytes were restimulated in vifm with pulsed DCs.

Development of clusters presenting expanded anti-HOX11 specific T cells were

observed in cultures containing cells derived from C57BU6 (B) and non-

transgenic (C) mouse, but in a culture containing T cells from HOX11 mouse no

clusters were noted (A). Picture D present T cell clusters expanded from

splenocytes from HOXl 1 transgenic mouse immunized with DCs pulsed with

OVA and restimulated in vitro.

Page 130: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS
Page 131: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Figure. 13 in vivo induction of HOXll RNA-specific CTL in HOXlI transgenic

and naïve (C57BU6 and non-transgenic) mice immunized with DCs pulsed with

IVT HOXl l RNA. Spleen and bone marrow derived DCs were pulsed with IVT

HOXI 1 RNA, irradiated and injected i.p. into the mice on two separate occasions

(with the interval of 7 days). Seven days after the second injection. splenocytes

were restimulated in vitro with IVT HOXl l RNA pulsed DCs and pementage of

lysis measured by JAM test. DC pulsed with IVR HOXl1 RNA. E.G.7-OVA and

EL4 cells were used as target cells. This experiment was done once with total of

four C57BU6, four non-transgenic and three HOXl1 transgenic mica. Error bars

present standard deviations within the each group.

Page 132: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

CTL ASSAY AFTER IMMUNIZATION WlTH IVT HOX11 RNA PULSED

DCS

DCHOXll RNA

.r

40% - -. - 30% - 0 .- X 0 20% - O CI Target: EL4

B

e h -

Wh - .% 40% O

*-

:: Ph- u O C

rnh-

looh

OO/o 1 1 1

501 B.1 1251

E

Page 133: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

3.6.3 Induction of CTL Response upon Stimulation witb Tumor Lysates

This experiment was designed to compare the ability of T cells isolated

from HOXll transgenic mice with lymphoma and naïve C57BU6 mice to

generate CTL responses in vitro after stimulation with lymphoma antigens.

Since the expansion of DCs from bone rnarrow cells requires 6 days. it was

necessary to cryopreserve spleen cells as a source of T cells. To ensure that

CTL responses were fymphoma antigen specific response and not an artifact

induced by the freezing procedure, fresh T cells were isolated from the spleen

of a syngeneic C57BU6 mouse on the day the CTL assay was set up. In so

doing, we were able to compare fresh and frozen T cell responses. As shown

in figurel4, presentation of tumor antigens by C57BU6 derived DCs resulted in

the induction of CTL activity as measured by percentage of lyses of dïfFerent

target cells (DCs pulsed with either IVT HOXl1 RNA or tumor cells lysats).

However, a lymphoma antigen specific response was seen only in assays

using C57BU6 derived T cells but not when HOX11 transgenic rnouse derived

T cells were used. These results lead us to several conclusions. First, they

suggested that HOXl 1 transgenic mouse derived lymphoma ceIl lysates were

a good source of tumor associated antigens, which when presented by DCs.

were sufficiently immunogenic to induce T cell response in naïve mice. Second,

the low percentage of killing found in HOXll derived T cells support findings

from the in vivo experiments using IVT HOXI 1 RNA suggesting that HOX11

transgenic T cells have been rendered non-responsive to HOXI 1 derived

peptides. Furthemore, since the T cells used in above experiment were

Page 134: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

isolated from the teminally il1 mouse in which the lymphoma developed, the

results provide preliminary data suggesting that the process of

lymphomagenesis in HOX11 transgenic mice is associated with the induction of

a nonresponsive state of T cells specific for HOX11 antigens.

Target

E:T

HOX11 frozen T

cells C57BU6 frozen T

cells C57BU6 fresh T cells

Table 8 In vitro CTL assay using DCs pulsed with lymphoma cell lysates. Ability

of cytotoxic T cells from teminally il1 HOXt l transgenic mouse and control C57BU6

mice to lyse specific target cells is presented (in percentage).

DC pulsed with IVT HOXl i RNA

20:L

2%

10%

13%

DC pulsed with Lymphoma Cell

Lysates 80:l 40:l

EL4

9%

28%

25%

20:l

5%

12%

15%

8O:L

16%

30%

33%

80:l

5%

7%

1

5%

15%

17%

40:l

10%

24%

21%

40: 1

3%

2%

/

Page 135: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Figure. 14 Induction of C I L response N, vitro by DCs pulsed with lymphoma ceIl

lysates. Bone mancw derived DCs from tenninally il1 HOXl1 mouse and control

C57BU6 mice were pulsed with lymphoma cell lysates and used as stimulator

cells in CTL assay. T cells were isolated from spleens and used as autologous

responder cells. CTL assay was done after two rounds of stimulation using DC

pulsed with tumor cell lysates, DCs pulsed with IVT HOXll RNA and unpuised

DCs as target cells.

Page 136: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

IN VITRO CTL ASSAY USlNG DCs PULSED WlTH LYMPHOMA CELL LYSATES

Target- DC-HOXI 1 RNA

w I rn 80: 1 40:1 20: 1

Target- DC-Tum-Lys.

-C- HOX11 T cells

+CS7BU6 T cells

+C57BU6 f'fesh T cells

80:1 40:1

Target- unpulsed DCs

Page 137: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Chapter 4

Discussion and Future Experiments

4.1 Discussion

Non-Hodgkin's lymphomas are a heterogeneous group of disorders

characterized by malignant proliferation of subpopulations of B or T lymphoid

cells. The nurnber of new cases of NHLs has increased in Western countries

since 1960. More then 85% of NHL patients have advanced disease at the time

of initial presentation. Aggressive chemotherapy is the primary form of therapy

for h ig h and intemediate-grade NHLs. Since lymphomas disseminate widely

by hematogenous routes, radiation therapy is usually considered an adjunct to

chernotherapy rather than a primary treatrnent. Relapse rates post bone

marrow transplantation are also high making the overall prognosis for patients

with NHL poor, despite improvements achieved by cornbined radiation and

chernotherapy.

Given the increasing evidence that chemotherapy, radiotherapy, and

stem cell transplantation approaches do not cure the majority of patients with 6

cell lymphoma, new investigative treatrnents are being consideredl? Several

studies have shown that the host defense against tumors is reliant on the

immune systern, supporting a potential for cancer immunotherapylm-1. Most

cancer irnmunotherapeutic approaches are based on modification of the host

immune system to promote tumor eradication. Further, since most tumors are

poorly immunogenic and escape host immune detection much research has

Page 138: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

been focused on the identification of tumor specific antigens and the

modification of tumor cells to make them more immunogenic and more

susceptible to endogenous effector mechanisrns. Studies using animal models

and pilot human clinical trials are underway to determine whether potential

tumor specific antigens can indeed be recognized by CTLs and, consequently,

induce tumor eradication.

Areas of immunotherapy specifically directed at the treatrnent of 6 cell

lymphoma, which are currently being evaluated in clinical trials, incfude: (1)

vaccination with idiotype derived peptide epitopes presented on professional

APC, (2) vaccination with CD40 activated lymphoma cells and (3) adoptive

transfer of lymphoma specific T cells 166.167.

In addition to these strategies for cancer immunotherapy, DC-based

immunotherapy is rapidly emerging as a potentially viable alternative.

Increased interest in DCs has been based on the hypothesis that only

optimized antigen presentation by APCs will induce T cell activation and

suffîcient anti tumor immune responses in vivo to eradicate tumor cells. The

pivota1 role of DCs in the induction of cellular immune responses has focused

attention on DCs as a potentially powerful tool for enhancing anti-tumor

immunity. Results obtained in animal models. using mouse transplantable

tumor cells have demonstrated the effectiveness of tumor peptide pulsed DCs

in stimulating both naïve CD4 and CD8 T cells in vitro and in vivo 102,168.

Furthemore, immunization of mice with DCs pulsed with tumor lysates are able

to induce protective imrnunity against subsequent lethaf challenges with live

Page 139: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

tumor cells '05. In animais model studies using transplantable 6 cell lyrnphoma

a single immunization with DCs pulsed with tumor specfic idiotype protein was

shown to induce regression of tumors as well as protecting mice against future

inoculations with lethal doses of tumor cells169.

Results from human clinical trials in which patients received several D G

idiotype infusions dernonstrated clinical responses. However the therapeutic

benefits were less dramatic than would have been predicted based on the

results from murine studies 121 36%

One possible reason for the poorer than expected results obtained in

pilot clinical trials of DCs based immunotherapy is the discrepant pattern of the

animal lymphoma models currently used to assess DC based therapeutic

modalities, compared to human lymphoma. The best responses in animal

studies were in fact obtained when non-tumor bearing animals were immunized

with tumor antigen presenting DCs, pnor to tumor ~halIenge103*~05. When

immunization occurred after the establishment of the tumor, outcomes were

significantly poorer. Therefore, these murine transplantable tumor model

systems are not reflective of the process of tumor progression that occurs in

patients with NHLs in which tumor cells arise spontaneously, acquire

mutations, escape immune detection and invade distant sites. Thus model

systems are required in which tumors arise spontaneously and in which tumor

progression is no separated from the host immune systern and the local tumor

environment.

Page 140: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

A long terni goal of our laboratory research is to assess the applicability -

of DC based immunotherapeutic protocols for the treatment of non-follicular B

cell lymphoma. To achieve our goals we used a HOX11 transgenic rnouse

model. which was produced in our laboratory by introdwing a transgenic

vector, consisting of the human HOX11 cDNA under the transcriptional control

of the murine IgH promoter and enhancer, thus driving the expression of the

HOX11 transgene during the B lineage development. HOX1 1 transgenic rnice

were healthy at birth and analysis of cells isolated from bone rnanow, thymus,

spleen and lymph nodes revealed normal B and T lymphopoiesis. However,

eig hty-five percent of mice spontaneously developed lymphoma during their

second year of He. Lymphomas were of a mature B cell phenotype with the

spleen being the main organ of tumor development. The lymphomas developed

in HOXll transgenic mice were low grade, indolent lymphomas with slow

progression to a hig hly malignant phenotype. Moreover, the lymphomas were

clinically and morphologically similar to human splenic marginal zone

lymphoma (SMZL) (as determined by Dr. Megan Lim, Sunnybrook 8 Women'

College Health Science Centre). Patients with SMZL are initially asymptomatic,

but at the tirne of clinical presentation, the disease has usually disseminated,

which is associated with a very poor prognosis even after aggressive

chemotherapy. Therefore, HOX1 1 transgenic mice provide a clinically relevant

model for a subtypa of human indolent lymphoma, which will be a powerful

systern to assess novel approaches for the treatment of NHL.

Page 141: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

Given the complexity of currently available animal models, the main

advantage of this model is the predictable spontaneous development of low to

intermediategrade mature 6 cell lymphomas with a premalignant phase k ing

detected as earîy as four weeks of age. This animal model may serve as a

unique pre-clinical model to study immunotherapeutic approaches to lymphoma

therapy.

Normal APC and T cell functioris are essential for induction of proper

antitumor responses in vivo. It was therefore necessary to carefully

characterize the functional properties of the immune system of HOXll

transgenic miœ before proceeding with studies designed to asses DC based

irnmunotherapeutic protocols for lymphoma therapy. Several reasons lead us

to select this strategy. First, it haî been well documented that although IgH

promoter and enhancer sequences prirnarily direct expression throughout B

cell developrnent, leaky expression in T lymphocytes and myeloid cells is

frequently seen. Therefore it was critical to assess whether HOX11 transgenic

T cells and DCs were functionally cornpetent. Second, the HOXI1 gene is

nonnally not expressed in T and B cells and the ectopic expression of this

transcription factor could activate expression of different cytokines or adhesion

molecules that could perturb the ability of HOXI1 transgenic mice to induce an

immune response. For example, abundant expression of 11-10, a potent

immunosuppressive cytokine, may prevent DC accumulation and antitumor

immunity, as demonstrated in some animal models170. Based on this

Page 142: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

knowledge. we analyzed the functional properties of HOX11 derived DCs and T

cells.

Characterization of HOXI 1 Derived DCs

DCs were isolated from spleen and bone rnarrow and ouf analysis

demonstrated that HOXI1 transgenic mice had nonal numbers of DCs. These

DCs could be expanded in culture suppleniented with GM-CSF and 11-4. Under

these conditions Î t was possible to reliably obtain large numben of HOXl l

DCs, further indicating that HOXll DCs showed a nomal proliferative

response to cytokines.

Phenotype Characterization

It is well established that proper antigen presentation and T cell

activation are dependent on the expression of specific cell surface antigens by

DCs. FACS analysis was therefore used to characterize the phenotype of

HOXl 1 transgenic mouse derived DCs171-173. Splenic DCs isolated from

HOX11 transgenic rnice of different ages showed a typical DCs surface antigen

phenotype when compared to DCs isolated from non-transgenic mice. These

cells were mainly mature DCs as demonstrated by the presence of suiface

markers specifically expressed at that stage (CDllc, CD80, CD86, CD40,

MHC class I and class II). Phenotypic analysis of bone mamw derived DCs

revealed several DCs subpopulations in both control and HOXI 1 transgenic

mice. These included subpopulations of DCs expressing high levels of MHC

class II and HSA molecules, moderate levels of MHC class 1, CD80 and CD86

Page 143: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

molecules. and low levels of CD1 l c and Mac-1. These expression profiles are

typical of an immature stage of DC development

These results are important with respect to both spleen and bone marrow

derived DCs. since MHC class I and class II molecules are necessary for the

antigen presentation and consequently activation of CD8 and CD4 T cells,

respectively. Furthemore. CD80 and CD86 are CO-stimulatory molecules also

required for T cell activation. while CD40 is required for DC activation and

maturation. Thus, the results document that the profile of antigens expressed

by HOXll mice DCs is consistent with a normal OC phenotype. Some

exampies of differences in levels of expression were seen among HOX11

transgenic mice, but this was also observed among normal mice, and therefore

probably represents normal variability of expression of cell surface antigens

detected on DCs isolated from different mice.

Presentation of MHC in Allogeneic MLR

The main objective of the next experiment was to evaluate the ability of

DCs to present antigens using allogeneic MLR. HOX11 derived DCs

(stimulator) were cultured with either C3H or C57BU6 derived T cells

(responder). The proliferation ability of responder cells were wmpared with

those obtained from cultures containing C3H derived DCs (stimulator. H-2k)

and C57BU6 T cells (responder. H-2b). When HOXll derived DCs were

compared with C3H denved DCs with respect to their ability to stimulate

C57BU6 T cells, similar results were observed. Since this assay requires OC to

be able to present their own MHC proteins and present them at the ce11 surface

Page 144: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

for the activation of allogeneic T cells, our resuîts indicate that HOX11 derived

DCs are functionally competent in their ability to process and present antigens.

Presentation of OVA Pmtein in MLR and CTL Assays

Both in vitro and in vivo expenmental systems were next used to assess

the ability of HOX11 derived DCs to present foreign antigens. Recently, several

studies showed that DCs can present antigenic epitopes denved from

exogenous antigens onto MHC class I molecules and induce an antigen

specific CTL response when pulsed with soluble proteins such as OVA

proteinla*.

The efficiency of HOXl1 denved DCs to present foreign antigens was

first tested in syngeneic MLR assays. in which OVA protein was presented by

HOXl 1 transgenic mice dedved DCs to autologous T cells. The results

demonstrated the ability of HOXll derived DCs to present OVA protein by

inducing 4-6 fold higher proliferation of stimulated T cells when compared to

non-stimulated T cells. Also, when these results were compared with those

obtained with DCs derived from non-transgenic mice, no differences were

found. Since protein loading of DCs requires uptake, intracellular processing

and presentation by both MHC class I and class II molecules. HOXI1

transgenic derived DCs demonstrated a nomal ability to present both class I

and class II restricted antigens and subsequently activate CD4 and CD8 T

cells.

In vitro CTL

Page 145: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

To assess the induction of MHC class I restricted CTL responses by

HOX11 derîved DCs, transgenic and control DCs were pulsed with OVA protein

and used to stimulate autologous T cells using in vito CTL assaysW

Expanded OVA specific T cells were able to lyse OVA antigen expressing

targets. whereas M e or no lysis were detected if T cells were expanded with

unpulsed DCs or when an irrelevant tumor cell line was used as a target cells.

Furthemore, anti-OVA cytolytic activity of these effedor T cells was blocked by

the addition of an t i -~ -2~ rnAb and blocking the presentation of OVA by DCs,

indicating that these CTLs were indeed MHC class I restricted. Therefore, our

results demonstrated that HOXI 1 derived DCs can be ef'ficiently pulsed with

OVA protein and used as antigen presenting cells to activate the cytolytic

activity of antigen specific CTL cells.

ln vivo CTL

In vivo experiments were undertaken to examine the capacity of HOX11

transgenic derived DCs ta present OVA protein and induce antigen specific,

primary immune responses. It is well demonstrated that bone rnarrow derived

DCs can present exogenous OVA protein and induce high level of CTL

responses in vivo and protective antitumor immunityl74. Moreover, the

identification of MHC class I (H-2b) restricted be presented by MHC class I

molecules and activated cytotoxic T cells10g. epitope that is recognized by CD8

CTL in C57BU6 mice indicated that OVA antigens can For the current study,

HOX11 transgenic, non-transgenic and C57BU6 mice were immunized on two

separate occasions with syngeneic spleen and bone marrow derived DCs

Page 146: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

pulsed with OVA protein. Similar levels of CTL adivity were observed among

HOXI 1, C57BU6 and non-transgenic mice. These results are in accordance

with our in vitro results and confimi that HOXi1 derived DCs are indeed

efficient antigen presenting cells. capable of inducing OVA specific CTL

responses in HOXl1 transgenic rnice.

Characterization of HOX11 Derived T Celis

Proliferative Response to PHA

For our studies it was neœssary to assess the functional properties of T

cells isolated from HOX11 transgenic mice, despite the fact that it was

previously reported that thymocyte development was unperturbed in HOXI1

mice with phenotypically normal T cell subpopulations in the thymus and

periphery. In wntrast to cytotoxicity, proliferation is not a specific effector

function of T lymphocytes, although it has been widely used to assess the

overall immunocompetence of an animal. One of the methods to study T cell

proliferative responses involves the stimulation of T cells with mitogenic

stimulators. T cells isolated from HOXI 1 transgenic and non-transgenic mice

were compared with respect to their proliferation responses to PHA. Our results

demonstrated no difference in the proliferative ability between non-transgenic

and HOXI 1 transgenic derived T cells.

Proliferative Response to Foreign MHC

To evafuate more specifically T cell proliferative responses, allogeneic

MLRs were used. in which T cell proliferation was assessed upon the

Page 147: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

stimulation with allogeneic DCs. HOX11 transgenic and C57BU6 (H-2b) derived

T cells were cultured with C3H (H-2k) derived DCs with similar levels of

proliferation between the two groups of mice being observed. Ttius, this result

further supports the conclusion that HOXI 1 transgenic T cells are functionally

competent.

ProMerative Responses to OVA Protein in MLR and CTL Assays

T cell proliferative responses were also evaluated using DCs pulsed with

OVA protein as a stimulator. This is a more specific assay than allogeneic MLR

because foreign protein presented by both classes of MHC molecules on DCs

must be recognized by autologous T cells for activation of CD4, which secrete

cytokines required for the activation CD8 T cells. HOXI 1 transgenic and non-

traiisgenic derived T cells were used as responder cells. which were stimulated

with OVA pulsed autologous DCs. The results demonstrated comparable

proliferative responses between HOX11 transgenic and control rnice. Thus

HOXl1 transgenic derived T cells are normal with respect to their proliferative

responses upon stimulation with foreign antigens.

In vitro CTL

CTL activity of HOX11 transgenic derived T cells was further detennined

by in vitro and in vivo assays. Since cytolyüc activity may be induced by a large

number of antigens, we used OVA protein rather than a single OVA peptide

epitope to pulse autologous DCs and stimulate production of OVA specific CTL

responses in vitro. HOXI 1 transgenic and non-transgenic derived T cells were

used to expand sufficient number of CTLs by Wo rounds of stimulation with

Page 148: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

pulsed DCs. All mice used in these assays had been backcrossed for more

then 4 generation ont0 C57BU6 background. since OVA protein is specific for

that strain of mice. The results indicated that primed cytotoxic T cells from

HOX11 transgenic mice had a similar ability to lyse OVA expressing target cells

as was found with normal and C57BU6 derived cytotoxic T cells. In contrast,

there was little or no response when irrelevant target cells were used.

Combined, these data support the conclusions drawn from allogeneic MLR,

that T cells derived from HOX11 transgenic mice are functionally competent to

induce an immune response against foreign antigens.

In vivo CTL

For in vivo assessrnent of the generation of anti-OVA CTL activity,

HOXl 1 transgenic derived T cell HOXI 1, non-transgenic and C57BU6 mice

were immunized on two separate occasions with spleen and bone marrow

derived DCs pulsed with OVA protein- These immunizations resulted in

activation of OVA specific T cells in each mouse group, which were shown in in

vitro assays to be capable to lysing different target cells expressing OVA. The

generation of OVA specific CTL activity requires the activation, in vivo, of both

CD4 and CD8 T cells by MHC class I and class II restricted peptides. These

observations reflect normal functions of HOXl 1 derived T cells.

Combined, these data indicate that HOXli DCs and T cells from healthy

mice are phenotypically and functionally normal in their ability to process.

present and respond to foreign antigens. Furthermore, the expression of the

HOX11 transgene and lymphoma development do not compromise these cells,

Page 149: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

supporting our hypothesis that HOX11 mice will provide a very powerful model

s ystem in wh ich to assess DCs based immunotherapeutic protocols.

Assessrnent of Immunogenicity of HOX11

Other critical questions addressed in this study are: (1) whether HOXj1

protein derived peptides are sufficiently immunogenic to induce HOXf 1 specific

immune response in naïve mice and (2) whether HOX11 iymphoma

progression in HOXl l transgenic mice is associated with the induction of

Iymphoma antigen-specific T cell tolerance.

Induction of Immune Responses in Naïve Mice

To address the first question, C57BU6 and non-transgenic mice were

injected on two separate occasions with bone manow or spleen denved DCs

pulsed with IVT HOX11 RNA. These RNA pulsed DCs were capable of

stirnulating primary CTL responses in vivo, inducing killing of target cells

expressing HOXl l (K3P) and autologous DCs pulsed wlh IVT HOX11 RNA.

This result indicates that HOXI 1 derived peptides are sufficiently immunogenic

to induce specific T cell responses. In addition to IV1 HOX11 RNA. tumor

lysates (from terminally il1 HOXI1 transgenic mouse) were used to stimulated

C57BU6 derived DCs. These tumor lysates presenting DCs induced a CTL

immune response as detemined by in vitro CTL assays. Data suggested that

CTL specific proliferative responses were induced and these cells were

capable of lysing autologous DCs pulsed with tumor Iysates. The main finding

of this expenment was that tumor cell developing in HOXI1 transgenic mice

Page 150: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

can serve as effective immunogens for the presentation of MHC class I and

class II restricted antigens. which may function for the activation of both CD4

and CD8 T cells. However. this was shown only in vitm using a small group of

C57BU6 mice, and additional in vivo expenments must be done to draw a firm

conclusion.

Induction of immune Responses in HOX1 i Transgenic Mice

To address the second question, similar experiments to those described

above were used to evaluate the induction of antigen specific CTL responses in

HOX11 transgenic mice upon the stimulation with IVT HOXll RNA and tumor

lysates pulsed DCs. For in vivo studies. HOXll transgenic mice were

immunized with IVT HOXI I RNA pulsed DCs on two separate occasions and

results indicated that HOXll derived T cells were not activated by DCs

presenting HOXl l protein derived antigens. with no induction of antigen-

specific CTL being observed. These findings were predictable given that the

HOX11 protein is constitutively expressed by B cells in transgenic mice and

would thus represent a 'self" protein. Since previous experiments

demonstrated the immunogenic~ of the HOXll protein, one possible

explanation for this result is that anti-HOX11 specific T cells were either deleted

during thymocyte development of rendered non-responsive.

To address the question of whether lymphoma progression in HOXl l

transgenic mice is associated with toleration of lymphoma antigen specific T

from a terminally il1 HOXl1 transgenic mouse with lymphoma were stimulated

in vitro with autologous tumor lysates pulsed DCs. There was no expansion of

Page 151: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

specific CTL in culture and antigen specific target cells lysis was not detected.

Although this expenment neeâs to be expanded to include a large? number of

HOXll transgenic mice with lymphoma with both in vitro and in vivo assays

used, this result is of significance because of its accordance with our in vitro

results. lt indicates again that lymphomagenesis in HOX11 miœ is associated

with the toleration of anti-lymphorna antigen-specific T cells. HOX11 mice thus

represent a clinically relevant madel in which to assess novel therapeutic

approaches for the treatrnent of indolent nonfollicular lymphoma.

Studies proposed by this master's thesis advanced our understanding

regarding the relationship between expression of HOX11 transgene and

inductior: of antitumor immune responses in HOX11 transgenic miœ. Also,

these studies helped us reveal that HOX11 protein derived peptides and

lymphoma antigens are sufficiently immunogenic to induce immune responses.

Finally, these studies have provided preliminary data with respect to a possible

mechanisrns by which lymphomas develop in HOXl1 transgenic mice leading

likely to non-responsive state in anti-tumor specific HOX11 derived T cells.

4.2 Future Studies

Numerous studies have shown that preventative imrnunization before the

inoculation of tumor cells into healthy rnice can be highly effective at inducing

tumor specific immune responses, resulting in rejection of tumor cells.

However, vaccination of a tumor-bearing host in the late stage of tumor

Page 152: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

developrnent usually fails to induce specific immunity. These findings lead to

the conclusion that specfic mechanisms are used by malignant cells, which are

responsible for a lack of efficient immune response. One possible mechanism

of tumor evasion is the development of antigen-specific T cell anergy, which

may occurs as an early event during tumar progression leading to a deficient

antigen-specîfic T cell responsesl?? Toleranœ to tumor antigens may result

from a decreased clonal frequency (peripheral deletion) or the induction of

proliferative unresponsiveness (clonal anergy). In addition, self-antigens may

be the most relevant and abundant antigens expressed by malignant cells and

the host's immune system must be tolerant of these antigens. The same

rnechanisms that mediate toleranœ to self-antigens may represent a potential

barrier for the full development of effective immune responses against antigens

expressed by tumors. In the current study, antigen-specific T cell toleranœ to

HOXI 1 derived peptides was observed since HOXl1 denved T cells were not

activated by autologous HOXI 1 RNA. In addition, DCs pulsed with lymphoma

cell lysates also failed to induce immune response. However, to better evaluate

the potential immunogenicity of tumor lysates isolated from HOX11 transgenic

mice with lymphoma, more studies need to be done.

(1) Future experiments will include injection of HOXl1 denved DCs pulsed with

lymphoma cell lysate into Young, healthy HOXI1 transgenic recipients. If

HOXI 1 lymphomagenesis is associated with the generation of lymphorna

associated antigens, recipient mice will mount Ipphoma-antigen specific

CTL responses. These studies will suggest the existence of different

Page 153: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

antigens. in addlion to HOX11, presented on lyrnphoma cells and that the

lack of an immune response in sick rnice is due to the toleranœ of

lymphoma antigen specific T cells.

(2) There is evidenœ to suggest that CTLA4 (negative regulator of T cell

function that binds to the 67 molecule) activation result in the down

regulation of an immune response. Furthemore, the blockade of C T W

can lower the fhreshold for T ceH activation. Moreover, in vivo blockade of

CTLA4 may induce the regression of established tumors. We will design

future studies using CTLA4 blocking antibodies in an attempt to reverse

the non-responsive state of lymphoma antigen specific T cells in HOX11

transgenic rnice. HOX11 transgenic mice will be injected with CTLA-4

blocking antibodies and then immunized with tumor lysate or IVT HOX11

RNA pulsed DCs, while some mice will receive only pulsed DCs. We

anticipate that these manipulations will also increased levels of anti

lymphoma immune responses in HOXI 1 transgenic mice receiving CTLA-4

and antigen pulsed DCs.

(3) Different experiments will be initiated to detenine whether the anergic

state in HOX11 transgenic mice with lymphoma can be reversad by

culturing tumor antigen presenting DCs and T cells with various cytokine

combination (11-2, 11-12 or INF-y). We will also detemine whether the

injection of CD40 activation antibody facilitates the in vivo presentation of

lymphoma associated antigens and the activation of specific T cells

Page 154: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

These experiments should provide important data, which wRI assist in the

developrnent of immunotherapeutic protocols for the treatrnent of NHL.

Page 155: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

4.3 Summary

We have demonstrated that DCs isolated from HOX11 transgenic mice

show a typical DCs surface antigen phenotype when cornpared to DCs isolated

from normal mice. In addition. we used two stringent experimental systems to

assess the antigen presenting ability of HOXII derived DCs: MLR and CTL

assays. The data obtained suggest that HOX11 derived DCs are able to

present both foreign MHC antigens and OVA-protein. Consistent with this are

the in vivo findings that HOXI 1 mice injected with OVA pulsed DCs are able to

activate both MHC class I and class II restricted OVA specific T cells. HOXl1

derived T cells show normal overall proliferative responses (PHA) and normal

proliferative responses to foreign MHC. These cells are also able to proliferate

upon stimulation with autologous OVA protein pulsed DCs.

We also demonstrated that HOX11 peptides or lymphoma cell lysates

are sufficiently immunogenic to induce specific immune responses in naïve

mice, since irnmunization with pulsed DCs induced proliferation of CTLs able to

lyse target cells. However, HOX11 derived T cells were not activated in vivo by

DCs pulsed with IVT HOX11 RNA. Moreover, DCs pulsed with lymphoma cell

lysates failed to activate autologous T cells isolated from the HOX11 mouse in

which the lymphoma developed. These data suggest that lymphoma

developrnent in HOX11 mice is associated with the tolerization of lymphorna

antigen specific T cells. Overall, these studies pave the way to use HOXll

transgenic mice modal to develop DC based immunotherapeutic protocols for

the treatrnent of spontaneously aflsing indolent nonfollicular lymphomas.

Page 156: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

References

1. Ogawa M: Differentiation and proliferation of hematopoietic stem cells.

Blood 81 : 2 W , 1993

2. Cohen JJ, al. e: Apoptosis and programmed cell death in immunology.

Ann Rev lmmunol 1 O:Z67,1992

3. Ames BN: Identifying enviromental chernicals causing mutations and

cancers. Science 204587, 1979

4. Kennedy AR. Cairns J, Little JB: Timing of the steps in transformation of

C3H 10TlQ by X-irradiation. Nature 307:85, 1984

5. Dalla-Favera R. Bregni M. Erikson J, Patterson D, Gallo RC. Croœ CM:

Human c-myc onc gene is located on the region of chromosome 8 that is

translocated in Burkitt's lymphorna cells. Proc Natl Acad Sci 79:7824, 1982

6. Pelicci PG. Knowles DMd, Magrath Il Dalla-Favera R: Chromosomal

breakpoints and structural alterations of the c-myc locus difier in endemic and

sporadic forms of Burkitt lymphoma. Proc Natl Acad Sci U S A 83:2984, 1986

7. Hirama Tl Koeffier HP: Role of the cyclin-dependent kinase inhibitors in

the development of cancer. Blood 86:841, 1995

8. Knowles DM: Neoplastic Hematopathology, Williams and Wilkins. 1992

9. Chappuis PO, Sappino AP: Update: lymphoid neoplasms-molecular

characterizations of non- Hodgkin's lymphomas: impact on patient

management. Sernin Hematol32:237, 1995

10. Knapp W, et. al: Flow cytometric analysis of dl-surface and intracellular

antigens in leukemia diagnosis. Cytometry 1 8: 1 87. 1 994

Page 157: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

11. Rappaport H: New concepts in the classification of malignant

hemopathies. Bull Cancer 61 :Il, 1974

12. Harris NI Jaffe El Stein H, Banks P. Chan JI CLeary M. Delson G, De

Wolf-Peeters C, Falini B. Gatter KI Grogan Tl lsaacson P. Knowles Dl Mason

D, Muller-Hemelic H, Pilery S, Piris M. Ralfkiaer E, Wamke R: A tevised

European-American classification of lymphoid neoplasms: a proposal from the

International Lyrnphoma Study Group. Blood 84: 1361, 1994

13. Borïng CC, Squires TS. Tong T, Montgomery S: Cancer statistics, 1994.

Cancer J Clin 447,1994

14. List A, Greer JI Cousar J. Stein R, Flexner J, Sinangil F, Davis J, Volsky

D. Purtilo D: Non-Hodgkin's lymphoma after treatrnent of Hodkin's disease:

association with Epstein-Barr virus. Ann lntern Med 105:668, 1986

15. Kinlen L: lmmunosuppressive therapy and acquired immunological

disorders. Cancer Res 52 (suppl):5474, 1992

16. Levine A: Aquired immunodeficiency syndrome-related lymphomas.

Blood 1 1 180:8, 1992

17. Mounter PJ, Lennard AL: Management of non-Hodgkin's lymphomas.

Postgrad Med 75:2, 1999

18. Rohatiner A, Lister TA: Management of follicular lymphoma. Curr Opin

Oncol 6:473, 1994

19. von Gunten CF, Gordon LI: Treatment of advanced, aggressive non-

Hodgkin's lymphoma. Curr Opin Oncol 6:480. 1993

Page 158: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

20. Paryani S. Hoppe RT, Burke JS. Sneed P. Dawley 0, Cox RS,

Rosenberg SA. Kaplan HS: Extralymphatic involvement in diffuse non-

Hodgkin's lymphoma. J Clin Oncol 1 :682, 1983

21. Philip Tl Guglielmi C. Hagenbeek A. Somen R, Van der Lelie Hl Bron Dl

Sonneveld P. Gisselbrecht Cl Cahn JY, Harousseau JL. et al.: Autologous

bone marrow transplantation as mmpared with salvage chernotherapy in

relapses of chemotherapy-sensitive non-Hodgkin's lymphoma. N Engi J Med

333:1540, 1995

22. van Besien K. Sobocinski KA, Rowlings PA. Murphy SC, Amitage JO,

Bishop MR, Chaekal OU. Gale RP. Klein JP. Lazarus HM, McCarthy PL. Jr.,

Raemaekers JM. Reiffers J. Phillips GL. Schattenberg AV, Verdonck LF. Vose

JM, Horowitz MM: Allogeneic bone manow transplantation for low-grade

lymphoma. Blood 923832. 1998

23. Prehn RT. Main MJ: lmmunity to methylcholanthrene-induced sarcorna.

J Natl Cancer lnst 1 8:769, 1957

24. Herlyn M. Koprowski H: Melanoma antigens: immunological and

biological characteriration and clinical significance. Ann Rev Immun 6283.

1988

25. Urban JL, Schreiber H: Tumor antigens. Ann Rev lmmunol lO:617. 1992

26. Tsang KY, Zaremba S. Nieroda CA. Zhu MZ. Hamilton JM. Schlom J:

Generation of humao cytotoxic T cells specific for human carcinoembryonic

antigen epîtopes from patients immunized with recombinant vaccinia-CEA

vaccine. J Natl Cancer lnst 87:982. 1995

Page 159: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

27. Teny WD, Henkart PA. Coligan JE, Todd CW: Carcinoembryonic

antigen: characterization and clinical applications. Transplant Rev 20:100, 1974

28. Bhatnagar J. Tewari HB. Bhatnagar M. Austin GE: Comparison of

carcinoernbryonic antigen in tissue and serum with grade and stage of colon

cancer. Anticancer Res l9:2I8l1 1999

29. lmada T, Rino Y, Takahashi M. Hatori S, Shiozawa M. Amano 1, Kondo

J: Serum CA 19-9, S M . STN and CEA levels of the peripheral and the dnining

venous blood in gastric cancer. Hepatogastroenterology 462086. 1999

30. Sutterlin M, Bussen S, Trott S. Caffier H: Predictive value of CEA and

CA 15-3 in the follow up of invasive breast cancer. Anticancer Res 19:2567.

1999

31. Kwak LW. Campbell MJ. Czerwinski DK, Hart S, Miller RA, Levy R:

Induction of immune responses in patients with 6-cell lymphoma against the

su rface-imm unoglobulin idiotype expressed by their tumors. N Engl J Med

327: 1209, 1 992

32. Burnet FM: The concept of immunological surveillance. Prog Exp Tum

Res 13:1, 1970

33. Kohler G, Milstein C: Continuous cultures of fus4 cells secreting

antibody of predefined specificity. Nature 256:495, 1975

34. Milstein C, Adetugbo K. Cowan NJ, Kohler G, Secher DS, Wlde CD:

Somatic cell genetics of antibody-secreting cells: studies of clonal

diversification and analysis by cell fusion. Cold Spnng Harb Symp Quant Biol

41:793, 1977

Page 160: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

35. Reth M: Antigen receptors on 6 lymphocytes. Ann Rev lmmunol 1 O:97,

1992

36. Clark EA. Lane PJL: Regulation of hurnan B e l l activation and

adhesion. Ann Rev lmmunol9:97,1991

37. Dillman RO: Antibodies as cytotoxic therapy. J Clin Oncol l2:l497, 1994

38. Vijayasarad hi S, Houghton AN: Purification of an autoantigenic 75-kDa

human melanosomal glywprotein. Int J Cancer 47:298. 1991

39. Goodman JW, Sercan EE: The complexity of structures involved in T

cell activation. Ann Rev lmmunol 1 :465, 1983

40. Janeway CA: The T cell receptor as a multicomponent signaling

machine: CD4lCD8 coreœptors and CD45 in T cell activation. Ann Rev

lmmunol 10:645.1992

41. Podack ER, Kupfer A: T cell effector functions: Mechanisms for delivery

of cytotoxicity and help. Ann Rev Cell Biol 7:479. 1991

42. El-Khatib M. Stanger BZ. Oogan Hl Cui H, Ju ST: The molecuiar

mechanism of FasL-mediated cytotoxicity by CD4+ TH1 clones. Cell

lmmunology 163:237,1995

43. Tschopp J, Nabholz M: Perforin-mediated target cell lysis by cytolytic T

lymphocytes. Annu Rev lmmunol8:279, 1990

44. Yuong LHY, Lui C-Cl Joag S. Rafii S. Yuong JD-E: How lymphocytes

kill. Ann Rev Med 41 :45. 1990

Page 161: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

45. van der Bruggen P. Traversari Cl Chomez P: A gene encoding an

antigen recognized by cytolytic T lymphocytes on a human melanoma. Science

254: 1643.l991

46. Yron 1. Wood TA. Spiess PJ. al. a: In vitro growth of murine T cells. The

isolation and growth of lymphoid cells infiltrating syngeneic solid tumors. J

lmmunol l25:238.1980

47. Roder J: Immune respons: killing cornes naturally. Cun Biol 1 :242, 1 991

48. Nathan CF, Murray HW. Cohn ZA: The macrophage as an effector cell.

N Engl J Med 303:622, 1980

49. Henans IF, Daish A. Yang JI Ritchie DS. Ronchese F: Antigen

expressed on tumor cells fails to elicit an immune response, even in the

presence of increased num bers of tumor-specific cytotoxic T lymphocyte

precursors. Cancer Res 58:3909. 1998

50. Restifo NP. et. al: Identification of human cancers deficient in antigen

processing. J Exp Med 177:265, 1993

51. Urban JL, Kripke ML. Schreiber H: Stepwise immunologie selection of

antigenic vatiants during tumor growth. J lmmunol 137:3036, 1986

52. Howard J: Supply and transport of peptides presented by class I MHC

molecules. Curr Opin lmmunol7:69. 1995

53. Hiltbold EM, Ciborowski P. Finn OJ: Naturally processed class II epitope

from the tumor antigen MUCI primes human CD4+ T celis. Cancer Res

58:5066, 1998

Page 162: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

54. Schultze J, Nadler LM, Gribben JG: B7-mediated costimulation and the

immune response. Blood Reviews 10:111, 1996

55. Green JM, et. al: Absence of 674ependent responses in CD28deficient

mice. lmmunity 1 501, 1994

56. Bluestone J: New perspectives of CD28-B7niediated T cell

costimulation. lmmunity 2:555. 1995

57. Zitvogel L, Mayordomo JI, Tjandrawan T, D e k o AB, Clarke MR. Lotze

MT, Storkus WJ: Therapy of murine tumors with tumor peptide-pulsed dendntic

cells: dependence on T cells. 87 costimulation, and T helper cell 1-associated

cytokines. J. Exp. Med. 183:87, 1996

58. Wahl SM: Transforming growth factor beta: The good. the bad, and the

ugly. J Exp Med 180: 1587, 1994

59. De Smedt T, van Mechelen M, De Becker G. Urbain JI Leo O, Moser M:

Effect of interleukin-1 O on dendritic cell maturation and function. Eur J lmmunol

27: 1229, 1997

60. Murase Tl Jirtle RL, B. MG: Transforming growth factor-beta plasma

concentrations in patients with leukemia and lymphoma receiving

chemoradiotherapy and marrow transplantation. Blood 83:2383, 1994

61. Benjamin O, Knobloch TJ, Dayton MA: Human B-cell interleukin-10: B-

cell lines derived from patients with acquired irnmunodeficiency syndrome and

Burkitt's lymphoma constitutively secrete large quantities of interleukin-10.

Blood 80: 1289, 1992

Page 163: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

62. Singer CR, Goldstone AH: Clinical studies of ABMT in non-Hodgkin's

lymphorna. Clin Haematol l5:l OS, 1986

63. Frei El Canellos GP: Dose: a critical factor in cancer chemotherapy. Am

J Med 69:585, 1980

64. Weber JS, Yang JC. Topalian SL, Schwartzentruber DJ. White DE,

Rosenberg SA: The use of interleukin-2 and lymphokine-activated killer cells

for the treatment of patients with non-Hodgkin's lymphoma. J Clin Onwi 10:33.

1992

65. Rosenberg SA. Packard BS, Aebersold PM. Solomon Dl Topalian SL,

Toy ST, Simon Pl Lotze MTl Yang JC, Seipp CA: Use of tumor-infiltrating

lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic

melanoma. A preliminary report. N Engl J Med 319:1676, 1988

66. Kaminski Ml Zasadny K, Francis 1, Milik A, Ross C, Moon SI Crawford S,

Burgess J, Petry NI Butchko G, Glenn S. Wahl R: Radioimrnunotherapy of B-

cell lymphoma with Anti-BI (Anti-CDZO) antibody. N Engl J Med 329:459, 1993

67. Kaminski MS, Zasadny KR, Francis IR, Milik AW, Ross CW, Moon SD,

Crawford SM, Burgess JM, Petry NA, Butchko GM, et al.: Radioimmunotherapy

of 6-cell lymphoma with [13'l]anti-~1 (anti-CDPO) antibody. N Engl J Med

329:459, 1993

68. KrolickKA,lsaksonPC,UhrJW,VitettaES: BCL1,amurinemodelfor

ch ronic lymphocytic leukemia: use of the surface immunoglobulin idiotype for

the detection and treatrnent of tumor. lmmunol Rev 48:81, 1979

Page 164: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

69. Kwak LW, Young HA, Pennington RW, Weeks SD: Vaccination with

syngeneic, lymphoma-derived immunoglobulin idiotype combined with

granulocyte/macrophage colony-stimulating factor primes mice for a protedive

T-cell response. Proc Natl Acad Sci 93:10972, 1996

70. George AJ. Folkard SG. Hamblin T J, K. SF: ldiotypic vaccination as a

treatment for B cell lymphoma. J lmmunol i41:2168, 1988

71. Allison JP, H u e AA, Leach DR: Manipulation of costimulatory signals

to enhance antitumor T- ce11 responses. Cun Opin lmmunol7:682, 1995

72. Banchereau JI Batan F, Blanchard Dl Briere F, Galizzi JP, Vankooten

Cl Liu YJ, Rousset F, Saeland S: The CD40 antigen and its ligand. Annu Rev

lmmunol 12:881,1994

73. Schultze JL, Michalak S. Seamon MJ. Dranoff G. Jung KI Daley J,

Delgado JC, Gribben JG, Nadler LM: CD4O-activated human B cells: an

alternative source of highly efficient antigen presenting cells to generate

autologous antigen-specific T cells for adoptive immunotherapy. J. Clin. Invest.

100:2757, 1997

74. Schultze JL, Seamon MJ, Michalak S, Gribben JG, Nadler LM:

Autologous tumor infiltrating T cells cytotoxic for follicular lymphoma cells can

be expanded in vitro. Blood 89:3806, 1997

75. Galea-Lauri J, Farzaneh F, Gaken J: Novel costimulators in the immune

gene therapy of cancer. Cancer Gene Therapy 3:202,1996

Page 165: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

76. Huang AYl Bruce AT. Pardoll DM, Levitsky HI: Does 87-1 expression

confer antigen-presenting cell capacity to tumors in vivo? J. Exp. Med.

l83:769, 1996

77. Smith M. Burchell JM, Graham RI Cohen EP, Taylor-Papadimitriou J:

Expression of 87.1 in a MUClexpressing mouse mammary epithelial tumour

cell line inhibits turnorigenicity but does not induce autoimmunity in MUC1

transgenic mice. immunology 97:648, 1999

78. Dranoff Gl Mulligan RC: Gene transfer as cancer therapy. Adv lmmunol

58:417, 1995

79. Levitsky HI, Montgomery JI Ahmadzadeh M, Staveley-OfCarroll K.

Guamieri F, Longo DL, Kwak LW: lmmunization with granulocyte-macrophage

colony-stimulating factor-transduced, but not 67-1-transduced, lymphoma cells

primes idiotype-specific T cells and generates potent systemic antitumor

immunity. J. Immunol. 156:3858, 1996

80. Tuting Tl OeLeo AB, Lotze MT, Storkus WJ: Genetically rnodified bone

marrow-derived dendritic cells expressing tumor-associated viral or "self"

antigens induce antitumor immunity in vivo. Eur. J. lmmunol27:2702, 1997

81. Koch F, Trockenbacher B. Schuler G, Romani N: Antigen processing

capacity of dendritic cells from mice of different MHC backgrounds: down-

regulation upon culture and evidenœ for heterogeneity of dendritic cell

populations. Advances in Experïmental Medicine & Biology 378:203, 1995

Page 166: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

82. Lenz P, Elbe A. Stingl G. Bergstresser PR: MHC class I expression on

dendritic cells is sufficient to sensitize for transplantation immunity. Journal of

Investigative Derrnatology 1 07:844, 1 996

83. Dubey CI Croft Ml Swain SL: Costimulatory requirements of naive CD4+

T cells. ICAM-1 or 67-1 can costimulate naive CD4 T cell activation but both

are required for optimum response. Joumal of lmmunology 155:45. 1995

84. Cohen PJ. Cohen PA. Rosenberg SA. Katz SI. Mule JJ: Murine

epidermal Langerhans cells and splenic dendritic cells present tumor-

associated antigens to primed T cells. Eur J Immun01 24131 5, 1994

85. Steinman RM. Cohn ZA: Identification of novel cell type in peripheral

lymphoid organs of mice. 1. Mophology. quantitation. tissue distribution. J Exp

Med 137:1142,1973

86. Steinman RM: The dendAic cell system and its role in immunogenicity.

Annu Rev lmmunol9:271,1991

87. Dubois B. Caux C: In vitro differenciation and functions of dendritic cells

obtained from CD34+ hematopoietic progenitors. Pathologie Biologie 43:829.

1995

88. Albert ML, Sayter 8, Bhardwaj N: Dendritic cells acquire antigen from

apoptotic cells and induce class 1-restricted CTLs. Nature 392:86. 1998

89. lnaba K. lnaba M. Naito M. Steinman RM: Dendritic cells progenitors

phagocytose perticulates, inclubing bacilus Calmette-Guerin organisms. and

sensitize mice to mywbacterial antigens in vivo. J Exp Med 178:479, 1993

Page 167: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

90. Gluckman JC, Canque BI Chapuis F, Rosenzwajg M: In vitro generation

of human dendritic cells and cell therapy. Cytokines. Cellular and Molecular

Therapy 3: 187, 1 997

91. Koch F, Stanzl U, Jennewein P. Janke K. Heufler C, Kampgen E,

Romani N. Schuler G: High level 11-12 production by murine dendritic cells:

upregulation via MHC class II and CD40 molecules and downregulation by II4

and 11-1 0. Journal of Experimental Medicine 184741, 1996

92. Dubois BI Vanbervliet 6, Fayette J: Dendritic cells enhance growth and

differentiation of CD40-activated 6 lymphocytes. J Exp Med 185:941, 1997

93. Saunders Dl Lucas K, Ismaili J, Wu L, Mataskovsky E, Dunn A,

Shortman K: Dendritic Cells Devetopment in Culture from Thymic Presursor

Cells in the Absence of Granulocyte/Macrophage Colony-stimulating Factor. J.

Exp. Med. 184:2185,1996

94. Saunders Dl Lucas K, Ismaili J, Wu L, Maraskovsky E, Dunn Al

Shortman K: Dendritic cell development in culture from thymic precursor cells

in the absence of granulocyte/macrophage colony-stimulating factor. J. Exp.

Med. l84:2185, 1996

95. Hart DN : Dendritic cells: unique leukocyte populations which control the

primary immune response. Blood 90:3245, 1997

96. lnaba K. inaba M. Romani NI Steinman RM: Generation of large

numbers of dendritic cells from mouse bone marrow cultures supplemented

with GM-CSF. J Exp Med 176:1693,1992

Page 168: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

97. Brodsky F. Guagliardi L: The cell biology of antigen proœssing and

presentation. Ann Rev lmmunol9:707, 1 991

98. Brocker T: Survival of mature CD4 T lymphocytes is dependent on major

histocompatibility complex dass l l-expressing dendntic cells. J. Exp. Med.

1 86:1223,1997

99. Guinan EC, Gribben JB, Boussiotis VA: Pivotal role of the B7:CD28

pathway in transplantation toleranœ and tumor immunity. Blood 84:3261, 1994

100. Mosmann TR. Sad S: The expanding universe of T-cell subsets: Thl,

Th2 and more. lmmunology Today 1 ?:l38,1996

101. Mumberg Dl Monach PA, Wanderling S. Philip M, Toledano AY.

Schreiber RD, Schreiber H: CD4(+) T cells eliminate MHC class Il-negative

cancer cells in vivo by indirect effects of IFN-gamma. Proc Natl Acad Sci

96:8633, 1999

102. Bhardwaj N, Seder RA. Reddy A, Feldman MV: 11-12 in conjunction with

dendritic cells enhanœs antiviral CD8+ CTL responses in vitro. J. Clin. Invest.

98:715, 1996

103. Boczkowski D, Nair SK, Snyder DI Gilboa E: Dendritic cells pulsed with

RNA are potent antigen-presenting cells in vitro and in vivo. J. Exp. Med.

1 84:465, 1996

104. Celiuui CM, Mayordomo JI, Storkus WJ, Lotze MT, Falo LD, Jr.:

Peptide-pulsed dendntic cells induœ antigen-specific CTL-mediated protective

tumor immunity [see cornments]. J. Exp. Med. l83:283, 1996

Page 169: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

105. Fields RC, Shimizu K, Mule JJ: Murine dendritic cells pulsed with whole

tumor lysates mediate potent antitumor immune respomses in vitro and in vivo.

Proc. Natl- Acad. Sci. 959482, 1998

106. lnaba K. Metlay JP, Crowley MT, Steinmân RM: Dendriiic Cells Pulsed

with Protein Antigens In Vitro Can Prime Antigen-specific. MHC-restricted T

Cells ln Situ. J. Exp. Med. 172:631, 1990

707. Nair SKI Boukowski D, Snyder 0, Gilboa E: Antigen-presenting cells

pulsed with unfractionated tumor-derived peptides are potent tumor vaccines.

Eur J lmmunol27:589,1997

108. Paglia P, Chiodoni Cl Rodolfo M, Colombo MP: Murine decdritic cells

loaded in vitro with soluble protein prime cytotoxic T lymphocytes against tumor

antigen in vivo. J Exp Med l83:317. 1996

109. Porgador A, Gilboa E: Bone marrow-generated dendritic cells pulsed

with a class 1-restncted peptide are potent inducers of cytotoxic T lymphocytes.

Journal of Experimental Medicine 182:255, 1995

1 1 0. Mayordomo JI, Zonna T, Storkus WJ, Zitvogel LI Celluui C, Falo CD,

MeIief CJ, lldstad ST, Kast WM, Deleo AB.: Bone rnanowdenved dendritic

cells pulsed with synthetic tumour peptides elicit protective and therapeutic

antitumour immunity. Nature Medicine 1:1297, 1995

141. Nair SU, Boczkowski D, Morse M, Cumming RI, Lyerly HK, Gilboa E:

Induction of primary carcinoembryonic antigen (CEA)-specific cytotoxic T

lymphocytes in vitro using human dendritic cells transfected with RNA. Nature

Biotechnol 16:364, 1998

Page 170: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

112. Nair SK, Hull S. Coleman D. Gilboa E. Lyeriy HK, Morse MA: Induction

of carcinoem bryonic antigen (CEA)-specific cytotoxic T- lymphocyte responses

in vitro using autologous dendritic cells loaded with CEA peptide or CEA RNA

in patients with metastatic malignancies expressing CEA. Int J Cancer 82:121.

1999

113. Arthur JF, Butterfield LH, Roth MD: A cornparison of gene transfer

methods in human dendritic cells. Cancer Gene Thet 4:17,1997

1 14. McArthur JG, Mulligan RC: Induction of protective anti-tumor immunity

by gene-rnodified dendritic cells. J. Immunot. 2l:4l, 1998

1 1 5. Gong J, Chen Dl Kashiwaba M. Kufe O: Induction of antitumor adivity

by immunization with fusions of dendritic and carcinoma cells. Nat Med 3:558,

1997

116. Brossart P, Goldrath AW, Bu& EA, Martin S. Bevan MJ: Virus-mediated

delivery of antigenic epitopes into dendritic cells as a means to induce CTL. J.

Immunol. 158:3270,1997

1 1 7. Girolornoni G. Ricciardi-Castagnoli P: Dendritic cells hold promise for

imrnunotherapy. Immunol. Tod. l8:lO2, 1997

118. Gong J, Chen L. Chen D, Kashiwaba M, Manome Y, Tanaka Tl Kufe D:

Induction of antigen-specific antitumor immunity with adenovirus-transduced

dendritic cells. Gene Therapy 4: 1023. 1997

119. Siena S. Di Nicola M, Bregni M. Mortanni R. Anichini A, Lombardi L,

Ravagnani F, Panniani G. Gianni AM: Massive ex vivo generation of functional

Page 171: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

dendritic cells from mobilized CD34+ blood progenitors for anticancer therapy.

Exp. Hemat. 233463, 1995

120. Romani NI Reider O. Heuer M. Ebner S. Kampgen E, Eibl BI

Niederwieser Dl Schuler G: Generation of mature dendritic cells from human

blood. An impmved method with special regard to clinical applicability. J.

Immunol. Meth. 1 g6:l37. 1996

121. Hsu FJ: Vaccination of patient with Bcell lymphorna using autologous

antigen-pulsed dendritic cells. Nature Med 2:52, 1996

122. Timmemran JM. Levy R: Linkage of foreign carrier protein or GM-CSF to

a tumor antigen improves the efficacy of pulsed dendritic cell vaccination.

BIood 90:514, 1997

123. Mukherji B. Chakraborty NG, Yamasaki S. Okino Tl Yamase H, Sporn

JR. Kurtzman SKI Ergin MT, Ozok J, Meehan J: Induction of antigen-specific

cytolytic T cells in situ in human melanoma by immunization with synthetic

peptide-pulsed autologous antigen presenting cells. Proc Natl Acad Sci U S A

92:8078, 1995

124. Nestle FOI Alijagic S. Gilliet M. Schadendore D: Vaccination of

melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat

Medicine 4:328. 1 998

125. Nestle FOI Alijagic S. Gilliet M. Sun Y. Grabbe S. Dummer R. Burg G.

Schadendorf D: Vaccination of melanoma patients with peptide- or tumor

lysate-pulsed dendritic cells [see wmments]. Nature Medicine 4:328, 1998

Page 172: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

126. Reichardt VL, Okada CY, Stockerl-Goldstein KE, Bogen 6, Levy R:

Rationale for adjuvant idiotypic vaccination after high-dose therapy for multiple

myeloma. Biol Blood Marrow Transplant 3:157, 1 997

127. Wen YJ, Ling M. Bailey-Wood RI Lim SH: ldiotypic protein-pulsed

adherent peripheral blood mononuclear dl- denved dendritic cells prime

immune system in multiple myelorna. Clin Cancer Res 4:957, 1998

128. Vaione F, Smail E, Peshwa MW Phase 1 trial of dendntic celtbased

immunotherapy with APC8015 for hormone-refractory prostate cancer (HRPC).

Proc Am Soc Cancer Res 39: 173,1998

129. Salgaller ML, Lodge PA, McLean JG, Tjoa BA, Loftus DJl Ragde Hl

Kenny GM, Rogers M. Boynton AL, Murphy GP: Report of immune monitoring

of prostate cancer patients undergoing T- cell therapy using dendritic cells

pu lsed with H LA-A2-specific peptides from prostate-specific membrane antigen

(PSMA). Prostate 35:144, 1998

130. Tjoa BA, Simmons SJ, Elgamal A, Rogers Ml Ragde Hl Kenny GM,

Troychak MJ, Boynton AL, Murphy GP: Follow-up evaluation of a phase II

prostate cancer vaccine trial. Prostate 40: 125, 1999

131. Holtl L, Rieser C, Papesh Cl Ramoner R, Bartsch Gl Thumher M:

CD83+ blood dendritic cells as a vaccine immunotherapy of metastatic renal-

cell cancer (letter). Lancet 352:1358, 1998

132. Scott MT, Tamcun JW, Hartzell GW: The structure and function of the

homeodomain. Biochim Biophys Acta 989:25, 1989

Page 173: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

133. Scheidereit C, Cromlish JA. Gerster T, Kawakami K, Balmaceda CG,

Currie RA. Roeder RG: A human lymphoid-specific transcription factor that

activates irnmunoglobulin genes is a homoeobox protein. Nature 336551. 1988

134. Boncinelli E, Acampora O, Pannese M, M DE, Somma R, Gaudino G,

Stomaiuolo A. Cafiero M. Faiella A, Simeone A: Organization of human class I

homeobox genes. Genome 31 :745,1989

135. Gehring WJ: Homeoboxes in the study of development Science

236: lZ45, l987

136. Lawrence HJ, Largman C: Homeobox genes in normal hematopoiesis

and leukemia. Blood 80:2445, 1992

137. Lawrence HJ. Sauvageau G, Humphries RK, Largman C: The role of

HOX homeobox genes in normal and leukernic hematopoiesis. Stem Cell

l4:28 1, 1 996

138. Celetti A, Barba P, Cillo Cl Rotoli B, Boncinelli El Magfi MC:

Characteristic patterns of HOX gene expression in different types of human

leukernia. Int J Cancer 53:237, 1993

139. Vieille-Grosjean 1, Roullot V, Courtois G: Identification of homeobox-

contaiining genes expressed in hematopoietic blast cells. Biochern Biophys

Res Commun 185:785. 1992

140. Shen WF, Largman C, Lowney P, Corral JC, Detmer K, Hauser CA,

Simonitch TA, Hack FM, Lawrence HJ: Lineage-restricted expression of

homeobox-containing genes in human hematopoietic cell lines. Proc Natl Acad

Sci 86:8536, 1989

Page 174: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

141. Care A, Testa U, Bassani A, Tritarelli E, Montesoro El Samoggia P.

Cianetti L, Peschle C: Coordinate expression and proliferative role of HOXB

genes in adivated adult T lymphocytes. Mol Cell Biol 14:4872, 1994

142. Petrini Ml Quaranta MT, Testa U, Samoggia Pl Tritarelli El Care A,

Cianetti LI Valtieri Ml Barletta Cl Peschle C: Expression of selected human

HOX-2 genes in B K acute lyrnphoid leukemia and interleukin-2/interleukin-1

beta-stimulated natural killer lymphocytes. Blood 80:185, 1992

143. Deguchi Y, Kehrl JH: Selective expression of two homeobox genes in

CD34-positive cells from human bone manow. Blood 78:323, 1991

144. Dube ID, Kamel-Reid S, Yuan CC, Lu M, Wu XI Corpus G, Raimondi

SC, Crist WM, Carroll AJ, Minowada JI Baker JB: A novel human homeobox

gene lies at the chromosome 10 breakpoint in lymphoid neoplasias with

chromosomal translocation t(10; 14). Blood 78:2996, 1991

145. Dube ID, Raimondi SC, Pi O, Kalousek DK: A new translocation,

t(10; l4)(q24;q 1 1 ), in T cell neoplasia. Blood 67:1181, 1986

146. Salvati PD, Ranford PR, Ford J. Kees UR: HOX11 expression in

pediatric acute lymphoblastic leukemia is associated with T-cell phenotype.

Oncogene 1 1 :1333,1995

147. Dear TN, Colledge WH, Carlton MB, Lavenir 1, Larron Tl Smith AJ,

Warren AJ, Evans MJ, Sofroniew MV, Rabbitts TH: The HOX11 gene is

essential for cell survival during spleen development. Development 121 :2909,

1995

Page 175: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

148. Roberts VJ. van Dijk MA. Mune C: Localization of Pbxl transcripts in

developing rat embryos. Mech Dev 51 :W3. 1995

149. Raju K. Tang S, Dube ID, Kamel-Reid S. Bryce DM, Breitrnan ML:

Characterization and developmental expression of Tlx-1. the murine homolog

of HOX11. Mech Dev44:51,1993

150. Moretti P. Simmons P, Thomas Pl Haylock Dl Rathjen P. Vadas M, R

DA: Identification of homeobox genes expressed in human haemopoietic

progenitor cells. Gene l44:2l3. 1994

151. Lu Ml Zhang N, Ho AD: Genomic organisation of the putative human

homeobox proto-oncogene HOX-11 (TCL-3) and its endogenous expression in

T cells. Oncogene 7:1325, 1992

152. Hatano M. Roberts CWM. Minden M. Crist WM, Korsmeyer SJ:

Deregulation of a homeobox gene, HOXI 1, by the t( l0; l l ) in T cell leukemia.

Science 253:79, 1 991

153. Hough MR, Reis MD, Singaraja RI Bruce Dl M., Kamel-Reid S. Dardik 1.

Breitman ML, Dube ID: A Model of spontaneous B-lineage lymphomas in lgiip-

HOXI 1 transgenic mice. Proc. Natl. Acad. Sci. 95. 1998

154. Sutcliffe SB, Belch AR, Connors JM, Gascoyne RD, Gospodarowic MK,

Huebsch LBl Lacroix L, Langley GR, Meyer RI Sawka C: Lymphoma and you:

A guide for patients living with Hodgkin's disease and non-Hodgkin's

lymphoma. Mississauga. Ontario, The Medicine Group Ltd., 1998

Page 176: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

155. DeMatos P. Abdel-Wahab 2, Vervaert Cl Seigler HF: Vaccination wïth

dendritic cells inhibits the growth of hepatic metastases in B6 mice. Cell

lmmunol 185:65,1998

156. Mayordomo JI, Zorina T, Storkus WJ. Zitvogel L, Garcia-Prats MD.

DeLeo AB, Lotze MT: Bone manow-derived dendritic cells serve as potent

adjuvants for peptide-based antitumor vaccines. Stem Cells 1 5:94. 1997

157. Nair SK. Snyder D. Rouse BT. Gilboa E: Regression of tumors in mice

vaccinated with professional antigen-presenting cells pulsed with tumor

extracts. International Journal of Cancer 70:706, 1997

158. Mulders P, Tso CL, Gitlitz 8, Kaboo R, Hinkel A, Frand S. Kiertscher S,

Roth MD, deKemion J, Figlin R, Belldegrun A: Presentation of renal tumor

antigens by human dendritic cells activates tumor-infiltrating lymphocytes

against autologous tumor: implications for live kidney cancer vaccines. Clin

Cancer Res 5:445,1999

159. Cayeux S, Richter G, Becker Cl Pezzutto A, Dorken B. Blankenstein T:

Direct and indirect T cell priming by dendritic ceIl vaccines. Eur J lrnmunol

29:225, 1999

160. Karasuyama Hl Melchers F: Establishment of mouse cell lines which

constitutively secrete large quantities of interieukin 2, 3, 4 or 5 using modified

cDNA expression vectors. Eur J lmmunol l8:Q7, 1988

161. Ralph P. Nakoinz I: lnhibitory effects of lecitins and lymphocyte

mitogens on rnurine lyrnphornas and myelomas. J Natl Cancer lnst 51:883,

1973

Page 177: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

162. Moore M. FR Cl MJ B: Introduction of soluble protein into the Class I

pathway of antigen processing and presentation. cell 54777, 1998

163. Smith SD, Morgan R, Link MP, McFall P. Hecht F: Cytogenetic and

immunophenotypic analysis of cell lines established from patients with T cell

leu kemiallymp homa. Blood 67:650, 1986

164. Lord EM, Frelinger JG: Tumor immunotherapy: cytokines and antigen

presentation. Cancer Immunology, lmrnunotherapy 46:75, 1998

165. Kawakami Y. Rosenberg SA: Human tumor antigens rewgnized by T-

cells. lmmunologic Research l6:313, 1997

166. Hsu FJ, Caspar CB, Czerwinski Dl Kwak LW, Liles TM, Syrengelas A,

Taidi-Laskowski 8, Levy R: Tumor-specific idiotype vaccines in the treatment of

patients with B- cell lymphoma: long-terni results of a clinical trial. Blood

89:3129, 1 997

167. French RR. Claude Chan HT, Tutt AL, Glennie MJ: CD40 antibody

evokes a cytotoxic T-cell response that eradicates lymphoma and bypasses T-

cell help. Nature Medicine 5548, 1999

168. Constant SL, Bottornly K: Induction of Th1 and Th2 CD4+ T cell

responses: the alternative approaches. Ann Rev lmmunol 15297. 1997

169. Flamand V, Somasse Tl Thielemans K, Demanet Cl Bakkus M. Bazin Hl

Tielemans F, leo O, Urbain J, Moser M: Murine dendritic cells pulsed in vitro

with tumor antigen induce tumor resistance in vivo. Eur J lmrnunol 24:605,

1994

Page 178: the - University of Toronto T-Space · 4 Table of Contents CHAPTER 1 1 LNTRODUCTION 1.1 HEIMATOPOIETTC CELL OEUGM, GROWTH AND DIFFERENTIATIoN 1.2 MALIGNANT HEMATOPOIESIS 1.3 NEOPLA~MS

170. Qui 2. Noffz G, Mohaupt M, Blankenstein T: Interleukin-10 prevents

dend ritic cell accumulation and vaccination with granulocyte-macrophage

colon y-stimulating factor gene-modified tumor cells. J lmmunol 1 59:770, 1 997

171. lnaba K. Metlay JP, Crowley MT, Witmer-Pack M, Steinman RM:

Dendritic cells as antigen presenting cells in vivo. Internat Rev lmrnunol 6:197,

1990

172. lnaba KI inaba M, Witmer-Pack M, Hatchcock K. Hodes R, Steinman

RM: Expression of 87 costimulator molecules on mouse dendritic cells. Adv

Exp Med & Biol378:65, 1995

173. lnaba K, Pack M. lnaba Ml Sakuta H, lsdell FI Steinman RM: High levels

of a major histocornpatibility complex Il-self peptide complex on dendritic cells

from the T cell areas of lymph nodes. J Exp Med 186:665,1997

174. Porgador A, Snyder D, Gilboa E: Induction of antitumor imrnunity using

bone marrow-generated dendritic cells. Journal of lmmunology l56:2918. 1996

175. Matzinger P: Tolerance, danger, and the extended family. Ann Rev

lmmunol12:991, 1994


Recommended