accelerated progression of chronic lymphocytic

216

Upload: others

Post on 25-Jul-2022

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC
Page 2: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC
Page 3: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

LEUKEMIA IN Eµ-TCL1 TRANSGENIC MICE EXPRESSING CATALYTICALLY-INACTIVE RECOMBINATION-ACTIVATING GENE 1

___________________________________

By

VINCENT KIHARA NGANGA

___________________________________

A DISSERTATION

Submitted to the Faculty of the Graduate School of the Creighton

University in Partial Fulfillment of the Requirements for the Degree of

Doctor of Philosophy in the Department of Medical Microbiology and

Immunology

_________________________________

Omaha, Nebraska

July, 31, 2013

Page 4: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC
Page 5: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

iii

ABSTRACT

Chronic lymphocytic leukemia (CLL) is the most prevalent adult leukemia

and mainly affects the elderly. CLL is a heterogeneous disease that is

characterized by progressive accumulation of small lymphocytes that express

CD19, CD5, and CD23 in the blood and lymphoid organs. The origins of CLL

remain unclear but emerging evidence suggests that CLL likely evolves from a

more benign biologically similar condition known as monoclonal B cell

lymphocytosis. However, what initiates this condition or how it evolves to CLL is

unknown.

Recently developed transgenic mice that express catalytically inactive

RAG1 in the periphery (dnRAG1 mice) develop an early-onset indolent CD5+ B

cell lymphocytosis, caused in part by a defect in secondary V(D)J

rearrangements initiated to alter autoreactive B cell receptor specificity.

Hypothesizing that the CD5+ B cells accumulating in these animals represent a

CLL precursor, dnRAG1 mice were crossed with CLL-prone Eµ-TCL1 mice,

which carry the T cell leukemia/lymphoma 1 (TCL1) transgene, to determine

whether dnRAG1 expression in Eµ-TCL1 mice accelerates the onset of CLL-like

disease. These experiments showed that CD5+ B cell expansion and CLL

progression occurred more rapidly and uniformly in double-transgenic mice (DTG

mice) compared to Eµ-TCL1 mice, but with similar phenotypic and leukemogenic

features. Interestingly, splenic CD3+TCRβ+CD4-CD8- (double-negative) and

CD4+CD25+ regulatory T cells increased in frequency as a function of age in

DTG mice, suggesting they may be linked to the expanding CD5+ B cells.

Page 6: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

iv

Comparative analysis of gene expression profiles in normal and

transgenic CD5+ B cells indicate that CLL may develop by distinct but convergent

pathways that may be nurtured by loss of tolerance. These studies also reveal a

possible role for prolactin signaling in the regulation of receptor editing.

The work in this dissertation suggests that the B cell antigen receptor

plays a significant role in CLL pathogenesis in that failure to remodel the receptor

in response to autoreactivity may promote the benign accumulation of CD5+ B

cells, which may then be subjected to secondary genetic lesions that promote

CLL progression.

Page 7: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

v

DEDICATION

This dissertation is dedicated to:

My father, John Nganga Manjie, and my mother, Elizabeth Wanjiru Nganga

Page 8: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

vi

ACKNOWLEDGEMENTS

First and foremost I would like to thank my advisor, Dr. Patrick Swanson,

for his guidance and relentless support during my graduate career. I would like to

thank him specifically for allowing me to pursue my education in his laboratory

and for his patience, encouragement, as well as willingness to entertain new

ideas. Secondly, I would like to thank the members of my committee, Dr. Jason

Bartz, Dr. Michael Belshan, Dr. Kristen Drescher, and Dr. Hina Naushad for their

continued direction and support. I acknowledge their genuine effort and expert

advice to mould me into a better student and scientist.

My experience as a student would not have been as exciting without the

wonderful working environment of the Swanson laboratory. I would like to

acknowledge former members, Aleksei Kriatchko, Ming Zhang, Prafulla Raval,

Sushil Kumar, and Dirk Anderson for initiating me into this laboratory and

teaching me most of the techniques I know. Furthermore, this work would not

have been possible without the excellent assistance of Dirk Anderson, Victoria

Palmer, Michele Kassmeier, and Nathan Schabla. I would also like to thank

Koushik Mondal, Mary Rothermund, and Razia-Aziz Seible for their valuable

input, especially to Koushik Mondal for many a discussions as well as his

friendship.

I would also like to extend my gratitude to the Creighton University

Medical Microbiology and Immunology department and its staff and faculty for

financial, administrative, and educational support. I wish to acknowledge the

outstanding flow cytometry services of Dr. Greg Perry as well as his technical

Page 9: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

vii

and scientific advice over the years. Additionally, I want to thank former and

currents students, who were a source of friendship and encouragement over the

years, particularly, Charles Schutt, Cameron Schweitzer, Philip Kurpiel, and Ming

Zhang.

Finally, I would like to thank my family for their unyielding moral and

financial support. To my parents, brothers and sisters, thank you for offering your

finances and moral support as well as your patience. This is also goes to the

extended Manjie family as well, including my grandmother, Zipporah Njeri Manjie,

for her never-ending encouragement. I also wish to extend my genuine

appreciation to my uncle Stephen Kirumba and his family for both financial and

moral encouragement throughout my school years, and to the Njenga family for

moral support as well as their friendship.

The work in this thesis was supported by the Health Future Foundation,

the Nebraska LB506 and LB595 Cancer and Smoking Disease Research

Programs, and the Nebraska LB692 Biomedical Research Program grants

awarded to Dr. Patrick Swanson.

Page 10: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

viii

TABLE OF CONTENTS

Abstract .............................................................................................................. iii

Dedication ........................................................................................................... v

Acknowledgements ........................................................................................... vi

Table of contents .............................................................................................. vii

List of Figures .................................................................................................. xiii

List of Tables .................................................................................................... xv

List of Abbreviations ....................................................................................... xvi

Chapter 1: Introduction ...................................................................................... 1

1.1 B-cell chronic lymphocytic leukemia history & epidemiology… ....... 2

1.1.1 A brief history of CLL ..................................................... 2

1.1.2 Descriptive epidemiology ............................................... 4

1.2 Clinical characteristics of CLL ......................................................... 7

1.2.1 Diagnosis and natural history .......................................... 7

1.2.2 Heterogeneity ................................................................ 14

1.2.3 Current therapy ............................................................. 17

1.3 Cellular and molecular characteristics of CLL ............................... 22

1.3.1 Normal B cell development and CLL cell of origin ......... 22

1.3.2 Genetic and chromosomal defects in CLL .................... 34

1.3.3 Role of the B cell receptor ............................................ 37

1.3.4 Role of the tumor microenvironment ............................ 42

Page 11: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

ix

1.4 Animal models of CLL ................................................................... 48

1.4.1 Xenograft mice. ............................................................. 48

1.4.2 New Zealand mice ........................................................ 49

1.4.3 Eμ-TCL1 transgenic mice.............................................. 52

1.4.4 APRIL transgenic mice ................................................. 55

1.4.5 TRAF2DN/BCL2 transgenic mice ................................. 56

1.4.6 PKCα-KR graft mice ...................................................... 58

1.4.7 SV40 large T antigen transgenic mice .......................... 59

1.4.8 DLEU2/miR15a/16-1 knock-out mice ............................ 60

1.4.9 miR29 transgenic mice ................................................. 61

1.5. Research goals and specific aims ................................................. 63

Chapter 2: Methods .......................................................................................... 67

2.1 Mice .............................................................................................. 67

2.2 Flow cytometry .............................................................................. 67

2.3 Adoptive transfer ........................................................................... 69

2.4 α-galactosylceramide treatment .................................................... 69

2.5 Cell culture .................................................................................... 69

2.6 Histopathology and WBC counts ................................................... 70

2.7 Clonality ........................................................................................ 71

2.8 Immunoglobulin gene sequencing and analysis ............................ 71

2.9 Immunoglobulin levels ................................................................... 73

2.10 Microarray ..................................................................................... 74

Page 12: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

x

2.11 Real-time quantitative PCR ........................................................... 76

2.12 Statistics ........................................................................................ 77

Chapter 3: Results..............................................................................................79

3.1 dnRAG1 expression accelerates leukemogenesis in

Eμ-TCL1 mice…….. ...................................................................... 79

3.1.1 Expression of dnRAG1 in Eμ-TCL1 mice shortens

survival .......................................................................... 79

3.1.2 Time course analyses of CLL-like disease progression

in DTG mice… ............................................................. 82

3.1.3 B cells of DTG mice are clonally transformed and bear

unmutated Ig genes ...................................................... 88

3.2 dnRAG1 expression in Eμ-TCL1 mice suppresses

immunoglobulin production ........................................................... 95

3.3 Distinct gene expression profiles in CD5+ B cells in both young

and old transgenic mice ................................................................ 97

3.3.1 Gene expression profiles in 12-week-old mice .............. 97

3.3.2 Gene expression profiles in ill DTG and age-matched

counterparts ................................................................ 102

3.3.3 Tolerogenic and BCR genes are perturbed in

transgenic mice .......................................................... 108

3.4 Alteration of the T cell compartment in transgenic mice .............. 110

3.4.1 Increase in CD4+CD25+ T cells in DTG mice and

Page 13: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

xi

CD3+TCRβ+CD4-CD8- (double-negative) T cells in

Eµ-TCL1 and DTG Mice ............................................ 110

3.4.2 The role double-negative T cells in transgenic mice .. 120

Chapter 4: Discussion .................................................................................... 129

4.1 Failure in self-tolerance promotes CLL progression .................... 129

4.1.1 Expression of dominant-negative RAG1 accelerates

CLL progression in Eµ-TCL1 mice ............................. 129

4.1.2 Breaching tolerance promotes CLL ............................. 132

4.1.1 Significance of Prl2a1 expression in dnRAG1 and

DTG mice .................................................................... 137

4.2 Role of regulatory T cells in CLL ................................................. 139

4.2.1 CD4+CD25+ T cells...................................................... 139

4.2.2 Double-negative T cells ............................................... 140

4.3 Lessons from murine models of benign and malignant CD5+

B cell expansion .......................................................................... 142

4.4 Future work ................................................................................. 145

4.4.1 Mechanisms of disease acceleration in DTG

Mice ............................................................................ 145

4.4.2 Role of hormone gene superfamily in normal and

leukemic B cells .......................................................... 146

4.4.3 Role of CD1d .............................................................. 148

4.4.4 Identitiy of double-negative T cells .............................. 149

Page 14: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

xii

4.5 Concluding remarks .................................................................... 150

Chapter 5: References…….. .......................................................................... 151

Page 15: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

xiii

LIST OF FIGURES

Figure 1.1 : B cell development and differentiation .......................................... 23

Figure 1.2 : Mechanisms of self- tolerance in B cells ........................................ 27

Figure 1.3 : Accumulation of B1-like CD5+ B cells with age in dnRAG1

mice ............................................................................................. 65 66

Figure 3.1 : dnRAG1 expression in Eµ-TCL1 mice enhances

lymphoproliferation and shortens lifespan .................................... 80

Figure 3.2 : dnRAG1 expression in Eµ-TCL1 mice accelerates accumulation

of CLL-like cells ............................................................................. 81

Figure 3.3 : Surface immunophenotype of CLL-like cells in DTG mice ............. 83

Figure 3.4 : Characterization of CLL-like disease progression in DTG mice .... 84

Figure 3.5 : DTG mice show evidence of CLL like disease at 36 weeks .......... 86

Figure 3.6 : Bone marrow and kidney histology of 36-week-old mice ............... 87

Figure 3.7: Engraftment of leukemic cells from DTG mice in SCID mice ......... 89

Figure 3.8 : Clonal transformation of B cells from DTG mice ............................ 96

Figure 3.10 : Identification of differentially expressed genes in WT

CD19+B220hiCD5- B cells and CD19+CD5+ B cells from

dnRAG1, Eµ-TCL1, and DTG mice at 12 weeks ......................... 98

Figure 3.11 : CD5+ B cells from dnRAG1, Eμ-TCL1, and DTG mice are most

similar to normal B1a B cells ..................................................... 100

Figure 3.12 : Identification of differentially expressed genes in WT

CD19+B220hiCD5- B cells and CD19+CD5+ B cells from ill

Page 16: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

xiv

DTG mice and age-matched dnRAG1 and Eµ-TCL1 mice ........ 103

Figure 3.13 : Validation of differentially expressed genes by quantitative

PCR .......................................................................................... 106

Figure 3.14 : CD23 expression on leukemic cells from an ill DTG mouse ...... 107

Figure 3.15 : Hierarchical clustering analysis of genes involved in B cell

receptor signaling from 12-week-old and older WT and

transgenic mice ........................................................................ 109

Figure 3.16 : Characterization of T cell subsets in transgenic mice .............. 111

Figure 3.17 : Characterization of lymph node T cells ...................................... 113

Figure 3.18 : Expansion of CD4+CD25+ T cells in DTG mice .......................... 115

Figure 3.19 : Expansion of CD3+TCRβ+CD8-CD4- (DN) T cells in transgenic

mice .......................................................................................... 116

Figure 3.20 : CD1d expression in CD5+ B cells of transgenic mice ............... 117

Figure 3.21 : α-GalCer treatment has little effect on CD5+ B cell

accumulation in DTG mice ......................................................... 118

Figure 3.22 : Expression of CD160, BTLA, HVEM, and DX5 in DN T or

CD5+ B cells ............................................................................... 119

Figure 3.23 : DN T cells inhibit accumulation of CD5+ cells after adoptive

transfer ....................................................................................... 121

Figure 3.24 : DN T cells inhibit expansion of CD5+ B cells in cell culture ....... 123

Figure 4.1 : Model for how defects in receptor editing can promote

development of MBL and CLL ..................................................... 133

Page 17: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

xv

LIST OF TABLES

Table 1.1 : Rai and Binet staging systems ........................................................ 11

Table 1.2 : Murine models of CLL ..................................................................... 51

Table 2.1 : Primers for RT-PCR ........................................................................ 78

Table 3.1 : Analysis of IgVH sequences from ill Eµ-TCL1 and DTG mice ......... 93

Table 3.2 : Analysis of IgVL sequences from ill Eµ-TCL1 and DTG mice ......... 94

Table 3.3 : Top 50 differentially expressed genes between 12-week dnRAG1

and DTG ....................................................................................... 125

Table 3.4 : Top common differentially expressed genes among older transgenic

mice compared to WT mice .......................................................... 126

Table 3.5 : Top 50 differentially expressed genes between ill DTG mouse

(M53) and Eµ-TCL1 mouse (M52) ................................................ 127

Table 3.6 : Top 50 differentially expressed genes between ill DTG mouse

(M57) and Eµ-TCL1 mouse (M52) ................................................ 128

Page 18: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

xvi

LIST OF ABBREVIATIONS

APRIL A proliferation-inducing ligand

BAFF B cell-activating factor of TNF family

BCR B cell receptor

BM bone marrow

CLL B cell chronic lymphocytic leukemia

CTLA4 cytotoxic T-lymphocyte antigen 4

dnRAG1 dominant-negative recombination activating gene 1

DN T double-negative T cell

DTG double-transgenic mice

FACS fluorescence-activated cell sorting

FO mature follicular B cell

Ig immunoglobulin

IgH immunoglobulin heavy chain

IgL immunoglobulin light chain

IL-10 interleukin-10

LN lymph node

MZ marginal zone B cell

NKT natural killer T cells

NZB/W New Zealand black/white

PB peripheral blood

PerC peritoneal cavity

Page 19: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

xvii

Prl2a1 prolactin family 2, subfamily a, member 1

Prl2a1 prolactin family 7, subfamily c, member 1

Prl8a2 prolactin family 8, subfamily a, member 2

RAG1 recombination activating gene 1

RAG2 recombination activating gene 2

RE receptor editing

SCID severe combined immunodeficiency

SHM somatic hypermutation

SLE systemic lupus erythematosus

SLL small lymphocytic lymphoma

T transitional B cell

TCL1 T cell leukemia/lymphoma 1

Treg regulatory T cell

V(D)J variable (diversity) joining

Page 20: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

1

CHAPTER I: INTRODUCTION

B-cell chronic lymphocytic leukemia (CLL) is the most prevalent adult

leukemia and mainly affects the elderly (Redaelli et al., 2004; Morton et al. 2007;

Watson et al., 2008). CLL is characterized by abnormal accumulation of small

lymphocytes that express CD19, CD5, and CD23 in blood and lymphoid tissues

such as bone marrow, lymph nodes, or spleen (Matutes and Polliack, 2000;

Dighiero and Hamblin, 2008). CLL is a heterogeneous disease that exhibits a

variable clinical course; some patients have an indolent disease that may not

require medical intervention while others suffer an aggressive disease associated

with short survival (Damle et al., 1999; Hamblin et al., 1999; Van Bockstaele et

al., 2009). Emerging evidence suggests that CLL likely evolves from a more

benign biologically similar condition known as monoclonal B cell lymphocytosis

(Rawstron et al., 2002; Rawstron et al., 2008; Lanasa et al., 2011; Kern et al.,

2012). However, what initiates this condition or why it evolves to CLL is unknown.

What is apparent is that the feedback between CLL cells and their environment

through various receptors, including those that bind antigens, is important but

how these interactions contribute to CLL pathogenesis is still debatable. CLL is

currently incurable.

Page 21: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

2

1.1 B-CELL CHRONIC LYMPHOCYTIC LEUKEMIA (CLL) HISTORY AND

EPIDEMIOLOGY

1.1.1 A Brief History of CLL

The understanding of what we now know as leukemia began earnestly in

Europe in the 19th century. Between 1811 and 1841, at least six physicians

published cases of their patients who had blood and spleen abnormalities that

may have been leukemia (Piller, 2001). However, John Hughes Bennett, an

English physician, and Rudolph Virchow, a German pathologist, were the first,

independent of each other, to identify leukemia as a distinct disease in 1845

(Piller, 2001; Marti and Zenger, 2004). Bennett described a disease of

“suppuration of the blood”, which he named leucocythemia, that was associated

with enlargement of the spleen and liver (Bennett, 1845; Bennett, 1851). Virchow

noted that in addition to an enlarged spleen, his patient had a reversed ratio of

pigmented to colorless corpuscles. He named this disease leukemia (Virchow,

1845; Virchow, 1847; Hamblin, 2000).

As interest in this new disease grew, others such as Ernst Neumann

showed that leukemia also involved the marrow, and supported Virchow in that

leukemia could be further divided into splenomedullary/pyoid (myeloid leukemias

today) and lymphatic/lymphadenoid forms (Rai, 1993; Hamblin, 2000). These two

forms of leukemia were formally confirmed with the advent of tri-acidic aniline

dye-based differential stains for blood cells in smears (Erlich, 1891). Additionally,

the terms “acute” and “chronic” would come into use to describe those leukemias

that progressed rapidly or gradually, respectively (Piller, 2001). As cases of new

Page 22: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

3

and old blood cell disorders continued to accumulate in the medical literature in

the latter half of the 19th century, it became necessary to categorize them.

Pathologist Hans Kundrat would introduce a systematic distinction approach

between leukemias, pseudoleukemias, and lymphosarcoma (lymphoma),

observing that the latter spread through connected nodes while sparing blood

and marrow (Marti and Zenger, 2004).

In the early 20th century, Turk (1903) and Osler (1909) offered the first

comprehensive description and classification of the entity we now know as B cell

chronic lymphocytic leukemia (CLL) (Hamblin, 2000; Marti and Zenger, 2004;

Linet et al., 2007). Turk would also note that CLL was indistinguishable from

some lymphomas (Hamblin, 2000). The most elaborate, and probably the most

accurate, description of CLL, however, would come in 1924 in a large by Minot

and Isaacs. In that study, they described the natural history of CLL in 98 patients

and attempted to treat the patients with radiation, which reduced tumor mass, but

failed to improve overall survival (Minot and Isaacs, 1924).

In the following decades there was substantial progress in understanding

and treating CLL. The most notable success in treating CLL was with the

alkylating agent Chlorambucil (Galton, 1961). Other progress came in refining the

description, diagnostic criteria, and prognosis of CLL as larger studies were

performed. It became quite clear that there was a wide ranging survival time

among patients, with some displaying stable disease while others had

progressive disease, leading to the proposal to stratify diagnostic criteria (Boggs,

1966; Galton, 1966; Dameshek, 1967).

Page 23: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

4

Shortly following the breakthrough using antibodies to identify and

distinguish B and T lymphocytes, it was shown that CLL was a disease of B

lymphocytes and that CLL cells additionally expressed the T lymphocyte antigen,

CD5 (Wilson and Nossal, 1971; Papamichail et al., 1971; Caligaris-Cappio et

al.,1982). This period also saw the development of two clinical staging systems

(Table 1) (Rai et al., 1975; Binet et al., 1981). These two systems, unlike earlier

ones, consisted of readily measurable clinical and laboratory parameters and

became widely adopted (Rai, 1993) and are still used today. Additional

prognostic factors together with the Rai/Binet staging systems, and improved

understanding of how CLL cells interact with their surroundings have allowed for

better prediction of clinical outcome and tailored therapy, which continue to

improve the management of CLL.

1.1.2 Descriptive Epidemiology

CLL is the most common adult leukemia in Australia, Europe, and North

America (Redaelli et al., 2004; Morton et al. 2007; Watson et al., 2008), including

the United States, where CLL cases account for over 30% of all leukemias (Yuille

et al., 2000; Hawlader et al., 2012). The Surveillance Epidemiology and End

Results (SEER) program of the National Cancer Institute, which has probably the

most comprehensive cancer registry, estimates that 16,060 Americans will be

diagnosed with CLL, and 4,580 will die from the disease in 2012 (Hawlader et al.,

2012). According to SEER, the overall CLL incidence is 4.2 per 100,000

Americans. Most CLL patients are men as they have a higher incidence of 5.8

Page 24: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

5

per 100,000 compared to 3.0 for women. CLL risk varies with race; the order of

decreasing risk is White > Black > Indian/Alaska Native > Hispanic >

Asian/Pacific Islander. CLL is a disease of older adults. Based on SEER data

from 2005-2009, only 1.8% of CLL patients were diagnosed before age 44 and

less than 9.8% of CLL patients before age 54. The median age at diagnosis is 72

years. The 5-year relative survival based on 2002-2008 data is 78.8%. The

lifetime risk of developing CLL based on 2007-2009 data is 0.49%; alternatively 1

in 202 Americans will develop CLL in their lifetime.

The cause of CLL remains unknown but environmental risk factors have

been sought. Factors such as radiation, industrial chemicals, and cigarette

smoke, which are known to contribute to other cancers, have been of particular

interest (Linet et al., 2007). Although radiation has been linked to other

leukemias, no strong link could be made for CLL in studies involving nuclear

facility/radiation workers (Gluzman et al., 2006; Muirhead et al., 2009). Moreover,

retrospective studies show the relative risk of CLL among Japanese atomic bomb

survivors remains low despite increases in other types of lymphoproliferative

diseases (Finch et al., 1969; Hsu et al., 2013). An extensive review by Blair et al.

(2007) on studies investigating CLL link to chemicals such as industrial solvents,

chemotherapy alkylating agents, pesticides, and cosmetic dyes found little

evidence for increased risk of CLL except possibly for certain pesticides and

chemicals such as benzene and butadiene, which are commonly used in the

petroleum and rubber occupations. A stronger occupational risk is for

Page 25: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

6

farming/agricultural industries, which has been linked to pesticide use and

possibly fertilizers (Blair et al., 2007; Linet et al., 2007).

Infectious agents have also been speculated as potential causes of CLL

(Hamblin, 2006). Landgren et al. (2007) suggested that pneumonia can possibly

trigger CLL based on their findings that previous pneumonia infection and

frequency was significantly correlated with CLL risk. Furthermore, CLL cases that

exclusively use IgHV4-34 antibodies were found to carry higher than normal

Epstein-Barr virus (EBV) and cytomegalovirus (CMV) nucleic acid material,

suggesting that viral antigens may be involved in the etiology of CLL in some

cases (Kostareli et al., 2004). These possibilities are also supported by Messmer

and colleagues (2004) who reported that CLL cells from unrelated patients in

different geographic regions share striking similarities in the structure of their

antigen receptors, which would not be expected to occur by chance, suggesting

that these receptors are selected by a limited set of common antigens. Bacterial

polysaccharide capsules, viral coats, fungal cell wall glucans, and even self-

antigens have all been speculated to contribute to CLL pathogenesis (Redaelli et

al., 2004; Ghiotto et al., 2004; Hoogebom et al., 2013; Herve et al., 2005).

Familial studies suggest that there is a stronger genetic risk for CLL as

compared to the environment. The risk of developing CLL is increased 2-8-fold

for first-degree relatives of CLL patients (Crowther-Swanepoel and Houlston,

2009; Di Bernardo et al., 2008). Based on both linkage and association studies,

the risk alleles identified for CLL include those related to cell death and immune

regulation (Crowther-Swanepoel and Houlston, 2009; Di Bernardo et al., 2008;

Page 26: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

7

Slager et al., 2010). However, others have noted that these risk genes are also

associated with other lymphoproliferative disorders, suggesting they are not

unique for CLL (Yuille et al., 2000). The role of genetics is further clarified by the

observation that CLL risk does not change with immigration. For example,

immigrants from Asia who move to the West where the incidence of CLL is

higher, remain at low risk for developing CLL as do their descendants (Herrinton

et al., 1996; Pan et al., 2002).

1.2 CLINICAL CHARACTERISTICS

1.2.1 Diagnosis and Natural History

CLL is characterized by the accumulation of small mature lymphocytes

typically with scanty cytoplasm, clumped chromatin, and inconspicuous nucleoli

with a CD19+CD20loCD5+CD23+IgMloIgDloCD22lo/-CD79loFMC7lo/-

immunophenotype. CLL cells tend to be fragile and form readily observable

smudges in blood smears (Matutes and Polliack, 2000; Dighiero and Hamblin,

2008; Nowakowski et al., 2007). Diagnosis of CLL is established by a finding of

an absolute peripheral blood (PB) lymphocytosis of > 5x109/L and lymphocytes

with at least one B cell marker, CD19 or CD20, that are monoclonal (commonly

determined by light chain restriction), and co-express CD5 and CD23 (Pangalis

et al., 2002; Redaeli et al., 2004). A finding of lymphadenopathy, splenomegaly,

or other organ infiltration by cells with a CLL immunophenotype but with an

absolute PB lymphocytosis below 5x109/L is considered a different manifestation

Page 27: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

8

of the disease, and is known as small lymphocytic lymphoma (SLL) (Harris et al.,

1999; Campo et al., 2011) and comprises 25% of all CLL/SLL cases (Dores et

al., 2007). While bone marrow (BM) examination is not required for all patients at

diagnosis, involvement is considered when there is at least 30% infiltration by

lymphoid cells (Redaeli et al., 2004). The most common considerations in the

differential diagnosis of CLL are: (i) mantle cell lymphoma (MCL), which

expresses CD5, but not CD23, and carries a recurrent (11;14)(q13;q32)

chromosomal translocation associated with cyclin D1 overexpression, or (ii) less

closely related follicular lymphoma, hairy-cell leukemia, and marginal-zone

lymphoma, which are CD5-. Follicular lymphoma, but not CLL, expresses CD10,

and hairy-cell leukemia has morphologically conspicuous hair-like projections

(Dighiero and Hamblin, 2008). The median age at CLL diagnosis is 72 years with

twice as many men as women (Hawlader et al., 2012). Some patients have no

symptoms at diagnosis, but lymphadenopathy (80%), followed by splenomegaly

(50%) are the most common findings. Those with advanced disease usually

present with constitutional symptoms, mainly fatigue, weight loss, infections,

bleeding, anemia, and thrombocytopenia (Redaeli et al., 2004).

Rai and Binet are two widely used clinical staging systems in CLL (Rai et

al., 1975; Binet et al., 1981). Rai and Binet systems are based on clinical and

hematological findings of lymphocytosis, lymphadenopathy and other

organomegaly, anemia, and thrombocytopenia which segregate patients into

three and five stages, respectively, with three main categories that reflect

differences in survival (Table 1). About 80% of patients have low tumor burden

Page 28: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

9

and fall in the first stage but those presenting with extensive lymphadenopathy or

hepatosplenomegaly, with anemia and/or thrombocytopenia have the worst

prognosis (Van Bockstaele et al., 2009).

Additional prognostic factors, which usually correlate with Rai and Binet

systems, relate to serum proteins and cellular characteristics. (i) Serum markers

such as elevated levels of lactate dehydrogenase, β2-microglobulin, soluble

CD23, CD27, CD44, and thrombopoietin are associated with shorter survival (Rai

et al., 2004; Van Bockstaele et al., 2009). (ii) Proliferation markers such as

absolute lymphocyte count doubling time (lymphocyte doubling time) of less than

12 months, elevated thymidine kinase, and shorter telomeres and high

telomerase activity are associated with progressive disease and shorter survival

(Rai et al., 2004; Van Bockstaele et al., 2009). (iii) Cytogenetic abnormalities,

typically detected by FISH, are present in 80% of cases (Zenz et al., 2011). The

most common of these is chromosome 13q14 deletion which is seen in 40-60%

of patients but with a median survival of 133 months these case have a relatively

better outcome even beyond those with a normal karyotype. Trisomy 12 (15-30%

of cases) has an equally fair outcome with a median survival of 114 months,

which is comparable that of a normal karyotype. 11q22-q23 and 17p13 deletion

are less common (~10-20%) but with survival times of 79 and 32 months,

respectively, these cases are associated with an aggressive disease and

refractory therapy (Dohner et al., 1995; Dohner et al., 1997; Dohner et al., 2000).

(iv) IgVH mutational status - Presence or absence of mutations in Ig genes is

based on greater or lesser than 98% homology to the closest germline. About

Page 29: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

10

half of CLL cases carry mutated IgVH and have good prognosis with a median

survival of ~24 years, whereas those with unmutated IgVH have a relatively

shorter survival of ~9 years (Damle et al., 1999; Hamblin et al., 1999).

A subset of patients carrying IgVH3-21 has poor survival irrespective of

mutational status (Ghia et al., 2008; Krober et al., 2006). ZAP-70 and/or CD38

expression correlate with mutational status and are associated with poor

prognosis (Damle et al., 1999; Crespo et al., 2003). Because of the relative ease

and low cost of measuring CD38 and ZAP-70 expression by flow cytometry,

these can act as surrogate markers for mutation status (Damle et al., 1999;

Crespo et al., 2003). However there is no absolute consensus on what fraction of

CLL should express these proteins to consider them positive (typically, cases are

considered positive if > 30% (CD38) or >20% (ZAP-70) of CLL express these

proteins). These surrogate markers for mutational status should also be used

carefully since ZAP-70 or CD38 expression levels can fluctuate in some patients

(Hamblin et al., 2002; Van Bockstaele et al., 2009). Expression of lipoprotein

lipase also correlates with lack of mutations and can similarly distinguish the two

clinical groups of CLL (Haslinger et al., 2004; Heintel et al., 2005).

Immunological dysfunction has been known in CLL for decades (Jim,

1956; Dameshek, 1967; Chiorazzi et al., 1979). Immunological defects primarily

involve the leukemic cells but also extend to other immune cells. The most

common is loss of immunocompetence by the B cells manifested by

hypogammaglobulinemia, poor response to vaccinations, and autoimmune

Page 30: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

11

Table 1.1 Rai and Binet staging systemse

Category Features

Median

survival

(years)

Rai stage

0 Low risk Lymphocytosisa 13+

I Intermediate risk Lymphocytosis and enlarged nodes 8

II Lymphocytosis and enlarged spleen or liver

III High risk Lymphocytosis and anemiab 2

IV Lymphocytosis and thrombocytopeniac

Binet stage

A Low risk Hemoglobin ≥ 100g/L, platelets ≥ 100x109/L, 15

and < 3 enlarged lymphoid areasd

B Intermediate risk Hemoglobin ≥ 100g/L, platelets ≥ 100x109/L, 5

and ≥ 3 enlarged lymphoid areas

C High risk Hemoglobin < 100g/L and or platelets < 100x109/L, 3

regardless of number of enlarged lymphoid areas

a Blood absolute lymphocyte count ≥ 5x10

9/L with monoclonal B cells with a CLL immunophenotype.

b Hemoglobin < 110g/L, with or without enlarged lymph nodes, spleen or liver.

c Platelets < 100x10

9/L, with or without anemia or enlarged lymph nodes, spleen or liver.

d One of five areas: cervival, axillary, and inguinal lymph nodes, spleen, and liver.

e Rai et al., 1975; Rai et al., 1987; Binet et al., 1981; Rai et al., 2004.

Page 31: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

12

phenomena (Dearden, 2008). Progressive hypogammaglobulinemia is a

common feature in CLL patients. Rozman et al. (1988) reported that 19.5% of

240 previously untreated patients had hypogammaglobulinemia at diagnosis and

severity was correlated to clinical stage and survival. About 70% of patients

develop hypogammaglobulinemia within 7 years of diagnosis and the probability

of having deficiency for at least one Ig class is even higher (Dearden, 2008).

Progressive hypogammaglobulinemia is not primarily due to BM failure since

reconstitution is rarely seen after therapy (Colovic et al., 2001; Dearden, 2008).

Serum IgG and IgA, but not IgM levels appear to be better predictors of survival

(Rozman et al., 1988). Ig production defects, which also lead to suboptimal

response to vaccinations in CLL patients (Sinisalo et al., 2001; Hartkamp, et al.,

2001), likely originate from intrinsic B cell defects as well leukemia-induced T cell

dysfunction.

Defects are also observed in other immune cells. T cells, for instance,

display altered function and perturbed frequencies. The ratio of circulating CD4+

to CD8+ T cells is inverted in PB but CD4+ T cells are increased in the LN (Kay

1981; Herrmann et al., 1982). Most of these T cells are oligoclonal or clonal,

defective in immunological synapse formation, and exhibit signs of anergy or

exhaustion (Farace et al., 1994; Serrano et al., 1997; Gorgun et al., 2009; Riches

et al., 2013). Tregs, which are immunosuppressive in nature, are also increased

in CLL (D’Arena et al., 2012). Reduced NK and monocyte cytotoxicity, neutrophil

chemotaxis and phagocytic activity, and complement activation defects all

contribute to systematic immune dysfunction in CLL (Dearden, 2008).

Page 32: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

13

Cytopenias are the most common autoimmune manifestations in CLL

(Zent and Kay, 2010). Autoimmune hemolytic anemia (AIHA) is the most

common of these and occurs in 5-10% of cases followed by immune

thrombocytopenia (ITP; 2%), and pure red cell aplasia and neutropenia, which

are the least common with each accounting for 0.5% of cases. AIHA and ITP

findings increase with clinical stage and indicate poor prognosis (Zent and Kay,

2010; Hodgson et al., 2011). Interestingly, autoantibodies in CLL cases are

mainly contributed by non-leukemic B cells, probably due to impaired peripheral

tolerance (Zent and Kay, 2010). Cytopenias are also induced frequently by

certain therapies such as purine analogues like fludarabine (Zent and Kay,

2010).

Other complications in CLL, which are primarily attributed to immune

dysfunction, include infections and secondary malignancies and are the most

common causes of death (Itala et al., 1992; Molica et al., 1993; Kyasa et al.

2004). The majority of infections are attributed to hypogammaglobulinemia and

are caused by common bacteria such as Streptococcus pneumoniae,

Haemophilus influenzae, Escherichia coli, Klebsiella pneumoniae, and

Pseudomonas aeruginosa (Morrison, 2010). Viral and fungal infections are less

common and are usually a side effect of certain therapies. Infections most

commonly involve herpes family viruses (herpes simplex, varicella zoster, CMV,

and EBV), and Aspergillus, Pneumocystis, and Candida (Morrison, 2010). The

mucosa is the most common site of involvement, and pulmonary diseases

Page 33: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

14

(overwhelmingly pneumonia) are the main cause of hospitalization and death

(Ahmed et al., 2003).

CLL patients are at increased risk of developing a second malignancy,

which are mainly non-hematological solid malignancies of skin and lung as well

as lymphomas, or other hematological malignancies (Maddocks-Christianson et

al., 2007; Hisada et al., 2001). Additionally CLL cells can transform into a high

grade lymphoproliferative disorder in 5-10% of cases (Tsimberidou et al., 2006;

Reiniger et al., 2006). Kyasa et al. (2004) found 16% of 132 CLL patients

developed an additional malignancy, which was the most common cause of

death followed by progressive CLL disease and infections. Although increased

risk of secondary malignancies is well documented following therapy, genetic

predisposition and loss of tumor surveillance are purported to play a role (Robak

et al., 2004; Maddocks-Christianson et al., 2007).

1.2.2 Heterogeneity

Using a wealth of CLL patient data gathered over twenty years, Boggs and

colleagues (Boggs et al., 1966) found variability in survival among patients,

confirming earlier hints that CLL was not a uniform disease. In the late 1990s, it

was established that mutational status of the IgVH genes was linked to clinical

outcome; leukemia cells that harbor mutated Ig genes (~50%) are associated

with indolent CLL, whereas lack of mutations in the Ig genes is associated with

aggressive disease and poor outcome (Damle et al., 1999; Hamblin et al., 1999).

The median survival for mutated is ~24 years compared to ~9 years for

Page 34: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

15

unmutated cases (Damle et al., 1999; Hamblin et al., 1999). Expression of other

markers, particularly CD38 and ZAP-70, has additionally been shown to correlate

with unmutated status but can also be independent prognosticators of disease

outcome (Hamblin et al., 2002; Crespo et al., 2003; Schroers et al., 2005).

Chromosomal defects, which are prognostic factors as well, may correspond to

mutational status. For example, cases with 13q14 deletion are also more likely to

carry mutations, and 11q22-q23 cases usually bear unmutated IgVH genes (Van

Bockstaele et al., 2009; Zenz et al., 2011). Unmutated cases also tend to have a

higher incidence of AIHA (Maura et al., 2013). Differences in clinical outcomes

can be traced to cellular properties, which show that unmutated CLL BCR are

more likely to bind self-antigens, and are more proficient at BCR signaling, partly

attributed to presence of ZAP-70 and/or CD38 (Herve et al., 2005; Guarini et al.,

2008).

Emerging evidence suggests that CLL likely evolves from a more benign

and biologically similar condition known as monoclonal B cell lymphocytosis

(MBL). Rawstron and co-workers (2002) sought to identify whether seemingly

healthy individuals carry CLL-like cells analogous to a monoclonal gammopathy

of undetermined significance (M-GUS) condition that precedes multiple myeloma.

They determined that ~3.5% of otherwise healthy adults over 40 years of age

carry monoclonal cells with a CLL immunophenotype. MBL is diagnosed (often

incidentally) by a finding of monoclonal B cell lymphoctosis < 5x109/L with CLL-

like cells in the absence of cytopenias, lymphadenopathy, or organomegaly

(Marti et al., 2005). First-degree relatives of CLL patients have a higher incidence

Page 35: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

16

of MBL (13.5 % - 18%) than controls (Shim et al., 2010). MBL with lymphocytosis

progresses to CLL requiring treatment at the rate of 1.1% per year (Rawstron et

al., 2008).

MBL has several features similar to CLL: the ratio of male to female

incidence is 1.9:1, the most common cytogenetic defects are 13q14 deletion

(48%) and trisomy 12 (20%), and there is a frequent increase in clonal T cell

populations (Rawstron et al., 2002; Rawstron et al., 2008; Fazi et al., 2011). In

addition, it has been reported that individuals with MBL have an increased risk of

developing bacterial infections. In one study, 16.2% of MBL and 18.4% of CLL

required hospitalization due to infections compared to 6.2% of controls (Moreira

et al., 2013). These similarities, taken together with gene profiling studies which

reveal that MBL has a characteristic profile of “good prognosis CLL”, has led to

the suggestion that MBL should be classified as early-stage CLL (Lanasa et al.,

2011; Kern et al., 2012). The observation that most MBL already harbor

cytogenetic defects of CLLyet progress slowly to CLL, suggests that these

defects may predispose individuals to CLL, but are not adequate for developing

frank CLL, which requires additional stochastic mutations to promote disease

progression.

About 5-10% of CLL cases transform into refractory high grade

lymphoproliferative disorders known as Richter’s transformation and

prolymphocytic transformation (Tsimberidou et al., 2006; Reiniger et al., 2006).

The appearance of large cells with reticular chromatin and prominent nucleoli

(mainly diffuse large B cell lymphoma) in CLL cases is termed Richter’s

Page 36: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

17

transformation. This transformation is usually accompanied by organomegaly

and constitutional symptoms, and reduced survival of less than 15 months

(Richter, 1928; Foucar and Rydell, 1980). In prolymphocytic transformation, the

CLL cells turn into immature-like lymphoblasts that retain the CLL

immunophenotype and survival ranges from a few months to 2 years (Enno et

al., 1979; Kjeldsberg and Marty, 1981; Ghani et al., 1986). The risk of developing

Richter’s is increased by a predominant nodal disease, CD38 expression, IgVH4

gene usage, and lack of 13q14 deletion (Rossi et al., 2008). Both Richter’s and

prolymphocytic transformations may be caused by miss-targeted AID, EBV

infection, and nucleoside analogue treatment (Robak et al., 2004; Thornton et al.,

2005).

1.2.3 Current Therapy

Early treatment of CLL, which was limited to radiation and chemotherapy

with arsenic compounds, was rarely effective (Minot and Isaacs, 1924; Wintrobe

and Hasenbush, 1939; Galton, 1959). However, advances came in the 1940s

and 1950s following a surge in development of chemotherapeutic compounds,

both new and those that had been successful in treating other cancers (Galton

1955; Galton, 1961; Shaw, 1961; Kaung et al., 1964). Of these, alkylating agents,

chlorambucil and cyclophosphamide, and the corticosteroid, prednisone, were

found to be fairly effective in treating CLL in clinical trials. Furthermore, these

drugs were easily administered orally and had modest side effects, except for

prednisone, which was associated with increased infections but was most

Page 37: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

18

effective for resolving hemolytic anemia and thrombocytopenia (Galton 1955;

Shaw, 1961; Galton, 1961; Kaung et al., 1964). These drugs became first-line

therapy for CLL for the following decades. Due to its low toxicity and convenient

administration, chlorambucil remains the best choice for elderly patients who are

unfit for standard therapy (Hallek, 2010).

Standard therapy for CLL today is based on chemoimmunotherapy.

Overall, CLL therapy currently draws from several drug classes that include

alkylating agents (chlorambucil, cyclophosphamide, and bendamustine), purine

nucleoside analogues (fludarabine, pentostatin, and cladribine), corticosteroids

such as prednisone, and monoclonal antibodies directed against CD20 (rituximab

and ofatumumab) and CD52 (alemtuzumab) (Halek et al., 2008; Gribben and

O’brien, 2011; Hallek, 2010). The purine analogues tend to be more effective

than alkylating agents or corticosteroids as monotherapies but they are more

myelotoxic (Keating et al., 1989; Hallek, 2010). However, combination therapies

are preferred in treating CLL because they provide superior overall responses

and survival. For instance, fludarabine plus cyclophosphamide can achieve an

overall response rate of over 80% and a complete remission rate of ~ 20%

(Catovsky et al., 2007; Hallek, 2010). Furthermore, addition of the monoclonal

antibody, rituximab, to the fludarabine and cyclophosphamide regimen is even

more effective with an overall response rate of over 90% and a complete

remission rate of over 40% in previously untreated patients (Keating et al., 2005;

Hallek, 2010). These monoclonal antibodies function through a variety of

mechanisms to achieve clearance of leukemic cells that include antibody-

Page 38: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

19

dependent cell cytotoxicity, complement-dependent cytotoxicity, and direct

signaling that promotes cell death (Jaglowski et al., 2010). They can be effective

even as single agents but function better in combination therapies, and are

particularly useful in treating autoimmune complications and relapsed/refractory

disease (Cheson, 2010; Jaglowski et al., 2010).

Based on variability in clinical outcomes in CLL, low-risk patients without

progressive disease are monitored rather than treated. In fact, studies have

shown that chemotherapy in early asymptomatic CLL does not alter survival

(Dighiero et al., 1998). Therefore, CLL patients in Rai stage 0/Binet stage A with

stable disease usually are not treated unless they show signs of progression

(Hallek, 2008). Symptomatic patients at any stage, or Rai stage III and IV/Binet

stage C patients are recommended for standard treatment, which generally

involves a combination of fludarabine, cyclophosphamide, and rituximab (Hallek,

2008). Refractory/relapsed or high-risk CLL, for example cases with 17p13

deletion or p53 mutations, can benefit from other regimens such as alemtuzumab

alone or in combination with fludarabine, or bendamustine with/without rituximab,

or the immunomodulator lenalidomide. CLL patients who fail these second-line

treatments may be eligible for a clinical trial or reduced-intensity allogeneic stem

cell transplant. Complications from AIHA or ITP are still best treated with

corticosteroids. Monoclonal antibodies specific for CD20 and CD52 have also

shown considerable effect (Gribben and O’brien, 2011). Otherwise, splenectomy

may be necessary (Hallek et al, 2008; Gribben and O’brien, 2011). There is no

Page 39: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

20

standard therapy for transformations, such as Richter’s, but it is often treated like

other high-grade non-Hodgkin’s lymphomas (Gribben and O’brien, 2011).

Newer therapies have greatly improved the management of CLL.

Monoclonal antibodies, for instance, have now become part of standard therapy

but they have also found utility in treating complicated disease as well. Their

success has inspired investigation on new and existing antibodies used in other

conditions for their effect on CLL. They include additional antibodies against

CD20 and to other markers expressed on CLL cells such as CD19, CD23, CD37,

CD38, CD40, and CD74 (Jaglowski et al. 2010; Cheson, 2010; Hallek, 2010).

Lenalidomide is a thalidomide-derived immunomodulatory agent with activity

against CLL (Kotla et al., 2009). This drug, which is in phase III clinical trials, has

been shown to be effective in treating relapsed and refractory CLL, and is one of

the few alternatives currently available where other therapy has failed (Chanan-

Khan et al., 2006; Ferrajoli et al., 2008; Hallek, 2010; Ramsay et al., 2008;

Schulz et al., 2013). Although the precise mechanism of action of lenalidomide in

CLL is unclear, this drug purportedly targets the tumor microenvironment through

multiple effects such as in reducing angiogenetic factors, reducing the interaction

of CLL cells with non-tumor support cells, and enhancing anti-tumor immune

responses through augmentation of NK and T cells activity as well as antigen

presentation by CLL cells (Kotla et al., 2009; Ramsay and Gribben, 2009).

Recently, lenalidomide has been shown to have the ability to block inhibitory

receptors on CLL cells thus restoring the formation of a functional immunological

synapse with T cells (Ramsay et al., 2012).

Page 40: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

21

Autologous stem cell transplant (SCT) in CLL is not curative but allogeneic

SCT has the potential of curing the disease. However, these therapies are only

indicated for a fraction of CLL patients (mainly younger patients with high-risk

disease) due to the advanced age and comorbidities in the typical CLL patient

(Toze et al., 2012). Due to high mortality rates associated with myeloablative

allogeneic SCT, even in younger patients, reduced-intensity conditioning (RIC)

allogeneic SCT is preferred for patients eligible for SCT (Toze et al., 2012;

Jaglowski and Byrd, 2012).

Our understanding of CLL biology has facilitated the development of

several novel targeted therapies that are very promising. These include: (i) Bcl2

family antagonists (ABT-263 and oblimersen) (Billard, 2012); (ii) cyclin-

dependent kinase inhibitors such as dinaciclib, and flavopiridol, an alternative

salvage therapy in phase III trials (Jaglowski and Byrd, 2012); and (iii) BCR

signaling inhibitors, mostly directed toward BCR kinases Syk (fostamatinib), Lyn

(dasatinib), and BTK (Ibrutinib), and PI3-kδ (GS1101) (Woyach et al., 2012).

Recently, T cells engineered to express a chimeric antigen receptor (CAR)

have shown promise in a small study. This CAR consists of CD19-specific

antigen-binding domains of an antibody coupled to T cell receptor signaling

domains to bypass major histocompatibility complex restriction. When autologous

CAR T cells were infused into three patients with advanced and chemorefractory

CLL (two patients had the 17p13 deletion), they expanded, persisted, and drove

leukemia into remission in two patients and induced partial response in the third

one. However, patients inevitably develop hypogammaglobulinemia because

Page 41: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

22

CAR T cells target leukemic and non-leukemic B cells all of which express CD19

(Kalos et al., 2011). Despite these major advances in treatment, CLL remains an

incurable disease.

1.3 CELLULAR AND MOLECULAR CHARACTERISTICS OF CLL

1.3.1 Normal B Cell Development and CLL Cell of Origin

B lymphocytes are white blood cells in the humoral arm of the adaptive

immune system where that produce antibodies to eliminate harmful antigens

(Cooper et al., 1965; Cooper, 1987). To generate an antibody repertoire with the

diversity required to indentify a potentially infinite number of foreign but not self-

antigens, these cells employ a number of diversification and selection

mechanisms in a highly ordered differentiation and developmental process

(Tonegawa, 1983; Hardy and Hayakawa, 2001; Glanville et al., 2009). Total or

partial failure in these mechanisms can lead to disorders ranging from

immunodeficiency and autoimmunity to B cell malignancies (Ballow, 2002; Sinha

et al., 1990; Goodnow, 1997; Shaffer et al., 2002).

B cell development after birth begins in hematopoietic progenitors found in

the BM and is completed in peripheral lymphoid organs (Figure 1.1). B cells arise

from the common lymphoid progenitor and proceed through several intermediate

differentiation stages to maturity as they gain immunocompetence. These

immature and mature B cell populations are distinguishable by expression of

Page 42: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

23

Figure 1.1 B cell development and differentiation. B cells arise from the common

lymphoid progenitor (CLP) in the bone marrow, where they develop initially in an

antigen-independent manner, and complete their maturation in secondary lymphoid

organs such as the spleen. The early B cell subsets are marked by formation of the B

cell receptor during three main waves of RAG1/2 expression to rearrange the

immunoglobulin heavy chain, light chain, and to remodel the light chain after recognizing

self-antigens (receptor editing), which is also postulated to occur in peripheral

transitional B cells that still retain autoreactivity and germinal center B cells that become

autoreactive after acquiring somatic hypermutations. Immature B cells migrate to the

spleen and progress through transitional (T) stages 1 and 2 to become follicular (FO)

mature B cells, which when stimulated by antigens differentiate into antibody-producing

plasma cells and memory B cells. Transitional B cells also give rise to marginal zone

(MZ) B cells, and to a lesser extent, to B1 B cells, which largely derive from liver

progenitors in the fetus and neonate (Adapted from Hardy, 2006 and Hardy et al.,

2007)specific markers, morphology, function, or a combination of these features (Hardy

and Hayakawa, 2001).

Page 43: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

24

Early B-lineage cells in mice express pan-B cell markers, namely B220

followed by CD19 and are marked by the ordered assembly of immunoglobulin

(Ig) gene segments, which are otherwise separated on the chromosome, to form

Ig heavy and light chain genes that encode the B cell receptor (BCR) (Tonegawa,

1983; Hardy et al., 2007). The process of Ig gene assembly, together with other

diversification mechanisms, has been estimated to yield B cells with an excess of

3.5x1010 unique specificities in humans (Glanville et al., 2009). The Ig gene

assembly process begins at the B220+CD43+ pro-B cell stage, where the

recombination activating genes 1 and 2 (RAG1 and RAG2) are expressed

(Oettinger et al., 1990). The RAG1 and RAG2 proteins together initiate site-

specific DNA recombination that joins variable (V), diversity (D), and joining (J)

immunoglobulin gene segments in a process known as V(D)J recombination

(Early et al., 1980; Oettinger et al., 1990). This process begins in the Ig heavy

chain (IgH) locus with the joining of a DH and JH gene segment, followed by

joining of an upstream VH segment to the rearranged DH-JH gene segments to

form the variable exon of the heavy chain gene. Imprecise joining of these gene

segments in developing lymphocytes during V(D)J recombination contributes

additional diversity to antigen receptors (Gersteins and Lieber, 1993; Lieber et

al., 2004).

Upon successful completion of rearrangement, transcription ensues and

the variable exon is joined to a µ constant region by RNA splicing to produce the

mature IgHµ mRNA (Ghia et al., 1998; Hardy et al., 2007). RAG1/2 expression is

extinguished and the rearranged IgHμ heavy chain pairs with a surrogate light

Page 44: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

25

chain, which is composed of the proteins VpreB and λ5, and is expressed on the

cell surface as the pre-B cell receptor (pre-BCR) (Tsubata and Reth, 1990). The

pre-BCR marks a critical checkpoint in determining the functionality of the

rearranged heavy chain; successful pre-BCR formation allows signaling that

leads to proliferation of late pro-B cells to become large pre-B cells. When large

pre-B cells cease proliferating, they become small pre-B cells at which point

RAG1 and RAG2 are re-expressed to assemble the Ig light chain (IgL). In mice

and humans, rearrangement begins at a single IgLκ allele, followed by the

second allele, and finally IgLλ until a productive light chain is achieved. Failure to

produce functional rearrangements in either the IgH or IgL loci leads the B cell to

undergo apoptosis. Successful pairing of IgHμ with a light chain leads to the

assembly of a BCR that is capable of antigen binding and is expressed on the

cell surface, which marks the immature B cell stage

(CD19+B220+CD43+IgMhiIgD) (Hardy et al. 2007).

Since “primary” V(D)J rearrangement is an antigen-independent process,

a large number (up to 75%) of newly-formed immature B cells are inherently

autoreactive (Wardemann et al., 2003; Nemazee, 2006). This immature B cell

stage therefore, marks a critical checkpoint in enforcement of central tolerance

by negative selection or “re-education” of B cells that bind self-antigens.

Depending on the anatomical location and the nature of self-antigens bound by

immature B cells, there are three possible outcomes (Figure 1.2): (i) clonal

deletion by programmed cell death, (ii) receptor editing, and (iii) functional

inactivation (Jankovic et al., 2004; Goodnow et al., 2005; Hillion et al., 2005;

Page 45: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

26

Nemazee, 2006). Receptor editing involves re-expression of RAG1 and RAG2

and subsequent rearrangement of the light chain in an attempt to remodel the

autoreactive BCR into an innocuous one (Tiegs et al., 1993; Girschick et al.,

2001; Nemazee, 2006). Mechanisms of inactivation include intrinsic programs

that downregulate the BCR and its positive regulators and upregulate its negative

regulators such as CD5 rendering the B cell anergic, or extrinsic factors such as

limitation of growth factors and costimulation/inflammatory signals, as well as the

activity of regulatory cells (Goodnow et al., 2005; Basten and Silveira, 2010).

Through alternative RNA splicing, immature B cells begin to express IgD

on the surface and migrate from the BM as transitional (T) B cells to continue

differentiation in peripheral lymphoid organs such as spleen and lymph nodes.

Functionality as well as variation in IgD, IgM, CD21, and CD23 levels can further

distinguish transitional B cells into T1, T2, and T3 subsets. A fraction of T1 B

cells may undergo receptor editing to remove remnant or new unwanted

specificities. T1 B cells progress to the T2 stage, and eventually to

CD19+B220+IgMintIgDhiCD21loCD23+ competent mature follicular (FO) B cells

(King and Monroe, 2000; Chung et al., 2003; Carsetti et al., 2004). In addition to

FO B cells, transitional B cells also give rise to other mature B cell populations

including marginal zone (MZ) and B1 B cells, but this appears to occur in a

ligand-dependent manner (Berland and Wortis, 2002; Pillai and Cariappa, 2009).

Naïve mature FO B cells that encounter a pathogen and receive costimulation

Page 46: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

27

Figure 1.2 Mechanisms of self-tolerance in B cells. To maintain tolerance, B cells

that bind self-antigens during development either re-express RAG1/2 and undergo

secondary V(D)J rearrangement (receptor editing) to remodel the B cell receptor,

become functionally inactivated through indiction of intrinsic programs or B cell extrinsic

factors, or are deleted by apoptosis (Adapted from Goodnow et al., 2005).

Page 47: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

28

from follicular helper T cells that recognize the cognate antigen form proliferation

structures in lymphoid follicles known as germinal centers (GCs) in secondary

lymphoid organs ( Shlomchik and Weisel, 2012; Victora and Nussenzweig,

2012). Between cycles of proliferation in the GC, point mutations are introduced

into the variable regions of the BCR genes by the enzyme activation-induced

cytidine deaminase (AID) in a process also known as somatic hypermutation

(SHM) (Muramatsu et al., 2000). Mutations that change the amino acid sequence

also alter the binding affinity of the receptor. This is followed by a series of

positive selection events and proliferation of B cells with high affinity BCRs in

what is known as affinity maturation (Rajewsky, 1996). B cells that receive

catastrophic mutations or gain autoreactivity through SHM generally undergo cell

death (Rajewsky, 1996). In some instances, B cells acquiring autoreactivity in

response to SHM or infection/immunization may undergo receptor revision, which

is similar to receptor editing but occurs in the periphery as opposed to the BM

(Noia and Neuberger, 2007; Victora and Nussenzweig, 2012; Hillion et al., 2005;

Nemazee and Weigert, 2000). AID activity can also lead to DNA double-strand

breaks that foster replacement of the µ heavy chain constant region genes with

other classes/isotypes of heavy chain constant region genes in what is known as

class switch recombination (CSR). CSR alters the constant and not variable

portion of the receptor, and therefore affects antibody function but not specificity

(Muramatsu et al., 2000; Stavnezer et al., 2008). B cells that survive these

germinal center reactions terminally differentiate into long-lived memory B cells

or antibody-producing plasma cells (Shlomchik and Weisel, 2012).

Page 48: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

29

Beside conventional B cells, innate-like MZ and B1 B cells contribute to

the pool of mature B cell subsets in the periphery. MZ and B1 B cells display a

limited repertoire of antigen receptors compared to mature FO B cells that are

believed to be selected by antigens during development. These cells however,

have evolved as such to provide rapid responses early during infection at the

expense of forming classical memory B cells (Martin et al., 2001; Bendelac et al.,

2001).

MZ B cells, which are distinguished by expression of high levels of CD21

and CD1d and low levels of CD23, are thought to originate by positive selection

from the conventional B2 lineage during development by antigens that induce

BCR signaling in a thymus independent (TI) manner (Wen et al., 2005; Pillai et

al., 2005). MZ B cells derive their name from splenic marginal zone areas in

follicles where they reside. This location allows them to be among the first B cells

to encounter particulate blood-borne antigens and thus they are important in

early control of pathogens during infection (Martin et al., 2001).

B1 B cells are long-lived self-renewing CD19+B220loIgMhiIgDloCD23- B

cells that can be either be CD5+ (B1a) or CD5- (B1b) that populate peritoneal and

pleural cavities (Berland and Wortis, 2002; Hardy, 2006). B1 B cells are thought

to originate primarily through a distinct lineage from neonatal/fetal precursors in

the liver and omentum but may also be derived through induced differentiation of

transitional B cells in the conventional B2 lineage via BCR cross-linking by

multivalent TI antigens (Hayakawa et al., 2003; Berland and Wortis, 2002; Hardy,

2006; Montecino-Rodriguez et al., 2006; Montecino-Rodriguez and Dorshkind

Page 49: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

30

2012). Consequently, B1 B cells express BCRs that have a limited repertoire

which lack mutations, and possess naturally low affinity and polyreactivity toward

foreign and self-antigens. In fact, many known specificities of B1 B cells are to

membrane lipids, oxidized lipids, and red blood cell epitopes. It is therefore not

surprising that B1 cells are implicated in several autoimmune conditions when

tolerance is breached (Jamin et al., 1993; Pao et al., 2007; Jellusova et al.,

2010). However, these B cells are maintained because their BCRs also cross-

react with epitopes found on common pathogens such as bacterial capsular

polysaccharides (Hardy, 2006; Berland and Wortis, 2002). Therefore, like MZ B

cells, these cells form a critical first line of defense by spontaneous production of

IgM antibodies (natural antibodies) particularly against encapsulated bacteria

(Bendelac et al., 2001; Carsetti et al., 2004).

The origins of a lymphoproliferative disorder are linked to the

differentiation stage at which it occurs (Kuppers, 2005). Identifying normal

counterparts, which are often blurred by atypical characteristics in malignant

cells, is critical to understanding the etiology and pathogenesis of these

diseases. Transformation by chromosomal translocations involving Ig loci and

(proto)oncogenes are molecular hallmarks of many types of B cell malignancies,

but such defining lesions are absent in CLL, leaving its origins unresolved

(Kuppers, 2005). However, based on the ability of CLL patients’ hematopoietic

stem cells (HSCs) to form unique CLL-like oligoclonal or monoclonal B cells

when transplanted in mice, Kikushige et al. (2011) suggest that transformation

events can also occur much earlier than perceived. Since CLL cases carrying

Page 50: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

31

mutated Ig variable heavy chain (IgVH) genes are associated with good

prognosis while those with unmutated IgVH show poor survival (Hamblin et al.,

1999), it is unclear whether these two clinically opposed groups develop by

divergent or convergent mechanisms.

Comparative gene expression profiling using oligonucleotide microrrays

has been a powerful tool in gaining insight into the origins of CLL. Several

studies utilizing microarrays show that CLL cells have a distinct gene expression

profile compared to normal B cell subsets or other malignant populations (Klein

et al., 2001; Rosenwald et al., 2001; Jelinek et al., 2003). Moreover, clinically

heterogeneous CLL cases distinguished on the basis of IgVH mutation status

display a common gene expression profile suggesting that they derive from the

same precursor or undergo similar transformation mechanisms (Klein et al.,

2001; Rosenwald et al., 2001). Interestingly, Klein et al. (2001) reported that CLL

cells are mostly related to normal memory B cells compared to normal naïve,

CD5+, or GC B cell subsets. This finding contradicted the previous view that CLL

was a disease of naïve B cells, but was supported by Damle and colleagues

(2002) by showing that CLL cells express CD23, CD25, CD69, CD71, and have

reduced expression of CD22, FcγRIIb, CD79b, and IgD, which is a surface

phenotype associated with antigen-experienced B cells. CLL cells additionally

express the memory marker, CD27, but this marker is also expressed on human

MZ B cells. MZ B cells can also express other memory B-like markers, and

because they can participate in both thymus-dependent and independent antigen

responses and bear BCR configurations that are either mutated or unmutated,

Page 51: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

32

this subset provides convenient possibilities for how mutated and unmutated CLL

may emerge. However, MZ B cells typically do not express CD5 or CD23

(Chiorazzi and Ferrarini, 2011; Weill et al., 2009).

More recently, Seifert et al. (2012) using comparative gene profiling of

CLL cells and sorted normal B cell subsets, reported that CLL originates from

CD5+ B cells. Unlike earlier studies, CD27+ memory B cells, which are not a

homogeneous population, were split into IgM+ and class-switched memory B

cells, and adult PB mature CD5+ B cell subsets were chosen instead of using

cells derived from umbilical cord blood, which is conveniently rich in CD5+ B cells

but are mainly immature. Gene expression profiles of CLL cells were compared

with that of normal mature CD5+, naïve, splenic MZ, and IgM+ and class-switched

memory B cells. This study found that while CLL cells have a distinct gene

expression signature compared to normal B cell subsets, the leukemic cells are

most closely related to CD5+ and naïve B cells. Furthermore, CLL with

unmutated IgVH most closely resemble mature CD5+CD27- PB B cells while CLL

with mutated IgVH genes most closely resemble a previously unknown

CD5+IgM+CD27+ post-GC B cell subset (Seifert et al., 2012).

CD5 is a hallmark of CLL cells as well as the murine B1a B cell.

Additionally, CLL and B1a B cells share other features including interleukin 10

(IL-10) production, and a limited BCR repertoire, leading to the hypothesis that

the natural counterpart for CLL is the human equivalent of the B1a B cell (Dono

et al., 2004; Nordgren and Joshi, 2010). Both CLL cells and murine B1 B cells

are known to produce IL-10, which they probably use as an autocrine growth

Page 52: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

33

factor or for immunosuppressive functions (O’Garra et al, 1992; Gary-Gouy et al.,

2002; 2010; DiLillo et al., 2013). CLL exhibits a restricted Ig gene repertoire and

displays an antibody reactivity profile skewed toward cytoskeletal and

membrane-associated self, modified self, and bacterial antigens (Rosen et al.,

2010). This reactivity profile is also shared by B1 and MZ B cells that are

positively selected for these specificities, as well as subsets of developing

(transitional) and extrafollicular B cells that exhibit self-reactivity produced by

primary Ig gene rearrangements, or as an unintended consequence of somatic

mutation, respectively (Rosen et al., 2010). The human B1 B cell, however, is

elusive since CD5 is not a B cell subset marker in humans. A recent description

of a CD20+CD27+CD43+CD70- human blood B1 B cell population that shares

properties with murine B1 cells such as unmutated repertoire, antigenic

specificity, BCR signaling, and natural antibody production was received with

controversy (Griffin et al., 2011, Griffin et al., 2012, Reynaud and Weill, 2012;

Descatoire et al., 2011).

Approximately 30% of circulating B cells in humans display CD5 on their

surface, which is expressed constitutively or upon induction (Youinou et al.,

1999). CD5 can be expressed in transitional or extrafollicular B cells undergoing

receptor editing or revision, respectively (Hillion et al., 2005; Sims et al., 2005). In

addition, CD5 expression may be induced on B cells rendered anergic by chronic

(auto)antigenic stimulation (Hippen et al., 2000). CD5 negatively regulates B cell

receptor signaling, which, given the proclivity of CD5+ B cells toward self-

reactivity, can serve a protective role by suppressing B cell activation to limit

Page 53: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

34

autoantibody production (Youinou et al., 2006). Thus, because CLL shares a

similar pattern of antigenic reactivity and several markers including CD5

expression with a variety of B cell subsets, all these may be considered potential

etiological sources of human CLL, which may partly explain the clinical

heterogeneity of this disease.

1.3.2 Genetic and Chromosomal Defects in CLL

Although there is no single lesion that defines all CLL, a series of

chromosomal abnormalities and deregulated genes are frequently detected

(Klein and Dalla-Favera, 2010; Kuppers, 2005. The first chromosomal defects in

CLL were found using conventional cytogenetics but this was complicated by the

fact that mitogens were required to induce mitosis since CLL cells divide slowly.

These methods found that ~50% of CLL cases carried chromosomal defects with

the most common being trisomy 12 and chromosome 13q deletion (Juliusson et

al., 1990). More sensitive methods utilizing comparative genomic hybridization or

fluorescence in-situ hybridization show that over 80% of CLL patients carry a

chromosomal abnormality (Bentz et al., 1995; Dohner et al., 2000). The most

common are 13q14 deletion (40-60%), 11q22-q23 deletion (15-20%), trisomy 12

(15-30%), 17p13 deletion (~10%), 6q deletion (6%), and rare abnormalities

(trisomy 3q, trisomy 8q, and translocation involving 14q32) accounting for 12% of

cases (Matutes et al., 1996; Dohner et al., 2000; Van Bockstaele et al., 2009).

Most chromosomal defects in CLL are prognostic factors that define

subgroups. Patients with CLL cells bearing 17p13 and 11q22-q23 deletion have

Page 54: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

35

the worst prognosis with median survival of 32 and 79 months, respectively

(Dohner et al., 2000). Interestingly, the most common defect (13q14 deletion) is

associated with a better outcome (133 months) compared to normal karyotype

(111 months) while trisomy 12, although associated with atypical morphology,

remains relatively stable with a median survival of 114 months (Dohner et al.,

2000; Matutes et al., 1996). Moreover, 17p13 and 11q22-q23 deletions are

associated with late diagnosis in addition to an aggressive nodal disease for

11q22-q23 deletions (Dohner et al., 1997), and refractory therapy in 17p13

deletions (Dohner et al., 1995).

That cytogenetic defects are related to clinical outcomes suggests that

certain genes in the affected loci are involved in CLL etiology or progression.

Some of these deletions are quite large and involve many genes. However, gene

expression profiling of CLL subgroups defined by chromosomal aberrations

confirms that many of the differentially expressed genes map to the deleted

regions in the respective chromosomes (Haslinger et al., 2004). Trisomy 12, for

example, implicates dosage alteration for an unknown number of genes. The

most common aberration, 13q14 deletion, involves a cluster of genes in the

minimally deleted region that include a potential tumor suppressor DLEU2, and

microRNAs miR15a and miR16-1 (Liu et al., 1997; Calin et al., 2002). These

miRNAs, which are known to play a role in prostate cancer, are reduced in ~68%

of CLL cases (Bonci et al., 2008; Calin et al., 2002). miR15a and miR16-1 targets

include CCND1, Wnt3a, and Bcl2 in prostate tumors (Bonci et al., 2008) and can

inhibit Bcl2 leading to apoptosis of leukemia cell lines (Cimmino et al., 2005).

Page 55: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

36

Deletion of this region in mice leads to a spectrum of CLL-like lymphoproliferative

diseases suggesting that it may be involved in pathogenesis of CLL (Klein et al.,

2010; Lia et al., 2011). The next common defect, 11q22-q23 deletion, is

associated with loss the DNA double-strand break sensing gene ataxia

telangiactasia mutated (ATM). Additionally, it is common to find mutations in the

remaining allele or mutations in both alleles without a deletion, suggesting that

ATM is important for driving pathogenesis in a subset of CLL cases (Schaffner et

al., 1999). Skowronska et al. (2011) analyzed ATM mutations and deletions in

CLL cells and found that a single loss of ATM at one allele was unlikely to initiate

CLL but rather influenced rapid disease progression through total loss of ATM

after a stochastic mutation in the remaining allele. 17p13 deletion leads to loss of

the tumor suppressor protein TP53 (p53). About 4.5% of CLL patients show loss

of p53 but without 17p deletion, which is associated with poor prognosis and is

attributed to mutation of this gene (Zenz et al., 2010).

Other gene defects occur frequently but are not associated with

chromosomal aberrations. For example miR-29 and miR-181, which target and

are inversely correlated with expression of the proto-oncogene T cell leukemia 1

(TCL1), are commonly downregulated in poor prognosis CLL (Pekarsky et al.,

2006). Bcl2 and other Bcl2-family genes are known to be overexpressed in CLL

and confer resistance to apoptosis (Hanada et al., 1993; Buggins and Pepper

2010). Other genes that are often overexpressed in CLL include Notch1, SF3B1,

and DAPK. Deregulation of many of these genes has been attributed to

epigenetics. For instance, abnormal methylation patterns have been shown to

Page 56: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

37

alter TCL1, DAPK1, and Bcl2 expression in CLL (Plass et al., 2007; Zenz 2010;

Mansouri et al., 2012).

1.3.3 The Role of the B Cell Receptor

Many B cell lymphoproliferative disorders, like normal B cells, depend on

the BCR expression for survival (Kuppers, 2005). There is increasing evidence

that CLL does not only depend on the BCR, but the BCR plays a central role in

pathogenesis. During analyses of CLL BCRs in the 1990s, it was discovered that

mutation status of IgVH genes separates CLL cases into two groups: those

bearing mutations (mutated) and those without (unmutated) (Schroeder and

Dighiero, 1994; Fais et al., 1998). Thereafter, it was shown that mutation status

(defined as greater or lesser than 98% homology to the nearest germline) is

related to clinical outcome; the BCRs in approximately half of CLL cases are

mutated and the patients have good prognosis while the remaining cases have

unmutated BCRs and the patients have poor prognosis (Hamblin et al., 1999;

Damle et al., 1999; Maura et al., 2013). In a clever experiment, Herve et al.

(2005) showed that unmutated CLL expressed polyreactive and autoreactive

antibodies that were not seen in mutated cases; however, in vitro reversion of

mutations back to the germline configuration in the latter cases restored capacity

to bind self-antigens, suggesting that signaling through the BCR may explain

clinical heterogeneity based on mutation status. These differences in mutation

status raise the question whether unmutated and mutated cases derive from two

precursors: naïve and post-GC B cells, respectively (Fais et al., 1998).

Page 57: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

38

Additional evidence that the BCR itself is involved in the etiology of CLL

stems from the observation that CLL has a biased repertoire, and 20-30% of

cases have highly similar or even identical BCRs based on complementarity

determining region 3 (CDR3) homology, which is referred to as stereotypy

(Messmer et al., 2004; Ghiotto et al., 2004; Widhopf et al., 2004; Stamatopoulos

et al., 2007; Agathangelidis et al., 2012). The CDR3 is a hypervariable region

formed at the V(D)J junction that determines the specificity of the receptor (Xu

and Davis , 2000). In humans, the probability of getting the same three gene

segments in a heavy chain is 1 in 7650 and over 1 in 1.5 million for both heavy

and κ light chain (Widhopf et al., 2004). The remarkable odds at which

stereotypic BCRs occur in CLL imply that selection of CLL cells is driven by a

limited group of common antigens (Ghiotto et al., 2004). The most frequently

used IgVH genes are VH1-69, VH3-07, and VH4-34. These genes preferentially

associate with a set of D and J genes resulting in similar structures. Typically,

VH1-69 has a long acidic CDR3 with many tyrosines and lacks mutations; VH3-

07 has a short CDR3 that is minimally acidic and generally mutated; VH4-34 also

has long acidic CDR3 with many tyrosines and may either be mutated or

unmutated (Chiorazzi and Ferrarini, 2003). Some CLL cases use VH3-21 which

tends to pair with Vλ2-14 and is associated with poor survival independent of

mutation status (Thorselius et al., 2006).

CLL cells have a restricted Ig repertoire that recognizes apoptotic cell

debris and microbial motifs (Rosen et al., 2010). Earlier knowledge that CLL

patients have a high incidence of autoimmune hemolytic anemia and

Page 58: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

39

thrombocytopenia and that CD5+ B cells are involved in autoimmune conditions

in mice has led to the finding that serum from CLL patients contains

crossreactive antibodies with specificities to formed blood elements and

cytoplasmic and nuclear antigens (Hamblin et al., 1986; Brooker et al., 1988;

Sthoeger et al., 1989). More recently, screening the specificities of expressed

and purified CLL antibodies found that antibodies bound neoantigens revealed

during apoptosis such as non-muscle myosin heavy chain IIA, vimentin, filamin

B, coffilin-1, cardiolipin, and proline-rich acidic protein-1, and modified epitopes

such as oxidized Low-density lipoprotein (Ox-LDL). CLL antibodies also bound

Streptococcus pneumoniae polysaccharide which like Ox-LDL, contains

phosphorylcholine (Catera et al., 2008; Myhrinder et al., 2008; Chu et al., 2010).

Reactivity to other microbial antigens have been reported by Hoogeboom et al.

(2013) who found that a subset of mutated CLL recognized fungal glycans, and

Kostareli et al. (2009) who found persistent EBV and CMV DNA in a subset of

patients using VH4-34, suggesting that fungal and viral antigens could be

involved in CLL pathogenesis in a subset of patients. This reactivity profile

skewed toward cytoskeletal and membrane-associated self, modified self, and

bacterial antigens is also shared by B1 and MZ B cells that are positively

selected for these specificities (Bendelac et al., 2001). Since natural antibodies

from healthy population also recognize most of these antigens (Catera et al.,

2008), Myhrinder et al., 2008), and Chu et al., 2010), it raises the possibility that

CLL derives from B cells endowed with scavenger ability for apoptotic debris and

removal of pathogenic bacteria.

Page 59: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

40

The BCR in CLL appears to be engaged by antigens but atypical

expression of signaling components indicate some slight differences from normal

BCR signaling (Woyach et al., 2012). BCR signal transduction occurs through a

multicomponent cascade regulated primarily by protein tyrosine kinases and

phosphatases. The BCR is linked to Igα (CD79a) and Igβ (CD79b), which contain

immunoreceptor tyrosine-based activation motifs (ITAMs). This BCR signaling

complex is also coupled with a number of positive (CD19, CD45, and CD21) and

negative (CD5, CD22, CD72, and FcγRIIB) regulators that balance immunity and

tolerance. Activation occurs when BCRs are crosslinked by antigens, leading to

oligomerization and phosphorylation of the ITAMs by the Src family protein

tryrosine kinase, Lyn. Lyn also phosphorylates the cytoplasmic tail of CD19 to

activate phosphatidylinositol 3-kinase (PI3-K) signaling. Spleen tyrosine kinase

(Syk) is recruited to the phosphorylated ITAMs and phosphorylates a number of

targets in turn setting off a cascade that activates NF-κB and MAP kinase

pathways (Goodnow, 2001; Xu et al., 2005; Kurosaki et al., 2010; Stevenson et

al., 2011). Expression patterns of BCR signaling components in CLL indicate

previous or on-going engagement of the antigen receptor. CD5 is a scavenger

receptor family glycoprotein normally expressed in T cells but also seen in a

subset of B cells. In T cells, CD5 acts as a negative regulator in fine-tuning T cell

receptor (TCR) signals in response to antigens (Azzam et al., 2001). In B cells,

CD5 appears to play the same role in negatively regulating BCR signals in

response to antigens since genetic disruption of CD5 leads to loss of tolerance in

mice (Bikah et al., 1996; Hippen et al., 2000). CD5 functions by recruiting the

Page 60: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

41

inhibitory protein tyrosine phosphatase SHP-1 to the BCR in B1 cells (Sen et al.,

1999; Ochi and Watanabe, 2000). Downregulation of B220, an isoform of CD45

(Ptprc) and pan-B cell marker that acts as a positive regulator of BCR signaling,

suggests a concerted effort with CD5 to quell antigen-induced BCR signals in

B1a B cells (Hardy and Hayakawa, 2001; Berland and Wortis, 2002). The surface

immunophenotype comprising of CD5, a hallmark of CLL, reduced expression of

IgM, IgD, and CD79b, and expression of activation markers postulate that these

cells derive or are driven by antigenic stimulation (Caligaris-Cappio et al., 1982;

Matutes and Polliack, 2000; Damle et al., 1999). Functionally, ligation of the

BCR in CLL cell has been shown to induce the expression of anti-apoptotic

molecules Bcl2 and Mcl1 through NF-κB leading to increased survival,

particularly in unmutated cases (Bernal et al., 2001; Guarini et al., 2008).

CLL cells oddly express TCR signaling components. Cytotoxic T-

lymphocyte antigen 4 (CTLA4) is expressed in T cells as an inhibitor of TCR

signals since mice with this deficiency have uncontrolled proliferation of T cells

and develop autoimmunity (Alegre et al., 2001; Goodnow et al., 2005). CTLA4 is

also expressed on CLL B cells but its expression correlates inversely with

disease progression, indicating that CTLA4 plays a similar role in B cells and

consequently, loss of BCR signal suppression leads to expansion of CLL cells

(Kosmaczewska et al., 2005; Joshi et al., 2007). ζ–chain-associated protein

kinase 70 (ZAP-70) is expressed in T cells and natural killer cells and is required

for propagating receptor signals (Au-Yeung et al., 2009). Rosenwald et al.

(2001), using microarrays, found the most reliable gene that discriminated

Page 61: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

42

unmutated and mutated CLL cases was ZAP-70. Expression of ZAP-70 in CLL is

associated with poor prognosis (Durig et al., 2003). In CLL cells, ZAP-70 plays a

similar role to Syk and enhances BCR signals (Stevenson et al., 2011; Woyach

et al., 2012). CD38 is highly expressed in activated B and T cells as well as a

subset of CLL cells (Malavasi et al., 2011). Moreover, CD38 expression, similar

to unmutated and ZAP-70+ CLL, is associated with a distinct gene expression

profile and poor prognosis, linking BCR signals to cell function and clinical

outcome (Damle et al., 1999, Deaglio et al., 2003; Joshi et al., 2007). Because of

the relative ease in determining CD38 and ZAP-70 expression by flow cytometry,

they can act as surrogate markers for mutation status (Hamblin et al., 2002;

Crespo et al., 2003). Inhibition of BCR signaling with small molecule inhibitors

that target Syk (fostamatinib) and phosphatidylinositol 3-kinase delta (PI3-kδ)

inhibitor (CAL-101) or deleting PKCβ blocks BCR signaling and CLL cell survival

and migration in vitro, and tumor growth in mice (Quiroga et al., 2009; Suljagic et

al., 2010; Hoellenriegel et al., 2011; Holler et al., 2009). Currently, there are nine

inhibitors that target BCR signaling in early phase clinical trials for B cell

malignancies that include four for CLL (fostamatinib, Dasatinib, CAL-101, and

Ibrutinib) (Woyach et al., 2012).

1.3.4 Role of theTumor Microenvironment

CLL cells undergo spontaneous apoptosis in vitro and are refractory to

long-term culture (Collins et al., 1989). This is remarkable since CLL cells often

overexpress anti-apoptotic molecules such as Bcl2 and Mcl1 (Buggins and

Page 62: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

43

Pepper 2010). In fact, for many years, CLL was regarded as a disease of

accumulation, rather than proliferation, of lymphocytes defective in apoptosis

(Dameshek, 1967; Andreeff et al., 1980; Caligaris-Cappio and Hamblin, 1999).

This later changed when in vivo labeling with heavy water showed that CLL cells

proliferate and have a much higher turnover than earlier perceived (Messmer et

al., 2005). This contradiction between in vivo and in vitro activity of CLL cells

strongly implicates the involvement of some extrinsic cellular or soluble factor(s)

that typical cultures do not recapitulate.

Infiltration of CLL cells disrupts and re-organizes the normal architecture

of lymphoid organs (Fecteau and Kipps, 2012). In the lymph node (LN), BM, and

spleen to a smaller extent, CLL cells can be found intermixed non-leukemic cells

such as T cells and follicular dendritic cells (FDCs) in histological structures

named pseudofollicles or proliferation centers (PCs) (Swerdlow et al., 1984;

Soma et al., 2006; Schmid and Isaacson, 2007). CLL cells in pseudofollicles

express the proliferation marker Ki67 and constitutively activate NF-κB (Herreros

et al., 2010). These specialized niches or microenvironments are common in

many tumors and provide support for tumor growth, escape from immune

surveillance, and chemoresistance (Whiteside, 2008).

The CLL tumor microenviroment is comprised of a feedback network

between leukemic and support cells that is mediated by cytokines/chemokines,

and direct interaction. The major cytokine/chemokine players are those of the

tumor necrosis factor (TNF) family cytokines such B cell-activating factor of TNF

family (BAFF) and a proliferation-inducing ligand (APRIL), which are produced by

Page 63: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

44

accessory cells (Fecteau and Kipps, 2012; Endo et al., 2007). BAFF and APRIL

have been reported to support survival of CLL cells in vitro by engaging their

receptors, B cell maturation antigen (BCMA) and transmembrane activator and

calcium modulator cyclophilin ligand-interactor (TACI) on CLL cells, to activate

canonical NFκ-B signaling (Endo et al., 2007). The chemokines CXCL12 (stromal

cell-derived factor-1, SDF-1) and CXCL13, also produced by support cells, attract

leukemic cells and other lymphocytes that express CXCR4 (Fecteau and Kipps,

2012). CLL cells can also receive direct signals, for example, through

engagement of CD40 on leukemia cells and CD40L on T cells. Stimulated CLL

cells, in turn, produce CCL3, CCL4 and CCL22 to recruit more T cells and

monocytes to the pseudofollicles (Endo et al., 2007; Fecteau and Kipps, 2012).

Co-culture experiments show that normal cells such as FDCs, endothelial

cells, and BM stromal cells (BMSCs) can enhance survival of CLL cells

(Pedersen et al. 2002; Maffei et al., 2011; Panayiotidis et al., 1996). BM stromal

cells (BMSCs), which naturally support developing B cell precursors, have been

found to prolong survival and prevent apoptosis of CLL cells in vitro, indicating

that these cells have the potential to protect CLL cells in the BM (Panayiotidis et

al., 1996). BMSCs produce SDF-1 to attract leukemic cells, which express

CXCR4, and tether them via VCAM-1 on their surface to CD49d on the leukemic

cells (Fecteau and Kipps, 2012). BMSCs can also induce resistance to

chemotherapeutic drugs in vitro in a mechanism that involves Notch signaling

(Kamdje et al., 2012). BMSCs may contribute to pathogenetic mechanisms since

Page 64: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

45

this interaction can induce genetic changes in leukemic cells including

upregulation of the TCL1, a known oncogene (Sivina et al., 2012).

In an effort to determine other cells that might provide support to cultured

CLL cells, Burger et al. (2000) found large, round, and adherent peripheral blood

mononuclear cells that protected co-cultured leukemic cells from apoptosis akin

to BSMCs. Because these cells were reminiscent of thymic nurse cells, they

were named nurse-like cells (NLCs). NLCs have also been found in spleens of

CLL patients (Burger et al., 2000; Tsukada et al., 2002). NLCs are CD14+

monocyte-derived cells that express SDF-1, normally produced by BM stroma to

support developing early B cell precursors, through which they protect CLL cells

from apoptosis. NLCs additionally secrete BAFF and APRIL, which promote

survival of CLL cells in a SDF-1-independent pathway that requires NF-κB to

induce Mcl-1. NLCs express CD31, a ligand for CD38, which is expressed in a

subset of CLL cases and may provide additional survival signals and modulate

cytokine production by CLL cells (Fecteau and Kipps, 2012). CLL cells not only

subvert NLCs to produce survival factors, but may use them to evade immune

detection, based on comparative gene expression profiling studies showing that

NLCs derived from co-cultures with CLL with CD14+ PBMCs from healthy or CLL

patients exhibit impaired immune competence (Bhattacharya et al., 2011). NLCs

produce chemokines SDF-1 and CXCL13, which recruit CLL cells since they

express their receptors, CXCR4 and CXCR5, respectively. CLL cells, in turn,

produce CCL21, CCL3 and CCL4 to recruit more monocytes and T cells to PCs

(Fecteau and Kipps, 2012).

Page 65: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

46

Abnormalities of the T cell compartment in CLL were first reported in

1980s. The ratio of circulating CD4+ T cells to CD8+ T cells is inverted, resulting

in an increase in CD8+ T cells (Kay 1981; Herrmann et al., 1982). However, CD4+

T cells are increased in LN and BM of CLL patients (Caligaris-Cappio and

Hamblin, 1999), suggesting that they are preferentially recruited to these sites.

Researchers have also observed frequent clonal CD4+ and CD8+ T cell

expansion in CLL patients reflecting potential selection by leukemic cells (Farace

et al., 1994; Serrano et al., 1997). CLL cells induce gene expression changes in

CD4+ and CD8+ T cells particularly in those that affect cytoskeleton

polymerization and immune response (Gorgun et al., 2005; Gorgun et al., 2009;

Kiaii et al., 2013). Other changes in T cells from CLL patients include the

expression of markers of anergy or exhaustion and cytokine/chemokine

programs that support CLL survival (Brusa et al., 2013; Riches et al., 2013)

(Gorgun et al., 2005; Gorgun et al., 2009; Brusa et al., 2013; Riches et al., 2013;

Kiaii et al., 2013). Functionally, these T cells are impaired in immunological

synapse formation. Interestingly, these changes are not permanent and can be

reversed by treatment with the immunomodulatory drug, lenalidomide, which

suggests an active interaction between leukemic cells and T cells (Gilling et al.,

2012; Ramsay et al., 2008; Gorgun et al., 2009). CLL cells produce CCL22 to

attract CD4+ T cells to PCs where ligation of CD40 on CLL cells and CD40L on T

cells enhances the survival of CLL cells. In fact, the CD40-CD40L interaction is

often exploited to prolong CLL cultures by propagating them on a layer of

CD40L-transfected fibroblasts (Kater et al., 2004; Ghia et al., 2002). In an

Page 66: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

47

investigation by Vogler et al. (2009), CLL cells cultured on a layer of CD40L-

expressing fibroblasts in the presence of IL-4 induced a 1000-fold resistance to a

Bcl2 family antagonist, suggesting that these interactions may contribute to the

chemoresistance often encountered in LNs.

Regulatory T cells (Tregs) are increased in CLL. In several studies

examining the Tregs subset, defined as either CD4+CD25+ or CD4+CD25+ with

Foxp3, or with low or absent CD127, absolute number of Tregs was found to be

increased compared to healthy controls and as a fraction of T cells in most cases

(D’Arena et al., 2012). Higher Treg numbers are associated with advanced

disease and are predictive of time-to-treatment in patients with early-stage CLL

(Weiss et al., 2011). These observations hint that immunosuppression by Tregs

contributes to CLL pathogenesis. However, CLL cells themselves produce

immunosuppressive cytokines, IL-10 and TGFβ. These cytokines skew immunity

from a Th1 to a Th2 profile. Furthermore, T cells from CLL patients express more

CTLA4, an inhibitor of TCR activation, than those in healthy individuals (Ramsay

et al., 2008; DiLillo et al., 2013; Motta et al., 2005). Collectively, this paints a

picture in which T cells in CLL have been subverted to favor tolerance while

neglecting immunity. The presence or role of other cells with anti-tumor activity

(natural killer (NK) and NK T cells (NKT) in pseudofollicles has not been

established. However, NK cell defects in killing and lytic granule production are

well known in CLL (Kay et al., 1984). On the other hand, NKT cells from PB of

CLL patients appear functionally competent for cytotoxicity, and CLL cells

Page 67: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

48

express CD1d and retain capacity to present CD1d-restricted antigens to NKT

cells (Guven et al., 2003; Fais et al., 2004).

1.4 ANIMAL MODELS OF CLL

1.4.1 Xenograft Mice

The difficulty in establishing long-term cultures of leukemic cells from CLL

patients has partly contributed to the prominence of CLL animal models (Table

1.2). One of the first approaches to generate CLL models was by xenogeneic

transplantation of primary CLL cells or cell lines into immunodeficient mice. Early

models were plagued by poor engraftment efficiency and retention of leukemic

cells at the inoculation site (Kobayashi et al., 1992; Hummel et al., 1996). The

failure of CLL cells to migrate to other lymphoid tissues may be the result of

compatibility issues with human cells in mice, but CLL cell-intrinsic factors cannot

be discounted because Shimoni et al. (1997) showed that engraftment ability

correlates with Rai tumor stage. CLL cell lines derived from more aggressive

disease (e.g. the JVM3 line derived from a CLL patient with prolymphocytic

transformation) engrafted with close to 100% efficiency in irradiated nude mice,

while a CLL line derived from a patient with less progressed disease, called

MEC2, failed to engraft (Loisel et al. (2005). Recently, Bertilaccio et al. (2009)

using Rag2-/-γc-/- immunodeficient mice, which lack NK cells (unlike earlier

models), found that the CLL cell line, MEC1, could be engrafted with 100%

Page 68: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

49

efficiency. Moreover, these cells infiltrated several organs after intravenous or

subcutaneous injection leading to an aggressive disease.

Although xenograft models are usually limited in application by the fact

that they typically rely on transplanting end stages of a disease, Kikushige et al.

(2011) exploited a xenogeneic model to show that oncogenic defects in CLL can

occur in HSCs. In this study, HSCs from CLL patients transferred to NOD/RAG1-/-

IL2rgnull mice gave rise to lymphoid cells including B cells that mature to

oligoclonal or MBL-like B cells. Nevertheless, due to the short generation time of

xenograft mice, they remain a vital tool in evaluating certain disease aspects

such as therapeutic strategies.

1.4.2 New Zealand Mice

New Zealand strains of mice are prone to autoimmunity and spontaneous

development of an age-dependent CLL-like lymphoproliferative disease (Stall et

al., 1988; Kono et al., 1994; Phillips et al., 1992; Raveche et al., 2007). CD5+ B

cells, the predominant subset in fetal and neonatal mice, decline days after birth

and remain relatively low except in the peritoneal and pleural cavities, where they

are abundant throughout life. Some mouse strains such as New Zealand black

(NZB) and New Zealand white (NZW) have a higher frequency of these cells.

For instance, CD5+ B cells make up 5-10% of splenic cells in NZB mice at 3

months of age compared to about 2% in other strains (Hayakawa et al., 1983;

Berland and Wortis, 2002). Interestingly, CD5+ B cells reappear in elderly mice,

and clonal populations capable of self-renewal can be detected in mice older

Page 69: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

50

than 15 months in most strains. However, clonal CD5+ B cell populations are

detectable much earlier (3-4 months) in the peritoneum, spleen, and other organs

in NZB, NZW, and are highest in NZB x NZW F1 mice (Stall et al., 1988). Older

NZB mice develop a CLL-like disease characterized by small mature hyperdiploid

CD5+IgM+B220lo B cells with unmutated Ig genes in the spleen, PB, peritoneal

cavity, and rarely in LN but not BM (Phillips et al., 1992; Mahboudi et al., 1992;

Raveche et al., 2007; Salerno et al., 2010). NZ mice have long been used to

study autoimmunity with NZW x NZW F1 hybrid mice being a model of systemic

lupus erythematosus (SLE). Dissection of such crosses found that MHC

haplotypes controlled whether the mice would develop SLE or CLL. Homozygous

H-2z/z mice had the highest frequency of CD5+ B cells and developed CLL while

H-2d/z heterozygotes were prone to SLE (Helyer and Howie, 1963; Okada et al.,

1991; Hamano et al., 1998). Two cytokines, IL-10 and IL-5, which can act as

growth factors for normal B1 cells (Gary-Gouy et al., 2002; Hitoshi et al., 1990),

are critical for CLL development in these mice. Loss of IL-10 in CD5+ B cells of

NZB leads to increased cell-cycle block and apoptosis in CD5+ B cells, and NZB

mice deficient in IL-10 fail to develop CLL. Interestingly, PerC B1 B cells were not

sensitive to loss of IL-10 and their frequency remained unaffected (McCarthy et

al., 2004; Czarneski et al., 2004). Expression of an IL-5 transgene in SLE-prone

NZB x NZW F1 mice, in contrast to normal B1 B cells, reduced production of

antibodies and development of SLE while favoring CLL progression (Wen et al.,

2004). Raveche et al. (2007), using NZB mice crossed to DBA/2 F1 mice in a

genome-wide linkage study to identify loci associated with CLL in these mice,

Page 70: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

51

Table 1.2 Murine models of CLL

Model Characteristic features Reference

NZB Spontaneous model; CD5+IgM

+B220

lo B cells in Sp and PerC Phillips et al., 1992;

at 4 mo; miR16-1 mutation; late-onset leukemia. Raveche et al., 2007

Eµ-TCL1 CD5+IgM

+B220

lo B cells first detected in the PerC at 2 mo; Bichi et al., 2002;

leukemia in 13-18 mo; frequent secondary malignancies. Zanesi et al., 2006

TRAF2DN/BCL2 CD5+IgM

hiB220

IntCD11b

+; splenomegaly, lymphadenopathy; Zapata et al., 2004

80% dead by 14 mo.

APRIL B1 neoplasm; CD5+IgM

loB220

lo, detectable in PerC at 4-5 mo; Planelles et al., 2004

pleural effusion, ascites.

PKCα-KR HPCs give rise to CD19hiCD5

+IgM

loCD23

+IgD

lo lymphocytes Nakagawa et al., 2006

in vitro and in Sp & LN of RAG1-/-

mice.

SV40 large T Ag Sporadic expression of SV40 large T antigen; ter Brugge et al., 2009

CD5+B220

+IgM

+CD43

+ in PB, other organs; leukemia by 10 mo.

miR15a/16-1-KO Low penetrance, late onset CD5+IgM

+B220

lo MBL, CLL/SLL in Klein et al., 2010

PerC, Sp, PB, BM, & LN; non-Hodgkin’s lymphoma.

miR29 Indolent leukemia; CD5+CD19

+IgM

+ B cells in Sp at 2 mo; Santanam et al., 2011

humoral defect.

Abbreviations: Perc, peritoneal cavity; Sp, spleen; LN, lymph node; PB, peripheral blood; BM;

bone marrow; HPCs, hematopoietic progenitor cells; mo, months.

Page 71: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

52

found previously undescribed risk loci including one with synteny to 13q14 in

humans. 13q14 deletion is the most common cytogenetic defect in human CLL

and implicates loss of miR15a and miR16-1, among other genes, in CLL

development (Matutes et al., 1996; Klein et al., 2010). These microRNA genes

are not deleted in mice but sequencing them revealed a point mutation in the 3'

flanking region of miR16-1 that corresponded with its downregulation in tissues

suggested a possible role in murine CLL as well. Exogenous introduction of

miR15-1 or miR16-1 into cells derived from NZB mice decreased cyclin D1

levels, and increased cell cycle G1 phase block and sensitivity to drug-induced

apoptosis indicating CLL disease in NZB mice is partly due to loss of cell cycle

regulation (Raveche et al., 2007; Salerno et al., 2009). Moreover, these mice

exhibit an inherent genomic instability linked to reduced fidelity in DNA repair

(Raveche et al., 1979; Reddy and Flalkow, 1980). The strong genetic component

in NZ mice is much like familial CLL. More importantly, this model is useful in

dissecting the relationship between CLL and autoimmunity.

1.4.3 Eμ-TCL1 Transgenic Mice

T cell leukemia/lymphoma 1 (TCL1) is a proto-oncogene that is commonly

overexpressed in many lymphoproliferative disorders, including CLL. In CLL,

TCL1 expression levels correlate with shorter time-to-treatment and aggressive

disease suggesting a possible role in pathogenesis (Narducci, et al., 2000;

Herling et al., 2006; Aggarwal et al., 2008). TCL1 was first identified as the gene

commonly involved in chromosomal translocations and inversions in mature T

Page 72: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

53

cell leukemias, namely, T cell prolymphocytic leukemia and T cell chronic

lympocytic leukemia (Virgilio et al., 1994). TCL1 is normally expressed early in

lymphocyte development and diminishes in later stages such as in mature T

cells, post-GC B cells, and plasma cells. TCL1 is required for lymphocyte

development sincedeficient mice show loss of both T and B cell numbers and

impaired function (Narducci, et al., 2000; Kang et al., 2005; Herling et al., 2007).

Virgilio et al. (1998) tested the oncogenic potential of TCL1 in mice by placing it

under the control of Lck proximal promoter for targeted overexpression in the T

cell compartment. TCL1 overexpression induced expansion of T cells in young

mice with CD8+ mature T cell leukemias developing in older mice. However,

TCL1 appears to be more tumorigenic in murine B cells since overexpression of

TCL1 in both B and T compartments predominantly generates a variety of mature

B cell lymphomas (Hoyer et al., 2002). The mechanisms of TCL1-mediated

transformation in B cells are still unfolding. Under normal conditions, TCL1 acts

as a co-activator of Akt by binding to its pleckstrin homology domain and

augmenting phosphorytion of Akt and its nuclear translocation to induce growth,

survival, and proliferation (Pekarsky, et al., 2000; Teitell et al., 2005). However,

several other functions of TCL1 have been noted in neoplastic cells such as

transcriptional regulation and inhibition of DNA methylation (Pekarsky, et al.,

2008; Motiwala et al., 2011; Gaudio et al., 2011; Palamarchuk et al., 2011).

Bichi et al. (2002), in a quest to elucidate the role of TCL1 specifically in B

cell malignancies, discovered that targeted overexpression of TCL1 in the B

lineage causes a CLL-like disease in elderly mice. These mice carry the human

Page 73: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

54

TCL1 transgene under the control of the VH promoter and IgH μ enhancer (Eμ-

TCL1 mice) for targeted expression in immature and mature B cells. Eμ-TCL1

are characterized by progressive accumulation of G0/G1-arrested

CD5+IgM+B220lo B cells first observed in the PerC at 2 months, in the spleen by

3-5 months, and in the bone marrow by 5-8 months. By 13-18 months, these

mice develop a CLL-like malignancy indicated by hepatosplenomagaly, high

white blood cell (WBC) counts (180x106 cells/mL versus 2.8x106 cells/mL in wild

type mice), and lymphadenopathy. Additionally, Zanesi et al. (2006) observed

that over a third of these mice develop secondary malignancies, a common

feature in human CLL as well, and these may be attributed to genetics or failed

tumor surveillance in untreated mice. Sequencing Ig genes from leukemic B cells

shows that they are mainly unmutated, a characteristic seen in aggressive CLL in

humans, and bear specificities akin to B1 cells directed toward lipid and

polysaccharide motifs common in self and microbial antigens (Yan et al., 2006).

Other features of human CLL demonstrated in these mice include the ability to

induce T cell dysfunction. Eμ-TCL1 leukemic cells induce clonal expansion of T

cells with defective immunological synapse formation and altered gene

expression. These T cell changes are temporary and can be reversed by

lenalidomide therapy (Gorgun et al., 2009; Hofbauer et al., 2011). Because of the

many similarities to aggressive human CLL, Eμ-TCL1 mice have become a

widely accepted pre-clinical tool (Zanesi et al, 2006; Johnson et al., 2006;

Suljagic et al., 2010), as well as a model for dissecting CLL biology (Enzler et al.,

Page 74: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

55

2009; Wu et al., 2009; Chen et al., 2009; Bertilaccio et al., 2011; Chen et al.,

2011).

1.4.4 APRIL Transgenic Mice

A proliferation-inducing ligand (APRIL) and B cell activating factor (BAFF)

are two cytokines of the TNF family that mediate survival and differentiation in

normal B cells and resistance to apoptosis in CLL cells (Kern et al., 2003). BAFF

is a critical survival factor for maturating B cells. Loss of BAFF or its receptors

interferes with B cell maturation and overexpression of BAFF is associated with

loss of B cell tolerance and development of autoimmunity. BAFF receptors

include the BAFF receptor, B cell maturation antigen (BMCA), and

transmembrane activator and calcium modulator cyclophilin ligand-interactor

(TACI), of which the latter two are also share with APRIL (Rickert et al., 2011).

Expression of APRIL can stimulate growth of tumor cells, and CLL patients have

increased serum APRIL levels that also correlates with survival implicating it in

the pathogenesis of CLL (Hahne et al., 1998; Planelles et al., 2007). APRIL

transgenic mice were developed by Stein et al., 2002) to investigate the role of

APRIL in lymphocytes. These mice carry a transgene for human APRIL driven by

Lck distal promoter for expression in T lymphocytes. Young APRIL transgenic

mice have no gross abnormalities but show increased T cell proliferation and

survival capacity, and display enhanced thymus-independent responses and

increased serum IgM (Stein et al., 2002). Importantly, older mice develop a B1 B

cell neoplasia that is similar to CLL (Planelles et al., 2004). These B1 cells are

Page 75: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

56

detectable in the Perc at 4-5 months and show an age-dependent accumulation.

By 9-12 months ~40% of mice develop tumors in mesenteric lymph nodes and

Peyer’s patches. Further analyses show these cells are CD5+IgMloB220loCD23-

and portray enhanced survival in culture. Leukemic cells progressively infiltrate

other organs including the spleen, leading to splenomegaly, and can be detected

in non-lymphoid tissues such as kidney and liver. These data show that APRIL

can promote development of CLL-like disease in mice. NLCs have been found to

be a source of BAFF and APRIL through which they enhance survival of CLL in

microenvironments (Nishio et al., 2005).

1.4.5 TRAF2DN/BCL2 Transgenic Mice

Bcl2 is an anti-apoptotic protein commonly overexpressed in several

cancers including follicular B cell lymphoma and CLL (Hockenberry et al., 1990;

Hanada et al., 1993). Transgenic overexpression of Bcl2 in mice enhances

lymphocyte survival and fosters development of a low-grade polyclonal

lymphoproliferative disease with lymphadenopathy and splenomegaly

(Kastumata et al., 1992). TNF receptor-associated factor 2 is a member of TNF

receptor-associated factors that link TNF family receptors to intracellular

signaling to regulate cell proliferation and survival in normal cells and in

pathological conditions including cancer, when deregulated (Arch et al., 1998;

Zapata et al., 2000). Mice engineered to overexpress a dominant-negative

TRAF2 mutant protein (TRAF2DN) develop a lymphoproliferative disease that is

characterized by lymphadenopathy and splenomegaly with associated polyclonal

Page 76: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

57

B cell expansion (Lee et al., 1997). This TRAF2 mutant lacks part of the N

terminus that contains the RING and zinc finger domains, and therefore mimics

TRAF1, which naturally lacks these domains (Bradley and Pober, 2001). Since

TRAF1 is overexpressed in CLL B cells (Zapata et al., 2000; Munzert et al.,

2002), Zapata and colleagues (2004) tested whether combined overexpression

of Bcl2 and TRAF2DN can promote development of CLL. Double transgenic mice

developed lymphadenopathy, splenomegaly, high WBC counts with common

occurrence of pleural effusions and ascites, and more than 80% of the mice died

by 14 months. FACS analysis and histopathological examination identified a

CLL/SLL-like disease that was defined by expansion of small- to medium-sized

non-cycling CD5+IgMhiB220IntIgDloCD11b+CD21loCD23- lymphocytes in the

spleen, BM, LN, PB and lungs. Lymphocytes from double transgenic mice

demonstrated increased survival in cultures and resistance to fludarabine or

dexamethasone-induced apoptosis suggesting that these lymphocytes

accumulate primarily due to enhanced survival, particularly since they exhibit

lower proliferation compared to those of wild type cells. Recent follow-up work by

the same group shows the TRAF2DN transgene is expressed at low levels and

induces degradation and loss of endogenous TRAF2 (Perez-Chacon et al.,

2012). Therefore, these works demonstrate that loss of TRAF2 and concomitant

overexpression of Bcl2 promotes a CLL/SLL-like disease in mice.

Page 77: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

58

1.4.6 PKCα-KR Graft Mice

Protein kinase C (PKC) encompasses multiple isoforms of ubiquitous

serine/threonine protein kinases that regulate cellular proliferation, differentiation,

and survival in normal and malignant conditions (Spitaler and Cantrell, 2004;

Michie and Nakagawa, 2005). PKCs are activated by Ca2+ and diacylglycerol

(which is mimicked by phorbol esters) upon antigen receptor stimulation (Spitaler

and Cantrell, 2004). Recent evidence suggests that CD5 can also activate PKC

in a subset of CLL patients increasing cell survival (Perez-Chacon et al., 2007).

Moreover, a specific inhibitor of PKCδ induces apoptosis of human CLL cells and

genetic deletion of PKCβ or specific inhibition thwarts development of murine

CLL (Ringshausen et al., 2006; Holler et al., 2009). Nakagawa et al. (2006)

tested the potential of PKCα and PKCδ isoforms to transform B cells by stably

expressing their dominant-negative forms (PKCα-KR and PKCδ-KR,

respectively) in murine fetal liver-derived hematopoietic progenitor cells (HPCs),

which were co-cultured with a BM stromal cell line together with growth factors.

PKCα-KR induced more proliferation of the cultured cells and was further

investigated. Flow cytometric analysis showed that greater than 95% of cells

carrying PKCα-KR expressed B cell markers, CD19 and B220, as well as CLL-

related markers, CD5 and CD23 suggesting that subversion of PKCα signaling

promotes the development of a CLL-like phenotype (Nakagawa et al., 2006).

Notably, the authors did not check whether CD23 was co-expressed by the CD5+

B cells. CD5+ B cells showed prolonged survival in in vitro cultures lacking

stroma and growth factors with concomitant expression of Bcl2. HPCs stably

Page 78: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

59

expressing PKCα-KR were transplanted into RAG1-/- mice to determine their fate

in vivo. Analyses of spleen and LNs detected cells with the same

immunophenotype as those found in vitro (CD19hiCD5+CD23+IgMloIgDlo).

Infiltration in other organs was not reported but non-CLL tumors were found to

form at injection site. Cells derived from a CLL tumor were mostly in the G0/G1

phase of cell cycle and appeared vulnerable to apoptosis, but resumed

proliferation when cultured on a stromal layer with growth factors. Collectively,

these studies demonstrated that altered PKCα function interferes with cell

survival and proliferation and promotes the outgrowth of CLL-like cells in mouse

tissues.

1.4.7 SV40 Large T Antigen Transgenic Mice

Simian virus 40 (SV40) large T antigen is a potent oncogene that

transforms cells by variety of mechanisms, most commonly, by inhibiting tumor

suppressors such as p53 and Rb (Ahuja et al., 2005). The finding of SV40 large

T antigen sequences human hematopoietic and lymphoid malignancies are

incriminating (Shivapurka et al., 2002; Shivapurkar et al., 2004). Ter Brugge et al.

(2009) investigated the effect of SV40 large T antigen in B lymphocytes with a

targeted SV40 large T antigen transgene that is expressed sporadically. This

transgene, under the control of IgHµ enhancer but without a promoter, was

inserted in the IgH locus between D and J gene segments and in the opposite

transcriptional orientation. This SV40 large T antigen, therefore, is predicted to

be expressed by antisense transcription and under two scenarios: early in

Page 79: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

60

development before D-J recombination, and later as long as the targeted allele is

unrearranged. Insertion of the transgene does not appear to hinder

recombination because allotypic markers do not show biased usage toward a

particular allele in young mice. However, analyses of older mice show an age-

dependent progressive increase of large atypical B cells with biased expression

of the non-targeted allele in PB leading to development of CLL-like leukemia by

10 months, which can be accelerated by loss of p53. These B cells were

monoclonal and CD5+B220+IgM+CD43+ and present in the LN, spleen, and BM.

Half of the sequenced tumors (4/8) used the B1 B cell-associated VH11 family

heavy chain and had no mutations while the rest used VHJ558 family and

showed either few or multiple mutations. SV40 large T antigen has not been

detected in CLL, therefore, it is unlikely to be involved in transformation in this

disease. Since SV40 large T antigen targets multiple pathways (Ahuja et al.,

2005) this intriguing model may be useful in identifying signaling mechanisms

that promote CLL development.

1.4.8 DLEU2/miR15a/16-1 Knockout Mice

In CLL, 13q14 deletion occurs in over half of cases, and is the most

common chromosomal aberration in MBL and CLL (Dohner et al., 2000;

Rawstron et al., 2008). This deletion is also common in several lymphoid and

non-hematological solid malignancies suggesting that loss of genes in this region

predisposes an individual to cancer. A 300 kilobase minimally deleted region

(MDR) has been identified (Lui et al., 1997; Migliazza et al., 2001). The MDR

Page 80: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

61

encodes for DLEU2, a sterile transcript from a class of long non-coding RNAs

that regulate gene expression, and microRNAs commonly downregulated in CLL

and prostate cancer, miR-15a and miR-16-1 (Calin et al., 2002; Bonci et al.,

2008). To test whether the MDR acts as a tumor suppressor, Klein et al. (2010)

generated mice with MDR deletion, or miR-15a/16-1 deletion only. Young MDR-/-

and miR-15a/16-1-/- appeared normal but older mice (> 9 months) showed

expansion of CD5+IgM+B220lo B cells than began in the PerC. Eventually, 42% of

MDR-/- and 26% of miR-15a/16-1-/- mice develop a spectrum of indolent

lymphoproliferative disorders that range from MBL, CLL/SLL, and non-Hodgkins

lymphomas. Although the role of DLEU2 in development of lymphoproliferative

disorders is unknown, miR-15a and miR-16-1 were found to regulate cell cycle

progression by targeting cyclins and cyclin-dependent kinases. Therefore, loss of

miR-15a and miR-16-1 leads to unchecked cell proliferation. Interestingly,

sequence analysis revealed that CD5+ B cells in the lymphoproliferative disorders

carried unmutated BCRs that shared ≥80% HCDR3 amino acid sequence

homology among different mice (stereotyped) suggesting a role for antigens in

pathogenesis akin to humans.

1.4.9 miR-29 Transgenic Mice

MicroRNA are modulators of gene expression, therefore, they are

implicated in a myriad of conditions when they are deregulated (Ambros et al.,

2004; Garzon et al., 2009). Using microRNA expression profiling, Pekarsky et al.

(2006) found miR-29 regulates TCL1 expression and was downregulated in

Page 81: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

62

aggressive compared indolent CLL. This group later found that when compared

to normal cord blood CD19+ B cells, miR-29 was significantly upregulated in

indolent CLL suggesting it could be involved in pathogenesis of these cases of

CLL (Santanam et al., 2010). To test this possibility, they generated mice that

express miR-29 in immature and mature B cells. The majority of miR-29

transgenic mice show expansion of CLL-like cells detectable as early as 2

months in the spleen. These cells were modestly proliferating clonal

CD5+CD19+IgM+ lymphocytes that can be found later in both PB and BM, and in

the liver and kidney in advanced disease. These cells expand slowly but

gradually suggesting an indolent disease. For instance, CD5+ B cells constitute

~20% of all B cells in mice younger than 15 months, ~40% by 15-20 months, and

over 65% by 20-26 months with 4/20 transgenic mice followed to 24-26 months

developing overt leukemia, which was also the cause of death. miR-29

transgenic mice B cells additionally resemble those of human CLL in

immunological incompetence marked by reduction in serum IgG and poor

response to immunization.

Santanam et al. (2010) contend that the mechanism of leukemogenesis

may involve loss of a p53 responsive gene known as peroxidasin (a miR-29

target that was validated in these cells), which is downregulated in acute myeloid

leukemia (AML) (Santanam et al., 2010). Interestingly, transplantation of HSCs

overexpressing miR-29 in mice leads to development of a myeloproliferative

disorder that resembles acute myeloid leukemia (AML) (Han et al., 2010).

Santanam et al. (2010) concluded that miR29 is involved in the pathogenesis of

Page 82: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

63

indolent CLL. This implied that the reduction of miR-29 (which also targets TCL1)

observed in aggressive CLL leads to upregulation of one of its targets, TCL1,

causing an aggressive disease.

1.5 RESEARCH GOALS AND SPECIFIC AIMS

CLL is a heterogeneous adult leukemia that is characterized by

progressive accumulation of small lymphocytes that express CD19, CD5, and

CD23 the blood and lymphoid organs (Matutes and Polliack, 2000; Dighiero and

Hamblin, 2008). The origins of CLL remain unclear but emerging evidence

suggests that CLL likely evolves from a more benign biologically similar condition

known as monoclonal B cell lymphocytosis (Rawstron et al., 2002; Rawstron et

al., 2008; Lanasa et al., 2011; Kern et al., 2012). However, what initiates this

condition or how it evolves to CLL is unknown. Accumulating evidence suggest

that that the feedback between CLL cells and their environment through various

receptors, including those that bind antigens partly contributes to CLL

pathogenesis.

CLL exhibits a restricted Ig gene repertoire and displays an antibody

reactivity profile shared by innate-like B1 and marginal zone MZ B, as well as

subsets of transitional and extrafollicular B cells that exhibit self-reactivity

produced by primary Ig gene rearrangements, or as an unintended consequence

of somatic mutation, respectively. CLL cells are also marked by CD5, which is

constitutively expressed on a subset of normal B1 cells and induced in B cells

undergoing receptor editing/revision, or those rendered anergic by chronic

Page 83: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

64

(auto)antigenic stimulation, where it may function to negatively regulate B cell

receptor signaling and suppress B cell activation to limit autoantibody production

(Hippen et al., 2000; Hillion et al., 2005; Sims et al., 2005; Youinou et al., 2006).

In principle, B cells undergoing receptor editing/revision may be blocked

from completing this process if a dominant negative form of RAG1 is expressed

in sufficient excess over endogenous RAG1. We recently generated dominant-

negative RAG1 (dnRAG1) mice expressing a catalytically inactive, but DNA

binding-competent, form of RAG1 in the periphery that show evidence of

impaired secondary V(D)J recombination that occurs in response to self-reactivity

(Hassaballa et al., 2011). Interestingly, these animals develop a progressive,

antigen-dependent, accumulation of CD5+ B cells (Figure 1.3) which are clonally

diverse, yet repertoire-restricted, and possess a splenic B1-like

immunophenotype. However, dnRAG1 mice do not develop CD5+ B cell

neoplasia. The indolent accumulation of CD5+ B cells in dnRAG1 mice is

reminiscent of MBL, but the lack of progression to CLL in this model suggests

additional factors are required to promote transformation. TCL1 was considered

a plausible factor because it is often overexpressed in CLL and leads to a CLL-

like disease in aging Eμ-TCL1 mice (Herling et al., 2006; Bichi et al., 2002).

Predicting that molecular defects which promote benign CD5+ B cell

accumulation, such as impaired receptor editing, are a rate-limiting step in CLL

progression, we hypothesized that dnRAG1 expression in Eµ-TCL1 would

accelerate CD5+ B cell accumulation and CLL onset compared to Eµ-TCL1 mice.

Page 84: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

65

Figure 1.3 Accumulation of B1-like CD5+ B cells with age in dnRAG1 mice. (A) Flow

cytometric analysis of splenic lymphocytes at 12 weeks in WT and dnRAG1 mice.

Numbers indicate the percentage all of cells represented in a given a gate. (B) The

percentage of splenic CD19+B220lo B cells was evaluated in WT and dnRAG1 mice as a

function of age from fetal day 17 (17dF) to 48 weeks. Differences are significant from 4

weeks of age onward (P<0.001).

Page 85: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

66

We proposed two specific aims to test this hypothesis:

1. Determine whether dnRAG1 expression in TCL1 transgenic mice

accelerates CLL development. dnRAG1 mice were bred to Eµ-TCL1 to

generate WT, dnRAG1, Eµ-TCL1, and dnRAG1/Eµ-TCL1 double transgenic

(DTG) mice, which were monitored for accumulation of CD5+ B cell and CLL

characteristics by flow cytometry. Eµ-TCL1 and DTG mice were monitored for

development of CLL and survival.

2. Identify candidate factors involved in CD5+ B cell accumulation in

transgenic mice. CD5+ B cells accumulate in both dnRAG1 and Eµ-TCL1 mice,

but they likely arise through distinct mechanisms. To identify factors that regulate

CD5+ B cell development in these animals, and determine the extent to which the

individual transgenes contribute to CD5+ B accumulation in DTG mice, we

proposed to compare patterns of gene expression in CD5+ B cells from dnRAG1,

Eµ-TCL1, and DTG mice to each other and to normal follicular B cells.

The origins of CLL remain unknown. However, the large parallels between

human and murine B cell development allow us to study its pathogenesis in

these models. Identification of factors that regulate malignant CD5+ B

accumulation could lead to potential biomarkers or therapeutic targets for this

incurable leukemia.

Page 86: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

67

CHAPTER 2: METHODS

2.1 MICE

Eµ-TCL1 mice (Bichi et al., 2002) were kindly provided by C. M. Croce

and Y. Pekarsky (Ohio State University, Columbus, Ohio). Eµ-TCL1 mice and

dnRAG1 mice (Hassaballa et al., 2011), both on C57BL/6 backgrounds, were

crossed to generate cohorts of wild type, dnRAG1, Eµ-TCL1, and DTG mice.

Genotyping was performed by polymerase chain reaction (PCR) as described

(Bichi et al., 2002; Hassaballa et al., 2011). Cohorts were euthanized by cervical

dislocation either at predetermined ages (6, 12, 24, and 36 weeks), or when they

became moribund. SCID mice (B6.CB17-prkdcscid/SzJ) were purchased from the

Jackson Laboratory (Bar Harbor, ME). All mice were housed in individually

ventilated microisolator cages in an Association for Assessment and

Accreditation of Laboratory Animal Care certified animal facility in accordance

with university and federal guidelines, and mouse protocols were approved by

the Creighton University Institutional Animal Care and Use Committee. Survival

data was obtained by monitoring a cohort of 11 Eµ-TCL1 and 14 DTG mice until

the animals showed signs of illness, which included hunched posture, labored

breathing, lethargy and reduced grooming, necessitating euthanasia.

2.2 FLOW CYTOMETRY

Lymphoid tissues were collected from ill mice or at predetermined time

points for fluorescence-activated cell sorting (FACS) analysis. Single-cell

suspensions were prepared from spleen, lymph node, and thymus in RF10

Page 87: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

68

medium [RPMI 1640 (Invitrogen, Carlsbad, CA) containing 10% (v/v) fetal bovine

serum (FBS, Invitrogen) by shredding with two needles and homogenizing with

syringe. Blood was collected in heparinized tubes either from tail veins of live

mice or by cardiac puncture after euthanasia. BM cells were collected by flushing

marrow from femurs and tibias with RF10 medium into a collection tube. Red

blood cells were depleted from all tissues by hypotonic lysis with ammonium

chloride (0.14M ammonium chloride, 17mM Tris-HCl, pH 7.2). The white blood

cells were centrifuged, resuspended in phosphate-buffered saline (PBS)

containing 4% (v/v) FBS (PBS4) and counted on a particle counter (Coulter

Counter Model Z1, Beckman Coulter, Fullerton, CA). 1x106 cells were stained on

ice for 30 minutes with the following fluorochrome-conjugated monoclonal

antibodies: BD Biosciences (San Jose, CA) anti-B220-PE-TXRD (RA3-6B2), anti-

CD19-APC-Cy7 (ID3), anti-CD5-biotin (53-7.3), anti-CD21/CD35-PE (7G6), anti-

CD11b-PE (M1/70), anti-CD23-Biotin (B3B4), anti-CD4-APC-Cy7 (GK1.5), and

anti-CD25-PE-Cy7 (PC61), and eBioscience (San Diego, CA) anti-CD5-PE (53-

7.3), anti-IgM-APC (II/41), anti-IgD-FITC (11-26c), anti-CD3-APC (145-2C11),

anti-CD8-A700 (53-6.7), anti-TCRβ-FITC (H57-597). Samples containing

biotinylated antibodies were further developed with streptavidin-Qdot585

(Invitrogen). Data collection and cell sorting was performed using a FACSAria

flow cytometer (BD Biosciences). Flow cytometric data was analyzed using the

FlowJo software (Tree Star, Inc. Ashland, OR).

Page 88: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

69

2.3 ADOPTIVE TRANSFER

CD19+CD5+ spleen lymphocytes alone or together with CD3+TCRβ+CD4-

CD8- or CD3+TCRβ+CD8+ spleen lymphocytes sorted from Eµ-TCL1 or DTG

mice by FACS were adoptively transferred into SCID mice. Sorted cells were

washed twice and resuspended in PBS. 2x104 to 1x106 sorted lymphocyte

populations (as indicated for specific experiments in the figures) in 200 µL of PBS

were injected into tail veins of 4- to 9-week-old recipient SCID mice. Control

SCID mice received PBS only. Grafting was monitored by FACS on blood

collected from tail veins every few weeks. The mice were euthanized at

experimental endpoint and their lymphoid organs were analyzed by FACS.

2.4 α-GALACTOSYLCERAMIDE TREATMENT

α-GalCer (KRN7000) (Avanti Polar Lipids, Alabaster, AL) stock was

prepared by resuspension at 1mg/mL in DMSO (Sigma, St. Louis, MO). 12-week-

old WT and DTG mice were treated with 5 µg of α-GalCer in 100µL of PBS with

0.025% Tween 20 or 100 µL of PBS with 0.025% Tween 20 by tail vein injection

once every week for 4 weeks. Mice were euthanized one week following the last

dose and their lymphoid organs were harvested and the abundance and

distribution of lymphocyte subsets was analyzed by FACS.

2.5 CELL CULTURE

CD19+CD5+ spleen lymphocytes sorted from DTG mice by FACS were

cultured with or without CD3+TCRβ+CD4-CD8- spleen lymphocytes from the

same mouse. Sorted cells were washed before resuspension with lymphocyte

Page 89: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

70

culture medium consisting of RPMI supplemented with 10% FBS, 1x10-5 M β-

mercaptoethanol, 2mM L-glutamine, and 100 μg/mL penicillin and streptomycin

(Takahashi and Strober, 2008). The cells were cultured in 96-well round-bottom

plates at 37oC in a humidified incubator with 5% CO2. Cells were counted on day

3, 7, and 10 in a hemacytometer and viability was determined by trypan blue

exclusion.

2.6 HISTOPATHOLOGY AND WBC COUNTS

Tissues were fixed in 10% buffered formalin, embedded in paraffin,

sectioned, and stained with hematoxylin and eosin. Blood smears were prepared

and stained with Wright-Giemsa. Tissue sections or blood smears of at least

three mice for each genotype were evaluated by light microscopy for changes in

architecture and lymphocyte infiltration. Spleen, BM, liver, and kidney sections,

and blood smears were evaluated with a Nikon i80 microscope (Nikon, Melville,

NY) with a DigiFire camera running ImageSys Digital Imaging System (Soft

Imaging Systems GmBH, Munster, Germany) or a Nikon Eclipse E400

(Nikon) with an Optronics camera running MagnaFire software (Optronics,

Goleta, CA). High magnification BM sections and blood smear images were

acquired with a Nikon i80 microscope and Nikon Digital Sight DS-F1 camera

running the NIS-Elements Imaging software version 2.33. White blood cell (WBC)

counts were performed automatically with a Cellometer Auto X4 cell counter

(Nexcelom Bioscience (Lawrence, MA).

Page 90: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

71

2.7 CLONALITY

Clonal V(D)J rearrangements were investigated by Southern blot and PCR

using spleen genomic DNA isolated with Wizard Genomic DNA Purification Kit

(Promega, Madison, WI). For Southern blot, 10-15 µg was digested overnight

with EcoR1 (New England Biolabs, Ipswich, MA), separated on a 0.8% agarose

gel, transferred to Amersham Hybond-N+ nylon membrane (GE Healthcare,

Piscataway, NJ), and hybridized with a 32P-labeled JH probe described by Bichi et

al., 2002). Phosphor images were acquired using a Typhoon 9410 variable mode

imager (GE Healthcare). To detect rearrangements by PCR, spleen genomic

DNA was amplified using primer sets for IgH (VHJ558 sense and 3′ JH4

antisense), Igκ (Vκ sense and 3’ Jκ5 antisense), and CD14 input control as

(Hassaballa et al., 2011). 100 ng of DNA was used in a 50 µL PCR reaction

containing 10 pmol of each primer, 0.2mM dNTPs, 20 mM Tris-HCl, pH 8.4, 50

mM KCl, 1.5 mM MgCl2, and 1.25 units of Taq polymerase. The DNA was

subjected to 30 cycles of amplification with 94oC for 30 sec denaturation, 58oC

for 1 min annealing, and 72oC for 2 min extension with a final extension of 10 min

at 72oC. 15 µL of PCR reactions was fractionated in a 1.5% agarose gel

containing ethidium bromide and imaged under ultra violet light.

2.8 IMMUNOGLOBULIN GENE SEQUENCING AND ANALYSIS

Total RNA was isolated from frozen spleen cells from ill Eµ-TCL1 and DTG mice

using Ambion RiboPure Kit (Ambion, Austin, TX). 5x106 spleen cells per 1 mL of

TRI Reagent were homogenized in 1.5 mL microcentrifuge tubes. After a 5 min

Page 91: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

72

incubation at room temperature, 100 µL of bromochloropropane was added and

mixed by vortexing at high speed for 15 sec and incubated at room temperature

for 5 min. Samples were centrifuged at 12,000 x g for 15 min at 4oC to allow

phase separation. The aqueous phase was transferred to a fresh 1.5 mL

microcentrifuge tubes to which 200 µL of 100% ethanol was added. Samples

were immediately mixed by a short vortex (5 sec) and transferred to a glass-fiber

filtration tube. The RNA was washed twice and eluted in 50 µL of elution buffer.

Complementary DNA (cDNA) was prepared from 1 μg of the purified RNA in a 20

μL reaction using Novagen First Strand cDNA Synthesis Kit (EMD Millipore,

Darmstadt, Germany) with 0.5 µg of oligo(dT) primers at 37oC for 1 hour. Ig

genes were amplified from 1-2 μL of the cDNA by PCR using Platinum Taq DNA

Polymerase High Fidelity (Invitrogen) in a 35-cycle PCR reaction with 94oC for 1

min denaturation, 55oC for 1 min annealing, and 68oC for 1 min extension with a

final extension of 10 min at 68oC using MuIgVH5′- A to -F forward and

MuIgMVH3′-1 reverse primers to amplify heavy chain sequences (Mouse Ig-

Primer Sets, Novagen), and mouse universal 5′ Mκ forward and 3′ κC reverse

primers to amplify light chain sequences. 30 µL of the PCR reactions was

fractionated in a1.5% agarose gel and the DNA was isolated using GeneJET Gel

extraction Kit (Fermentas, Thermo Scientific, Waltham, MA). 2 µL of the purified

PCR products was ligated into the PCR 2.1 TOPO TA plasmid vector

(Invitrogen), which was used to transform development and diminishes in later

stages such as in mature T cells, post-GC B cells, and plasma cells. TCL1 is

required for lymphocyte development since Top10 chemically-competent E. coli

Page 92: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

73

cells (Invitrogen). Isolated colonies were grown in mini-cultures overnight and

plasmid DNA was recovered using the E.Z.N.A Plasmid Mini Kit I (Omega Bio-

Tek, Norcross, GA). For both heavy and light chain genes, at least 10

independent clones per mouse were sequenced using a commercial vendor

(ACGT, Inc., Wheeling, IL). Sequences, with primer sites omitted, were analyzed

using IMGT/V-QUEST tool (http://www.imgt.org) to identify Ig gene usage,

mutations, CDR3 composition, and isoelectric point.

2.9 IMMUNOGLOBULIN LEVELS

Serum was prepared from blood collected from 12- and 36-week-old mice.

The blood was collected in tubes without anti-coagulants and allowed to clot for

30 minutes then centrifuged at 2000 x g for 10 minutes to obtain the serum.

Serum Igs were measured by ELISA using IMMUNO-TEK mouse IgM and IgG

kits (ZeptoMetrix, Buffalo, NY). Serum was diluted between 1:15,000 to

1:100,000 using the diluent (PBS, Triton X-100, 2-chloroacetamide) and added to

the ELISA plate in duplicate wells of 100 µL volume each. The plate was

incubated at 37oC for 30 min to allow the mouse antibodies to bind, after which

unbound material was removed through four washes with PBS containing Tween

20 and 2-chloroacetamide. 100 µL of anti-mouse IgG or IgM antibodies that are

conjugated to horseradish peroxidase were added to the plate, which was

incubated at 37oC for 30 min. After washing the plate, 100 µL of the substrate,

tetramethyl benzidine, was added and incubated at room temperature for 30 min.

The colorimetric reaction was quenched and optical density was measured at

Page 93: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

74

450 nM with a VersaMax microplate reader (Molecular Devices, Sunnyvale, CA).

Serum IgM or IgG concentration was determined based on the optical densities

of IgM or IgG standards, respectively run alongside the test serum.

2.10 MICROARRAY

Total RNA was isolated from sorted splenic CD19+B220hiD5- B cells

obtained from wild type mice and splenic CD19+CD5+ B cells obtained from

transgenic mice using the Ribopure kit (Ambion) as described above. Biotin-end

labeled cDNA was prepared from 100-200 ng of total RNA using whole transcript

labeling kits from either Affymetrix (Affymetrix, Santa Clara, CA) or Ambion.

Resultant cDNA was hybridized overnight to Mouse Gene 1.0 ST Arrays and

washed, stained, and scanned using the Affymetrix GeneChip system with a

3000 7G Affymetrix scanner at the University Nebraska Medical Center

Microarray Core Facility. All procedures were conducted following Affymetrix

suggested protocols. Arrays were normalized using the Robust Multichip

Average algorithm (Irizarry et al., 2003; Irizarry et al., 2006) included in the

Affymetrix Expression Console software. The data is deposited in NCBI’s Gene

Expression Omnibus under series accession number GSE44940.

Unsupervised clustering and principal component analyses of genes

showing the highest variation among arrays were performed on Log2-transformed

expression values using dChip software (Li & Wong, 2001). These genes were

determined using the filtering criteria of a standard deviation for logged data

threshold between 0.85 and 1000 (12-week mice arrays) or 1.40 and 1000 (older

Page 94: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

75

mice arrays) and an expression level greater than 5.65 in 25% of the samples.

Supervised clustering was performed on genes involved in regulating BCR

signaling (Kurosaki, 1999; Xu et al., 2005; Stevenson et al., 2011). Pair-wise

comparisons of the differentially expressed genes between genotypes was also

performed on dChip with linear expression values using the lower 90%

confidence bound interval for genes with ±1.2 fold change filtering criterion (12-

week mice) or ±1.5 fold change (older mice). Genes with a mean expression

value of less than 50 in both genotypes in a comparison pair were considered

absent and therefore not included.

To compare transgenic mice CD19+CD5+ B cells with normal B cell

subsets, data sets from 12-week-old mice and those obtained from splenic

transitional, MZ, B1a, mature follicular, and GC, and peritoneal B1a B cells

subsets available within the NCBI GEO series GSE15907 through the

Immunological Genome Project Consortium (ImmGen) (Heng et al., 2008) were

normalized together using the RMA algorithm in the Affymetrix Expression

Console. Batch effects were adjusted using ComBat (Johnson et al., 2007)

before clustering and PCA analysis by dChip. Unsupervised clustering was

performed on 102 genes which showed the highest variation that were selected

by the filtering criterion of a standard deviation for logged data threshold between

1.10 and 1000, and an expression level greater than or equal to 5.65 in 25% of

samples. Principal component analysis was also performed using the filtered

gene set.

Page 95: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

76

2.11 REAL-TIME QUANTITATIVE PCR

RNA was isolated from frozen spleen tissue using Ambion Ribopure Kit

(Ambion) as described in the previous sections. RNA was treated with DNAse1

(Sigma) to degrade any DNA carried over by incubating all the purified RNA (~50

µL) with 6 µL of DNAse1 for 15 min with 6 µL of the DNAse1 reaction buffer (20

mM Tris-HCl, pH 8.3, and 2 mM MgCl2). The reaction was terminated by adding

6 µL of 50 mM EDTA and heating at 70oC for 10 min. 1 µg of RNA was used to

generate cDNA in a 50 µL reaction using TaqMan Reverse Transcription

Reagents (Applied Biosystems, Foster City, CA) These reactions contained the

1X reaction buffer (50 mM KCl and 10 mM Tris-HCl, pH 8.3), 5.5 mM MgCl2, 500

µM dNTP mix, 2. 5 µM Random hexamer primers, 0.4 U RNase inhibitor, and

1.25 U reverse transcriptase. Reverse transcription was carried out under the

the following thermal cycling conditions: 25oC (10 min), 48oC (30 min), 95oC (5

min). Control samples were prepared similary but with the omission of reverse

transcriptase. 2 µL of the cDNA reactions was amplified in a 20 µL SYBR Green-

based real-time quantitative PCR reaction (Applied Biosystems) on an ABI 7500

Fast instrument (Applied Biosystems) using the primer sets for Prl2a1, Il10,

Sox4, Rgs13, Sell, and Actb (Table 2.1) under the following thermal cycling

parameters: 50oC for 2 min followed by 35 cycles of 95oC for15 sec and 60oC for

1 min. Relative gene expression levels were calculated using the comparative

threshold cycle method (Livak and Schmittgen, 2001), with expression

normalized to β-actin.

Page 96: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

77

2.12 STATISTICS

Statistics were performed using PASW software version 19 (SPSS, Inc.,

Chicago, IL) Differences among groups were determined by one-way ANOVA. A

p value equal to or less than 0.05 was considered significant. Data is presented

in bar graphs as mean ± the standard error. Survival was measured by Kaplan-

Meier method and the curves were compared using the log-rank test.

Page 97: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

78

Table 2.1 Primers for RT-PCR

Gene Forward primer sequence (5′ - ′3) Reverse primer sequence (5′ - ′3)

Prl2a1 GGAAAAGAGCAATGGACTCCTGG CAGTCTCTGACTTCAAGGATGCC

Il10 CGGGAAGACAATAACTGCACCC CGGTTAGCAGTATGTTGTCCAGC

Sox4 GCCTCCATCTTCGTACAACC AGTGAAGCGCGTCTACCTGT

Rgs13 CTACATCCAGCCACAGTCTCCT TGAGCTTCTTCAAAGCATGTTTGAG

Sell ATGGTGAGCATCCCAGCCTA CCCCTTCCAGCATTCCATCA

Actb AGAGGGAAATCGTGCGTGAC CAATAGTGACCTGGCCGT

Page 98: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

79

CHAPTER 3: RESULTS

3.1 dnRAG1 EXPRESSION ACCELERATES LEUKEMOGENESIS IN Eµ-

TCL1 MICE

3.1.1 Expression of dnRAG1 in Eμ-TCL1 Mice Shortens Survival

To test the hypothesis that dnRAG1 transgene expression accelerates

disease progression in Eµ-TCL1 mice, we crossed Eµ-TCL1 and dnRAG1

transgenic mice and performed survival analysis on a cohort of Eµ-TCL1 (n=11)

and DTG (n=14) mice. Consistent with our hypothesis, DTG mice showed a

significantly shorter lifespan compared to Eµ-TCL1 mice (p= 0.004), with median

survival times of 9.1 and 11.3 months, respectively (Figure 3.1A). At endpoint, all

DTG mice were visibly ill, showing lethargy and a hunched posture with labored

breathing, and evidence of organomegaly that necessitated euthanasia.

Splenomegaly was confirmed on necropsy in and a mixed population of small

and large lymphocytes was evident in peripheral blood smears in all mice (Figure

3.1B). About a quarter of Eµ-TCL1 or DTG mice developed lymphadenopathy

and/or hepatomegaly. Two DTG mice additionally presented with pleural

effusions and/or ascites. All ill Eµ-TCL1 and DTG mice showed an abundance of

CLL-like CD19+CD5+ B cells in the spleen (Figure 3.2A) that displayed an

IgM+IgD+B220+CD11blo/- phenotype, although the relative expression of these

antigens varied slightly between individuals (Figure 3.2B).

Page 99: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

80

Figure 3.1 dnRAG1 expression in Eµ-TCL1 mice enhances lymphoproliferation and

shortens lifespan. (A) Kaplan-Meier survival curves of Eµ-TCL1 (n=11) and DTG

(n=14) mice. Statistical analysis between groups was performed using the log-rank test

(median survival: 9.1 and 11.3 months for DTG and Eµ-TCL1 mice; p=0.004). (B)

Peripheral blood smears of ill Eµ-TCL1 and DTG mice at end point were stained with

Wright-Giemsa and imaged at 1000x magnification using Nikon i80 microscope with

Nikon Digital Sight DS-F1 camera running the NIS-Elements Imaging software version

2.33.

Page 100: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

81

Figure 3.2 dnRAG1 expression in Eµ-TCL1 mice accelerates accumulation of CLL-

like cells. Flow cytometry was used to analyze splenic lymphocytes from ill mice. (A)

The percentage of splenic CD19+CD5+ B cells among gated lymphocytes is shown at

endpoint for two representative Eµ-TCL1 and DTG mice. (B) Expression of sIgM, sIgD,

B220, and CD11b on CD19+CD5+ B cells from three ill Eμ-TCL1 mice (top row) and

three ill DTG mice (bottom row) is shown in (B). Histograms for the three different mice

are shown as solid, dashed, and dotted lines.

Page 101: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

82

3.1.2 Time Course Study of CLL-like Disease Progression in DTG Mice

Since survival analysis suggested that dnRAG1 expression in Eµ-TCL1

mice accelerates disease progression, we analyzed cohorts of wild-type, Eµ-

TCL1, dnRAG1, and DTG mice at 6, 12, 24, and 36 weeks of age to more

carefully compare CD5+ B cell accumulation rates using flow cytometry. The last

time point was chosen because it was close to the median survival of DTG mice.

At 36 weeks, CD19+CD5+ (or alternatively CD19+B220lo) B cells represented a

small fraction of total splenic B cells in WT mice, but were abundant in dnRAG1,

Eµ-TCL1, and DTG mice (Figure 3.3A). Further analysis of the CD19+B220lo

population from dnRAG1, Eµ-TCL1, and DTG mice showed uniform CD5+CD21-

CD23- staining and similar sIgM levels (Figure 3.3B); sIgD levels were

comparable between dnRAG1 and DTG mice, but slightly higher than in Eµ-

TCL1 mice (Figure 3.3B).

As expected, DTG mice showed more rapid CD5+ B cell accumulation

compared to either dnRAG1 or Eµ-TCL1 mice in all tissues tested (lymph nodes,

bone marrow, peripheral blood, and spleen) (Figure 3.4A). However, the

frequency of CD5+ B cells differed substantially between the various tissues

analyzed. For example, at 36 weeks of age, CD5+ B cells comprised on average

5% of lymphocytes in lymph nodes of DTG mice, compared to 1.1%, 2.4%, and

1.5% for dnRAG1, Eµ-TCL1, and WT mice, respectively. In spleen, by contrast,

CD5+ B cells represented on average 67% of lymphocytes in DTG mice,

compared to 33%, 22%, and 2.7% for dnRAG1 Eµ-TCL1, and WT mice,

Page 102: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

83

Figure 3.3 Surface immunophenotype of CLL-like cells in DTG mice. Flow

cytometric analysis of splenic lymphocytes from 36-week-old WT, dnRAG1, Eµ-TCL1,

and DTG mice showing the expression of CD19 and either CD5 (top row) or B220

(bottom row). The percentage of CD19+CD5+, CD19+CD5-, CD19+B220hi, CD19+B220lo

cells is shown for representative animals. Flow cytometry was used to compare the

expression of CD5, sIgM, sIgD, CD21, CD23, and CTLA4 expression on splenic WT

CD19+B220hi B cells to splenic CD19+B220lo B cells from dnRAG1, Eμ-TCL1 mice and

DTG mice.

Page 103: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

84

Figure 3.4 Characterization of CLL-like disease progression in DTG mice. (A) The

percentage of CD19+CD5+ cells in lymph node (LN), bone marrow (BM), peripheral blood

(PB), and spleen (SPL) of 6, 12, 24, and 36 week-old WT, dnRAG1, Eµ-TCL1, and DTG

mice (n=5-14) animals per genotype per time point) was determined using flow

cytometry as in Figure 3.3A and presented in bar graph format. Error bars represent the

standard error of the mean. Statistically significant differences (p<0.05) between values

obtained for DTG mice relative to WT (a), dnRAG1 (b), or Eµ-TCL1 (c) mice are

indicated. (B) Spleens from representative animals at 36 weeks are shown at left, and

the mean weights of 5-14 animals of each genotype at each time point are presented in

bar graph format at right. Spleen cellularities were used to determine the absolute

number of CD19+CD5+ cells for each genotype at 12, 24, and 36 weeks of age (C).

White blood cell (WBC) counts (D) were compared for WT, dnRAG1, Eμ-TCL1 mice and

DTG mice at 36 weeks of age. Error bars in all graphs represent the standard error of

the mean. *, p<0.05; ***, p< 0.001.

Page 104: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

85

respectively. A similar pattern was observed in the values obtained for peripheral

blood and bone marrow, which were intermediate between those determined in

lymph node and spleen (Figure 3.4A). By 36 weeks, all DTG mice showed

massive splenomegaly, with average spleen sizes ranging from 3-4-fold higher

than dnRAG1 and Eµ-TCL1 mice, to over 10-fold greater than WT mice (Figure

3.4B). At all time points tested, DTG mice showed a higher absolute number of

splenic CD5+ B cells compared to the other genotypes (Figure 3.4C).

All 36-week-old DTG mice showed evidence of lymphocytosis by white

blood cells counts and histopathological evaluation of blood smears (Figures

3.4D and 3.5); smudge cells similar to those observed in CLL patients were also

frequently detected (Figure 3.5). In the spleen, extensive lymphocytic infiltrates

were observed in DTG mice, with a corresponding loss of splenic architecture

(Figure 3.5). Abnormal lymphocytic infiltrates were clearly apparent in the livers

of all 36 week-old DTG mice, but bone marrow and kidneys were unremarkable

(Figures 3.5 and 3.6).

These data show that CD5+ B cells in dnRAG1, Eµ-TCL1, DTG mice are

phenotypically similar in terms of surface marker expression and organ

involvement; however, they emerge and accumulate with different kinetics in all

transgenic mice. These cells emerge earliest and accumulate more dramatically

in DTG mice compared to either dnRAG1 or Eµ-TCL1 mice. While they are

detectable earlier in dnRAG1 compared to Eµ-TCL1 mice, they accumulate at a

slower rate since dnRAG1 mice rarely develop frank leukemia.

Page 105: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

86

Figure 3.5 DTG mice show evidence of CLL-like disease at 36 weeks. (A) Blood

smears were developed with Wright-Giemsa stain. Images (200x and 1000x) were

acquired using Nikon Eclipse E400 microscope (Nikon, Melville, NY) and

Optronics camera running MagnaFire software (Optronics, Goleta, CA), and a Nikon i80

microscope and Nikon Digital Sight DS-F1 camera running the NIS-Elements Imaging

software version 2.33, respectively. Paraffin-embedded spleen and kidney sections

were developed with hematoxylin and eosin. Images (100x for spleen and 200x for liver)

were acquired using a Nikon i80 microscope and DigiFire camera running ImageSys

digital imaging software (Soft Imaging Systems GmBH, Munster, Germany). DTG mice

at this age show peripheral blood lymphocytosis with frequent occurrence of smudge

cells (arrow, see inset), loss of splenic architecture, and infiltration of leukemic cells in

liver (arrows). Scale bars: blood and liver 100 µM; spleen, 200 µM.

Page 106: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

87

Figure 3.6 Bone marrow and kidney histology of 36-week-old mice. Paraffin-

embedded bone marrow and kidney sections were developed with hematoxylin and

eosin. Images in columns 1 and 3 (100x for bone marrow, and 200x for kidney) were

acquired using a Nikon i80 microscope and DigiFire camera running ImageSys digital

imaging software (Soft Imaging Systems GmBH, Munster, Germany). Bone marrow

images in column 2 (1000x) were acquired using a Nikon i80 microscope and Nikon

Digital Sight DS-F1 camera running the NIS-Elements Imaging software version 2.33.

Bone marrow and kidney show little or no abnormal infiltration at this time point. Scale

bars: bone marrow, 200 μM; kidney, 100 μM.

Page 107: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

88

3.1.3 B Cells of DTG Mice are Clonally Transformed and Bear Unmutated

Ig Genes

The CLL-like B cells emerging in Eµ-TCL1 mice are known to be

leukemogenic (Zanesi et al., 2006). To test whether CLL-like cells in DTG mice

are also leukemogenic, 106 splenic CD19+CD5+ B cells from two different 36-

week-old DTG mice were injected i.v. into SCID recipient mice. In both cases,

successful engraftment was indicated by flow cytometry six weeks after adoptive

transfer (Figure 3.7A). By three months, all SCID mice receiving CD5+ B cells

from DTG mice developed splenomegaly (Figure

3.7B), peripheral blood lymphocytosis (Figure 3.7C), and infiltration of spleen,

bone marrow, and lymph nodes by leukemic cells (Figure 3.7D), which indicates

that these cells are malignantly transformed.

We previously showed that the CD5+ B cells accumulating in dnRAG1

mice are clonally diverse, but repertoire restricted (Hassaballa et al., 2011). To

determine whether the accumulating CD5+ B cells in DTG mice are also clonally

diverse, or have instead undergone oligoclonal or monoclonal expansion

indicative of transformation, we analyzed genomic DNA prepared from spleens of

36 week-old WT, dnRAG1, Eµ-TCL1, and DTG mice by Southern hybridization to

detect clonal rearrangements in the IgH locus (Figure 3.8A). This analysis was

also performed on DTG donor animals and SCID recipient mice at experimental

endpoint (3 months). We found that about half of the dnRAG1 (1/2), Eµ-TCL1

(2/4), and DTG (4/7) mice analyzed showed the presence of non-germline bands,

indicative of clonal expansion (Figure 3.7A, lanes 2-14). Notably, the pattern of

Page 108: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

89

Figure 3.7 Engraftment of leukemic cells from DTG mice in SCID mice. CD19+CD5+

splenocytes (106 cells) were purified by FACS from two different 36 week-old DTG mice

and injected into the tail veins of two recipient SCID mice for each donor; n=4 total.

Normal C57BL/6 mice and sham-injected SCID mice were included as controls.

CD19+CD5+ B cells were readily detected in SCID recipients but not control animals at 6

weeks post-transfer (A). The SCID mice were sacrificed 3 months post-transfer. Spleen

weights were measured (B), and the percentage (C) and/or absolute number (D) of

CD19+IgM+ B cells were determined in spleen, bone marrow (BM), lymph node (LN), and

peripheral blood (PB).

Page 109: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

90

Figure 3.8 Clonal transformation of B cells from DTG mice. (A) Genomic DNA

prepared from spleens of 36 week-old WT, dnRAG1, Eµ-TCL1, and DTG mice (lanes 2-

14), as well as DTG donor and SCID recipient mice in panel A (lanes 15-18), was

subjected to Southern hybridization using a JH-probe. Non-germline bands are indicated

by an asterisk. The JH probe contains a portion of the Eµ enhancer, and therefore also

detects the dnRAG1 transgene which contains this element (Hassaballa et al., 2011).

(B) VH-to-DJH and Vκ-to-Jµ rearrangements were amplified by PCR from genomic DNA

samples described in panel B. Rearrangements to a given J segment are shown at

right. The non-rearranging CD14 locus was amplified from the diluted genomic DNA

templates as a loading control.

Page 110: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

91

non-germline bands observed in samples prepared from donor DTG mice and

the corresponding SCID recipient mice were identical (Figure 3.8A, lanes 15-18),

suggesting the leukemic cells are stable and do not undergo further IgH gene

rearrangement after adoptive transfer.

Since some Eµ-TCL1 and DTG mice did not show obvious evidence of

clonal outgrowth by Southern blotting, we alternatively used a PCR-based

approach to compare the pattern of IgH and Igκ gene rearrangements between

the four genotypes of animals analyzed in Figure 3.8A. As expected from our

earlier studies (Hassaballa et al., 2011), WT and dnRAG1 mice showed a

clonally diverse B cell repertoire that includes rearrangements to all four

functional JH and Jκ gene segments, but dnRAG1 mice showed a bias toward

Jκ1 and Jκ2 gene usage consistent with a defect in secondary V(D)J

recombination (Figure 3.8B, lanes 3-6). By contrast, all Eµ-TCL1 and DTG mice

showed a highly restricted pattern of V(D)J rearrangements (Figure 3.7B,

compare lanes 3-6 to 7-15). Taken together, these data suggest that CD5+ B

cells accumulating in DTG mice represent a clonal expansion of leukemic B cells.

Previous studies have shown that leukemic cells in Eµ-TCL1 mice harbor

largely unmutated Ig genes that encode CDR3 sequences enriched in serine,

tyrosine, and charged residues, features which are also observed in aggressive

CLL in humans (Yan et al., 2006). To determine whether leukemic cells from

DTG mice share these characteristics, we analyzed IgH and Igκ sequences from

three ill Eµ-TCL1 and DTG mice (Tables 3.1 and 3.2). We found that, like Eµ-

TCL1 mice, Ig gene sequences in DTG mice are mostly unmutated and encode

Page 111: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

92

CDR3 regions enriched in serine, tyrosine, and charged residues. Indeed, some

of the same genes and CDR3 sequences are recovered from both genotypes.

For example, sequences utilizing the VH segments 1-55, 5-6, and 5-12 and/or

containing the VH CDR3 sequence encoding “YYGSS” were amplified from both

Eµ-TCL1 and DTG animals. We also detected recurrent Vκ4-91 and Vκ14-126

usage in Eµ-TCL1 and DTG mice. Several of these VH and Vκ sequences were

also represented among those amplified from Eµ-TCL1 mice by Yan et al.

(2006). Many of these gene sequences are associated with reactivity toward

autoantigens such as DNA, cardiolipin, and phosphatidyl choline (Tables 3.1 and

3.2).

Page 112: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

93

Page 113: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

94

Page 114: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

95

3.2 dnRAG1 EXPRESSION IN Eµ-TCL1 MICE SUPPRESSES

IMMUNOGLOBULIN PRODUCTION

Human CLL patients typically develop progressive

hypogammaglobulinemia (Rozman et al., 1988; Caligaris-Cappio and Hamblin,

1999). Our previous studies showed that dnRAG1 mice exhibit defective natural

IgM and IgG antibody production correlated with an intrinsic B cell

hyporesponsiveness toward antigenic stimulation (Hassaballa et al., 2011). To

determine whether Eµ-TCL1 expression influences antibody production in

dnRAG1 mice, we compared serum IgM and IgG levels in WT, dnRAG1, Eµ-

TCL1, and DTG mice at 12 and 36 weeks (Figure 3.9). Consistent with previous

findings (Hassaballa et al., 2011), IgM and IgG levels were reduced 2-3-fold in

dnRAG1 mice compared to WT mice at 12 weeks; at 36 weeks, this reduction

was even more pronounced (4-8 fold). By contrast, IgM and IgG levels in Eµ-

TCL1 and WT mice were quite similar at 12 weeks, but at 36 weeks, Eµ-TCL1

mice showed elevated IgM levels and decreased IgG levels compared to WT

mice. Interestingly, antibody levels in DTG mice resembled dnRAG1 mice, but

showed slightly lower serum IgM and IgG levels at both time points. Thus, these

data suggest that the suppressive effect of dnRAG1 expression on antibody

production in mice is not overcome by concomitant Eµ-TCL1 expression.

Page 115: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

96

Figure 3.9 DTG mice show evidence of severe hypogammaglobulinemia. Serum

IgM and IgG levels were measured by ELISA for groups of 5-7 12- and 36-week-old WT,

dnRAG1, Eµ-TCL1, or DTG mice. Individual values and means (indicated by horizontal

bar) are shown for each group; statistically significant differences between groups are

also depicted.

Page 116: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

97

3.3 DISTINCT GENE EXPRESSION PROFILES IN CD5+ B CELLS IN BOTH

YOUNG AND OLD TRANSGENIC MICE

To gain insight into the relative contribution of the individual dnRAG1 and

Eµ-TCL1 transgenes in promoting CLL-like disease in DTG mice, and identify

genes that are differentially expressed between the animals as function of age

and disease progression, we compared the gene expression profiles of splenic

CD19+CD5+ B cells from 12-week-old dnRAG1, Eµ-TCL1, and DTG mice and

splenic CD19+B220hiCD5- B cells from two age-matched WT mice, and repeated

this analysis using ill DTG mice, which was evidenced by lethargy accompanied

with organomegally, and their age-matched counterparts.

3.3.1 Gene Expression Profiles in 12-week-old Mice

To determine how closely the B cell populations from WT and transgenic

mice resemble each another, we initially performed unsupervised hierarchical

clustering analysis on expression data sets from sorted B cell populations from

12-week-old mice (Figure 3.10A). Perhaps not surprisingly, CD19+CD5+ B cell

gene expression profiles from transgenic mice were more similar to each other

than to the profiles obtained for WT CD19+B220hiCD5- B cell, which was also true

in older mice (Figure 3.12A). These experiments also revealed that at 12 weeks

of age, CD5+ B cells from DTG mice resembled those from dnRAG1 mice more

closely than Eµ-TCL1 mice (Figure 3.10A). Indeed, only two genes (Neto2 and

Lmo7) were differentially expressed by more than 3-fold between dnRAG1 and

DTG mice (Table 3.3). Additional comparison to gene expression profiles

Page 117: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

98

Page 118: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

99

Figure 3.10 Identification of differentially expressed genes in WT CD19+B220hiCD5-

B cells and CD19+CD5+ B cells from dnRAG1, Eµ-TCL1, and DTG mice at 12 weeks.

(A) Unsupervised hierarchical clustering of a set of 107 showing the most variation. The

Red and green intensities indicate high to low expression values, respectively. The

dendrogram shows the relationships between the arrays. (B) A list of common genes

among the 50 genes that display the greatest differential expression in WT

CD19+B220hiCD5- B cells relative to CD19+CD5+ B cells from all transgenic mice is

shown in the top panel. A list of common genes among the 50 genes that display the

greatest differential expression in CD19+CD5+ B cells from dnRAG1 and DTG mice

relative to Eµ-TCL1 mice is also shown in the bottom panel.

Page 119: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

100

WT (M188)

dnRAG1 (M175)

dnRAG1 (M194)DTG (M176)

DTG (M187)

DTG (M199)

Eµ-TCL1 (M167)Eµ-TCL1 (M173)

WT (M183)

MZ_Sp (1)MZ_Sp (2)

MZ_Sp (3)

T1_Sp (1)

T1-Sp (2)T1_Sp (3)

T2_Sp (1)T2_Sp (2)

T2_Sp (3)

T3_Sp (1)

T3_Sp (2)T3_Sp (3)

B1a_PC (1)

B1a_PC (2)B1a_PC (3)

B1a_Sp (1)B1a_Sp (2)

B1a_Sp (3)

FO_Sp (1)FO_Sp (2)

FO_Sp (3)

GC_Sp (1)

GC_Sp (2)

GC_Sp (3)

MZ

_S

pM

Z_

Sp

MZ

_S

pT

1_

Sp

T1

_S

pT

1_

Sp

Fo

_S

pF

o_

Sp

Fo

_S

pW

TW

TT

2_

Sp

T2

_S

pT

2_

Sp

T3

_S

pT

3_

Sp

T3

_S

pG

C_

Sp

GC

_S

pG

C_

Sp

dn

RA

G1

Eu

-TC

L1

Eu

-TC

L1

B1

a_

PC

B1

a_

PC

B1

a_

PC

B1

a_

Sp

B1

a_

Sp

B1

a_

Sp

DT

GD

TG

dn

RA

G1

DT

G

Cd1d1Cd36S1pr3MyofCeacam2I830077J02RikLOC674190Gm4759BC094916E030037K03RikZfp318Slfn8Cnn3Serpinb1aSellDpp4S1pr1If i203Tlr7S100a10A630033H20RikCd55Stat4Ms4a4cCcr6Gpr174Rasgrp3Fcer2aCr2Satb1Kmo1300014I06RikLrrk2Bcl6CpmCd93Serinc5Rgs13Rassf6Hist1h2bbCenpeFasAicdaHist1h1bMki67C330027C09Rik2810417H13RikCcnb1F630043A04RikGatmPrc1Cep55Kif11Ccna2E2f8Kif2cStilNcapg2Nuf2Plk1Bub1bNcapgTpx2Gcet2Gm600IgjNt5eAnxa2Nek2Racgap1LifrFxyd5Plac8Fcrl5Zc3h12cGm10879Lmo7Prkar2bIl6stNid1Ctla4S100a6Zbtb32Cd80Rbm47AdmFzd61810046K07RikPstpip2Ptpn22Tmem154Pde8aCd9Atxn1Gm10786Cd300lfCcr1Csf2rbDnase1l3Bhlhe41Hepacam2Il5ra

MZ

_S

p_#2

MZ

_S

p_#1

MZ

_S

p_#3

T1_S

p_#1

T1_S

p_#2

T1_S

p_#3

Fo_S

p_#1

Fo_S

p_#2

Fo_S

p_#31

WT

(M

188)

WT

(M

183)

T2_S

p_#1

T2_S

p_#2

T2_S

p_#3

T3_S

p_#1

T3_S

p_#2

T3_S

p_#3

GC

_S

p_#1

GC

_S

p_#2

GC

_S

p_#3

dnR

AG

1 (

M175)

Eu-T

CL1 (

M173)

Eu-T

CL1 (

M167)

B1a_P

C_#3

B1a_P

C_#1

B1a_P

C_#2

B1a_S

p_#2

B1a_S

p_#1

B1a_S

p_#3

DT

G (

M176)

DT

G (

M199)

dnR

AG

1 (

M194)

DT

G (

M187)

Cd1d1Cd36S1pr3MyofCeacam2I830077J02RikLOC674190Gm4759BC094916E030037K03RikZfp318Slfn8Cnn3Serpinb1aSellDpp4S1pr1Ifi203Tlr7S100a10A630033H20RikCd55Stat4Ms4a4cCcr6Gpr174Rasgrp3Fcer2aCr2Satb1Kmo1300014I06RikLrrk2Bcl6CpmCd93Serinc5Rgs13Rassf6Hist1h2bbCenpeFasAicdaHist1h1bMki67C330027C09Rik2810417H13RikCcnb1F630043A04RikGatmPrc1Cep55Kif11Ccna2E2f8Kif2cStilNcapg2Nuf2Plk1Bub1bNcapgTpx2Gcet2Gm600IgjNt5eAnxa2Nek2Racgap1LifrFxyd5Plac8Fcrl5Zc3h12cGm10879Lmo7Prkar2bIl6stNid1Ctla4S100a6Zbtb32Cd80Rbm47AdmFzd61810046K07RikPstpip2Ptpn22Tmem154Pde8aCd9Atxn1Gm10786Cd300lfCcr1Csf2rbDnase1l3Bhlhe41Hepacam2Il5ra

-3.0 -2.8 -2.5 -2.3 -2.1 -1.8 -1.6 -1.4 -1.2 -0.9 -0.7 -0.5 -0.2 0 0.2 0.5 0.7 0.9 1.2 1.4 1.6 1.8 2.1 2.3 2.5 2.8 3.0

A B

Figure 3.11 CD5+ B cells from dnRAG1, Eμ-TCL1, and DTG mice are most similar

to normal B1a B cells. (A) The gene expression profiles of sorted B cells from 12 week-

old mice in this study were compared to those from normal developing, mature, and

activated B cell subsets by unsupervised hierarchical clustering analysis. Clustering was

performed on batch effect-corrected log2 expression values for 102 genes (rows) and 33

arrays (columns), which includes those from sorted splenic wild type CD19+B220hiCD5-

(WT) and transgenic CD19+B220loCD5+ (dnRAG1, Eμ-TCL1, and DTG) B cells from 12

week-old mice reported here, and those obtained from sorted splenic and peritoneal B1a

cells (B1a-Sp and B1a-PC), splenic transitional B cells (T1-T3-Sp), splenic marginal

zone (MZ-Sp) and follicular B cells (FO-Sp), and splenic germinal center (GC-Sp) B cells

available through the ImmGen database (Heng et al., 2008). Red and green intensities

indicate high to low gene expression, respectively. The dendrogram shows the

relationships between the populations. (B) Principal component analysis (PCA) of the

102 genes identified in (A).

Page 120: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

101

available for normal B1 cells and splenic developing, mature, and activated B cell

subsets using hierarchical clustering and principal component analyses (PCA)

showed that the CD5+ B cell gene expression profiles from the transgenic mice

studied here most closely resembled the signature for normal splenic B1a cells

by both analyses, but also shared some features of MZ B cells by PCA (Figure

3.11).

To identify specific genes that are differentially expressed between the

sorted B cell populations in the 12-week-old group, we performed a series of

pair-wise comparisons between the microarray data sets. First, we compiled the

50 genes showing the greatest differential expression for each pair-wise

comparison (25 up and 25 down), and generated a list of common genes that are

differentially expressed between transgenic CD5+ B cells and WT B cells in all

three comparisons (Figure 3.10B). Among the genes included on this list are

cytotoxic T-lymphocyte-associated protein 4 (Ctla4; 8.4-16.9 fold increase),

complement receptor 2 (Cr2 [CD21]; 4.5- 5.4 fold decrease) and Fcer2a (CD23;

12-25 fold decrease). These differences were validated at the protein level by

flow cytometry (Figure 3.3B). Several other genes also displayed a consistent

pattern of differential expression, including Pstpip2 (+11-14 fold), Cpm (-11-12

fold), Sell (-4-14 fold), and Lrrk2 (-18-24 fold).

Using the same approach, we next compared the gene expression profiles

from dnRAG1 and DTG mice to those obtained from Eµ-TCL1 mice (Figure

3.10B). In this comparison, most genes showed less than a 10-fold difference in

relative expression. Among the 17 common upregulated genes, two prolactin

Page 121: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

102

family members, Prl2a1 and Prl7c1, showed increased expression in CD5+ B

cells from dnRAG1 mice (57 and 3.6 fold, respectively) and DTG mice (53 and

4.4 fold, respectively) relative to Eµ-TCL1 mice. In addition, the gene identified

as LOC100047053 showed elevated expression in CD5+ B cells from both

dnRAG1 (12.07 fold) and DTG (7.01 fold) mice relative to Eµ-TCL1 mice. This

gene is synonymous with Igκv6-32 (Igκv19-32) and was previously shown to be

highly represented in the light chain repertoire of CD19+B220lo B cells in dnRAG1

mice (Hassaballa et al., 2011), which likely explains its apparent upregulation in

CD5+ B cells from dnRAG1 and DTG mice relative to Eµ-TCL1 mice. None of

the 17 downregulated genes on this list showed >10-fold reduction in both pair-

wise comparisons.

3.3.2 Gene Expression Profiles in Ill DTG Mice and Age-matched

Counterparts

Unsupervised hierarchical clustering analysis of array sets of sorted B cell

populations from ill DTG mice (37 & 43 weeks) and their age-matched

transgenic and WT counterparts shows that CD5+ B cells from transgenic mice

are very similar to each other compared to the WT controls (Figure 3.12A).

However, the gene expression profiles obtained from DTG mice showed less

similarity to those from dnRAG1 mice than at 12 weeks, and instead clustered

more closely with the Eµ-TCL1 profile (Figure 3.12A). To identify specific

differentially expressed genes, we repeated pair-wise comparisons as described

for 12-week-old arrays focusing on differences between the transgenic CD5+

Page 122: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

103

Page 123: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

104

Figure 3.12 Identification of differentially expressed genes in WT CD19+B220hiCD5-

B cells and CD19+CD5+ B cells from ill DTG mice and age-matched dnRAG1 and

Eµ-TCL1 mice. (A) Unsupervised hierarchical clustering of a set of 102 genes showing

the most variation. The Red and green intensities indicate high to low expression values,

respectively. The dendrogram shows the relationships between the arrays. (B) A list of

common genes among the 50 genes that display the greatest differential expression in

CD19+CD5+ B cells from dnRAG1 and DTG mice relative to Eµ-TCL1 mice is shown in

the upper panel, and a list of 24 genes that display the greatest differential expression

(>2-fold) in CD19+CD5+ B cells DTG mice compared to both dnRAG1 and Eµ-TCL1 mice

is shown in the bottom panel.

Page 124: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

105

populations, although comparisons to WT CD19+B220hiCD5- B cells were also

made (Table 3.4). First, we compared gene expression data obtained for CD5+ B

cells from ill DTG and age-matched dnRAG1 mice to an older Eµ-TCL1 mouse

(Figure 3.12B). As observed at 12 weeks, Prl2a1 was highly upregulated in

CD5+ B cells from older dnRAG1 and ill DTG mice relative to the Eµ-TCL1

mouse (277 and 235-fold, respectively). Three additional genes, Sox4, Gpr177,

and Prl8a2, were also upregulated more than 10-fold in both dnRAG1 and DTG

CD5+ B cells in this comparison. Quantitative PCR (qPCR) analysis of Prl2a1

and Sox4 expression in whole spleen tissue obtained from 36-week-old WT and

transgenic mice confirmed that both genes were overexpressed in dnRAG1 and

DTG mice relative to both WT and Eµ-TCL1 mice (Figure 3.13). By contrast, 12

genes were downregulated by more than 10-fold among older dnRAG1 and DTG

mice relative to Eµ-TCL1 mice (Figure 3.12B); only one non-Ig-related gene,

Lgals1, appeared on the list of downregulated genes at both time points. We also

performed individual pair-wise comparisons between the CD5+ B cell gene

expression data from the older DTG and Eµ-TCL1 mice (Tables 3.5 and 3.6).

This exercise unexpectedly revealed that CD5+ B cells from one of the DTG mice

highly upregulated CD23 expression (32-fold) relative to the Eµ-TCL1 mouse

(Table 3.6). This observation was confirmed by flow cytometry (Figure 3.14), and

raises the possibility that CD23 expression may be induced in CD5+ B cells in

some cases when DTG mice become ill.

Page 125: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

106

Figure 3.13 Validation of differentially expressed genes by quantitative PCR. The

relative expression of Prl2a1, Il10, Sox4, Rgs13, and Sell in whole spleen tissue of 36-

week-old mice (n=3/ genotype) was measured by real-time quantitative PCR. Data is

presented as mean fold change relative to WT mice; error bars represent the standard

error of the mean. Significant differences between genotypes are indicated (*, p<0.05;

**, p< 0.01; ***, p< 0.001).

Page 126: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

107

Figure 3.14 CD23 expression on leukemic cells from an ill DTG mouse. Flow

cytometry was used to compare CD23 expression on CD19+B220lo B cells from the Eμ-

TCL1 mouse (M52) and the ill DTG mouse (F57) used for the comparative gene

expression analysis found in Table 3.6, which showed that Fcer2a (CD23) was

unregulated ~32-fold on CD19+CD5+ B cells from the DTG mouse relative to those from

the Eμ-TCL1 mouse.

Page 127: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

108

Finally, we compared the gene expression data from ill DTG mice to age-

matched dnRAG1 and Eµ-TCL1 mice to identify genes that were up- or down-

regulated >2-fold in both pair-wise comparisons which distinguish DTG mice from

both dnRAG1 and Eµ-TCL1 mice (Figure 3.12B). The expression of several of

the identified genes in spleen tissue obtained from 36-week-old WT and

transgenic mice was analyzed by qPCR. These studies confirmed that Rgs13

and Sell expression was significantly different between DTG mice and one or

both of the single-transgenic mice (Figure 3.13). The expression of other genes

tested, including Dpp4, Ccl22, and Plaur, showed no significant differences

between the transgenic animals.

3.3.3 Tolerogenic and BCR Signaling Genes are Perturbed in Transgenic

Mice

We evaluated the expression of genes involved in BCR signaling

(Kurosaki, 1999; Xu et al., 2005; Stevenson et al., 2011) and immune

suppression (Alegre et al., 2001; Goodnow et al., 2005; Moore et al., 2001),

some of which are reportedly differentially expressed in CLL. Supervised

hierarchical clustering analysis of BCR signaling genes showed that these genes

clearly differentiate WT CD19+B220hiCD5- B cells from transgenic CD19+CD5+ B

cells at both time points (Figure 3.15). One marker that obviously discriminates

CLL cells from normal adult follicular B cells is CD5. Although Cd5 was not

among the top 50 differentially expressed genes in any of the pair-wise

comparisons earlier, its expression was nevertheless consistently higher among

Page 128: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

109

Figure 3.15 Hierarchical clustering analysis of genes involved in B cell receptor

signaling from 12-week-old and older WT and transgenic mice. Supervised

hierarchical clustering analysis was performed using a set of 22 genes known to play a

role in B cell receptor signaling pathways. The relative expression of these genes in WT

CD19+B220hiCD5- B cells compared to transgenic CD19+CD5+ B cells is shown at right

for 12-week-old (top panel) and older animals (bottom panel).

Page 129: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

110

transgenic CD5+ B cells compared to WT CD19+B220hiCD5- B cells at 12 weeks

(~1.9-fold), and increased slightly in older mice (2.2-3.3-fold). Several other

genes involved in modulating BCR signaling known to be dysregulated on CLL

cells, including Nfatc1 (Le Roy et al., 2012), and Ptpn22 (Negro et al., 2012),

were also found to be dysregulated on all transgenic CD19+CD5+ B cells

compared to WT CD19+B220hiCD5- B cells. CD22, a negative regulator of BCR

signaling that is also underexpressed in CLL (Damle et al., 2002; Jelinek et al.,

2003), was downregulated at least 2.6-fold in CD5+ B cells at both time points,

except in older Eµ-TCL1 mice where the difference was more modest (-1.2 fold).

Some genes comprising a “tolerogenic signature” for CLL (Gilling et al., 2012;

DiLillo et al., 2012) were also upregulated on transgenic CD19+CD5+ B cells at

both time points and were increased in older mice, including Ctla4 (8.5-22 fold)

and Il10 (4.9-14 fold) (Figure 3.11 and (Table 3.4). Increased expression of both

genes has been confirmed either at the protein level on CD5+ B cells (Ctla4;

Figure 3.3), or at the transcript level in whole spleen tissue from transgenic mice

(Il10; Figure 3.13).

3.4 ALTERATION OF THE T CELL COMPARTMENT IN TRANSGENIC

MICE

3.4.1 Increase in CD4+CD25+ T cells in DTG Mice and CD3+TCRβ+CD4-CD8-

(Double-negative) T Cells in Eµ-TCL1 and DTG Mice

Because abnormalities in the T cell compartment are prevalent in CLL

(Kay 1981; Farace et al., 1994; D’Arena et al., 2012) and they have been

observed in Eµ-TCL1 mice (Hofbauer et al., 2011), we considered that DTG

Page 130: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

111

Page 131: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

112

Figure 3.16 Characterization of T cell subsets in transgenic mice. (A) T cells were

identified by co-expression of CD3 and TCRβ by flow cytometry (top). T cells were

further gated based on expression of CD4 and CD8 (bottom) and the percentages of

these subsets in these representative mice are indicated. (B-C) The percentage (B) and

absolute number (C) of CD4+ and CD8+ T cells in the spleen is shown at each time point

and genotype (n=5-14 mice per genotype per time point). Error bars represent the

standard error of the mean. Statistically significant differences (p<0.05) between values

obtained for DTG mice relative to WT (a), dnRAG1 (b), or Eμ-TCL1 (c) mice are

indicated.

Page 132: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

113

Figure 3.17 Characterization of lymph node T cells. Lymph node T cell subsets

[CD4+ and CD8+ (A), DN (B), and CD4+CD25+ (C)] were identified by flow cytometry at

36 weeks as in figures 13.16 and 3.18 (n=3-14 mice per genotype). Error bars represent

the standard error of the mean. Statistically significant differences (p<0.05) between

values obtained for DTG mice relative to WT (a), dnRAG1 (b), or Eμ-TCL1 (c);

significant differences between Eμ-TCL1 mice relative to WT (d), dnRAG1 (e), or DTG

(f) mice are also indicated.

Page 133: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

114

mice harbor these abnormalities as well. To characterize T cell frequency and

abundance, we used flow cytometry to analyze cohorts of WT, dnRAG1, Eµ-

TCL1, and DTG mice at 6, 12, 24, and 36 weeks of age. This analysis detected a

slight increase in the CD4+ T cell fraction in DTG mice at early time points but a

shift toward CD8+ T cell increase at 36 weeks in the spleen (Figure 3.16A & B).

Increase in CD8+ T cells and subsequent inversion of CD4+ and CD8+ T cell

ratios, which has been reported in Eµ-TCL1 mice (Hofbauer et al., 2011), was

noticeable in both Eµ-TCL1 and DTG mice at 36 weeks in spleen and LN, but is

more pronounced in Eµ-TCL1 mice (Figures 3.16B, C, and 3.17A). Although

absolute numbers of both CD4+ and CD8+ T cells was higher in transgenic

compared to WT mice at 36 weeks in spleen and LN, significant differences were

only established for DTG mice (Figures 3.16C and 3.17A). Further gating of

CD4+ T cells based on CD25 expression showed a significant increase in splenic

CD4+CD25+ T cells in DTG mice by percentage at 36 weeks and in absolute

numbers at 24 and 36 weeks (Figure 3.18). This population of T cells,

presumably Tregs, is commonly expanded in CLL (D’Arena et al., 2012).

Interestingly, we noticed that T cells that were doubly-negative for CD4 or

CD8 expression [CD3+TCRβ+CD8-CD4- (DN T cells)] were increased in

transgenic compared to WT mice particularly at later time points in Eµ-TCL1 and

DTG mice (Figures 3.16A, 3.17B, and 3.19). While the abundance of peripheral

DN T in WT mice is very low (Ford et al., 2006), these cells were significantly

increased in Eµ-TCL1 mice in LN and DTG mice in spleen (Figures 3.17B and

3.19). We hypothesized that DN T cells were a kind of regulatory cell such as

Page 134: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

115

Figure 3.18 Expansion of CD4+CD25+ T cells in DTG mice. (A) The percentage of

gated CD4+CD25+ T cells in the spleen identified by flow cytometry is shown. The mean

percentage of these cells for each genotype at 6, 12, 24, and 36 weeks (B), and

absolute numbers at 12, 24, and 36 weeks (C) (n=3-8 mice per genotype per time point)

are represented by bar graphs. Error bars represent the standard error of the mean.

Statistically significant differences (p<0.05) between values obtained for DTG mice

relative to WT (a), dnRAG1 (b), or Eμ-TCL1 (c) mice are indicated.

Page 135: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

116

Figure 3.19 Expansion of CD3+TCRβ+CD8-CD4- (DN) T cells in transgenic mice.

Double-negative (DN) T cells are CD3+TCRβ+CD8-CD4- T cells indentified by flow

cytometry as shown in figure 3.16A. The mean percentage of these cells for each

genotype at 6, 12, 24, and 36 weeks (A), and absolute numbers at 12, 24, and 36 weeks

(B) (n=5-8 mice per genotype per time point) are represented by bar graphs. Error bars

represent the standard error of the mean. Statistically significant differences (p<0.05)

between values obtained for DTG mice relative to WT (a), dnRAG1 (b), or Eμ-TCL1 (c);

significant differences between Eμ-TCL1 mice relative to WT (d), dnRAG1 (e), or DTG

(f) mice are also indicated.

Page 136: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

117

Figure 3.20 CD1d expression in CD5+ B cells of transgenic mice. (A) Comparison of

CD1d transcript levels between older WT CD19+B220hiCD5- and transgenic CD19+CD5+

B cells determined by gene expression profiling. (B) Flow cytometry was used to

compare the expression of CD1d in WT CD19+CD5- (filled histogram) and CD19+CD5+ B

cells from dnRAG1 (dotted), Eμ-TCL1 (dashed), and DTG mice (solid) at 36 weeks. (C)

The mean fluorescence intensities from B for 5-6 animals of each genotype are

presented in bar graph format. Error bars represent the standard error of the mean. ***,

p< 0.001.

Page 137: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

118

Figure 3.21 α-GalCer treatment has little effect on CD5+ B cell accumulation in

DTG mice. 12-week-old WT and DTG mice (n=4 each) were treated with 5µg of α-

GalCer by tail vein injection once every week for 4 weeks and sacrificed one week

following the final treatment. (A-B) The percentage (A) and absolute number (B) of

splenic CD19+CD5+ B cells identified by flow cytometry are shown for representative

animals (A) or in bar graph format (B). (C) Absolute numbers of DN T cells in the spleen,

dentified by flow cytometry, are shown in bar graph format.

Page 138: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

119

Figure 3.22 Expression of CD160, BTLA, HVEM, and DX5 in DN T or CD5+ B cells.

Expression levels of CD160, BTLA, HVEM, and DX5 in DN T cells (A), and CD160,

BTLA, and HVEM in WT CD19+CD5- and CD19+CD5+ B cells from transgenic mice (B)

was determined by FACS at 36 weeks for 5-6 mice per genotype. Error bars represent

the standard error of the mean. Statistically significant differences (p<0.05) between

values obtained for DTG mice relative to WT (a), dnRAG1 (b), or Eμ-TCL1 (c); Eμ-TCL1

mice relative to WT (d), dnRAG1 (e), or DTG (f) mice are indicated.

Page 139: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

120

NKT cells. However it is was unclear whether these cells were expanding to

inhibit or promote the leukemic cells. NKT cells, unlike conventional T cells,

recognize lipid antigens presented by the non-classical major histocompatibility

complex class 1-like molecule, CD1d (Wu et al., 2004). NKT cells mediate broad

functions ranging from immune protection in infections, maintaining tolerance,

and tumor surveillance (Godfrey and Kronenberg, 2004). Moreover, CLL cells

express CD1d (Fais et al., 2002) and in mice, NKT cells can stimulate B1 and MZ

B cells to augment antibody production in a CD1d-dependent manner (Takahashi

and Strober, 2008).

3.4.2 The Role of Double-negative T Cells in Transgenic Mice

To determine the role of DN T cells in transgenic mice, we initially checked

for CD1d expression in CD5+ B cells. We find that CD5+ B cells of dnRAG1 and

DTG mice had elevated CD1d transcript levels by gene expression arrays as well

as high cell surface expression as assessed by flow cytometry (Figure 3.20).

CD1d in Eµ-TCL1, however, was reduced compared to WT mice. Next, we tested

the role of CD1d in the expansion of leukemic and/or DN T cells in DTG mice.

Using α-galactosylceramide (α-GalCer), a potent agonist for CD1d-restricted

NKT cells (Wu et al., 2004), we found that α-GalCer treatment had little effect on

the number of CD5+ B cells or DN T cells in DTG mice (Figure 3.21). In fact, a

modest decrease was observed in CD5+ B cell numbers in α-GalCer-treated

compared to untreated DTG mice, suggesting an antagonistic effect. Based on

these findings, we reconsidered whether the DN T cells are in fact NKT cells, and

Page 140: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

121

Figure 3.23 DN T cells inhibit accumulation of CD5+ cells after adoptive transfer.

(A-B) SCID mice were injected with the indicated number of CD5+ B with or without DN T

cells sorted from the same donors indicated. The SCID mice were sacrificed 8 weeks

after transfer and the percentage (A) and absolute number (B) of CD5+ B cells was

determined by flow cytometry.

Page 141: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

122

thus explored alternative pathways that could mediate interactions between CD5+

B cells and DNT cells with relevance to CLL. An emerging pathway involves

CD160, B- and T- lymphocyte attenuator (BTLA), and herpes-virus entry

mediator (HVEM) (CD160-BTLA-HVEM signaling), a bi-directional switch that

can mediate inhibitory or stimulatory effects on immune cells (Cai and Freeman,

2009). CLL cells express CD160, normally expressed in NK, NKT, and T cells, to

promote survival (Liu et al., 2010) as well as BTLA, and HVEM (M’Hidi et al.,

2009). We used flow cytometry to define the expression of these markers and the

pan-NK cell marker CD49 (DX5) on CD5+ B cells and DNT cells. These

experiments show that DN T cells from DTG mice express higher levels of

CD160 and BTLA compared to DN T cells from other mice. All DN T cells from all

genotypes uniformly express low levels of HVEM (Figure 3.22A). On the other

hand, all CD5+ B cells express little CD160 and HVEM, and have reduced BTLA

levels compared to WT CD5- B cells (Figure 3.22B). That we did not observe

expression of DX5 in DN T cells from transgenic mice suggests that these cells

may not be NKT cells (Figure 3.22A).

Finally, we investigated the functional interaction between CD5+ B cells

and DN T cells. These experiments revealed that sorted DN T cells, when

adoptively co-transferred with sorted CD5+ B cells from Eµ-TCL1 mice into SCID

mice, inhibit the expansion of the leukemic cells (Figure 3.23). When co-cultured

together, DN T cells similarly inhibit the expansion of leukemic cells (Figure 3.24)

suggesting this effect is direct. Taken together, the data suggest that DN T cells

Page 142: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

123

Figure 3.24 DN T cells inhibit expansion of CD5+ B cells in cell culture. 1x105

CD19+CD5+ spleen lymphocytes sorted from DTG mice by FACS were cultured in

duplicates with or without 5x104 spleen DN T cells from the same mouse. Cells were

counted on day 3, 7, and 10 in a hemacytometer and viability was determined by trypan

blue exclusion.

Page 143: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

124

expanding in Eµ-TCL1 and DTG mice do not show clear NKT cell properties, but

nevertheless inhibit the expansion of CD5+ B cells both in vitro and in SCID mice.

Page 144: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

125

Table 3.3 Top 50 differentially expressed genes between 12-week dnRAG1 and DTG mice

Gene symbol DTG/dnRAG1 Gene description

Neto2 5.26 neuropilin (NRP) and tolloid (TLL)-like 2

Vash2 1.88 vasohibin 2

Tubb3 1.86 tubulin, beta 3

Crip1 1.69 cysteine-rich protein 1 (intestinal)

Pdcd1lg2 1.69 programmed cell death 1 ligand 2

Myadm 1.67 myeloid-associated differentiation marker

Fjx1 1.66 four jointed box 1 (Drosophila)

Tagln2 1.59 transgelin 2

Mid1 1.53 midline 1

Murc 1.48 muscle-related coiled-coil protein

Vim 1.48 vimentin

Olfr767 1.46 olfactory receptor 767

Tyrobp 1.45 TYRO protein tyrosine kinase binding protein

Cln3 1.44

ceroid lipofuscinosis, neuronal 3, juvenile (Batten, Spielmeyer-Vogt

disease)

Lrrc8c 1.38 leucine rich repeat containing 8 family, member C

2010204K13Rik 1.34 RIKEN cDNA 2010204K13 gene

Nid1 1.34 nidogen 1

Ptk2 1.34 PTK2 protein tyrosine kinase 2

Gpr176 1.32 G protein-coupled receptor 176

Mad2l2 1.31 MAD2 mitotic arrest deficient-like 2 (yeast)

Rtn4rl1 1.31 reticulon 4 receptor-like 1

Ppp3ca 1.30 protein phosphatase 3, catalytic subunit, alpha isoform

Mirhg1 1.27 microRNA host gene 1 (non-protein coding)

Jup 1.26 junction plakoglobin

Vapb 1.23 vesicle-associated membrane protein, associated protein B and C

Slamf6 -1.33 SLAM family member 6

Cyb5b -1.35 cytochrome b5 type B

Il2rb -1.36 interleukin 2 receptor, beta chain

Trim7 -1.39 tripartite motif-containing 7

Hmgn3 -1.41 high mobility group nucleosomal binding domain 3

LOC635992 -1.44 similar to ubiquitin-conjugating enzyme E2 variant 2

Olfr99 -1.44 olfactory receptor 99

Rnu12 -1.45 RNA U12, small nuclear // RNA U12, small nuclear

Gm4383 -1.49 predicted gene 4383

4933404M02Rik -1.52 RIKEN cDNA 4933404M02 gene

Pcp4 -1.6 Purkinje cell protein 4

Mpeg1 -1.63 macrophage expressed gene 1

Fcrl5 -1.70 Fc receptor-like 5

Gm7285 -1.71 predicted gene 7285

V165-D-J-C mu -1.76 IgM variable region

Mctp2 -1.77 multiple C2 domains, transmembrane 2

Ipcef1 -1.78 interaction protein for cytohesin exchange factors 1

Clec2d -1.83 C-type lectin domain family 2, member d

Gm9912 -1.89 predicted gene 9912

Zfp420 -1.92 zinc finger protein 420

Slc15a2 -2.06 solute carrier family 15 (H+/peptide transporter), member 2

EG665955 -2.25 predicted gene, EG665955

Gm5571 -2.27 predicted gene 5571 // predicted gene 5571

Igj -2.35 immunoglobulin joining chain

Lmo7 -3.14 LIM domain only 7

Page 145: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

126

Table 3.4 Top common differentially expressed genes among older transgenic mice relative to WT

mice

Fold change

Gene symbol dnRAG1/WT TCL1/WT DTG/WT Gene description

1810046K07Rik 27.28 31.4 38.48 RIKEN cDNA 1810046K07 gene

Cyp11a1 11.31 14.22 26.34

cytochrome P450, family 11, subfamily a,

polypeptide 1

Adm 36.41 25.5 23.38 adrenomedullin

Bmpr1a 10.04 17.82 19.52

bone morphogenetic protein receptor, type

1A

Pstpip2 25.95 53.52 18.61

proline-serine-threonine phosphatase-

interacting protein 2

Bhlhe41 14.04 25.41 17.97 basic helix-loop-helix family, member e41

Ctla4 14.74 21.66 16.77

cytotoxic T-lymphocyte-associated protein

4

Vpreb3 -8.90 -13.56 -9.12 pre-B lymphocyte gene 3

Cr2 -12.25 -26.54 -10.4 complement receptor 2

Gm4955 -8.73 -12.58 -12.38 predicted gene 4955

Satb1 -9.08 -38.65 -13.07 special AT-rich sequence binding protein 1

Gpr171 -22.08 -24.98 -19.47 G protein-coupled receptor 171

Stat4 -25.50 -24.02 -21.45

signal transducer and activator of

transcription 4

Lrrk2 -32.24 -37.39 -34.47 leucine-rich repeat kinase 2

Il10* 11.93 12.70 13.91 interleukin 10

*Il10 was not among the top 50 common differentially expressed genes in all pair-wise comparisons.

Page 146: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

127

Table 3.5 Top 50 differentially expressed genes between ill DTG mouse (M53) & Eµ-TCL1mouse

(M52)

Gene symbol*

DTG M53/TCL1 M52

Fold change Gene description

Prl2a1 269.40 prolactin family 2, subfamily a, member 1

Sox4 42.75 SRY-box containing gene 4

Prl8a2 21.45 prolactin family 8, subfamily a, member 2

Dpp4 18.78 dipeptidylpeptidase 4

Kcnj5 16.97

potassium inwardly-rectifying channel, subfamily J,

member 5

1300014I06Rik 14.56 RIKEN cDNA 1300014I06 gene

Rpl39l 12.40 ribosomal protein L39-like

Rgs13 11.34 regulator of G-protein signaling 13

Xlr3b 11.27 X-linked lymphocyte-regulated 3B

Atp1b1 10.38 ATPase, Na+/K+ transporting, beta 1 polypeptide

Gm10561 10.21 predicted gene 10561

Tet1 10.18 tet oncogene 1

Scamp1 9.92 secretory carrier membrane protein 1

Gpr177 9.48 G protein-coupled receptor 177

Gm10384 9.15 predicted gene 10384

Wdfy3 9.00 WD repeat and FYVE domain containing 3

Atrnl1 8.60 attractin like 1

Dusp6 8.36 dual specificity phosphatase 6

Rgs18 8.26 regulator of G-protein signaling 18

1110032E23Rik 8.24 RIKEN cDNA 1110032E23 gene

Xlr3c 8.24 X-linked lymphocyte-regulated 3C

Xlr3a 7.85 X-linked lymphocyte-regulated 3A

F11r 7.68 F11 receptor

Tes 7.44 testis derived transcript

Ccdc125 7.25 coiled-coil domain containing 125

Zdhhc2 -8.43 zinc finger, DHHC domain containing 2

Cd68 -9.13 CD68 antigen

Cd3g -9.21 CD3 antigen, gamma polypeptide

Gm410 -9.22 predicted gene 410

Kcnn4 -9.25

potassium intermediate/small conductance calcium-

activated channel, subfamily N, member 4

Fcgrt -10.94 Fc receptor, IgG, alpha chain transporter

Igk-V28 -12.16 immunoglobulin kappa chain variable 28 (V28)

Ighv1-72 -12.48 immunoglobulin heavy variable V1-72

Apobec2 -12.89

apolipoprotein B mRNA editing enzyme, catalytic

polypeptide 2

Fxyd5 -13.16 FXYD domain-containing ion transport regulator 5

Prkar2b -13.75

protein kinase, cAMP dependent regulatory, type

II beta

Fbxw13 -13.79 F-box and WD-40 domain protein 13

Ccbp2 -14.01 chemokine binding protein 2

Emr1 -14.46

EGF-like module containing, mucin-like, hormone

receptor-like sequence 1

Slamf9 -15.19 SLAM family member 9

Gm5486 -17.14 predicted gene 5486

Gstt1 -17.57 glutathione S-transferase, theta 1

Anxa2 -20.23 annexin A2

Fgl2 -20.71 fibrinogen-like protein 2

LOC100047053 -21.50 similar to monoclonal antibody kappa light chain

Pdlim1 -27.54 PDZ and LIM domain 1 (elfin)

LOC674190 -30.31 similar to Ig heavy chain V region IR2 precursor

Igh -33.42 immunoglobulin heavy chain complex

LOC100046894 -112.64 similar to Igk-C protein

V165-D-J-C mu -317.51 IgM variable region

*Items listed in bold are found in both Tables 3.5 and 3.6.

Page 147: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

128

Table 3.6 Top 50 differentially expressed genes between ill DTG mouse (F57) & Eµ-TCL1 mouse

(M52)

Gene symbol*

DTG F57/ TCL1 M52

Fold change Gene description

Prl2a1 200.97 prolactin family 2, subfamily a, member 1

Xist 95.10 inactive X specific transcripts

Prl8a2 66.42 prolactin family 8, subfamily a, member 2

Sox4 54.87 SRY-box containing gene 4

Car2 53.97 carbonic anhydrase 2

Gpr126 39.55 G protein-coupled receptor 126

Pfn2 35.98 profilin 2

Cp 34.95 ceruloplasmin

Tnip3 32.26 TNFAIP3 interacting protein 3

Fcer2a 31.59 Fc receptor, IgE, low affinity II, alpha polypeptide

Cpm 30.20 carboxypeptidase M

Gm10879 29.01 predicted gene 10879

Anxa1 25.91 annexin A1

Dusp4 25.75 dual specificity phosphatase 4

Abp1 23.25

amiloride binding protein 1 (amine oxidase, copper-

containing)

Id2 21.62 inhibitor of DNA binding 2

Rnf128 18.68 ring finger protein 128

Fabp7 18.40 fatty acid binding protein 7, brain

Dusp6 17.93 dual specificity phosphatase 6

Slc22a21 15.49

solute carrier family 22 (organic cation transporter), member

21

Akt3 14.84 thymoma viral proto-oncogene 3

Scin 14.54 scinderin

Azgp1 13.03 alpha-2-glycoprotein 1, zinc

Igh-6 12.75 immunoglobulin heavy chain 6 (heavy chain of IgM)

Parp8 12.59 poly (ADP-ribose) polymerase family, member 8

Cln8 -6.79 ceroid-lipofuscinosis, neuronal 8

Neurod4 -7.26 neurogenic differentiation 4

Lrrc25 -7.52 leucine rich repeat containing 25

Gp49a -7.74 glycoprotein 49 A

Tmprss13 -7.96 transmembrane protease, serine 13

Prkar2b -8.03 protein kinase, cAMP dependent regulatory, type II beta

Emr1 -8.05

EGF-like module containing, mucin-like, hormone

receptor-like sequence 1

Kdm5d -8.51 lysine (K)-specific demethylase 5D

Sell -9.15 selectin, lymphocyte

Cd9 -9.97 CD9 antigen

Fgl2 -10.09 fibrinogen-like protein 2

Cd3g -10.35 CD3 antigen, gamma polypeptide

Uty -10.71

ubiquitously transcribed tetratricopeptide repeat gene, Y

chromosome

Fbxw13 -12.03 F-box and WD-40 domain protein 13

Ccbp2 -13.49 chemokine binding protein 2

Gstt1 -13.80 glutathione S-transferase, theta 1

Hpse -13.87 heparanase

Lgals1 -14.20 lectin, galactose binding, soluble 1

V165-D-J-C mu -14.31 IgM variable region

Igh -16.44 immunoglobulin heavy chain complex

Cd36 -16.52 CD36 antigen

Ddx3y -24.47 DEAD (Asp-Glu-Ala-Asp) box polypeptide 3, Y-linked

Eif2s3y -24.72

eukaryotic translation initiation factor 2, subunit 3, structural

gene Y-linked

LOC674190 -37.70 similar to Ig heavy chain V region IR2 precursor

LOC100046894 -48.43 similar to Igk-C protein

*Items listed in bold are found in both Tables 3.5 and 3.6.

Page 148: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

129

CHAPTER 4: DISCUSSION

4.1 FAILURE IN SELF-TOLERANCE PROMOTES CLL PROGRESSION

4.1.1 Expression of Dominant-negative RAG1 Accelerates CLL

Progression in Eµ-TCL1 Mice

An emerging model for the etiology of CLL suggests that this disease

originates through the monoclonal expansion of an antigen-experienced

CD5+ B cell displaying poly-reactivity and/or autoreactivity toward molecular

motifs present on and often shared between apoptotic cells and bacteria

(Myhrinder et al., 2008). CD5+ B cells possessing this reactivity profile may

develop through positive selection, such as peritoneal B1 cells and MZ B

cells (Montecino-Rodriguez et al., 2006). Alternatively, they may emerge

from conventional B2 lineage cells that are driven to express CD5 as a

mechanism to attenuate chronic (auto)antigenic BCR signaling in response to

failed attempts at silencing or reprogramming fortuitous germline-encoded

BCR autoreactivity (Jankovic et al., 2004; Hippen et al., 2000). If immune

suppression is breached, the autoreactive B cell may enter the germinal

center reaction, giving rise to somatically mutated B cells with a more

restricted reactivity profile (Herve et al., 2005). In this way, both unmutated

and mutated CLL may be traced to a common polyreactive/autoreactive adult

B lineage precursor (Herve et al., 2005), which is consistent with the

observation that unmutated and mutated CLL show similar gene expression

profiles (Seifert et al., 2012).

Page 149: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

130

Receptor editing initiated in response to autoantigen exposure is

thought to occur primarily during the immature/transitional T1 stage of B cell

development (Wang et al., 2007; Keifer et al., 2008). We speculate that a

stochastic mutation which impairs receptor editing induced in response to

self-reactivity may enforce B cell autoreactivity. Based on our previous work

showing that dnRAG1 mice accumulate CD5+ B cells without progressing to

overt CLL (Hassaballa et al., 2011), we propose that autoreactivity enforced

by a receptor editing defect could permit continued autoantigenic stimulation,

and thereby promote sustained CD5 expression and persistence of a CD5+ B

cell clone. This possibility is consistent with the observation that CLL is often

preceded by an indolent monoclonal B cell lymphocytosis (MBL) that is

phenotypically and cytogenetically similar to CLL (Scarfo et al., 2010;

Rawstron et al., 2008). Although the initiating genetic mutation enforcing B

cell autoreactivity would normally expected to be rare and restricted to a

single B cell (thereby driving emergence of an MBL clone), developing B cells

undergoing receptor editing (estimated at 25-75% of B cells (Wardemann et

al., 2003) in dnRAG1 mice may frequently be blocked from successful editing

by dnRAG1 expression. Thus, by facilitating a rate-limiting step in the

emergence of CLL precursors, dnRAG1 expression may accelerate CLL

progression in Eµ-TCL1 mice. If so, one might predict that the clonal B cell

repertoire in ill DTG mice should be more diverse than ill Eµ-TCL1 mice.

Indeed, the observation that DTG mice show a higher number of unique

productively rearranged heavy and light chain genes amplified by PCR lends

Page 150: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

131

support to this idea (Table 3.1 and 3.2).

We recognize that the absence of recurrent RAG mutations in CLL

(Puente et al., 2011) argues that CLL does not arise through defects in RAG

activity. However, we emphasize that autoreactivity may be enforced by

RAG-independent mutations which disrupt any of the pathways involved in

initiating or completing receptor editing, or eliminating autoreactive B cells

that fail to successfully edit. The same principle also applies for defects in

receptor revision postulated to occur in B cells in response to acquiring

autoreactivity by somatic hypermutation. Once enforced autoreactivity is

established, subsequent genetic lesions (modeled here by Eµ-TCL1

expression) may promote further expansion and/or transformation of the

CD5+ B cell. A simplified model summarizing the etiology and progression of

CLL promoted by impaired receptor editing is shown in Figure 4.1. While this

model suggests that autoantigenic stimulation induces receptor editing, cell-

autonomous signaling mediated by certain intrinsically

autoreactive/polyreactive heavy chains showing recurrent usage in CLL might

also initiate this process (Duhren-von Minden et al., 2012). One way cell-

autonomous signaling could normally be attenuated is through light chain

receptor editing to permit selection of “editor” light chains that neutralize

interactions mediated by HCDR3. Studies of mice expressing an IgH

transgene specific for dsDNA provide precedence for this possibility (Li et al,

2001). We speculate that CLL cells may retain cell-autonomous signaling

capacity because non-editor light chains have either been positively selected

Page 151: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

132

or failed to be replaced by receptor editing.

4.1.2 Breaching Tolerance Promotes CLL

Previous studies have shown that CLL emergence in Eµ-TCL1 mice is

accelerated when B cell tolerance is breached (Enzler et al., 2009; Bertilaccio

et al., 2011). Transgene expression of the pro-survival factor for normal and

leukemic B cells, BAFF (Enzler et al., 2009), and loss of expression of TIR8

(type I transmembrane inhibitory receptor 8; SIGIRR), a negative regulator of

Toll-like receptor and interleukin receptor signaling (Bertilaccio et al., 2011)

both accelerate the onset and progression of murine CLL in Eµ-TCL1 mice.

Both BAFF and TIR8 are implicated in the regulation of B cell tolerance since

autoimmunity develops in BAFF transgenic mice (Mackay et al., 1999; Khare

et al., 2000) as well as in TIR8-/- mice bred onto a lupus-prone strain

background (Lech et al., 2008). That dnRAG1 expression also appears to

interfere with this process (Hassaballa et al., 2011), and similarly accelerates

disease progression in Eµ-TCL1 mice, argues that mechanisms required to

enforce self-tolerance are common targets for disruption in CLL etiology

and/or progression. The connection between loss of self-tolerance,

autoimmunity, and CLL is further underscored by observation that CLL BCRs

bind a multitude of autoantigens and about a quarter of CLL patients develop

autoimmune manifestations during the course of the disease (Kipps et al.,

1988; Rosen et al., 2010; Caligaris-Cappio et al., 1994; Zent and Kay, 2010).

Our studies also show that CLL cells in Eµ-TCL1 and DTG mice carry

Page 152: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

133

Page 153: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

134

Figure 4.1 Model for how defects in receptor editing can promote development

of MBL and CLL. Non-self-reactive immature B cells arriving from the bone marrow

progress normally through the immature/transitional stages of B cell development

where receptor editing normally occurs. An immature/T1 B cell possessing an

autoreactive specificity normally subjected to editing may acquire a spontaneous

mutation that impairs receptor editing (RE), (modeled here by dnRAG1 expression),

which enforces receptor specificity and leads the B cell to retain or adopt a CD5+ B1-

like (and CLL-like) phenotype. The cell may persist or accumulate as an MBL, which

may or may not require other mutations, such as those causing TCL1 over-

expression. Additional genetic lesions may promote MBL evolution to CLL and

perhaps subsequent transformation to high-grade malignancy such as in Richter’s

syndrome (RS) (Tsimberidou et al., 2006). A similar series of events may also occur

in post-germinal center B cells attempting to undergo receptor revision after having

acquired autoreactivitiy during hypermutation (not shown).

Page 154: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

135

BCRs with specificities toward autoantigens such as DNA, membrane

phospholipids, and RBC epitopes, implicating loss of tolerance in the initiation

or progression of leukemia. Additionally, the expression pattern of BCR

components and signaling genes in all transgenic mice suggest that these

cells have encountered antigens. CD5 expression, for example, may be

induced on B cells rendered anergic by chronic antigenic stimulation (Hippen

et al., 2000) which, in principle, would be expected if CLL was driven by self-

antigens which are ubiquitous in nature. Indeed, B cells in dnRAG1 mice

show some evidence of anergy, as they are intrinsically hyporesponsive

toward anti-IgM crosslinking and mitogenic stimulation in vitro and mount

poorer immune responses toward thymus-independent antigens compared to

wild-type controls (Hassaballa et al., 2011). Additional support for the

induction of functional anergy in dnRAG1 and DTG mice comes from the

observation that, serum IgM and IgG levels in these animals was significantly

lower compared to wild-type mice at both 12 and 36

weeks of age (Figure 3.9).

CLL B cells are antigen-experienced with cells from subset of patients

displaying signs of exhaustion (Mockridge et al., 2007; Muzio et al., 2008;

Stevenson et al., 2011). CD5 expression, which can be a marker for anergic

B cells (Hippen et al., 2000), may be induced after prolonged antigenic

exposure through the transcription factor Nfatc1, which itself has been found

Page 155: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

136

to be constitutively expressed in CLL or to be upregulated after BCR ligation

more so in cases with unmutated Ig VH genes (Guarini et al., 2008; Le Roy et

al., 2012). CD22, a negative regulator of BCR signaling that is downregulated

in CLL (Jelinek et al., 2003; Damle et al., 2002), was also found to be

downregulated in CD5+ B cells of transgenic mice, suggesting a previous

encounter with antigens (Lajaunias et al., 2002; Damle et al., 2002). The

upregulation of CTLA4, an inhibitory receptor normally expressed in T cells,

in CD5+ B cells from all transgenic mice and human CLL (Rosenwald et al.,

2001), implicates an adaptation to quench BCR signals since CLL cases

where CTLA4 is downregulated are associated with poor prognosis (Joshi et

al., 2007). The expression patterns of these genes may explain why a subset

of CLL cells respond poorly to antigen stimulation (Mockridge et al., 2007;

Muzio et al., 2008), which could be a contributing factor to the

hypogammaglobulinemia seen in CLL patients (Rozman et al., 1988; Colovic

et al., 2001).

Of particular interest is the finding that CD5+ B cells from all transgenic

mice, like CLL cells (Kitabayashi et al., 1995; DiLillo et al., 2013), produce the

anti-inflammatory/immunosuppressive cytokine, IL-10 (Moore et al., 2001).

This supports a similar cell of origin for both species since CD5+ B cells are

the primary source of B cell-derived IL-10 in mice and humans (O’Garra et al,

1992; Gary-Gouy et al., 2002). What is unclear is whether IL-10 is produced

to suppress inflammation and/or as a survival factor for leukemic cells. IL-10

expression has been shown to correlate with clinical stage and CLL

Page 156: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

137

progression (Kamper et al., 1999; Karmali et al., 2013), suggesting that it

plays a significant role in pathogenesis. IL-10 protects CLL cells from

apoptosis in culture (Kitabayashi et al., 1995) and loss of IL-10 blocks

leukemia development in NZB mouse strain susceptible to CLL, which

suggests that this cytokine may act as an autocrine growth factor for CLL

cells (Ramachandra et al., 1996; Czarneski et al., 2004). Additionally, IL-10

may contribute to the immunosuppressive phenotype observed in CLL. Since

IL-10 has a direct effect on T cells (Moore et al., 2001), it is possible that a

generalized increase in IL-10 expression limits T cell activation during

infection and even tumor surveillance.

Of note, IL-10 is also produced by regulatory B (Breg) cells, which

reportedly display a CD5+CD1dhiCD21+ phenotype (Mauri and Bosma, 2012).

However, since the splenic CD5+ B cells accumulating in dnRAG1, Eµ-TCL1,

and DTG mice are clearly CD21- (Figure 3.3), we conclude that these cells

are unlikely to be Bregs.

4.1.3 Significance of Prl2a1 Expression in dnRAG1 and DTG Mice

Gene expression profiling shows that one gene which distinguishes dnRAG1

and DTG mice from Eµ-TCL1 mice is Prl2a1, which is highly expressed in

CD5+ B cells from dnRAG1 and DTG mice (up to several hundred-fold)

compared with Eµ-TCL1 mice (Figures 3.10, 3.12, and 3.13). Prl2a1 is a

prolactin paralogue and a member of the hormone gene superfamily which

consists of growth hormone, prolactin, prolactin-like proteins, and placental

Page 157: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

138

lactogens (Goffin et al., 1996; Wiemers et al., 2003; Simmons et al., 2008).

The hormone gene superfamily members are related, based on sequence

analysis, by sequence and are thought to have originated through gene

duplications. While most mammals, including humans, have a single prolactin

gene, rodents have a least two dozen prolactin paralogues that arose by

divergence after duplication of prolactin (Wiemers et al., 2003; Li and Zhang,

2006). Expression of prolactin and related proteins is regulated both spatially

and temporally, and control a wide range of biological functions ranging from

reproduction and lactation to immunoregulation (Freeman et al., 2000).

Prolactin acts as a cytokine in immune cells by signaling through several

pathways including Jak2/Stat5, MAP kinase, or by activating protein tyrosine

kinases such Src, Fyn, and Tec to induce growth and survival (Clevenger et

al., 1998). Little is known about the role of Prl2a1 especially in the immune

system but the effect of prolactin on lymphocytes is well recognized

(Hiestand et al., 1986; Clevenger et al., 1998; Peeva et al., 2003; Saha et al.,

2009; Shelly et al., 2012). Prolactin or prolactin receptor deficient mice show

normal development of lymphocytes (Bouchard et al., 1999; Dorshkind and

Horseman, 2000), but hyperprolactinemia promotes autoimmunity in both

humans and mice. For instance, hyperprolactenemia is associated with

systemic lupus erythematosus, Sjogren’s syndrome, and rheumatoid arthritis

(Shelly et al., 2012) and prolactin induction exacerbates lupus in susceptible

mouse strains such as NZB/NZW F1 mice, which can be ameliorated by

treatment with bromocriptine (Saha et al., 2011). Bromocriptine is a

Page 158: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

139

dopamine receptor agonist (Waddington, 1986), which is thought to function

like dopamine in inhibiting the production of second messengers such as ca2+

and cyclic AMP, which are necessary for prolactin secretion (Enjalbert et al.,

1986; Schettini et al., 1986), Additional evidence suggests prolactin signaling

may regulate B cell tolerance mechanisms (Saha et al., 2011) by promoting

survival of T1 transitional B cells that are normally subjected to negative

selection at this stage (Peeva et al., 2003; Saha et al., 2009; Ledesma-Soto

et al., 2012). We speculate that murine autoreactive B cells transiently

produce Prl2a1 as a pro-survival factor to facilitate receptor editing, but in

dnRAG1 and DTG mice, Prl2a1 expression is sustained because receptor

editing is blocked by dnRAG1 expression. Because humans do not have a

Prl2a1 homologue (Li and Zhang, 2006), its role in B cell development and

CLL cannot be ascertained; whether an orthologous prolactin/growth

hormone family member assumes its function in human B cells remains

unclear.

4.2 ROLE OF REGULATORY T CELLS IN CLL

4.2.1 CD4+CD25+ T cells

Regulatory T cells (Tregs), normally defined as either CD4+CD25+ or

CD4+CD25+ with Foxp3, or with low or absent CD127, are increased in CLL

(D’Arena et al., 2012). The normal role of these cells is to maintain peripheral

tolerance (Sakaguchi et al., 2008) but their expansion in many hematological

Page 159: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

140

and solid malignancies suggests that they may also facilitate tumor escape.

This concept is supported by the observation that depletion of Tregs or all

CD4+ T cells restores anti-tumor immunity (Beyer and Schultze, 2006).

The mechanisms of Treg expansion in CLL are unclear. Although

Tregs may be formed through stimulation by tumor antigens, this does not

appear to be the case in CLL (Jak et al., 2009). Malignant, but not normal, B

cells can induce Treg differentiation from CD4+CD25- through direct contact

(Han et al., 2011). Because changes induced on CD4+ and CD8+ T cells by

CLL cells are mediated by direct contact (Gorgun et al., 2005), it is plausible

that a similar mechanism could also induce differentiation to Tregs.

Alternatively, differentiation of Tregs may be fostered by IL-10 and TGF-β

(Zheng et al., 2004), cytokines that are produced by CLL cells (Kitabayashi et

al., 1995; DiLillo et al., 2013; Kremer et al., 1992; Lotz et al., 1994; Schuler et

al., 1999).

The immunosuppressive properties of Tregs in conjunction with CD4+

and CD8+ T cell dysfunction and CLL-derived IL-10 presumably contribute to

the systemic immunodeficiency manifested in CLL patients. This could

explain the high incidence of autoimmune phenomena, infections, and risk of

secondary malignancies in CLL (Zent and Kay, 2010; Itala et al., 1992;

Molica et al., 1993; Kyasa et al. 2004; Maddocks-Christianson et al., 2007).

4.2.2 Double-negative (DN) T Cells

We found an age-dependent increase in splenic DN T cells in both Eµ-

Page 160: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

141

TCL1, and DTG mice. While these cells do not show clear NKT cell

properties, they promote clearance of CD5+ B cells both in vitro and when

they are co-transferred with CLL cells into immunodeficient mice (Figures

3.24 and 3.23). Whether these cells play the same role in their natural setting

in ill mice is unclear because CLL cells have a demonstrated proclivity to

inhibit or subvert the function of other cells in tumor microenvironments

(Fecteau and Kipps, 2012). For example, CD4+CD25+ T cells that also

expand in these mice, and are absent in SCID mice, could suppress DN T

cells. It would not be surprising if DN T cells from DTG mice are not active in

their natural setting since they express high levels of BTLA, which can be a

marker for anergy in T cells (Murphy et al., 2006).

Mature T cells that don’t express CD4 or CD8 encompass several

subsets including NKT cells (Godfrey et al., 2010). The role of NKT cells in

CLL is unclear. NKT cells do not appear to be expanded in CLL compared to

healthy population (Gibson et al., 2011) but their frequency is reduced in

patients with progressive compared to indolent disease (Jadidi-Niaragh et al.,

2012). However, NKT cells can be primed by CLL cells through CD1d

resulting in killing of the CLL cells in vitro (Fais et al., 2004). Since CLL cells

can induce functional defects in NK cells (Kay et al., 1984) and T cells

(Ramsey et al., 2008; Riches et al., 2013), it is not clear whether the same is

true for NKT cells in vivo particularly since there is a lack of autologous anti-

tumor immune responses even though CLL cells express tumor antigens

(Krackhardt et al., 2002).

Page 161: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

142

The possibility that NKT cells play a contributing role in CLL

pathogenesis is intriguing, but has not been fully investigated. CD1d, which

presents antigens to NKT cells, appears to be elevated in a subset of CLL

cases (Kotsianidis et al., 2011; Anastasiadis et al., 2013; Fais et al., 2004) as

well as in dnRAG1 and DTG mice (Figure 3.20). Although CD1d expression

is higher in B cell lymphoproliferative diseases originating in cells that

naturally express high levels of CD1d, such as splenic MZ lymphoma

compared to CLL (Amano et al., 1998; Kotsianidis et al., 2011), this marker is

nevertheless increased in CD38+ CLL cases and those bearing unmutated Ig

genes (Kotsianidis et al., 2011; Anastasiadis et al., 2013; Fais et al., 2004).

4.3 LESSONS FROM MURINE MODELS OF BENIGN AND

MALIGNANT CD5+ B CELL EXPANSION

Gene expression profiling studies provide evidence of distinct but

convergent pathways for CLL development. The finding that DTG mice more

closely resemble dnRAG1 mice than Eµ-TCL1 mice with respect to

expression profiles at 12 weeks and serum Ig levels at both time points

argues that CD5+ B cell accumulation in DTG mice is driven primarily by

dnRAG1 expression. Thus, CD5+ B cells in Eµ-TCL1 mice and dnRAG1/DTG

mice appear to initially emerge by slightly different mechanisms, yet their

gene expression patterns remain more similar to one another than to other

normal splenic B cell populations.

Page 162: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

143

Interestingly, this pattern may be also hold true for other mouse strains

that show evidence of CD5+ B cell accumulation (Hayakawa et al., 1983;

Berland and Wortis, 2002; Hassaballa et al., 2011) including murine models

of CLL (Table 1.2). For instance, most of the CLL models reportedly share an

immunophenotype that is centered on lower expression of B220 than

conventional B cells and lack of CD23, which is expressed on human CLL

cells (Table 1.2; Damle et al., 2002). This would suggest that either the same

B cell/stage is involved in transformation in these models or that the

transforming mechanism itself is similar. However, it is more likely that the

same B cell subset is affected in these models since the original molecular

defects lie in distinct pathways. This possibility is supported partly by the fact

that murine MZ and B1 B cells do not express CD23 (Waldschimidt et al.,

1992; Best et al., 1995). Additionally, in the case of NZB and Eµ-TCL1 mice

for instance, the leukemic cells bear other properties of B1 B cells such as IL-

10 expression, and BCR characteristics (O’Garra et al, 1992; Gary-Gouy et

al., 2002; Czarneski et al., 2004; DiLillo et al., 2013; Phillips et al., 1992; Yan

et al., 2006). Furthermore, the distinct patterns of organ involvement suggest

that leukemia in NZB, APRIL, and Eµ-TCL1 mice most likely originates from

peritoneal B1 cells, while the others may originate from similar cells found in

other organs such as the spleen (Table 1.2). The relatively long generation

time in most of the models suggests that additional defects are necessary to

develop frank leukemia. This possibility is supported by TCL1 expression in

the dnRAG1 mouse.

Page 163: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

144

Why do some B cell subsets appear more vulnerable to perturbation

than others? The answer may lie in the different pathways by which these

cells develop and function. B1 and MZ B cells, unlike follicular B cells, are

positively selected by self-antigens such as those resulting from apoptosis

and oxidative processes (Bendelac et al., 2001). In deed, even the

expression of Ig transgenes with specificities toward phosphatidyl choline,

thy-1, RBCs, and Sm, antigens that are principally bound by B1 cells,

expands the B1 compartment in mice (Berland and Wortis, 2002). These

specificities, while seemingly unwanted, are tolerated because they are

beneficial to the host in providing rapid protection against pathogenic

microbes and in clearing of apoptotic cellular debris (Bendelac et al., 2001;

Baumgarth, 2011). B1 cells, in particular, are crucial in these tissue

homeostatic functions since they express several scavenger receptors, of

which CD5 is one (Rodriguez-Manzanet et al., 2010; Jordo et al., 2011).

Therefore, when these cells become diseased in autoimmunity or neoplasia,

they are difficult to purge because they are naturally designed to be

maintained at a cost to the host. That CLL cells are difficult to maintain in

culture (Collins et al., 1989) would support the notion that these cells are

actively maintained in vivo, in essence, by persistent antigen stimulation.

Berland and Wortis (2002), in discussing the origins of B1 cells note

that defects in BCR signaling fundamentally affect the B1 lineage while

sparing conventional B cells for the most part. Deletion or defects in a variety

of positive regulators of BCR signals, namely, Btk, PKCβ, PLCγ, p85α

Page 164: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

145

subunit of PI3-K, CD19, BLNK, CD21/35, and vav-1 lead to loss of CD5+ B

cells in mice. On the other hand, defects targeting negative regulators of BCR

signaling, namely, SHP-1, CD22, Lyn, CD72, and more recently, Siglec-G

(Hoffmann et al., 2007; Ding et al., 2007) result in accumulation of CD5+ B

cells. Thus, the CD5 compartment in mice may be a repository for B cells

with certain related defects (Hassaballa et al., 2011). These observations

suggest that regulation of BCR signaling threshold is crucial in the generation

of CD5+ B cells. Collectively and in theory, these studies in murine models

suggest that there are multiple defects that can potentially lead to the

accumulation of CD5+ B cells, which could explain CLL heterogeneity in

humans.

4.4 FUTURE WORK

4.4.1 Mechanisms of Disease Acceleration in DTG Mice

Although we have not established specific factors responsible for

accelerating disease progression in DTG mice presumably because CD5+ B

cells in dnRAG1 and Eµ-TCL1 mice develop in overlapping pathways, we

have identified genes that are differentially expressed in CD5+ B cells from

DTG mice relative to their single-transgene counterparts which may warrant

further study. For example, both Dpp4 upregulation and Sell downregulation

reportedly correlate with poor prognosis in human CLL (Cro et al., 2009;

Stratowa et al., 2001), which is consistent with the earlier onset of disease

Page 165: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

146

observed in DTG mice relative to Eµ-TCL1 mice. Another interesting gene

that warrants further attention is PTPN22. PTPN22, which acts as a negative

regulator of BCR signaling in B cells (Arechiga et al., 2009), is upregulated in

CLL (Wang et al., 2004; Negro et al., 2013) and has polymorphisms that are

associated with multiple autoimmune conditions (Bottini et al., 2006; Dai et

al., 2013). Since this gene plays a significant role in BCR signaling fate

(Arechiga et al., 2009) and we found it to be upregulated least 2.5-fold at all

time points in all transgenic mice (Figure 3.15), it is possible that it plays a

significant role in CLL development in mice and humans.

4.4.2 Role of Hormone Gene Superfamily in Normal and Leukemic B

Cells

Besides Prl2a1, we found Prl7c1 and Prl8a2 to be upregulated in

dnRAG1 and DTG mice, implicating prolactin family genes in B cell

development and murine leukemia. Although prolactin and its receptor are

reportedly expressed in lymphocytes and other immune cells (Clevenger et

al., 1998; Peeva et al., 2003), this is the first report, to our knowledge, of

expression of these prolactin-like genes in lymphocytes. It would be

interesting to investigate whether these genes are expressed in normal

murine B and T lymphocytes, and if so, at what stages. Based on this study

and previous studies on prolactin (Peeva et al., 2003; Saha et al., 2009;

Ledesma-Soto et al., 2012), it is likely that these genes would be expressed

in transitional stages or in cells undergoing receptor editing/revision possibly

Page 166: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

147

to facilitate survival during selection processes. First and foremost, it would

be important to establish whether prolactin-like proteins have bioactivity on

lymphocytes. Loss of prolactin or its receptor has little effect on lymphocyte

development, but overexpression of prolactin aggravates autoimmune

conditions (Bouchard et al., 1999; Dorshkind and Horseman, 2000; Saha et

al., 2011; Shelly et al., 2012). If prolactin-like proteins also signal through the

prolactin receptor in lymphocytes, then it can be presumed that Prl2a1 knock-

out mice would not have a clear phenotype as well except owing to the fact

others have noted that hormones could act differently under stressful

conditions (Dorshkind and Horseman, 2000). Nevertheless blocking prolactin-

like proteins, by analogy to experiments using bromocriptine to inhibit

prolactin secretion in mice (Peeva et al., 2003), would help to establish

whether these prolactins are required for development of leukemia in mice.

Most importantly, are similar hormones involved in CLL pathogenesis?

Although humans lack these prolactin-like genes, orthologous genes within

the hormone gene superfamily, namely prolactin and growth hormone (Goffin

et al., 1996; Wiemers et al., 2003; Simmons et al., 2008), may assume a

similar function in human B cells. Interestingly, prolactin is involved in the

pathogenesis of some autoimmune conditions such as SLE (Gutierrez et al.,

1995; Saha et al., 2011), and CLL patients are also prone to autoimmunity

including SLE (Zent and Kay, 2010; Lugassy et al., 1992). The possibility that

there exists an intersection between autoimmunity and CD5+ B cell neoplasia

is illustrated by New Zealand mice, which serve as of model for SLE as well

Page 167: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

148

as CLL (Stall et al., 1988; Kono et

al., 1994; Phillips et al., 1992; Raveche et al., 2007). Thus, it is possible that

a subset of CLL patients may also have underlying hyperprolactinemia.

4.4.3 Role of CD1d in CLL

Since we found CD1d to be upregulated in dnRAG1 and DTG mice, it

would be interesting to dissect its role in leukemia development in light of the

recent findings that CD1d expression is elevated in aggressive CLL cases

(Fais et al., 2004; Kotsianidis et al., 2011; Anastasiadis et al., 2013). Even

though treatment of mice with α-GalCer seemed to have little effect on

accumulating CD5+ B cells in DTG mice, these experiments could be

extended to include CD1d antagonists or a different agonist (Lombardi et al.,

2010; Szatmari et al., 2006). However, targeted deletion of CD1d in B cells

would reveal whether or not it is required for accumulation of CD5+ B cells in

transgenic mice. Along with these experiments, one might monitor NKT cells

by using CD1d tetramers (Matsuda et al., 2000; Gumperz et al., 2002) to

determine their role in accumulation of CD5+ B cells in these mice. While the

identity of DN T cells found in Eµ-TCL1 and DTG mice remain unclear at

present, it could be determined if they are similar to other known T cell

subsets such as those described by Zhang et al. (2000) and Chen et al.

(2004) based on immunophenotype and secreted molecules.

Of particular interest is the nature and fate of antigens bound by B1

and CLL cells. These cells bear specificities toward peculiar antigens such as

Page 168: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

149

carbohydrates and lipids (Bendelac et al., 2001; Catera et al., 2008;

Myhrinder et al., 2008; Chu et al., 2010). That CD1d presents lipid and

glycolipid antigens (Porcelli et al., 1999; Godfrey et al., 2010) raises the

question whether there is a link between these activities in normal or

diseased B cells. For instance, how frequently do lipid-based antigens

internalized with the BCR end up being processed and loaded on CD1d, and

would this be relevant to normal B cell function and disease?

4.4.4 Identity of double-negative T cells

The double-negative (DN) T cells found to be expanding in Eµ-TCL1

and DTG mice do not appear to be NK T cells. There exist other less

common DN regulatory T cells. These cells make 1-5% of peripheral T cells

in mice and humans (Strober et al., 1996; Chen et al., 2004) but similar cells

have been found to expand be expanded in pathological conditions such as

SLE (Shivakumar et al., 1989; Crispin et al., 2008). The DN regulatory T cells

may not express NK cell markers such as NK1.1, as is the case in DTG mice,

but they additionally express markers of activation such as CD25 and CD69

but do not express CD44, and secrete α, β, and γ interferons (Strober et al.,

1996; Zhang et al., 2000; Chen et al., 2004). Functionally, DN T cell normally

regulate tolerance and are potent suppressors of other effector T cells and

thus have been shown to be critical in preventing allograft rejection (Zhang et

al., 2000; Chen et al., 2004). However, these cells can contribute to

pathological conditions such as SLE by producing inflammatory cytokines

Page 169: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

150

and enhancing production of autoantibodies (Shivakumar et al., 1989; Crispin

et al., 2008). Recently, a subset of DN T cells has also been reported to be

increased in the peripheral blood in CLL but its role in this disease remains to

be explored (Weinkove et al., 2013).

4.5 CONCLUDING REMARKS

Majority of the work presented in this dissertation (sections 3.1 to 3.3)

is published in Nganga et al. Blood. (2013) 121(19): 3855-66, S1-16. This

dissertation asserts that the BCR is not just a bystander but plays an active

role in CLL. In fact, studies in dnRAG1 and DTG mice imply that failure in

remodeling the BCR in response to autoreactivity can lead to the initiation of

clonal B cells that evolve to CLL by gathering genetic lesions. Moreover,

gene expression profiling studies with dnRAG1, Eµ-TCL1, and DTG mice

provide evidence of distinct but convergent pathways for CLL development.

However, the pathogenesis of murine CLL involves multiple factors including

potentially, prolactin-like hormones such a Prl2a1, and regulatory T cells.

Even though the role of the BCR in CLL is strongly suggested by the

significance of mutational status in clinical course among other factors, these

studies provide additional justification for considering the BCR signaling

pathway and its associated regulatory mechanisms as rational targets in CLL

treatment.

Page 170: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

151

CHAPTER 5: REFERENCES

Agathangelidis, A., Darzentas, N., Hadzidimitriou, A., Brochet, X., Murray, F., Yan, X. J., . . . Stamatopoulos, K. (2012). Stereotyped B-cell receptors in one-third of chronic lymphocytic leukemia: a molecular classification with implications for targeted therapies. Blood, 119(19), 4467-4475. doi: 10.1182/blood-2011-11-393694

Aggarwal, M., Villuendas, R., Gomez, G., Rodriguez-Pinilla, S. M., Sanchez-

Beato, M., Alvarez, D., . . . Piris, M. A. (2009). TCL1A expression delineates biological and clinical variability in B-cell lymphoma. Mod Pathol, 22(2), 206-215. doi: 10.1038/modpathol.2008.148

Ahmed, S., Siddiqui, A. K., Rossoff, L., Sison, C. P., & Rai, K. R. (2003).

Pulmonary complications in chronic lymphocytic leukemia. Cancer, 98(9), 1912-1917. doi: 10.1002/cncr.11736

Ahuja, D., Sáenz-Robles, M. T., & Pipas, J. M. (2005). SV40 large T antigen

targets multiple cellular pathways to elicit cellular transformation. Oncogene, 24(52), 7729-7745. doi: 10.1038/sj.onc.1209046

Alegre, M. L., Frauwirth, K. A., & Thompson, C. B. (2001). T-cell regulation by

CD28 and CTLA-4. Nat Rev Immunol, 1(3), 220-228. doi: 10.1038/35105024

Amano, M., Baumgarth, N., Dick, M. D., Brossay, L., Kronenberg, M.,

Herzenberg, L. A., & Strober, S. (1998). CD1 expression defines subsets of follicular and marginal zone B cells in the spleen: beta 2-microglobulin-dependent and independent forms. J Immunol, 161(4), 1710-1717.

Anastasiadis, A., Kotsianidis, I., Papadopoulos, V., Spanoudakis, E.,

Margaritis, D., Christoforidou, A., . . . Tsatalas, C. (2013). CD1d expression as a prognostic marker for chronic lymphocytic leukemia. Leuk Lymphoma. doi: 10.3109/10428194.2013.803222

Anderson, M. A., Huang, D. C., & Roberts, A. W. (2013). BH3 mimetic

therapy: an emerging and promising approach to treating chronic lymphocytic leukemia. Leuk Lymphoma. doi: 10.3109/10428194.2013.769223

Andreeff, M., Darzynkiewicz, Z., Sharpless, T. K., Clarkson, B. D., &

Melamed, M. R. (1980). Discrimination of human leukemia subtypes by flow cytometric analysis of cellular DNA and RNA. Blood, 55(2), 282-293.

Page 171: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

152

Arch, R. H., Gedrich, R. W., & Thompson, C. B. (1998). Tumor necrosis factor receptor-associated factors (TRAFs)--a family of adapter proteins that regulates life and death. Genes Dev, 12(18), 2821-2830.

Arechiga, A. F., Habib, T., He, Y., Zhang, X., Zhang, Z. Y., Funk, A., & Buckner, J. H. (2009). Cutting edge: the PTPN22 allelic variant associated with autoimmunity impairs B cell signaling. J Immunol, 182(6), 3343-3347. doi: 10.4049/jimmunol.0713370

Au-Yeung, B. B., Deindl, S., Hsu, L. Y., Palacios, E. H., Levin, S. E., Kuriyan,

J., & Weiss, A. (2009). The structure, regulation, and function of ZAP-70. Immunol Rev, 228(1), 41-57. doi: 10.1111/j.1600-065X.2008.00753.x

Azzam, H. S., DeJarnette, J. B., Huang, K., Emmons, R., Park, C. S.,

Sommers, C. L., . . . Love, P. E. (2001). Fine tuning of TCR signaling by CD5. J Immunol, 166(9), 5464-5472.

Ballow, M. (2002). Primary immunodeficiency disorders: antibody deficiency.

J Allergy Clin Immunol, 109(4), 581-591. Basten, A., & Silveira, P. A. (2010). B-cell tolerance: mechanisms and

implications. Curr Opin Immunol, 22(5), 566-574. doi: 10.1016/j.coi.2010.08.001

Bendelac, A., Bonneville, M., & Kearney, J. F. (2001). Autoreactivity by

design: innate B and T lymphocytes. Nat Rev Immunol, 1(3), 177-186. doi: 10.1038/35105052

Bennett, J. H. (1845). Case of hypertrophy of the spleen and liver, in which

death took place from suppuration of the blood. Edinburgh Medical and Surgical Journal, 64, 413-423.

Bennett, J. H. (1851). On leukocythemia, or blood containing an unusual

number of colourless corpuscles. Monthly Journal of Medical Sciences, 12, 17-38.

Bentz, M., Huck, K., du Manoir, S., Joos, S., Werner, C. A., Fischer, K., . . .

Lichter, P. (1995). Comparative genomic hybridization in chronic B-cell leukemias shows a high incidence of chromosomal gains and losses. Blood, 85(12), 3610-3618.

Berland, R., & Wortis, H. H. (2002). Origins and functions of B-1 cells with

notes on the role of CD5. Annu Rev Immunol, 20, 253-300. doi: 10.1146/annurev.immunol.20.100301.064833

Berland, R., & Wortis, H. H. (2003). Normal B-1a cell development requires B

Page 172: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

153

cell-intrinsic NFATc1 activity. Proc Natl Acad Sci U S A, 100(23), 13459-13464. doi: 10.1073/pnas.2233620100

Bernal, A., Pastore, R. D., Asgary, Z., Keller, S. A., Cesarman, E., Liou, H.

C., & Schattner, E. J. (2001). Survival of leukemic B cells promoted by engagement of the antigen receptor. Blood, 98(10), 3050-3057.

Bertilaccio, M. T., Scielzo, C., Simonetti, G., Ponzoni, M., Apollonio, B., Fazi,

C., . . . Ghia, P. (2010). A novel Rag2-/-gammac-/--xenograft model of human CLL. Blood, 115(8), 1605-1609. doi: 10.1182/blood-2009-05-223586

Bertilaccio, M. T., Simonetti, G., Dagklis, A., Rocchi, M., Rodriguez, T. V.,

Apollonio, B., . . . Muzio, M. (2011). Lack of TIR8/SIGIRR triggers progression of chronic lymphocytic leukemia in mouse models. Blood, 118(3), 660-669. doi: 10.1182/blood-2011-01-329870

Best, C. G., Kemp, J. D., & J., W. T. (1995). Murine B-cell subsets defined by

CD23. Methods, 8(1), 3-10. Bhattacharya, N., Diener, S., Idler, I. S., Rauen, J., Häbe, S., Busch, H., . . .

Mertens, D. (2011). Nurse-like cells show deregulated expression of genes involved in immunocompetence. Br J Haematol, 154(3), 349-356. doi: 10.1111/j.1365-2141.2011.08747.x

Bichi, R., Shinton, S. A., Martin, E. S., Koval, A., Calin, G. A., Cesari, R., . . .

Croce, C. M. (2002). Human chronic lymphocytic leukemia modeled in mouse by targeted TCL1 expression. Proc Natl Acad Sci U S A, 99(10), 6955-6960. doi: 99/10/6955 [pii]10.1073/pnas.102181599

Bikah, G., Carey, J., Ciallella, J. R., Tarakhovsky, A., & Bondada, S. (1996).

CD5-mediated negative regulation of antigen receptor-induced growth signals in B-1 B cells. Science, 274(5294), 1906-1909.

Billard, C. (2012). Design of novel BH3 mimetics for the treatment of chronic

lymphocytic leukemia. Leukemia, 26(9), 2032-2038. doi: 10.1038/leu.2012.88

Binet, J. L., Auquier, A., Dighiero, G., Chastang, C., Piguet, H., Goasguen, J.,

. . . Gremy, F. (1981). A new prognostic classification of chronic lymphocytic leukemia derived from a multivariate survival analysis. Cancer, 48(1), 198-206.

Blair, A., Purdue, M. P., Weisenburger, D. D., & Baris, D. (2007). Chemical

exposures and risk of chronic lymphocytic leukaemia. Br J Haematol, 139(5), 753-761. doi: 10.1111/j.1365-2141.2007.06874.x

Page 173: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

154

Boggs, D. R., Sofferman, S. A., Wintrobe, M. M., & Cartwright, G. E. (1966).

Factors influencing the duration of survival of patients with chronic lymphocytic leukemia. Am J Med, 40(2), 243-254.

Bonci, D., Coppola, V., Musumeci, M., Addario, A., Giuffrida, R., Memeo, L., . . . De Maria, R. (2008). The miR-15a-miR-16-1 cluster controls prostate cancer by targeting multiple oncogenic activities. Nat Med, 14(11), 1271-1277. doi: 10.1038/nm.1880

Bottini, N., Vang, T., Cucca, F., & Mustelin, T. (2006). Role of PTPN22 in

type 1 diabetes and other autoimmune diseases. Semin Immunol, 18(4), 207-213. doi: 10.1016/j.smim.2006.03.008

Bouchard, B., Ormandy, C. J., Di Santo, J. P., & Kelly, P. A. (1999). Immune

system development and function in prolactin receptor-deficient mice. J Immunol, 163(2), 576-582.

Bradley, J. R., & Pober, J. S. (2001). Tumor necrosis factor receptor-

associated factors (TRAFs). Oncogene, 20(44), 6482-6491. doi: 10.1038/sj.onc.1204788

Brusa, D., Serra, S., Coscia, M., Rossi, D., D'Arena, G., Laurenti, L., . . .

Deaglio, S. (2013). The PD-1/PD-L1 axis contributes to T cell dysfunction in chronic lymphocytic leukemia. Haematologica. doi: 0.3324/haematol.2012.077537

Bröker, B. M., Klajman, A., Youinou, P., Jouquan, J., Worman, C. P., Murphy,

J., . . . Collins, P. (1988). Chronic lymphocytic leukemic (CLL) cells secrete multispecific autoantibodies. J Autoimmun, 1(5), 469-481.

Buggins, A. G., & Pepper, C. J. (2010). The role of Bcl-2 family proteins in

chronic lymphocytic leukaemia. Leuk Res, 34(7), 837-842. doi: 10.1016/j.leukres.2010.03.011

Burger, J. A., Tsukada, N., Burger, M., Zvaifler, N. J., Dell'Aquila, M., &

Kipps, T. J. (2000). Blood-derived nurse-like cells protect chronic lymphocytic leukemia B cells from spontaneous apoptosis through stromal cell-derived factor-1. Blood, 96(8), 2655-2663.

Cai, G., & Freeman, G. J. (2009). The CD160, BTLA, LIGHT/HVEM pathway:

a bidirectional switch regulating T-cell activation. Immunol Rev, 229(1), 244-258. doi: 10.1111/j.1600-065X.2009.00783.x

Caligaris-Cappio, F. (1996). B-chronic lymphocytic leukemia: a malignancy of

anti-self B cells. Blood, 87(7), 2615-2620.

Page 174: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

155

Caligaris-Cappio, F., Bertero, M. T., & Bergui, L. (1994). Autoimmunity, autoimmune diseases and lymphoproliferative disorders. Haematologica, 79(6), 487-492.

Caligaris-Cappio, F., Gobbi, M., Bofill, M., & Janossy, G. (1982). Infrequent normal B lymphocytes express features of B-chronic lymphocytic leukemia. J Exp Med, 155(2), 623-628.

Caligaris-Cappio, F., & Hamblin, T. J. (1999). B-cell chronic lymphocytic

leukemia: a bird of a different feather. J Clin Oncol, 17(1), 399-408. Calin, G. A., Dumitru, C. D., Shimizu, M., Bichi, R., Zupo, S., Noch, E., . . .

Croce, C. M. (2002). Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A, 99(24), 15524-15529. doi: 10.1073/pnas.242606799

Campo, E., Swerdlow, S. H., Harris, N. L., Pileri, S., Stein, H., & Jaffe, E. S.

(2011). The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications. Blood, 117(19), 5019-5032. doi: 10.1182/blood-2011-01-293050

Carsetti, R., Rosado, M. M., & Wardmann, H. (2004). Peripheral development

of B cells in mouse and man. Immunol Rev, 197, 179-191. Catera, R., Silverman, G. J., Hatzi, K., Seiler, T., Didier, S., Zhang, L., . . .

Chiorazzi, N. (2008). Chronic lymphocytic leukemia cells recognize conserved epitopes associated with apoptosis and oxidation. Mol Med, 14(11-12), 665-674. doi: 10.2119/2008-00102.Catera

Catovsky, D., Richards, S., Matutes, E., Oscier, D., Dyer, M. J., Bezares, R.

F., . . . Group, N. C. L. L. W. (2007). Assessment of fludarabine plus cyclophosphamide for patients with chronic lymphocytic leukaemia (the LRF CLL4 Trial): a randomised controlled trial. Lancet, 370(9583), 230-239. doi: 10.1016/S0140-6736(07)61125-8

Cerutti, A., Cols, M., & Puga, I. (2013). Marginal zone B cells: virtues of

innate-like antibody-producing lymphocytes. Nat Rev Immunol, 13(2), 118-132. doi: 10.1038/nri3383

Chanan-Khan, A., Miller, K. C., Musial, L., Lawrence, D., Padmanabhan, S.,

Takeshita, K., . . . Czuczman, M. S. (2006). Clinical efficacy of lenalidomide in patients with relapsed or refractory chronic lymphocytic leukemia: results of a phase II study. J Clin Oncol, 24(34), 5343-5349. doi: 10.1200/JCO.2005.05.0401

Chen, S. S., Claus, R., Lucas, D. M., Yu, L., Qian, J., Ruppert, A. S., . . .

Page 175: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

156

Byrd, J. C. (2011). Silencing of the inhibitor of DNA binding protein 4 (ID4) contributes to the pathogenesis of mouse and human CLL. Blood, 117(3), 862-871. doi: 10.1182/blood-2010-05-284638

Chen, S. S., Raval, A., Johnson, A. J., Hertlein, E., Liu, T. H., Jin, V. X., . . . Plass, C. (2009). Epigenetic changes during disease progression in a murine model of human chronic lymphocytic leukemia. Proc Natl Acad Sci U S A, 106(32), 13433-13438. doi: 10.1073/pnas.0906455106

Chen, W., Ford, M. S., Young, K. J., & Zhang, L. (2004). The role and

mechanisms of double negative regulatory T cells in the suppression of immune responses. Cell Mol Immunol, 1(5), 328-335.

Cheson, B. D. (2010). Monoclonal antibody therapy of chronic lymphocytic

leukaemia. Best Pract Res Clin Haematol, 23(1), 133-143. doi: 10.1016/j.beha.2010.01.006

Chiorazzi, N., & Ferrarini, M. (2003). B cell chronic lymphocytic leukemia:

lessons learned from studies of the B cell antigen receptor. Annu Rev Immunol, 21, 841-894. doi: 10.1146/annurev.immunol.21.120601.141018

Chiorazzi, N., & Ferrarini, M. (2011). Cellular origin(s) of chronic lymphocytic

leukemia: cautionary notes and additional considerations and possibilities. Blood, 117(6), 1781-1791. doi: 10.1182/blood-2010-07-155663

Chiorazzi, N., Fu, S. M., Montazeri, G., Kunkel, H. G., Rai, K., & Gee, T.

(1979). T cell helper defect in patients with chronic lymphocytic leukemia. J Immunol, 122(3), 1087-1090.

Chu, C. C., Catera, R., Zhang, L., Didier, S., Agagnina, B. M., Damle, R. N., .

. . Chiorazzi, N. (2010). Many chronic lymphocytic leukemia antibodies recognize apoptotic cells with exposed nonmuscle myosin heavy chain IIA: implications for patient outcome and cell of origin. Blood, 115(19), 3907-3915. doi: 10.1182/blood-2009-09-244251

Chung, J. B., Silverman, M., & Monroe, J. G. (2003). Transitional B cells: step

by step towards immune competence. Trends Immunol, 24(6), 343-349.

Chung, S. A., & Criswell, L. A. (2007). PTPN22: its role in SLE and

autoimmunity. Autoimmunity, 40(8), 582-590. doi: 10.1080/08916930701510848

Cimmino, A., Calin, G. A., Fabbri, M., Iorio, M. V., Ferracin, M., Shimizu, M., .

. . Croce, C. M. (2005). miR-15 and miR-16 induce apoptosis by

Page 176: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

157

targeting BCL2. Proc Natl Acad Sci U S A, 102(39), 13944-13949. doi: 10.1073/pnas.0506654102

Clevenger, C. V., Freier, D. O., & Kline, J. B. (1998). Prolactin receptor signal

transduction in cells of the immune system. J Endocrinol, 157(2), 187-197.

Collins, R. J., Verschuer, L. A., Harmon, B. V., Prentice, R. L., Pope, J. H., &

Kerr, J. F. (1989). Spontaneous programmed death (apoptosis) of B-chronic lymphocytic leukaemia cells following their culture in vitro. Br J Haematol, 71(3), 343-350.

Colovic, N., Bogdanovic, A., Martinovic-Cemerikic, V., & Jankovic, G. (2001).

Prognostic significance of serum immunoglobulins in B-chronic lymphocytic leukemia. Archive of Oncology, 9(2), 79-82.

Cooper, M. D. (1987). Current concepts. B lymphocytes. Normal

development and function. N Engl J Med, 317(23), 1452-1456. doi: 10.1056/NEJM198712033172306

Crespo, M., Bosch, F., Villamor, N., Bellosillo, B., Colomer, D., Rozman, M., .

. . Montserrat, E. (2003). ZAP-70 expression as a surrogate for immunoglobulin-variable-region mutations in chronic lymphocytic leukemia. N Engl J Med, 348(18), 1764-1775. doi: 10.1056/NEJMoa023143

Crispín, J. C., Oukka, M., Bayliss, G., Cohen, R. A., Van Beek, C. A.,

Stillman, I. E., . . . Tsokos, G. C. (2008). Expanded double negative T cells in patients with systemic lupus erythematosus produce IL-17 and infiltrate the kidneys. J Immunol, 181(12), 8761-8766.

Cro, L., Morabito, F., Zucal, N., Fabris, S., Lionetti, M., Cutrona, G., . . .

Baldini, L. (2009). CD26 expression in mature B-cell neoplasia: its possible role as a new prognostic marker in B-CLL. Hematol Oncol, 27(3), 140-147. doi: 10.1002/hon.888

Crowther-Swanepoel, D., & Houlston, R. S. (2009). The molecular basis of

familial chronic lymphocytic leukemia. Haematologica, 94(5), 606-609. doi: 10.3324/haematol.2009.006296

Czarneski, J., Lin, Y. C., Chong, S., McCarthy, B., Fernandes, H., Parker, G.,

. . . Raveche, E. (2004). Studies in NZB IL-10 knockout mice of the requirement of IL-10 for progression of B-cell lymphoma. Leukemia, 18(3), 597-606. doi: 10.1038/sj.leu.2403244

D'Arena, G., Rossi, G., Vannata, B., Deaglio, S., Mansueto, G., D'Auria, F., . .

Page 177: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

158

. Musto, P. (2012). Regulatory T-Cells in Chronic Lymphocytic Leukemia and Autoimmune Diseases. Mediterr J Hematol Infect Dis, 4(1), e2012053. doi: 10.4084/MJHID.2012.053

Dai, X., James, R. G., Habib, T., Singh, S., Jackson, S., Khim, S., . . .

Rawlings, D. J. (2013). A disease-associated PTPN22 variant promotes systemic autoimmunity in murine models. J Clin Invest, 123(5), 2024-2036. doi: 10.1172/JCI66963

Dameshek, W. (1967). Chronic lymphocytic leukemia--an accumulative

disease of immunologically incompetent lymphocytes. Blood, 29(4), Suppl:566-584.

Damle, R. N., Ghiotto, F., Valetto, A., Albesiano, E., Fais, F., Yan, X. J., . . .

Chiorazzi, N. (2002). B-cell chronic lymphocytic leukemia cells express a surface membrane phenotype of activated, antigen-experienced B lymphocytes. Blood, 99(11), 4087-4093.

Damle, R. N., Wasil, T., Fais, F., Ghiotto, F., Valetto, A., Allen, S. L., . . .

Chiorazzi, N. (1999). Ig V gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia. Blood, 94(6), 1840-1847.

Deaglio, S., Capobianco, A., Bergui, L., Dürig, J., Morabito, F., Dührsen, U.,

& Malavasi, F. (2003). CD38 is a signaling molecule in B-cell chronic lymphocytic leukemia cells. Blood, 102(6), 2146-2155. doi: 10.1182/blood-2003-03-0989

Dearden, C. (2008). Disease-specific complications of chronic lymphocytic

leukemia. Hematology Am Soc Hematol Educ Program, 450-456. doi: 10.1182/asheducation-2008.1.450

Descatoire, M., Weill, J. C., Reynaud, C. A., & Weller, S. (2011). A human

equivalent of mouse B-1 cells? J Exp Med, 208(13), 2563-2564; author reply 2566-2569. doi: 10.1084/jem.20112232

Di Bernardo, M. C., Crowther-Swanepoel, D., Broderick, P., Webb, E.,

Sellick, G., Wild, R., . . . Houlston, R. S. (2008). A genome-wide association study identifies six susceptibility loci for chronic lymphocytic leukemia. Nat Genet, 40(10), 1204-1210. doi: 10.1038/ng.219

Di Noia, J. M., & Neuberger, M. S. (2007). Molecular mechanisms of antibody

somatic hypermutation. Annu Rev Biochem, 76, 1-22. doi: 10.1146/annurev.biochem.76.061705.090740

Page 178: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

159

Dighiero, G., & Hamblin, T. J. (2008). Chronic lymphocytic leukaemia. Lancet, 371(9617), 1017-1029. doi: 10.1016/S0140-6736(08)60456-0

Dighiero, G., Maloum, K., Desablens, B., Cazin, B., Navarro, M., Leblay, R., .

. . Travade, P. (1998). Chlorambucil in indolent chronic lymphocytic leukemia. French Cooperative Group on Chronic Lymphocytic Leukemia. N Engl J Med, 338(21), 1506-1514. doi: 10.1056/NEJM199805213382104

DiLillo, D. J., Matsushita, T., & Tedder, T. F. (2010). B10 cells and regulatory B cells balance immune responses during inflammation, autoimmunity, and cancer. Ann N Y Acad Sci, 1183, 38-57. doi: 10.1111/j.1749-6632.2009.05137.x

DiLillo, D. J., Weinberg, J. B., Yoshizaki, A., Horikawa, M., Bryant, J. M.,

Iwata, Y., . . . Tedder, T. F. (2013). Chronic lymphocytic leukemia and regulatory B cells share IL-10 competence and immunosuppressive function. Leukemia, 27(1), 170-182. doi: 10.1038/leu.2012.165

Ding, C., Liu, Y., Wang, Y., Park, B. K., Wang, C. Y., & Zheng, P. (2007).

Siglecg limits the size of B1a B cell lineage by down-regulating NFkappaB activation. PLoS One, 2(10), e997. doi: 10.1371/journal.pone.0000997

Dono, M., Cerruti, G., & Zupo, S. (2004). The CD5+ B-cell. Int J Biochem Cell

Biol, 36(11), 2105-2111. doi: 10.1016/j.biocel.2004.05.017 Dores, G. M., Anderson, W. F., Curtis, R. E., Landgren, O., Ostroumova, E.,

Bluhm, E. C., . . . Linet, M. S. (2007). Chronic lymphocytic leukaemia and small lymphocytic lymphoma: overview of the descriptive epidemiology. Br J Haematol, 139(5), 809-819. doi: 10.1111/j.1365-2141.2007.06856.x

Dorshkind, K., & Horseman, N. D. (2000). The roles of prolactin, growth

hormone, insulin-like growth factor-I, and thyroid hormones in lymphocyte development and function: insights from genetic models of hormone and hormone receptor deficiency. Endocr Rev, 21(3), 292-312.

Döhner, H., Fischer, K., Bentz, M., Hansen, K., Benner, A., Cabot, G., . . .

Stilgenbauer, S. (1995). p53 gene deletion predicts for poor survival and non-response to therapy with purine analogs in chronic B-cell leukemias. Blood, 85(6), 1580-1589.

Döhner, H., Stilgenbauer, S., Benner, A., Leupolt, E., Kröber, A., Bullinger, L.,

. . . Lichter, P. (2000). Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med, 343(26), 1910-1916. doi:

Page 179: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

160

10.1056/NEJM200012283432602 Döhner, H., Stilgenbauer, S., James, M. R., Benner, A., Weilguni, T., Bentz,

M., . . . Lichter, P. (1997). 11q deletions identify a new subset of B-cell chronic lymphocytic leukemia characterized by extensive nodal involvement and inferior prognosis. Blood, 89(7), 2516-2522.

Dühren-von Minden, M., Übelhart, R., Schneider, D., Wossning, T., Bach, M.

P., Buchner, M., . . . Jumaa, H. (2012). Chronic lymphocytic leukaemia is driven by antigen-independent cell-autonomous signalling. Nature, 489(7415), 309-312. doi: 10.1038/nature11309

Dürig, J., Nückel, H., Cremer, M., Führer, A., Halfmeyer, K., Fandrey, J., . . . Dührsen, U. (2003). ZAP-70 expression is a prognostic factor in chronic lymphocytic leukemia. Leukemia, 17(12), 2426-2434. doi: 10.1038/sj.leu.2403147

Early, P., Huang, H., Davis, M., Calame, K., & Hood, L. (1980). An

immunoglobulin heavy chain variable region gene is generated from three segments of DNA: VH, D and JH. Cell, 19(4), 981-992.

Ehrlich, P. (1891). Farbenanalytische Untersuchungen Zur Histologie und

Klinik Des Blutes. Hirschwald, Berlin. Endo, T., Nishio, M., Enzler, T., Cottam, H. B., Fukuda, T., James, D. F., . . .

Kipps, T. J. (2007). BAFF and APRIL support chronic lymphocytic leukemia B-cell survival through activation of the canonical NF-kappaB pathway. Blood, 109(2), 703-710. doi: 10.1182/blood-2006-06-027755

Enjalbert, A., Sladeczek, F., Guillon, G., Bertrand, P., Shu, C., Epelbaum, J.,

. . . Kordon, C. (1986). Angiotensin II and dopamine modulate both cAMP and inositol phosphate productions in anterior pituitary cells. Involvement in prolactin secretion. J Biol Chem, 261(9), 4071-4075.

Enno, A., Catovsky, D., O'Brien, M., Cherchi, M., Kumaran, T. O., & Galton,

D. A. (1979). 'Prolymphocytoid' transformation of chronic lymphocytic leukaemia. Br J Haematol, 41(1), 9-18.

Enzler, T., Kater, A. P., Zhang, W., Widhopf, G. F., Chuang, H. Y., Lee, J., . .

. Kipps, T. J. (2009). Chronic lymphocytic leukemia of Emu-TCL1 transgenic mice undergoes rapid cell turnover that can be offset by extrinsic CD257 to accelerate disease progression. Blood, 114(20), 4469-4476. doi: 10.1182/blood-2009-06-230169

Fais, F., Ghiotto, F., Hashimoto, S., Sellars, B., Valetto, A., Allen, S. L., . . .

Chiorazzi, N. (1998). Chronic lymphocytic leukemia B cells express restricted sets of mutated and unmutated antigen receptors. J Clin

Page 180: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

161

Invest, 102(8), 1515-1525. doi: 10.1172/JCI3009 Fais, F., Morabito, F., Stelitano, C., Callea, V., Zanardi, S., Scudeletti, M., . . .

Grossi, C. E. (2004). CD1d is expressed on B-chronic lymphocytic leukemia cells and mediates alpha-galactosylceramide presentation to natural killer T lymphocytes. Int J Cancer, 109(3), 402-411. doi: 10.1002/ijc.11723

Farace, F., Orlanducci, F., Dietrich, P. Y., Gaudin, C., Angevin, E., Courtier,

M. H., . . . Triebel, F. (1994). T cell repertoire in patients with B chronic lymphocytic leukemia. Evidence for multiple in vivo T cell clonal expansions. J Immunol, 153(9), 4281-4290.

Fazi, C., Scarfò, L., Pecciarini, L., Cottini, F., Dagklis, A., Janus, A., . . . Ghia,

P. (2011). General population low-count CLL-like MBL persists over time without clinical progression, although carrying the same cytogenetic abnormalities of CLL. Blood, 118(25), 6618-6625. doi: 10.1182/blood-2011-05-357251

Fecteau, J. F., & Kipps, T. J. (2012). Structure and function of the

hematopoietic cancer niche: focus on chronic lymphocytic leukemia. Front Biosci (Schol Ed), 4, 61-73.

Ferrajoli, A., Lee, B. N., Schlette, E. J., O'Brien, S. M., Gao, H., Wen, S., . . .

Keating, M. J. (2008). Lenalidomide induces complete and partial remissions in patients with relapsed and refractory chronic lymphocytic leukemia. Blood, 111(11), 5291-5297. doi: 10.1182/blood-2007-12-130120

Finch, S. C., Hoshino, T., Itoga, T., Ichimaru, M., & Ingram, R. H. (1969).

Chronic lymphocytic leukemia in Hiroshima and Nagasaki, Japan. Blood, 33(1), 79-86.

Ford, M. S., Zhang, Z. X., Chen, W., & Zhang, L. (2006). Double-negative T

regulatory cells can develop outside the thymus and do not mature from CD8+ T cell precursors. J Immunol, 177(5), 2803-2809.

Foucar, K., & Rydell, R. E. (1980). Richter's syndrome in chronic lymphocytic

leukemia. Cancer, 46(1), 118-134. Freeman, M. E., Kanyicska, B., Lerant, A., & Nagy, G. (2000). Prolactin:

structure, function, and regulation of secretion. Physiol Rev, 80(4), 1523-1631.

Galton, D. A. (1966). The pathogenesis of chronic lymphocytic leukemia. Can

Med Assoc J, 94(19), 1005-1010.

Page 181: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

162

Galton, D. A., Wiltshaw, E., Szur, L., & Dacie, J. V. (1961). The use of

chlorambucil and steroids in the treatment of chronic lymphocytic leukaemia. Br J Haematol, 7, 73-98.

Gary-Gouy, H., Harriague, J., Bismuth, G., Platzer, C., Schmitt, C., & Dalloul,

A. H. (2002). Human CD5 promotes B-cell survival through stimulation of autocrine IL-10 production. Blood, 100(13), 4537-4543. doi: 10.1182/blood-2002-05-1525

Garzon, R., Calin, G. A., & Croce, C. M. (2009). MicroRNAs in Cancer. Annu

Rev Med, 60, 167-179. doi: 10.1146/annurev.med.59.053006.104707 Gaudio, E., Spizzo, R., Paduano, F., Luo, Z., Efanov, A., Palamarchuk, A., . .

. Croce, C. M. (2012). Tcl1 interacts with Atm and enhances NF-κB activation in hematologic malignancies. Blood, 119(1), 180-187. doi: 10.1182/blood-2011-08-374561

Gellert, M. (2002). V(D)J recombination: RAG proteins, repair factors, and

regulation. Annu Rev Biochem, 71, 101-132. doi: 10.1146/annurev.biochem.71.090501.150203

Gerstein, R. M., & Lieber, M. R. (1993). Extent to which homology can

constrain coding exon junctional diversity in V(D)J recombination. Nature, 363(6430), 625-627. doi: 10.1038/363625a0

Ghani, A. M., Krause, J. R., & Brody, J. P. (1986). Prolymphocytic

transformation of chronic lymphocytic leukemia. A report of three cases and review of the literature. Cancer, 57(1), 75-80.

Ghia, E. M., Jain, S., Widhopf, G. F., Rassenti, L. Z., Keating, M. J., Wierda,

W. G., . . . Kipps, T. J. (2008). Use of IGHV3-21 in chronic lymphocytic leukemia is associated with high-risk disease and reflects antigen-driven, post-germinal center leukemogenic selection. Blood, 111(10), 5101-5108. doi: 10.1182/blood-2007-12-130229

Ghia, P., Strola, G., Granziero, L., Geuna, M., Guida, G., Sallusto, F., . . .

Caligaris-Cappio, F. (2002). Chronic lymphocytic leukemia B cells are endowed with the capacity to attract CD4+, CD40L+ T cells by producing CCL22. Eur J Immunol, 32(5), 1403-1413. doi: 10.1002/1521-4141(200205)32:5<1403::AID-IMMU1403>3.0.CO;2-Y

Ghia, P., ten Boekel, E., Rolink, A. G., & Melchers, F. (1998). B-cell

development: a comparison between mouse and man. Immunol Today, 19(10), 480-485.

Page 182: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

163

Ghiotto, F., Fais, F., Valetto, A., Albesiano, E., Hashimoto, S., Dono, M., . . . Chiorazzi, N. (2004). Remarkably similar antigen receptors among a subset of patients with chronic lymphocytic leukemia. J Clin Invest, 113(7), 1008-1016. doi: 10.1172/JCI19399

Ghosn, E. E., Yamamoto, R., Hamanaka, S., Yang, Y., Herzenberg, L. A., &

Nakauchi, H. (2012). Distinct B-cell lineage commitment distinguishes adult bone marrow hematopoietic stem cells. Proc Natl Acad Sci U S A, 109(14), 5394-5398. doi: 10.1073/pnas.1121632109

Gilling, C. E., Mittal, A. K., Chaturvedi, N. K., Iqbal, J., Aoun, P., Bierman, P.

J., . . . Joshi, S. S. (2012). Lymph node-induced immune tolerance in chronic lymphocytic leukaemia: a role for caveolin-1. Br J Haematol, 158(2), 216-231. doi: 10.1111/j.1365-2141.2012.09148.x

Girschick, H. J., Grammer, A. C., Nanki, T., Mayo, M., & Lipsky, P. E. (2001).

RAG1 and RAG2 expression by B cell subsets from human tonsil and peripheral blood. J Immunol, 166(1), 377-386.

Glanville, J., Zhai, W., Berka, J., Telman, D., Huerta, G., Mehta, G. R., . . .

Pons, J. (2009). Precise determination of the diversity of a combinatorial antibody library gives insight into the human immunoglobulin repertoire. Proc Natl Acad Sci U S A, 106(48), 20216-20221. doi: 10.1073/pnas.0909775106

Nadgornaya, V., Zavelevich, M., & Machilo, V. (2006). Patterns of

hematological malignancies in Chernobyl clean-up workers (1996-2005). Exp Oncol, 28(1), 60-63.

Godfrey, D. I., & Kronenberg, M. (2004). Going both ways: immune regulation

via CD1d-dependent NKT cells. J Clin Invest, 114(10), 1379-1388. doi: 10.1172/JCI23594

Godfrey, D. I., Stankovic, S., & Baxter, A. G. (2010). Raising the NKT cell

family. Nat Immunol, 11(3), 197-206. doi: 10.1038/ni.1841 Goffin, V., Shiverick, K. T., Kelly, P. A., & Martial, J. A. (1996). Sequence-

function relationships within the expanding family of prolactin, growth hormone, placental lactogen, and related proteins in mammals. Endocr Rev, 17(4), 385-410.

Goodnow, C. C. (1997). Balancing immunity, autoimmunity, and self-

tolerance. Ann N Y Acad Sci, 815, 55-66. Goodnow, C. C. (2001). Pathways for self-tolerance and the treatment of

autoimmune diseases. Lancet, 357(9274), 2115-2121.

Page 183: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

164

Goodnow, C. C., Sprent, J., Fazekas de St Groth, B., & Vinuesa, C. G.

(2005). Cellular and genetic mechanisms of self tolerance and autoimmunity. Nature, 435(7042), 590-597. doi: 10.1038/nature03724

Gorgun, G., Ramsay, A. G., Holderried, T. A., Zahrieh, D., Le Dieu, R., Liu,

F., . . . Gribben, J. G. (2009). E(mu)-TCL1 mice represent a model for immunotherapeutic reversal of chronic lymphocytic leukemia-induced T-cell dysfunction. Proc Natl Acad Sci U S A, 106(15), 6250-6255. doi: 10.1073/pnas.0901166106

Gribben, J. G., & O'Brien, S. (2011). Update on therapy of chronic

lymphocytic leukemia. J Clin Oncol, 29(5), 544-550. doi: 10.1200/JCO.2010.32.3865

Griffin, D. O., Holodick, N. E., & Rothstein, T. L. (2011). Human B1 cells in

umbilical cord and adult peripheral blood express the novel phenotype CD20+ CD27+ CD43+ CD70-. J Exp Med, 208(1), 67-80. doi: 10.1084/jem.20101499

Griffin, O. D., Quach, T., Batliwalla, F., Andreopoulos, D., Holodick, N. E., &

Rothstein, T. L. (2012). Human CD11b+ B1 cells are not monocytes: a reply to "gene profiling of CD11b+ and CD11b- B1 cell subsets reveals potential cell sorting artifacts". J Exp Med, 209(3), 434-436.

Guarini, A., Chiaretti, S., Tavolaro, S., Maggio, R., Peragine, N., Citarella, F.,

. . . Foà, R. (2008). BCR ligation induced by IgM stimulation results in gene expression and functional changes only in IgV H unmutated chronic lymphocytic leukemia (CLL) cells. Blood, 112(3), 782-792. doi: 10.1182/blood-2007-12-127688

Gumperz, J. E., Miyake, S., Yamamura, T., & Brenner, M. B. (2002).

Functionally distinct subsets of CD1d-restricted natural killer T cells revealed by CD1d tetramer staining. J Exp Med, 195(5), 625-636.

Gutiérrez, M. A., Molina, J. F., Jara, L. J., Cuéllar, M. L., García, C.,

Gutiérrez-Ureña, S., . . . Espinoza, L. R. (1995). Prolactin and systemic lupus erythematosus: prolactin secretion by SLE lymphocytes and proliferative (autocrine) activity. Lupus, 4(5), 348-352.

Görgün, G., Holderried, T. A., Zahrieh, D., Neuberg, D., & Gribben, J. G.

(2005). Chronic lymphocytic leukemia cells induce changes in gene expression of CD4 and CD8 T cells. J Clin Invest, 115(7), 1797-1805. doi: 10.1172/JCI24176

Page 184: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

165

Hahne, M., Kataoka, T., Schröter, M., Hofmann, K., Irmler, M., Bodmer, J. L., . . . Tschopp, J. (1998). APRIL, a new ligand of the tumor necrosis factor family, stimulates tumor cell growth. J Exp Med, 188(6), 1185-1190.

Hallek, M. (2010). Therapy of chronic lymphocytic leukaemia. Best Pract Res

Clin Haematol, 23(1), 85-96. doi: 10.1016/j.beha.2009.12.002 Hallek, M., Cheson, B. D., Catovsky, D., Caligaris-Cappio, F., Dighiero, G.,

Döhner, H., . . . Leukemia, I. W. o. C. L. (2008). Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines. Blood, 111(12), 5446-5456. doi: 10.1182/blood-2007-06-093906

Hamano, Y., Hirose, S., Ida, A., Abe, M., Zhang, D., Kodera, S., . . . Shirai, T.

(1998). Susceptibility alleles for aberrant B-1 cell proliferation involved in spontaneously occurring B-cell chronic lymphocytic leukemia in a model of New Zealand white mice. Blood, 92(10), 3772-3779.

Hamblin, T. (2000). Historical aspects of chronic lymphocytic leukaemia. Br J

Haematol, 111(4), 1023-1034. Hamblin, T. (2006). Is chronic lymphocytic leukemia a response to infectious

agents? Leuk Res, 30(9), 1063-1064. doi: 10.1016/j.leukres.2005.11.022

Hamblin, T. J., Davis, Z., Gardiner, A., Oscier, D. G., & Stevenson, F. K.

(1999). Unmutated Ig V(H) genes are associated with a more aggressive form of chronic lymphocytic leukemia. Blood, 94(6), 1848-1854.

Hamblin, T. J., Orchard, J. A., Ibbotson, R. E., Davis, Z., Thomas, P. W.,

Stevenson, F. K., & Oscier, D. G. (2002). CD38 expression and immunoglobulin variable region mutations are independent prognostic variables in chronic lymphocytic leukemia, but CD38 expression may vary during the course of the disease. Blood, 99(3), 1023-1029.

Han, Y., Wu, J., Bi, L., Xiong, S., Gao, S., Yin, L., . . . Zhang, S. (2011).

Malignant B cells induce the conversion of CD4+CD25- T cells to regulatory T cells in B-cell non-Hodgkin lymphoma. PLoS One, 6(12), e28649. doi: 10.1371/journal.pone.0028649

Han, Y. C., Park, C. Y., Bhagat, G., Zhang, J., Wang, Y., Fan, J. B., . . . Gu,

H. (2010). microRNA-29a induces aberrant self-renewal capacity in

Page 185: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

166

hematopoietic progenitors, biased myeloid development, and acute myeloid leukemia. J Exp Med, 207(3), 475-489. doi: 10.1084/jem.20090831

Hanada, M., Delia, D., Aiello, A., Stadtmauer, E., & Reed, J. C. (1993). bcl-2

gene hypomethylation and high-level expression in B-cell chronic lymphocytic leukemia. Blood, 82(6), 1820-1828.

Hardy, R. R. (2006). B-1 B cell development. J Immunol, 177(5), 2749-2754. Hardy, R. R., & Hayakawa, K. (2001). B cell development pathways. Annu

Rev Immunol, 19, 595-621. doi: 10.1146/annurev.immunol.19.1.595 Hardy, R. R., Kincade, P. W., & Dorshkind, K. (2007). The protean nature of

cells in the B lymphocyte lineage. Immunity, 26(6), 703-714. doi: 10.1016/j.immuni.2007.05.013

Harris, N. L., Jaffe, E. S., Diebold, J., Flandrin, G., Muller-Hermelink, H. K.,

Vardiman, J., . . . Bloomfield, C. D. (1999). World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues: report of the Clinical Advisory Committee meeting-Airlie House, Virginia, November 1997. J Clin Oncol, 17(12), 3835-3849.

Hartkamp, A., Mulder, A. H., Rijkers, G. T., van Velzen-Blad, H., & Biesma,

D. H. (2001). Antibody responses to pneumococcal and haemophilus vaccinations in patients with B-cell chronic lymphocytic leukaemia. Vaccine, 19(13-14), 1671-1677.

Haslinger, C., Schweifer, N., Stilgenbauer, S., Döhner, H., Lichter, P., Kraut,

N., . . . Abseher, R. (2004). Microarray gene expression profiling of B-cell chronic lymphocytic leukemia subgroups defined by genomic aberrations and VH mutation status. J Clin Oncol, 22(19), 3937-3949. doi: 10.1200/JCO.2004.12.133

Hassaballa, A. E., Palmer, V. L., Anderson, D. K., Kassmeier, M. D., Nganga,

V. K., Parks, K. W., . . . Swanson, P. C. (2011). Accumulation of B1-like B cells in transgenic mice over-expressing catalytically inactive RAG1 in the periphery. Immunology, 134(4), 469-486. doi: 10.1111/j.1365-2567.2011.03509.x

Hayakawa, K., Asano, M., Shinton, S. A., Gui, M., Wen, L. J., Dashoff, J., &

Hardy, R. R. (2003). Positive selection of anti-thy-1 autoreactive B-1 cells and natural serum autoantibody production independent from bone marrow B cell development. J Exp Med, 197(1), 87-99.

Hayakawa, K., Hardy, R. R., Parks, D. R., & Herzenberg, L. A. (1983). The

Page 186: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

167

"Ly-1 B" cell subpopulation in normal immunodefective, and autoimmune mice. J Exp Med, 157(1), 202-218.

Heintel, D., Kienle, D., Shehata, M., Kröber, A., Kroemer, E., Schwarzinger,

I., . . . Group, C. S. (2005). High expression of lipoprotein lipase in poor risk B-cell chronic lymphocytic leukemia. Leukemia, 19(7), 1216-1223. doi: 10.1038/sj.leu.2403748

Helyer, B. J., & Howie, J. B. (1963). Renal disease associated with positive

lupus erythematosus tests in a cross-bred strain of mice. Nature, 197, 197.

Heng, T. S., Painter, M. W., & Consortium, I. G. P. (2008). The

Immunological Genome Project: networks of gene expression in immune cells. Nat Immunol, 9(10), 1091-1094. doi: 10.1038/ni1008-1091

Herling, M., Patel, K. A., Hsi, E. D., Chang, K. C., Rassidakis, G. Z., Ford, R.,

& Jones, D. (2007). TCL1 in B-cell tumors retains its normal b-cell pattern of regulation and is a marker of differentiation stage. Am J Surg Pathol, 31(7), 1123-1129. doi: 10.1097/PAS.0b013e31802e2201

Herling, M., Patel, K. A., Khalili, J., Schlette, E., Kobayashi, R., Medeiros, L.

J., & Jones, D. (2006). TCL1 shows a regulated expression pattern in chronic lymphocytic leukemia that correlates with molecular subtypes and proliferative state. Leukemia, 20(2), 280-285. doi: 10.1038/sj.leu.2404017

Herman, S. E., & Johnson, A. J. (2012). Molecular pathways: targeting

phosphoinositide 3-kinase p110-delta in chronic lymphocytic leukemia. Clin Cancer Res, 18(15), 4013-4018. doi: 10.1158/1078-0432.CCR-11-1402

Herreros, B., Rodríguez-Pinilla, S. M., Pajares, R., Martínez-Gónzalez, M. A.,

Ramos, R., Munoz, I., . . . Piris, M. A. (2010). Proliferation centers in chronic lymphocytic leukemia: the niche where NF-kappaB activation takes place. Leukemia, 24(4), 872-876. doi: 10.1038/leu.2009.285

Herrinton, L. J., Goldoft, M., Schwartz, S. M., & Weiss, N. S. (1996). The

incidence of non-Hodgkin's lymphoma and its histologic subtypes in Asian migrants to the United States and their descendants. Cancer Causes Control, 7(2), 224-230.

Herrmann, F., Lochner, A., Philippen, H., Jauer, B., & Rühl, H. (1982).

Imbalance of T cell subpopulations in patients with chronic lymphocytic leukaemia of the B cell type. Clin Exp Immunol, 49(1), 157-162.

Page 187: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

168

Hervé, M., Xu, K., Ng, Y. S., Wardemann, H., Albesiano, E., Messmer, B. T.,

. . . Meffre, E. (2005). Unmutated and mutated chronic lymphocytic leukemias derive from self-reactive B cell precursors despite expressing different antibody reactivity. J Clin Invest, 115(6), 1636-1643. doi: 10.1172/JCI24387

Hiestand, P. C., Mekler, P., Nordmann, R., Grieder, A., & Permmongkol, C.

(1986). Prolactin as a modulator of lymphocyte responsiveness provides a possible mechanism of action for cyclosporine. Proc Natl Acad Sci U S A, 83(8), 2599-2603.

Hillion, S., Saraux, A., Youinou, P., & Jamin, C. (2005). Expression of RAGs

in peripheral B cells outside germinal centers is associated with the expression of CD5. J Immunol, 174(9), 5553-5561.

Hippen, K. L., Tze, L. E., & Behrens, T. W. (2000). CD5 maintains tolerance

in anergic B cells. J Exp Med, 191(5), 883-890. Hisada, M., Biggar, R. J., Greene, M. H., Fraumeni, J. F., & Travis, L. B.

(2001). Solid tumors after chronic lymphocytic leukemia. Blood, 98(6), 1979-1981.

Hitoshi, Y., Yamaguchi, N., Mita, S., Sonoda, E., Takaki, S., Tominaga, A., &

Takatsu, K. (1990). Distribution of IL-5 receptor-positive B cells. Expression of IL-5 receptor on Ly-1(CD5)+ B cells. J Immunol, 144(11), 4218-4225.

Hockenbery, D., Nuñez, G., Milliman, C., Schreiber, R. D., & Korsmeyer, S. J.

(1990). Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death. Nature, 348(6299), 334-336. doi: 10.1038/348334a0

Hodgkin. (1832). On some Morbid Appearances of the Absorbent Glands and

Spleen. Med Chir Trans, 17, 68-114. Hodgson, K., Ferrer, G., Montserrat, E., & Moreno, C. (2011). Chronic

lymphocytic leukemia and autoimmunity: a systematic review. Haematologica, 96(5), 752-761. doi: 10.3324/haematol.2010.036152

Hodgson, K., Ferrer, G., Pereira, A., Moreno, C., & Montserrat, E. (2011).

Autoimmune cytopenia in chronic lymphocytic leukaemia: diagnosis and treatment. Br J Haematol, 154(1), 14-22. doi: 10.1111/j.1365-2141.2011.08707.x

Hoellenriegel, J., Meadows, S. A., Sivina, M., Wierda, W. G., Kantarjian, H.,

Page 188: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

169

Keating, M. J., . . . Burger, J. A. (2011). The phosphoinositide 3'-kinase delta inhibitor, CAL-101, inhibits B-cell receptor signaling and chemokine networks in chronic lymphocytic leukemia. Blood, 118(13), 3603-3612. doi: 10.1182/blood-2011-05-352492

Hofbauer, J. P., Heyder, C., Denk, U., Kocher, T., Holler, C., Trapin, D., . . .

Egle, A. (2011). Development of CLL in the TCL1 transgenic mouse model is associated with severe skewing of the T-cell compartment homologous to human CLL. Leukemia, 25(9), 1452-1458. doi: 10.1038/leu.2011.111

Hoffmann, A., Kerr, S., Jellusova, J., Zhang, J., Weisel, F., Wellmann, U., . . .

Nitschke, L. (2007). Siglec-G is a B1 cell-inhibitory receptor that controls expansion and calcium signaling of the B1 cell population. Nat Immunol, 8(7), 695-704. doi: 10.1038/ni1480

Holler, C., Piñón, J. D., Denk, U., Heyder, C., Hofbauer, S., Greil, R., & Egle,

A. (2009). PKCbeta is essential for the development of chronic lymphocytic leukemia in the TCL1 transgenic mouse model: validation of PKCbeta as a therapeutic target in chronic lymphocytic leukemia. Blood, 113(12), 2791-2794. doi: 10.1182/blood-2008-06-160713

Howlader, N., Noone, A., Krapcho, M., Neyman, N., Aminou, R., Waldron,

W., . . . Cronin, K. (2012). SEER Cancer Statistics Review, 1975-2009 (Vintage 2009 Populations). Retrieved 9-4-12, 2012, from http://seer.cancer.gov/csr/1975_2009_pops09/

Hoyer, K. K., French, S. W., Turner, D. E., Nguyen, M. T., Renard, M.,

Malone, C. S., . . . Teitell, M. A. (2002). Dysregulated TCL1 promotes multiple classes of mature B cell lymphoma. Proc Natl Acad Sci U S A, 99(22), 14392-14397. doi: 10.1073/pnas.212410199

Hsu, W. L., Preston, D. L., Soda, M., Sugiyama, H., Funamoto, S., Kodama,

K., . . . Mabuchi, K. (2013). The Incidence of Leukemia, Lymphoma and Multiple Myeloma among Atomic Bomb Survivors: 1950-2001. Radiat Res. doi: 10.1667/RR2892.1

Hummel, J. L., Lichty, B. D., Reis, M., Dubé, I., & Kamel-Reid, S. (1996).

Engraftment of human chronic lymphocytic leukemia cells in SCID mice: in vivo and in vitro studies. Leukemia, 10(8), 1370-1376.

Irizarry, R. A., Hobbs, B., Collin, F., Beazer-Barclay, Y. D., Antonellis, K. J.,

Scherf, U., & Speed, T. P. (2003). Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics, 4(2), 249-264. doi: 10.1093/biostatistics/4.2.249

Page 189: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

170

Irizarry, R. A., Wu, Z., & Jaffee, H. A. (2006). Comparison of Affymetrix GeneChip expression measures. Bioinformatics, 22(7), 789-794. doi: 10.1093/bioinformatics/btk046

Itälä, M., Helenius, H., Nikoskelainen, J., & Remes, K. (1992). Infections and

serum IgG levels in patients with chronic lymphocytic leukemia. Eur J Haematol, 48(5), 266-270.

Jadidi-Niaragh, F., Jeddi-Tehrani, M., Ansaripour, B., Razavi, S. M.,

Sharifian, R. A., & Shokri, F. (2012). Reduced frequency of NKT-like cells in patients with progressive chronic lymphocytic leukemia. Med Oncol, 29(5), 3561-3569. doi: 10.1007/s12032-012-0262-4

Jaffe, E. S., Harris, N.L., Stein, H. & Vardiman, J.W. (eds.). (2001). World

Health Organization classification of tumours: pathology and genetics of tumours of haematopoietic and lymphoid tissues. Lyon: IARC Press.

Jaglowski, S. M., Alinari, L., Lapalombella, R., Muthusamy, N., & Byrd, J. C.

(2010). The clinical application of monoclonal antibodies in chronic lymphocytic leukemia. Blood, 116(19), 3705-3714. doi: 10.1182/blood-2010-04-001230

Jaglowski, S. M., & Byrd, J. C. (2012). Novel therapies and their integration

into allogeneic stem cell transplant for chronic lymphocytic leukemia. Biol Blood Marrow Transplant, 18(1 Suppl), S132-138. doi: 10.1016/j.bbmt.2011.11.018

Jak, M., Mous, R., Remmerswaal, E. B., Spijker, R., Jaspers, A., Yagüe, A., .

. . Van Oers, M. H. (2009). Enhanced formation and survival of CD4+ CD25hi Foxp3+ T-cells in chronic lymphocytic leukemia. Leuk Lymphoma, 50(5), 788-801. doi: 10.1080/10428190902803677

Jamin, C., Lamour, A., Pennec, Y. L., Hirn, M., Le Goff, P., & Youinou, P.

(1993). Expression of CD5 and CD72 on T and B cell subsets in rheumatoid arthritis and Sjögren's syndrome. Clin Exp Immunol, 92(2), 245-250.

Jankovic, M., Casellas, R., Yannoutsos, N., Wardemann, H., & Nussenzweig,

M. C. (2004). RAGs and regulation of autoantibodies. Annu Rev Immunol, 22, 485-501. doi: 10.1146/annurev.immunol.22.012703.104707

Jelinek, D. F., Tschumper, R. C., Stolovitzky, G. A., Iturria, S. J., Tu, Y.,

Lepre, J., . . . Kay, N. E. (2003). Identification of a global gene expression signature of B-chronic lymphocytic leukemia. Mol Cancer Res, 1(5), 346-361.

Page 190: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

171

Jellusova, J., Wellmann, U., Amann, K., Winkler, T. H., & Nitschke, L. (2010).

CD22 x Siglec-G double-deficient mice have massively increased B1 cell numbers and develop systemic autoimmunity. J Immunol, 184(7), 3618-3627. doi: 10.4049/jimmunol.0902711

Jim, R. T. (1957). Serum gamma globulin levels in chronic lymphocytic

leukemia. Am J Med Sci, 234(1), 44-47. Johnson, A. J., Lucas, D. M., Muthusamy, N., Smith, L. L., Edwards, R. B.,

De Lay, M. D., . . . Byrd, J. C. (2006). Characterization of the TCL-1 transgenic mouse as a preclinical drug development tool for human chronic lymphocytic leukemia. Blood, 108(4), 1334-1338. doi: 10.1182/blood-2005-12-011213

Johnson, W. E., Li, C., & Rabinovic, A. (2007). Adjusting batch effects in

microarray expression data using empirical Bayes methods. Biostatistics, 8(1), 118-127. doi: 10.1093/biostatistics/kxj037

Jordö, E. D., Wermeling, F., Chen, Y., & Karlsson, M. C. (2011). Scavenger

receptors as regulators of natural antibody responses and B cell activation in autoimmunity. Mol Immunol, 48(11), 1307-1318. doi: 10.1016/j.molimm.2011.01.010

Joshi, A. D., Hegde, G. V., Dickinson, J. D., Mittal, A. K., Lynch, J. C., Eudy,

J. D., . . . Joshi, S. S. (2007). ATM, CTLA4, MNDA, and HEM1 in high versus low CD38 expressing B-cell chronic lymphocytic leukemia. Clin Cancer Res, 13(18 Pt 1), 5295-5304. doi: 10.1158/1078-0432.CCR-07-0283

Juliusson, G., Oscier, D. G., Fitchett, M., Ross, F. M., Stockdill, G., Mackie,

M. J., . . . Gahrton, G. (1990). Prognostic subgroups in B-cell chronic lymphocytic leukemia defined by specific chromosomal abnormalities. N Engl J Med, 323(11), 720-724. doi: 10.1056/NEJM199009133231105

Kalos, M., Levine, B. L., Porter, D. L., Katz, S., Grupp, S. A., Bagg, A., &

June, C. H. (2011). T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med, 3(95), 95ra73. doi: 10.1126/scitranslmed.3002842

Kamper, E. F., Papaphilis, A. D., Angelopoulou, M. K., Kopeikina, L. T.,

Siakantaris, M. P., Pangalis, G. A., & Stavridis, J. C. (1999). Serum levels of tetranectin, intercellular adhesion molecule-1 and interleukin-10 in B-chronic lymphocytic leukemia. Clin Biochem, 32(8), 639-645.

Page 191: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

172

Kang, S. M., Narducci, M. G., Lazzeri, C., Mongiovì, A. M., Caprini, E.,

Bresin, A., . . . Russo, G. (2005). Impaired T- and B-cell development in Tcl1-deficient mice. Blood, 105(3), 1288-1294. doi: 10.1182/blood-2004-04-1453

Karmali, R., Paganessi, L. A., Frank, R. R., Jagan, S., Larson, M. L.,

Venugopal, P., . . . Christopherson, K. W. (2013). Aggressive disease defined by cytogenetics is associated with cytokine dysregulation in CLL/SLL patients. J Leukoc Biol, 93(1), 161-170. doi: 10.1189/jlb.0612301

Kater, A. P., Evers, L. M., Remmerswaal, E. B., Jaspers, A., Oosterwijk, M.

F., van Lier, R. A., . . . Eldering, E. (2004). CD40 stimulation of B-cell chronic lymphocytic leukaemia cells enhances the anti-apoptotic profile, but also Bid expression and cells remain susceptible to autologous cytotoxic T-lymphocyte attack. Br J Haematol, 127(4), 404-415. doi: 10.1111/j.1365-2141.2004.05225.x

Kay, N. E. (1981). Abnormal T-cell subpopulation function in CLL: excessive

suppressor (T gamma) and deficient helper (T mu) activity with respect to B-cell proliferation. Blood, 57(3), 418-420.

Keating, M. J., Kantarjian, H., Talpaz, M., Redman, J., Koller, C., Barlogie, B.,

. . . McCredie, K. B. (1989). Fludarabine: a new agent with major activity against chronic lymphocytic leukemia. Blood, 74(1), 19-25.

Keating, M. J., O'Brien, S., Albitar, M., Lerner, S., Plunkett, W., Giles, F., . . .

Kantarjian, H. (2005). Early results of a chemoimmunotherapy regimen of fludarabine, cyclophosphamide, and rituximab as initial therapy for chronic lymphocytic leukemia. J Clin Oncol, 23(18), 4079-4088. doi: 10.1200/JCO.2005.12.051

Kern, C., Cornuel, J. F., Billard, C., Tang, R., Rouillard, D., Stenou, V., . . .

Kolb, J. P. (2004). Involvement of BAFF and APRIL in the resistance to apoptosis of B-CLL through an autocrine pathway. Blood, 103(2), 679-688. doi: 10.1182/blood-2003-02-0540

Kern, W., Bacher, U., Haferlach, C., Dicker, F., Alpermann, T., Schnittger, S.,

& Haferlach, T. (2012). Monoclonal B-cell lymphocytosis is closely related to chronic lymphocytic leukaemia and may be better classified as early-stage CLL. Br J Haematol, 157(1), 86-96. doi: 10.1111/j.1365-2141.2011.09010.x

Khare, S. D., Sarosi, I., Xia, X. Z., McCabe, S., Miner, K., Solovyev, I., . . .

Hsu, H. (2000). Severe B cell hyperplasia and autoimmune disease in

Page 192: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

173

TALL-1 transgenic mice. Proc Natl Acad Sci U S A, 97(7), 3370-3375. doi: 10.1073/pnas.050580697

Kiaii, S., Kokhaei, P., Mozaffari, F., Rossmann, E., Pak, F., Moshfegh, A., . . .

Mellstedt, H. (2013). T cells from indolent CLL patients prevent apoptosis of leukemic B cells in vitro and have altered gene expression profile. Cancer Immunol Immunother, 62(1), 51-63. doi: 10.1007/s00262-012-1300-y

Kiefer, K., Nakajima, P. B., Oshinsky, J., Seeholzer, S. H., Radic, M., Bosma,

G. C., & Bosma, M. J. (2008). Antigen receptor editing in anti-DNA transitional B cells deficient for surface IgM. J Immunol, 180(9), 6094-6106.

Kikushige, Y., Ishikawa, F., Miyamoto, T., Shima, T., Urata, S., Yoshimoto,

G., . . . Akashi, K. (2011). Self-renewing hematopoietic stem cell is the primary target in pathogenesis of human chronic lymphocytic leukemia. Cancer Cell, 20(2), 246-259. doi: 10.1016/j.ccr.2011.06.029

King, L. B., & Monroe, J. G. (2000). Immunobiology of the immature B cell:

plasticity in the B-cell antigen receptor-induced response fine tunes negative selection. Immunol Rev, 176, 86-104.

Kipps, T. J., Robbins, B. A., Kuster, P., & Carson, D. A. (1988). Autoantibody-

associated cross-reactive idiotypes expressed at high frequency in chronic lymphocytic leukemia relative to B-cell lymphomas of follicular center cell origin. Blood, 72(2), 422-428.

Kitabayashi, A., Hirokawa, M., & Miura, A. B. (1995). The role of interleukin-

10 (IL-10) in chronic B-lymphocytic leukemia: IL-10 prevents leukemic cells from apoptotic cell death. Int J Hematol, 62(2), 99-106.

Kjeldsberg, C. R., & Marty, J. (1981). Prolymphocytic transformation of

chronic lymphocytic leukemia. Cancer, 48(11), 2447-2457. Klein, U., & Dalla-Favera, R. (2010). New insights into the pathogenesis of

chronic lymphocytic leukemia. Semin Cancer Biol, 20(6), 377-383. doi: 10.1016/j.semcancer.2010.10.012

Klein, U., Lia, M., Crespo, M., Siegel, R., Shen, Q., Mo, T., . . . Dalla-Favera,

R. (2010). The DLEU2/miR-15a/16-1 cluster controls B cell proliferation and its deletion leads to chronic lymphocytic leukemia. Cancer Cell, 17(1), 28-40. doi: 10.1016/j.ccr.2009.11.019

Klein, U., Tu, Y., Stolovitzky, G. A., Mattioli, M., Cattoretti, G., Husson, H., . . .

Dalla-Favera, R. (2001). Gene expression profiling of B cell chronic

Page 193: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

174

lymphocytic leukemia reveals a homogeneous phenotype related to memory B cells. J Exp Med, 194(11), 1625-1638.

Kobayashi, R., Picchio, G., Kirven, M., Meisenholder, G., Baird, S., Carson,

D. A., . . . Kipps, T. J. (1992). Transfer of human chronic lymphocytic leukemia to mice with severe combined immune deficiency. Leuk Res, 16(10), 1013-1023.

Kono, D. H., Burlingame, R. W., Owens, D. G., Kuramochi, A., Balderas, R.

S., Balomenos, D., & Theofilopoulos, A. N. (1994). Lupus susceptibility loci in New Zealand mice. Proc Natl Acad Sci U S A, 91(21), 10168-10172.

Kosmaczewska, A., Ciszak, L., Suwalska, K., Wolowiec, D., & Frydecka, I.

(2005). CTLA-4 overexpression in CD19+/CD5+ cells correlates with the level of cell cycle regulators and disease progression in B-CLL patients. Leukemia, 19(2), 301-304. doi: 10.1038/sj.leu.2403588

Kostareli, E., Hadzidimitriou, A., Stavroyianni, N., Darzentas, N.,

Athanasiadou, A., Gounari, M., . . . Stamatopoulos, K. (2009). Molecular evidence for EBV and CMV persistence in a subset of patients with chronic lymphocytic leukemia expressing stereotyped IGHV4-34 B-cell receptors. Leukemia, 23(5), 919-924. doi: 10.1038/leu.2008.379

Kotla, V., Goel, S., Nischal, S., Heuck, C., Vivek, K., Das, B., & Verma, A.

(2009). Mechanism of action of lenalidomide in hematological malignancies. J Hematol Oncol, 2, 36. doi: 10.1186/1756-8722-2-36

Kotsianidis, I., Nakou, E., Spanoudakis, E., Bouchliou, I., Moustakidis, E.,

Miltiades, P., . . . Tsatalas, C. (2011). The diagnostic value of CD1d expression in a large cohort of patients with B-cell chronic lymphoproliferative disorders. Am J Clin Pathol, 136(3), 400-408. doi: 10.1309/AJCP2F2DOXOTXHZA

Krackhardt, A. M., Harig, S., Witzens, M., Broderick, R., Barrett, P., &

Gribben, J. G. (2002). T-cell responses against chronic lymphocytic leukemia cells: implications for immunotherapy. Blood, 100(1), 167-173.

Kremer, J. P., Reisbach, G., Nerl, C., & Dörmer, P. (1992). B-cell chronic

lymphocytic leukaemia cells express and release transforming growth factor-beta. Br J Haematol, 80(4), 480-487.

Kröber, A., Bloehdorn, J., Hafner, S., Bühler, A., Seiler, T., Kienle, D., . . .

Stilgenbauer, S. (2006). Additional genetic high-risk features such as

Page 194: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

175

11q deletion, 17p deletion, and V3-21 usage characterize discordance of ZAP-70 and VH mutation status in chronic lymphocytic leukemia. J Clin Oncol, 24(6), 969-975. doi: 10.1200/JCO.2005.03.7184

Kurosaki, T. (1999). Genetic analysis of B cell antigen receptor signaling.

Annu Rev Immunol, 17, 555-592. doi: 10.1146/annurev.immunol.17.1.555

Kurosaki, T., Shinohara, H., & Baba, Y. (2010). B cell signaling and fate

decision. Annu Rev Immunol, 28, 21-55. doi: 10.1146/annurev.immunol.021908.132541

Kyasa, M. J., Hazlett, L., Parrish, R. S., Schichman, S. A., & Zent, C. S.

(2004). Veterans with chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) have a markedly increased rate of second malignancy, which is the most common cause of death. Leuk Lymphoma, 45(3), 507-513.

Küppers, R. (2005). Mechanisms of B-cell lymphoma pathogenesis. Nat Rev

Cancer, 5(4), 251-262. doi: 10.1038/nrc1589 Lajaunias, F., Nitschke, L., Moll, T., Martinez-Soria, E., Semac, I.,

Chicheportiche, Y., . . . Izui, S. (2002). Differentially regulated expression and function of CD22 in activated B-1 and B-2 lymphocytes. J Immunol, 168(12), 6078-6083.

Lanasa, M. C., Allgood, S. D., Slager, S. L., Dave, S. S., Love, C., Marti, G.

E., . . . Caporaso, N. E. (2011). Immunophenotypic and gene expression analysis of monoclonal B-cell lymphocytosis shows biologic characteristics associated with good prognosis CLL. Leukemia, 25(9), 1459-1466. doi: 10.1038/leu.2011.117

Lanemo Myhrinder, A., Hellqvist, E., Sidorova, E., Söderberg, A., Baxendale,

H., Dahle, C., . . . Rosén, A. (2008). A new perspective: molecular motifs on oxidized LDL, apoptotic cells, and bacteria are targets for chronic lymphocytic leukemia antibodies. Blood, 111(7), 3838-3848. doi: 10.1182/blood-2007-11-125450

Le Roy, C., Deglesne, P. A., Chevallier, N., Beitar, T., Eclache, V., Quettier,

M., . . . Varin-Blank, N. (2012). The degree of BCR and NFAT activation predicts clinical outcomes in chronic lymphocytic leukemia. Blood, 120(2), 356-365. doi: 10.1182/blood-2011-12-397158

Lech, M., Kulkarni, O. P., Pfeiffer, S., Savarese, E., Krug, A., Garlanda, C., . .

. Anders, H. J. (2008). Tir8/Sigirr prevents murine lupus by suppressing the immunostimulatory effects of lupus autoantigens. J

Page 195: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

176

Exp Med, 205(8), 1879-1888. doi: 10.1084/jem.20072646 Ledesma-Soto, Y., Blanco-Favela, F., Fuentes-Pananá, E. M., Tesoro-Cruz,

E., Hernández-González, R., Arriaga-Pizano, L., . . . Chávez-Rueda, A. K. (2012). Increased levels of prolactin receptor expression correlate with the early onset of lupus symptoms and increased numbers of transitional-1 B cells after prolactin treatment. BMC Immunol, 13, 11. doi: 10.1186/1471-2172-13-11

Lee, S. Y., Reichlin, A., Santana, A., Sokol, K. A., Nussenzweig, M. C., &

Choi, Y. (1997). TRAF2 is essential for JNK but not NF-kappaB activation and regulates lymphocyte proliferation and survival. Immunity, 7(5), 703-713.

Li, C., & Hung Wong, W. (2001). Model-based analysis of oligonucleotide

arrays: model validation, design issues and standard error application. Genome Biol, 2(8), RESEARCH0032.

Li, H., Jiang, Y., Prak, E. L., Radic, M., & Weigert, M. (2001). Editors and

editing of anti-DNA receptors. Immunity, 15(6), 947-957. Li, Y., & Zhang, Y. (2006). Molecular evolution of prolactin gene family in

rodents. Acta Genetica Sinica, 33(7), 590-597. Lia, M., Carette, A., Tang, H., Shen, Q., Mo, T., Bhagat, G., . . . Klein, U.

(2012). Functional dissection of the chromosome 13q14 tumor-suppressor locus using transgenic mouse lines. Blood, 119(13), 2981-2990. doi: 10.1182/blood-2011-09-381814

Lieber, M. R., Ma, Y., Pannicke, U., & Schwarz, K. (2004). The mechanism of

vertebrate nonhomologous DNA end joining and its role in V(D)J recombination. DNA Repair (Amst), 3(8-9), 817-826. doi: 10.1016/j.dnarep.2004.03.015

Linet, M. S., Schubauer-Berigan, M. K., Weisenburger, D. D., Richardson, D.

B., Landgren, O., Blair, A., . . . Dores, G. M. (2007). Chronic lymphocytic leukaemia: an overview of aetiology in light of recent developments in classification and pathogenesis. Br J Haematol, 139(5), 672-686. doi: 10.1111/j.1365-2141.2007.06847.x

Liu, F. T., Giustiniani, J., Farren, T., Jia, L., Bensussan, A., Gribben, J. G., &

Agrawal, S. G. (2010). CD160 signaling mediates PI3K-dependent survival and growth signals in chronic lymphocytic leukemia. Blood, 115(15), 3079-3088. doi: 10.1182/blood-2009-08-239483

Liu, Y., Corcoran, M., Rasool, O., Ivanova, G., Ibbotson, R., Grandér, D., . . .

Page 196: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

177

Oscier, D. (1997). Cloning of two candidate tumor suppressor genes within a 10 kb region on chromosome 13q14, frequently deleted in chronic lymphocytic leukemia. Oncogene, 15(20), 2463-2473. doi: 10.1038/sj.onc.1201643

Livak, K. J., & Schmittgen, T. D. (2001). Analysis of relative gene expression

data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods, 25(4), 402-408. doi: 10.1006/meth.2001.1262

Loisel, S., Ster, K. L., Quintin-Roue, I., Pers, J. O., Bordron, A., Youinou, P.,

& Berthou, C. (2005). Establishment of a novel human B-CLL-like xenograft model in nude mouse. Leuk Res, 29(11), 1347-1352. doi: 10.1016/j.leukres.2005.04.017

Lombardi, V., Stock, P., Singh, A. K., Kerzerho, J., Yang, W., Sullivan, B. A., .

. . Akbari, O. (2010). A CD1d-dependent antagonist inhibits the activation of invariant NKT cells and prevents development of allergen-induced airway hyperreactivity. J Immunol, 184(4), 2107-2115. doi: 10.4049/jimmunol.0901208

Lotz, M., Ranheim, E., & Kipps, T. J. (1994). Transforming growth factor beta

as endogenous growth inhibitor of chronic lymphocytic leukemia B cells. J Exp Med, 179(3), 999-1004.

Lugassy, G., Lishner, M., & Polliack, A. (1992). Systemic lupus

erythematosus and chronic lymphocytic leukemia: rare coexistence in three patients, with comments on pathogenesis. Leuk Lymphoma, 8(3), 243-245. doi: 10.3109/10428199209054911

M'Hidi, H., Thibult, M. L., Chetaille, B., Rey, F., Bouadallah, R., Nicollas, R., .

. . Xerri, L. (2009). High expression of the inhibitory receptor BTLA in T-follicular helper cells and in B-cell small lymphocytic lymphoma/chronic lymphocytic leukemia. Am J Clin Pathol, 132(4), 589-596. doi: 10.1309/AJCPPHKGYYGGL39C

Mackay, F., Woodcock, S. A., Lawton, P., Ambrose, C., Baetscher, M.,

Schneider, P., . . . Browning, J. L. (1999). Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations. J Exp Med, 190(11), 1697-1710.

Maddocks-Christianson, K., Slager, S. L., Zent, C. S., Reinalda, M., Call, T.

G., Habermann, T. M., . . . Shanafelt, T. D. (2007). Risk factors for development of a second lymphoid malignancy in patients with chronic lymphocytic leukaemia. Br J Haematol, 139(3), 398-404. doi: 10.1111/j.1365-2141.2007.06801.x

Page 197: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

178

Maffei, R., Fiorcari, S., Bulgarelli, J., Martinelli, S., Castelli, I., Deaglio, S., . . . Marasca, R. (2012). Physical contact with endothelial cells through β1- and β2- integrins rescues chronic lymphocytic leukemia cells from spontaneous and drug-induced apoptosis and induces a peculiar gene expression profile in leukemic cells. Haematologica, 97(6), 952-960. doi: 10.3324/haematol.2011.054924

Mahboudi, F., Phillips, J., & Raveche, E. (1992). Immunoglobulin gene

sequence-analysis of B-1 (cd5+b) cell clones in a murine model of chronic lymphocytic-leukemia and richters syndrome. Int J Oncol, 1(4), 459-465.

Malavasi, F., Deaglio, S., Damle, R., Cutrona, G., Ferrarini, M., & Chiorazzi,

N. (2011). CD38 and chronic lymphocytic leukemia: a decade later. Blood, 118(13), 3470-3478. doi: 10.1182/blood-2011-06-275610

Mansouri, L., Cahill, N., Gunnarsson, R., Smedby, K. E., Tjönnfjord, E.,

Hjalgrim, H., . . . Rosenquist, R. (2013). NOTCH1 and SF3B1 mutations can be added to the hierarchical prognostic classification in chronic lymphocytic leukemia. Leukemia, 27(2), 512-514. doi: 10.1038/leu.2012.307

Marti, G. E., Rawstron, A. C., Ghia, P., Hillmen, P., Houlston, R. S., Kay, N., .

. . Consortium, I. F. C. (2005). Diagnostic criteria for monoclonal B-cell lymphocytosis. Br J Haematol, 130(3), 325-332. doi: 10.1111/j.1365-2141.2005.05550.x

Marti, G. E., & Zenger, V. (2004). The natural history of CLL: historical

perspective. In G. Faguet (Ed.), Contemporary Hematology (pp. 3-54). Totowa, NJ: Humana Press.

Martin, F., Oliver, A. M., & Kearney, J. F. (2001). Marginal zone and B1 B

cells unite in the early response against T-independent blood-borne particulate antigens. Immunity, 14(5), 617-629.

Matsuda, J. L., Naidenko, O. V., Gapin, L., Nakayama, T., Taniguchi, M.,

Wang, C. R., . . . Kronenberg, M. (2000). Tracking the response of natural killer T cells to a glycolipid antigen using CD1d tetramers. J Exp Med, 192(5), 741-754.

Matutes, E., Oscier, D., Garcia-Marco, J., Ellis, J., Copplestone, A.,

Gillingham, R., . . . Catovsky, D. (1996). Trisomy 12 defines a group of CLL with atypical morphology: correlation between cytogenetic, clinical and laboratory features in 544 patients. Br J Haematol, 92(2), 382-388.

Page 198: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

179

Matutes, E., & Polliack, A. (2000). Morphological and immunophenotypic features of chronic lymphocytic leukemia. Rev Clin Exp Hematol, 4(1), 22-47.

Maura, F., Visco, C., Falisi, E., Reda, G., Fabris, S., Agnelli, L., . . .

Cortelezzi, A. (2013). B-cell receptor configuration and adverse cytogenetics are associated with autoimmune hemolytic anemia in chronic lymphocytic leukemia. Am J Hematol, 88(1), 32-36. doi: 10.1002/ajh.23342

Mauri, C., & Bosma, A. (2012). Immune regulatory function of B cells. Annu

Rev Immunol, 30, 221-241. doi: 10.1146/annurev-immunol-020711-074934

Mauri, C., & Ehrenstein, M. R. (2008). The 'short' history of regulatory B cells.

Trends Immunol, 29(1), 34-40. doi: 10.1016/j.it.2007.10.004 McCarthy, B. A., Mansour, A., Lin, Y. C., Kotenko, S., & Raveche, E. (2004).

RNA interference of IL-10 in leukemic B-1 cells. Cancer Immun, 4, 6. Messmer, B. T., Albesiano, E., Efremov, D. G., Ghiotto, F., Allen, S. L., Kolitz,

J., . . . Chiorazzi, N. (2004). Multiple distinct sets of stereotyped antigen receptors indicate a role for antigen in promoting chronic lymphocytic leukemia. J Exp Med, 200(4), 519-525. doi: 10.1084/jem.20040544

Messmer, B. T., Messmer, D., Allen, S. L., Kolitz, J. E., Kudalkar, P., Cesar,

D., . . . Chiorazzi, N. (2005). In vivo measurements document the dynamic cellular kinetics of chronic lymphocytic leukemia B cells. J Clin Invest, 115(3), 755-764. doi: 10.1172/JCI23409

Michie, A. M., & Nakagawa, R. (2005). The link between PKCalpha regulation

and cellular transformation. Immunol Lett, 96(2), 155-162. doi: 10.1016/j.imlet.2004.08.013

Migliazza, A., Bosch, F., Komatsu, H., Cayanis, E., Martinotti, S., Toniato, E.,

. . . Dalla-Favera, R. (2001). Nucleotide sequence, transcription map, and mutation analysis of the 13q14 chromosomal region deleted in B-cell chronic lymphocytic leukemia. Blood, 97(7), 2098-2104.

Minot, G. B., & Isaacs, R. (1924). Lymphatic Leukemia; Age Incidence,

Duration, and Benefit Derived from Irradiation. Boston Med Surg J, 191(1), 1-9.

Molica, S., Levato, D., & Levato, L. (1993). Infections in chronic lymphocytic

leukemia. Analysis of incidence as a function of length of follow-up.

Page 199: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

180

Haematologica, 78(6), 374-377. Montecino-Rodriguez, E., & Dorshkind, K. (2012). B-1 B cell development in

the fetus and adult. Immunity, 36(1), 13-21. doi: 10.1016/j.immuni.2011.11.017

Montecino-Rodriguez, E., Leathers, H., & Dorshkind, K. (2006). Identification

of a B-1 B cell-specified progenitor. Nat Immunol, 7(3), 293-301. doi: 10.1038/ni1301

Moore, K. W., de Waal Malefyt, R., Coffman, R. L., & O'Garra, A. (2001).

Interleukin-10 and the interleukin-10 receptor. Annu Rev Immunol, 19, 683-765. doi: 10.1146/annurev.immunol.19.1.683

Moreira, J., Rabe, K. G., Cerhan, J. R., Kay, N. E., Wilson, J. W., Call, T. G., .

. . Shanafelt, T. D. (2013). Infectious complications among individuals with clinical monoclonal B-cell lymphocytosis (MBL): a cohort study of newly diagnosed cases compared to controls. Leukemia, 27(1), 136-141. doi: 10.1038/leu.2012.187

Morrison, V. A. (2010). Infectious complications of chronic lymphocytic

leukaemia: pathogenesis, spectrum of infection, preventive approaches. Best Pract Res Clin Haematol, 23(1), 145-153. doi: 10.1016/j.beha.2009.12.004

Morton, L. M., Turner, J. J., Cerhan, J. R., Linet, M. S., Treseler, P. A.,

Clarke, C. A., . . . Weisenburger, D. D. (2007). Proposed classification of lymphoid neoplasms for epidemiologic research from the Pathology Working Group of the International Lymphoma Epidemiology Consortium (InterLymph). Blood, 110(2), 695-708. doi: 10.1182/blood-2006-11-051672

Motiwala, T., Zanesi, N., Datta, J., Roy, S., Kutay, H., Checovich, A. M., . . .

Jacob, S. T. (2011). AP-1 elements and TCL1 protein regulate expression of the gene encoding protein tyrosine phosphatase PTPROt in leukemia. Blood, 118(23), 6132-6140. doi: 10.1182/blood-2011-01-323147

Motta, M., Rassenti, L., Shelvin, B. J., Lerner, S., Kipps, T. J., Keating, M. J.,

& Wierda, W. G. (2005). Increased expression of CD152 (CTLA-4) by normal T lymphocytes in untreated patients with B-cell chronic lymphocytic leukemia. Leukemia, 19(10), 1788-1793. doi: 10.1038/sj.leu.2403907

Munzert, G., Kirchner, D., Stobbe, H., Bergmann, L., Schmid, R. M., Döhner,

H., & Heimpel, H. (2002). Tumor necrosis factor receptor-associated

Page 200: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

181

factor 1 gene overexpression in B-cell chronic lymphocytic leukemia: analysis of NF-kappa B/Rel-regulated inhibitors of apoptosis. Blood, 100(10), 3749-3756. doi: 10.1182/blood.V100.10.3749

Muramatsu, M., Kinoshita, K., Fagarasan, S., Yamada, S., Shinkai, Y., &

Honjo, T. (2000). Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme. Cell, 102(5), 553-563.

Murphy, K. M., Nelson, C. A., & Sedý, J. R. (2006). Balancing co-stimulation

and inhibition with BTLA and HVEM. Nat Rev Immunol, 6(9), 671-681. doi: 10.1038/nri1917

Nakagawa, R., Soh, J. W., & Michie, A. M. (2006). Subversion of protein

kinase C alpha signaling in hematopoietic progenitor cells results in the generation of a B-cell chronic lymphocytic leukemia-like population in vivo. Cancer Res, 66(1), 527-534. doi: 10.1158/0008-5472.CAN-05-0841

Narducci, M. G., Pescarmona, E., Lazzeri, C., Signoretti, S., Lavinia, A. M.,

Remotti, D., . . . Russo, G. (2000). Regulation of TCL1 expression in B- and T-cell lymphomas and reactive lymphoid tissues. Cancer Res, 60(8), 2095-2100.

Negro, R., Gobessi, S., Longo, P. G., He, Y., Zhang, Z. Y., Laurenti, L., &

Efremov, D. G. (2012). Overexpression of the autoimmunity-associated phosphatase PTPN22 promotes survival of antigen-stimulated CLL cells by selectively activating AKT. Blood, 119(26), 6278-6287. doi: 10.1182/blood-2012-01-403162

Nemazee, D. (2006). Receptor editing in lymphocyte development and

central tolerance. Nat Rev Immunol, 6(10), 728-740. doi: 10.1038/nri1939

Nemazee, D., & Weigert, M. (2000). Revising B cell receptors. J Exp Med,

191(11), 1813-1817. Nganga, V. K., Palmer, V. L., Naushad, H., Kassmeier, M. D., Anderson, D.

K., Perry, G. A., . . . Swanson, P. C. (2013). Accelerated progression of chronic lymphocytic leukemia in Eμ-TCL1 mice expressing catalytically inactive RAG1. Blood, 121(19), 3855-3866, S3851-3816. doi: 10.1182/blood-2012-08-446732

Nishio, M., Endo, T., Tsukada, N., Ohata, J., Kitada, S., Reed, J. C., . . .

Kipps, T. J. (2005). Nurselike cells express BAFF and APRIL, which can promote survival of chronic lymphocytic leukemia cells via a

Page 201: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

182

paracrine pathway distinct from that of SDF-1alpha. Blood, 106(3), 1012-1020. doi: 10.1182/blood-2004-03-0889

Nordgren, T. M., & Joshi, S. S. (2010). The etiology of chronic lymphocytic

leukemia: another look at the relationship between B1 and cells and CLL. Open Leuk J, 3, 69-73.

Nowakowski, G. S., Hoyer, J. D., Shanafelt, T. D., Geyer, S. M., LaPlant, B.

R., Call, T. G., . . . Kay, N. E. (2007). Using smudge cells on routine blood smears to predict clinical outcome in chronic lymphocytic leukemia: a universally available prognostic test. Mayo Clin Proc, 82(4), 449-453. doi: 10.4065/82.4.449

Nwabo Kamdje, A. H., Bassi, G., Pacelli, L., Malpeli, G., Amati, E., Nichele, I.,

. . . Krampera, M. (2012). Role of stromal cell-mediated Notch signaling in CLL resistance to chemotherapy. Blood Cancer J, 2(5), e73. doi: 10.1038/bcj.2012.17

O'Garra, A., Chang, R., Go, N., Hastings, R., Haughton, G., & Howard, M.

(1992). Ly-1 B (B-1) cells are the main source of B cell-derived interleukin 10. Eur J Immunol, 22(3), 711-717. doi: 10.1002/eji.1830220314

Ochi, H., & Watanabe, T. (2000). Negative regulation of B cell receptor-

mediated signaling in B-1 cells through CD5 and Ly49 co-receptors via Lyn kinase activity. Int Immunol, 12(10), 1417-1423.

Oettinger, M. A., Schatz, D. G., Gorka, C., & Baltimore, D. (1990). RAG-1

and RAG-2, adjacent genes that synergistically activate V(D)J recombination. Science, 248(4962), 1517-1523.

Okada, T., Takiura, F., Tokushige, K., Nozawa, S., Kiyosawa, T., Nakauchi,

H., . . . Shirai, T. (1991). Major histocompatibility complex controls clonal proliferation of CD5+ B cells in H-2-congenic New Zealand mice: a model for B cell chronic lymphocytic leukemia and autoimmune disease. Eur J Immunol, 21(11), 2743-2748. doi: 10.1002/eji.1830211114

Palamarchuk, A., Yan, P. S., Zanesi, N., Wang, L., Rodrigues, B., Murphy,

M., . . . Pekarsky, Y. (2012). Tcl1 protein functions as an inhibitor of de novo DNA methylation in B-cell chronic lymphocytic leukemia (CLL). Proc Natl Acad Sci U S A, 109(7), 2555-2560. doi: 10.1073/pnas.1200003109

Pan, J. W., Cook, L. S., Schwartz, S. M., & Weis, N. S. (2002). Incidence of

leukemia in Asian migrants to the United States and their

Page 202: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

183

descendants. Cancer Causes Control, 13(9), 791-795. Panayiotidis, P., Jones, D., Ganeshaguru, K., Foroni, L., & Hoffbrand, A. V.

(1996). Human bone marrow stromal cells prevent apoptosis and support the survival of chronic lymphocytic leukaemia cells in vitro. Br J Haematol, 92(1), 97-103.

Pangalis, G. A., Vassilakopoulos, T. P., Dimopoulou, M. N., Siakantaris, M.

P., Kontopidou, F. N., & Angelopoulou, M. K. (2002). B-chronic lymphocytic leukemia: practical aspects. Hematol Oncol, 20(3), 103-146. doi: 10.1002/hon.696

Pao, L. I., Lam, K. P., Henderson, J. M., Kutok, J. L., Alimzhanov, M.,

Nitschke, L., . . . Rajewsky, K. (2007). B cell-specific deletion of protein-tyrosine phosphatase Shp1 promotes B-1a cell development and causes systemic autoimmunity. Immunity, 27(1), 35-48. doi: 10.1016/j.immuni.2007.04.016

Papamichail, M., Brown, J. C., & Holborow, E. J. (1971). Immunoglobulins on

the surface of human lymphocytes. Lancet, 2(7729), 850-852. Pedersen, I. M., Kitada, S., Leoni, L. M., Zapata, J. M., Karras, J. G.,

Tsukada, N., . . . Reed, J. C. (2002). Protection of CLL B cells by a follicular dendritic cell line is dependent on induction of Mcl-1. Blood, 100(5), 1795-1801.

Peeva, E., Michael, D., Cleary, J., Rice, J., Chen, X., & Diamond, B. (2003).

Prolactin modulates the naive B cell repertoire. J Clin Invest, 111(2), 275-283. doi: 10.1172/JCI16530

Pekarsky, Y., Koval, A., Hallas, C., Bichi, R., Tresini, M., Malstrom, S., . . .

Croce, C. M. (2000). Tcl1 enhances Akt kinase activity and mediates its nuclear translocation. Proc Natl Acad Sci U S A, 97(7), 3028-3033. doi: 10.1073/pnas.040557697

Pekarsky, Y., Palamarchuk, A., Maximov, V., Efanov, A., Nazaryan, N.,

Santanam, U., . . . Croce, C. M. (2008). Tcl1 functions as a transcriptional regulator and is directly involved in the pathogenesis of CLL. Proc Natl Acad Sci U S A, 105(50), 19643-19648. doi: 10.1073/pnas.0810965105

Pekarsky, Y., Santanam, U., Cimmino, A., Palamarchuk, A., Efanov, A.,

Maximov, V., . . . Croce, C. M. (2006). Tcl1 expression in chronic lymphocytic leukemia is regulated by miR-29 and miR-181. Cancer Res, 66(24), 11590-11593. doi: 10.1158/0008-5472.CAN-06-3613

Page 203: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

184

Perez-Chacon, G., Vargas, J. A., Jorda, J., Morado, M., Rosado, S., Martin-Donaire, T., . . . Perez-Aciego, P. (2007). CD5 provides viability signals to B cells from a subset of B-CLL patients by a mechanism that involves PKC. Leuk Res, 31(2), 183-193. doi: 10.1016/j.leukres.2006.03.021

Phillips, J. A., Mehta, K., Fernandez, C., & Raveché, E. S. (1992). The NZB

mouse as a model for chronic lymphocytic leukemia. Cancer Res, 52(2), 437-443.

Pillai, S., & Cariappa, A. (2009). The follicular versus marginal zone B

lymphocyte cell fate decision. Nat Rev Immunol, 9(11), 767-777. doi: 10.1038/nri2656

Pillai, S., Cariappa, A., & Moran, S. T. (2005). Marginal zone B cells. Annu

Rev Immunol, 23, 161-196. doi: 10.1146/annurev.immunol.23.021704.115728

Piller, G. (2001). Leukaemia - a brief historical review from ancient times to

1950. Br J Haematol, 112(2), 282-292. Planelles, L., Carvalho-Pinto, C. E., Hardenberg, G., Smaniotto, S., Savino,

W., Gómez-Caro, R., . . . Hahne, M. (2004). APRIL promotes B-1 cell-associated neoplasm. Cancer Cell, 6(4), 399-408. doi: 10.1016/j.ccr.2004.08.033

Planelles, L., Castillo-Gutiérrez, S., Medema, J. P., Morales-Luque, A.,

Merle-Béral, H., & Hahne, M. (2007). APRIL but not BLyS serum levels are increased in chronic lymphocytic leukemia: prognostic relevance of APRIL for survival. Haematologica, 92(9), 1284-1285.

Plass, C., Byrd, J. C., Raval, A., Tanner, S. M., & de la Chapelle, A. (2007).

Molecular profiling of chronic lymphocytic leukaemia: genetics meets epigenetics to identify predisposing genes. Br J Haematol, 139(5), 744-752. doi: 10.1111/j.1365-2141.2007.06875.x

Ponader, S., Chen, S. S., Buggy, J. J., Balakrishnan, K., Gandhi, V., Wierda,

W. G., . . . Burger, J. A. (2012). The Bruton tyrosine kinase inhibitor PCI-32765 thwarts chronic lymphocytic leukemia cell survival and tissue homing in vitro and in vivo. Blood, 119(5), 1182-1189. doi: 10.1182/blood-2011-10-386417

Porcelli, S. A., & Modlin, R. L. (1999). The CD1 system: antigen-presenting

molecules for T cell recognition of lipids and glycolipids. Annu Rev Immunol, 17, 297-329. doi: 10.1146/annurev.immunol.17.1.297

Page 204: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

185

Puente, X. S., Pinyol, M., Quesada, V., Conde, L., Ordóñez, G. R., Villamor, N., . . . Campo, E. (2011). Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature, 475(7354), 101-105. doi: 10.1038/nature10113

Pérez-Chacón, G., Llobet, D., Pardo, C., Pindado, J., Choi, Y., Reed, J. C., &

Zapata, J. M. (2012). TNFR-associated factor 2 deficiency in B lymphocytes predisposes to chronic lymphocytic leukemia/small lymphocytic lymphoma in mice. J Immunol, 189(2), 1053-1061. doi: 10.4049/jimmunol.1200814

Quiroga, M. P., Balakrishnan, K., Kurtova, A. V., Sivina, M., Keating, M. J.,

Wierda, W. G., . . . Burger, J. A. (2009). B-cell antigen receptor signaling enhances chronic lymphocytic leukemia cell migration and survival: specific targeting with a novel spleen tyrosine kinase inhibitor, R406. Blood, 114(5), 1029-1037. doi: 10.1182/blood-2009-03-212837

Rai, K. R. (1987). A critical analysis of staging in CLL Chronic lymphocytic

leukemia: recent progress and future direction (Vol. 59, pp. 253-264). New York.

Rai, K. R. (1993). Progress in chronic lymphocytic leukaemia: a historical

perspective. Baillieres Clin Haematol, 6(4), 757-765. Rai, K. R., Sawitsky, A., Cronkite, E. P., Chanana, A. D., Levy, R. N., &

Pasternack, B. S. (1975). Clinical staging of chronic lymphocytic leukemia. Blood, 46(2), 219-234.

Rajewsky, K. (1996). Clonal selection and learning in the antibody system.

Nature, 381(6585), 751-758. doi: 10.1038/381751a0 Ramachandra, S., Metcalf, R. A., Fredrickson, T., Marti, G. E., & Raveche, E.

(1996). Requirement for increased IL-10 in the development of B-1 lymphoproliferative disease in a murine model of CLL. J Clin Invest, 98(8), 1788-1793. doi: 10.1172/JCI118978

Ramsay, A. G., Clear, A. J., Fatah, R., & Gribben, J. G. (2012). Multiple

inhibitory ligands induce impaired T-cell immunologic synapse function in chronic lymphocytic leukemia that can be blocked with lenalidomide: establishing a reversible immune evasion mechanism in human cancer. Blood, 120(7), 1412-1421. doi: 10.1182/blood-2012-02-411678

Ramsay, A. G., & Gribben, J. G. (2009). Immune dysfunction in chronic

lymphocytic leukemia T cells and lenalidomide as an immunomodulatory drug. Haematologica, 94(9), 1198-1202. doi:

Page 205: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

186

10.3324/haematol.2009.009274 Ramsay, A. G., Johnson, A. J., Lee, A. M., Gorgün, G., Le Dieu, R., Blum,

W., . . . Gribben, J. G. (2008). Chronic lymphocytic leukemia T cells show impaired immunological synapse formation that can be reversed with an immunomodulating drug. J Clin Invest, 118(7), 2427-2437. doi: 10.1172/JCI35017

Raveche, E. S., Salerno, E., Scaglione, B. J., Manohar, V., Abbasi, F., Lin, Y.

C., . . . Marti, G. E. (2007). Abnormal microRNA-16 locus with synteny to human 13q14 linked to CLL in NZB mice. Blood, 109(12), 5079-5086. doi: 10.1182/blood-2007-02-071225

Raveché, E. S., Tjio, J. H., & Steinberg, A. D. (1979). Genetic studies in NZB

mice. II. Hyperdiploidy in the spleen of NZB mice and their hybrids. Cytogenet Cell Genet, 23(3), 182-193.

Rawstron, A. C., Bennett, F. L., O'Connor, S. J., Kwok, M., Fenton, J. A.,

Plummer, M., . . . Hillmen, P. (2008). Monoclonal B-cell lymphocytosis and chronic lymphocytic leukemia. N Engl J Med, 359(6), 575-583. doi: 10.1056/NEJMoa075290

Rawstron, A. C., Green, M. J., Kuzmicki, A., Kennedy, B., Fenton, J. A.,

Evans, P. A., . . . Hillmen, P. (2002). Monoclonal B lymphocytes with the characteristics of "indolent" chronic lymphocytic leukemia are present in 3.5% of adults with normal blood counts. Blood, 100(2), 635-639.

Redaelli, A., Laskin, B. L., Stephens, J. M., Botteman, M. F., & Pashos, C. L.

(2004). The clinical and epidemiological burden of chronic lymphocytic leukaemia. Eur J Cancer Care (Engl), 13(3), 279-287. doi: 10.1111/j.1365-2354.2004.00489.x

Reddy, A. L., & Fialkow, P. J. (1980). Chromosome fragility in New Zealand

black mice: effect of ultraviolet and gamma radiations on fetal fibroblasts in vitro. J Natl Cancer Inst, 64(4), 939-941.

Reiniger, L., Bödör, C., Bognár, A., Balogh, Z., Csomor, J., Szepesi, A., . . .

Matolcsy, A. (2006). Richter's and prolymphocytic transformation of chronic lymphocytic leukemia are associated with high mRNA expression of activation-induced cytidine deaminase and aberrant somatic hypermutation. Leukemia, 20(6), 1089-1095. doi: 10.1038/sj.leu.2404183

Riches, J. C., Davies, J. K., McClanahan, F., Fatah, R., Iqbal, S., Agrawal, S.,

. . . Gribben, J. G. (2013). T cells from CLL patients exhibit features of

Page 206: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

187

T-cell exhaustion but retain capacity for cytokine production. Blood, 121(9), 1612-1621. doi: 10.1182/blood-2012-09-457531

Richter, M. N. (1928). Generalized Reticular Cell Sarcoma of Lymph Nodes

Associated with Lymphatic Leukemia. Am J Pathol, 4(4), 285-292.287. Rickert, R. C., Jellusova, J., & Miletic, A. V. (2011). Signaling by the tumor

necrosis factor receptor superfamily in B-cell biology and disease. Immunol Rev, 244(1), 115-133. doi: 10.1111/j.1600-065X.2011.01067.x

Ringshausen, I., Oelsner, M., Weick, K., Bogner, C., Peschel, C., & Decker,

T. (2006). Mechanisms of apoptosis-induction by rottlerin: therapeutic implications for B-CLL. Leukemia, 20(3), 514-520. doi: 10.1038/sj.leu.2404113

Robak, T., Blonski, J. Z., Gora-Tybor, J., Kasznicki, M., Konopka, L.,

Ceglarek, B., . . . Całbecka, M. (2004). Second malignancies and Richter's syndrome in patients with chronic lymphocytic leukaemia treated with cladribine. Eur J Cancer, 40(3), 383-389.

Rodriguez-Manzanet, R., Sanjuan, M. A., Wu, H. Y., Quintana, F. J., Xiao, S.,

Anderson, A. C., . . . Kuchroo, V. K. (2010). T and B cell hyperactivity and autoimmunity associated with niche-specific defects in apoptotic body clearance in TIM-4-deficient mice. Proc Natl Acad Sci U S A, 107(19), 8706-8711. doi: 10.1073/pnas.0910359107

Rosenwald, A., Alizadeh, A. A., Widhopf, G., Simon, R., Davis, R. E., Yu, X., .

. . Staudt, L. M. (2001). Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia. J Exp Med, 194(11), 1639-1647.

Rossi, D., Cerri, M., Capello, D., Deambrogi, C., Rossi, F. M., Zucchetto, A., .

. . Gaidano, G. (2008). Biological and clinical risk factors of chronic lymphocytic leukaemia transformation to Richter syndrome. Br J Haematol, 142(2), 202-215. doi: 10.1111/j.1365-2141.2008.07166.x

Rosén, A., Murray, F., Evaldsson, C., & Rosenquist, R. (2010). Antigens in

chronic lymphocytic leukemia--implications for cell origin and leukemogenesis. Semin Cancer Biol, 20(6), 400-409. doi: 10.1016/j.semcancer.2010.09.004

Rozman, C., & Montserrat, E. (1995). Chronic lymphocytic leukemia. N Engl

J Med, 333(16), 1052-1057. doi: 10.1056/NEJM199510193331606 Rozman, C., Montserrat, E., & Viñolas, N. (1988). Serum immunoglobulins in

Page 207: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

188

B-chronic lymphocytic leukemia. Natural history and prognostic significance. Cancer, 61(2), 279-283.

Ruchlemer, R., & Polliack, A. (2012). Geography, ethnicity and "roots" in

chronic lymphocytic leukemia. Leuk Lymphoma. doi: 10.3109/10428194.2012.740670

Saha, S., Gonzalez, J., Rosenfeld, G., Keiser, H., & Peeva, E. (2009).

Prolactin alters the mechanisms of B cell tolerance induction. Arthritis Rheum, 60(6), 1743-1752. doi: 10.1002/art.24500

Saha, S., Tieng, A., Pepeljugoski, K. P., Zandamn-Goddard, G., & Peeva, E.

(2011). Prolactin, systemic lupus erythematosus, and autoreactive B cells: lessons learnt from murine models. Clin Rev Allergy Immunol, 40(1), 8-15. doi: 10.1007/s12016-009-8182-6

Sakaguchi, S., Yamaguchi, T., Nomura, T., & Ono, M. (2008). Regulatory T

cells and immune tolerance. Cell, 133(5), 775-787. doi: 10.1016/j.cell.2008.05.009

Salerno, E., Scaglione, B. J., Coffman, F. D., Brown, B. D., Baccarini, A.,

Fernandes, H., . . . Raveche, E. S. (2009). Correcting miR-15a/16 genetic defect in New Zealand Black mouse model of CLL enhances drug sensitivity. Mol Cancer Ther, 8(9), 2684-2692. doi: 10.1158/1535-7163.MCT-09-0127

Salerno, E., Yuan, Y., Scaglione, B. J., Marti, G., Jankovic, A., Mazzella, F., .

. . Raveche, E. (2010). The New Zealand black mouse as a model for the development and progression of chronic lymphocytic leukemia. Cytometry B Clin Cytom, 78 Suppl 1, S98-109. doi: 10.1002/cyto.b.20544

Santanam, U., Zanesi, N., Efanov, A., Costinean, S., Palamarchuk, A.,

Hagan, J. P., . . . Pekarsky, Y. (2010). Chronic lymphocytic leukemia modeled in mouse by targeted miR-29 expression. Proc Natl Acad Sci U S A, 107(27), 12210-12215. doi: 10.1073/pnas.1007186107

Scarfò, L., Dagklis, A., Scielzo, C., Fazi, C., & Ghia, P. (2010). CLL-like

monoclonal B-cell lymphocytosis: are we all bound to have it? Semin Cancer Biol, 20(6), 384-390. doi: 10.1016/j.semcancer.2010.08.005

Schaffner, C., Stilgenbauer, S., Rappold, G. A., Döhner, H., & Lichter, P.

(1999). Somatic ATM mutations indicate a pathogenic role of ATM in B-cell chronic lymphocytic leukemia. Blood, 94(2), 748-753.

Schettini, G., Hewlett, E. L., Cronin, M. J., Koike, K., Yasumoto, T., &

Page 208: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

189

MacLeod, R. M. (1986). Dopaminergic inhibition of anterior pituitary adenylate cyclase activity and prolactin release: the effects of perturbing calcium on catalytic adenylate cyclase activity. Neuroendocrinology, 44(1), 1-7.

Schmid, C., & Isaacson, P. G. (1994). Proliferation centres in B-cell malignant

lymphoma, lymphocytic (B-CLL): an immunophenotypic study. Histopathology, 24(5), 445-451.

Schroeder, H. W., & Dighiero, G. (1994). The pathogenesis of chronic

lymphocytic leukemia: analysis of the antibody repertoire. Immunol Today, 15(6), 288-294. doi: 10.1016/0167-5699(94)90009-4

Schroers, R., Griesinger, F., Trümper, L., Haase, D., Kulle, B., Klein-Hitpass,

L., . . . Dürig, J. (2005). Combined analysis of ZAP-70 and CD38 expression as a predictor of disease progression in B-cell chronic lymphocytic leukemia. Leukemia, 19(5), 750-758. doi: 10.1038/sj.leu.2403707

Schuler, M., Tretter, T., Schneller, F., Huber, C., & Peschel, C. (1999).

Autocrine transforming growth factor-beta from chronic lymphocytic leukemia-B cells interferes with proliferative T cell signals. Immunobiology, 200(1), 128-139.

Schulz, A., Dürr, C., Zenz, T., Döhner, H., Stilgenbauer, S., Lichter, P., &

Seiffert, M. (2013). Lenalidomide reduces survival of chronic lymphocytic leukemia cells in primary cocultures by altering the myeloid microenvironment. Blood, 121(13), 2503-2511. doi: 10.1182/blood-2012-08-447664

Seifert, M., Sellmann, L., Bloehdorn, J., Wein, F., Stilgenbauer, S., Dürig, J.,

& Küppers, R. (2012). Cellular origin and pathophysiology of chronic lymphocytic leukemia. J Exp Med, 209(12), 2183-2198. doi: 10.1084/jem.20120833

Sen, G., Bikah, G., Venkataraman, C., & Bondada, S. (1999). Negative

regulation of antigen receptor-mediated signaling by constitutive association of CD5 with the SHP-1 protein tyrosine phosphatase in B-1 B cells. Eur J Immunol, 29(10), 3319-3328. doi: 10.1002/(SICI)1521-4141(199910)29:10<3319::AID-IMMU3319>3.0.CO;2-9

Serrano, D., Monteiro, J., Allen, S. L., Kolitz, J., Schulman, P., Lichtman, S.

M., . . . Gregersen, P. K. (1997). Clonal expansion within the CD4+CD57+ and CD8+CD57+ T cell subsets in chronic lymphocytic leukemia. J Immunol, 158(3), 1482-1489.

Page 209: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

190

Shaffer, A. L., Rosenwald, A., & Staudt, L. M. (2002). Lymphoid malignancies: the dark side of B-cell differentiation. Nat Rev Immunol, 2(12), 920-932. doi: 10.1038/nri953

Shelly, S., Boaz, M., & Orbach, H. (2012). Prolactin and autoimmunity.

Autoimmun Rev, 11(6-7), A465-470. doi: 10.1016/j.autrev.2011.11.009 Shim, Y. K., Middleton, D. C., Caporaso, N. E., Rachel, J. M., Landgren, O.,

Abbasi, F., . . . Vogt, R. F. (2010). Prevalence of monoclonal B-cell lymphocytosis: a systematic review. Cytometry B Clin Cytom, 78 Suppl 1, S10-18. doi: 10.1002/cyto.b.20538

Shimoni, A., Marcus, H., Canaan, A., Ergas, D., David, M., Berrebi, A., &

Reisner, Y. (1997). A model for human B-chronic lymphocytic leukemia in human/mouse radiation chimera: evidence for tumor-mediated suppression of antibody production in low-stage disease. Blood, 89(6), 2210-2218.

Shivakumar, S., Tsokos, G. C., & Datta, S. K. (1989). T cell receptor

alpha/beta expressing double-negative (CD4-/CD8-) and CD4+ T helper cells in humans augment the production of pathogenic anti-DNA autoantibodies associated with lupus nephritis. J Immunol, 143(1), 103-112.

Shivapurkar, N., Harada, K., Reddy, J., Scheuermann, R. H., Xu, Y.,

McKenna, R. W., . . . Gazdar, A. F. (2002). Presence of simian virus 40 DNA sequences in human lymphomas. Lancet, 359(9309), 851-852. doi: 10.1016/S0140-6736(02)07921-7

Shivapurkar, N., Takahashi, T., Reddy, J., Zheng, Y., Stastny, V., Collins, R.,

. . . Gazdar, A. F. (2004). Presence of simian virus 40 DNA sequences in human lymphoid and hematopoietic malignancies and their relationship to aberrant promoter methylation of multiple genes. Cancer Res, 64(11), 3757-3760. doi: 10.1158/0008-5472.CAN-03-3307

Shlomchik, M. J., & Weisel, F. (2012). Germinal center selection and the

development of memory B and plasma cells. Immunol Rev, 247(1), 52-63. doi: 10.1111/j.1600-065X.2012.01124.x

Siegel, R., Naishadham, D., & Jemal, A. (2013). Cancer statistics, 2013. CA

Cancer J Clin, 63(1), 11-30. doi: 10.3322/caac.21166 Simmons, D. G., Rawn, S., Davies, A., Hughes, M., & Cross, J. C. (2008).

Spatial and temporal expression of the 23 murine Prolactin/Placental Lactogen-related genes is not associated with their position in the

Page 210: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

191

locus. BMC Genomics, 9, 352. doi: 10.1186/1471-2164-9-352 Sims, G. P., Ettinger, R., Shirota, Y., Yarboro, C. H., Illei, G. G., & Lipsky, P.

E. (2005). Identification and characterization of circulating human transitional B cells. Blood, 105(11), 4390-4398. doi: 10.1182/blood-2004-11-4284

Sinha, A. A., Lopez, M. T., & McDevitt, H. O. (1990). Autoimmune diseases:

the failure of self tolerance. Science, 248(4961), 1380-1388. Sinisalo, M., Aittoniemi, J., Oivanen, P., Käyhty, H., Olander, R. M., & Vilpo,

J. (2001). Response to vaccination against different types of antigens in patients with chronic lymphocytic leukaemia. Br J Haematol, 114(1), 107-110.

Sivina, M., Hartmann, E., Vasyutina, E., Boucas, J. M., Breuer, A., Keating,

M. J., . . . Burger, J. A. (2012). Stromal cells modulate TCL1 expression, interacting AP-1 components and TCL1-targeting micro-RNAs in chronic lymphocytic leukemia. Leukemia, 26(8), 1812-1820. doi: 10.1038/leu.2012.63

Skowronska, A., Austen, B., Powell, J. E., Weston, V., Oscier, D. G., Dyer, M.

J., . . . Stankovic, T. (2012). ATM germline heterozygosity does not play a role in chronic lymphocytic leukemia initiation but influences rapid disease progression through loss of the remaining ATM allele. Haematologica, 97(1), 142-146. doi: 10.3324/haematol.2011.048827

Slager, S. L., Goldin, L. R., Strom, S. S., Lanasa, M. C., Spector, L. G.,

Rassenti, L., . . . Cerhan, J. R. (2010). Genetic susceptibility variants for chronic lymphocytic leukemia. Cancer Epidemiol Biomarkers Prev, 19(4), 1098-1102. doi: 10.1158/1055-9965.EPI-09-1217

Soma, L. A., Craig, F. E., & Swerdlow, S. H. (2006). The proliferation center

microenvironment and prognostic markers in chronic lymphocytic leukemia/small lymphocytic lymphoma. Hum Pathol, 37(2), 152-159. doi: 10.1016/j.humpath.2005.09.029

Spitaler, M., & Cantrell, D. A. (2004). Protein kinase C and beyond. Nat

Immunol, 5(8), 785-790. doi: 10.1038/ni1097 Stall, A. M., Fariñas, M. C., Tarlinton, D. M., Lalor, P. A., Herzenberg, L. A., &

Strober, S. (1988). Ly-1 B-cell clones similar to human chronic lymphocytic leukemias routinely develop in older normal mice and young autoimmune (New Zealand Black-related) animals. Proc Natl Acad Sci U S A, 85(19), 7312-7316.

Page 211: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

192

Stamatopoulos, K., Belessi, C., Moreno, C., Boudjograh, M., Guida, G., Smilevska, T., . . . Davi, F. (2007). Over 20% of patients with chronic lymphocytic leukemia carry stereotyped receptors: Pathogenetic implications and clinical correlations. Blood, 109(1), 259-270. doi: 10.1182/blood-2006-03-012948

Stavnezer, J., Guikema, J. E., & Schrader, C. E. (2008). Mechanism and

regulation of class switch recombination. Annu Rev Immunol, 26, 261-292. doi: 10.1146/annurev.immunol.26.021607.090248

Stein, J. V., López-Fraga, M., Elustondo, F. A., Carvalho-Pinto, C. E.,

Rodríguez, D., Gómez-Caro, R., . . . Hahne, M. (2002). APRIL modulates B and T cell immunity. J Clin Invest, 109(12), 1587-1598. doi: 10.1172/JCI15034

Stevenson, F. K., Krysov, S., Davies, A. J., Steele, A. J., & Packham, G.

(2011). B-cell receptor signaling in chronic lymphocytic leukemia. Blood, 118(16), 4313-4320. doi: 10.1182/blood-2011-06-338855

Sthoeger, Z. M., Wakai, M., Tse, D. B., Vinciguerra, V. P., Allen, S. L.,

Budman, D. R., . . . Chiorazzi, N. (1989). Production of autoantibodies by CD5-expressing B lymphocytes from patients with chronic lymphocytic leukemia. J Exp Med, 169(1), 255-268.

Stratowa, C., Löffler, G., Lichter, P., Stilgenbauer, S., Haberl, P., Schweifer,

N., . . . Wilgenbus, K. K. (2001). CDNA microarray gene expression analysis of B-cell chronic lymphocytic leukemia proposes potential new prognostic markers involved in lymphocyte trafficking. Int J Cancer, 91(4), 474-480.

Suljagic, M., Longo, P. G., Bennardo, S., Perlas, E., Leone, G., Laurenti, L.,

& Efremov, D. G. (2010). The Syk inhibitor fostamatinib disodium (R788) inhibits tumor growth in the Eμ- TCL1 transgenic mouse model of CLL by blocking antigen-dependent B-cell receptor signaling. Blood, 116(23), 4894-4905. doi: 10.1182/blood-2010-03-275180

Swerdlow, S. H., Murray, L. J., Habeshaw, J. A., & Stansfeld, A. G. (1984).

Lymphocytic lymphoma/B-chronic lymphocytic leukaemia--an immunohistopathological study of peripheral B lymphocyte neoplasia. Br J Cancer, 50(5), 587-599.

Szatmari, I., Pap, A., Rühl, R., Ma, J. X., Illarionov, P. A., Besra, G. S., . . .

Nagy, L. (2006). PPARgamma controls CD1d expression by turning on retinoic acid synthesis in developing human dendritic cells. J Exp Med, 203(10), 2351-2362. doi: 10.1084/jem.20060141

Page 212: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

193

Takahashi, T., & Strober, S. (2008). Natural killer T cells and innate immune B cells from lupus-prone NZB/W mice interact to generate IgM and IgG autoantibodies. Eur J Immunol, 38(1), 156-165. doi: 10.1002/eji.200737656

Teitell, M. A. (2005). The TCL1 family of oncoproteins: co-activators of

transformation. Nat Rev Cancer, 5(8), 640-648. doi: 10.1038/nrc1672 ter Brugge, P. J., Ta, V. B., de Bruijn, M. J., Keijzers, G., Maas, A., van Gent,

D. C., & Hendriks, R. W. (2009). A mouse model for chronic lymphocytic leukemia based on expression of the SV40 large T antigen. Blood, 114(1), 119-127. doi: 10.1182/blood-2009-01-198937

Thornton, P. D., Bellas, C., Santon, A., Shah, G., Pocock, C., Wotherspoon,

A. C., . . . Catovsky, D. (2005). Richter's transformation of chronic lymphocytic leukemia. The possible role of fludarabine and the Epstein-Barr virus in its pathogenesis. Leuk Res, 29(4), 389-395. doi: 10.1016/j.leukres.2004.09.008

Thorsélius, M., Kröber, A., Murray, F., Thunberg, U., Tobin, G., Bühler, A., . .

. Rosenquist, R. (2006). Strikingly homologous immunoglobulin gene rearrangements and poor outcome in VH3-21-using chronic lymphocytic leukemia patients independent of geographic origin and mutational status. Blood, 107(7), 2889-2894. doi: 10.1182/blood-2005-06-2227

Tiegs, S. L., Russell, D. M., & Nemazee, D. (1993). Receptor editing in self-

reactive bone marrow B cells. J Exp Med, 177(4), 1009-1020. Tonegawa, S. (1983). Somatic generation of antibody diversity. Nature,

302(5909), 575-581. Toze, C. L., Dalal, C. B., Nevill, T. J., Gillan, T. L., Abou Mourad, Y. R.,

Barnett, M. J., . . . Shepherd, J. D. (2012). Allogeneic haematopoietic stem cell transplantation for chronic lymphocytic leukaemia: outcome in a 20-year cohort. Br J Haematol, 158(2), 174-185. doi: 10.1111/j.1365-2141.2012.09170.x

Tsimberidou, A. M., & Keating, M. J. (2006). Richter's transformation in

chronic lymphocytic leukemia. Semin Oncol, 33(2), 250-256. doi: 10.1053/j.seminoncol.2006.01.016

Tsubata, T., & Reth, M. (1990). The products of pre-B cell-specific genes

(lambda 5 and VpreB) and the immunoglobulin mu chain form a complex that is transported onto the cell surface. J Exp Med, 172(3), 973-976.

Page 213: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

194

Tsukada, N., Burger, J. A., Zvaifler, N. J., & Kipps, T. J. (2002). Distinctive

features of "nurselike" cells that differentiate in the context of chronic lymphocytic leukemia. Blood, 99(3), 1030-1037.

Van Bockstaele, F., Verhasselt, B., & Philippé, J. (2009). Prognostic markers

in chronic lymphocytic leukemia: a comprehensive review. Blood Rev, 23(1), 25-47. doi: 10.1016/j.blre.2008.05.003

Victora, G. D., & Nussenzweig, M. C. (2012). Germinal centers. Annu Rev

Immunol, 30, 429-457. doi: 10.1146/annurev-immunol-020711-075032 Virchow, R. (1845). Weisses Blut. Neue Notizen aus dem Gebiete der Natur-

und Heilkunde, 36, 151-156. Virchow, R. (1847). Zur pathologischen Physiologie des Bluts. II. Weisses

Blut (Leukämie). Virchows Arch. Path. Anat, 1, 563-572. Virgilio, L., Lazzeri, C., Bichi, R., Nibu, K., Narducci, M. G., Russo, G., . . .

Croce, C. M. (1998). Deregulated expression of TCL1 causes T cell leukemia in mice. Proc Natl Acad Sci U S A, 95(7), 3885-3889.

Virgilio, L., Narducci, M. G., Isobe, M., Billips, L. G., Cooper, M. D., Croce, C.

M., & Russo, G. (1994). Identification of the TCL1 gene involved in T-cell malignancies. Proc Natl Acad Sci U S A, 91(26), 12530-12534.

Vogler, M., Butterworth, M., Majid, A., Walewska, R. J., Sun, X. M., Dyer, M.

J., & Cohen, G. M. (2009). Concurrent up-regulation of BCL-XL and BCL2A1 induces approximately 1000-fold resistance to ABT-737 in chronic lymphocytic leukemia. Blood, 113(18), 4403-4413. doi: 10.1182/blood-2008-08-173310

Waddington, J. L. (1986). Bromocriptine, selective D2 dopamine receptor

agonists and D1 dopaminergic tone. Trends Pharmacol Sci., 7, 223-224.

Waldschmidt, T., Snapp, K., Foy, T., Tygrett, L., & Carpenter, C. (1992). B-

cell subsets defined by the Fc epsilon R. Ann N Y Acad Sci, 651, 84-98.

Wang, H., Feng, J., Qi, C. F., Li, Z., Morse, H. C., & Clarke, S. H. (2007).

Transitional B cells lose their ability to receptor edit but retain their potential for positive and negative selection. J Immunol, 179(11), 7544-7552.

Wang, J., Coombes, K. R., Highsmith, W. E., Keating, M. J., & Abruzzo, L. V.

Page 214: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

195

(2004). Differences in gene expression between B-cell chronic lymphocytic leukemia and normal B cells: a meta-analysis of three microarray studies. Bioinformatics, 20(17), 3166-3178. doi: 10.1093/bioinformatics/bth381

Wardemann, H., Yurasov, S., Schaefer, A., Young, J. W., Meffre, E., &

Nussenzweig, M. C. (2003). Predominant autoantibody production by early human B cell precursors. Science, 301(5638), 1374-1377. doi: 10.1126/science.1086907

Watson, L., Wyld, P., & Catovsky, D. (2008). Disease burden of chronic

lymphocytic leukaemia within the European Union. Eur J Haematol, 81(4), 253-258. doi: 10.1111/j.1600-0609.2008.01114.x

Weill, J. C., Weller, S., & Reynaud, C. A. (2009). Human marginal zone B

cells. Annu Rev Immunol, 27, 267-285. doi: 10.1146/annurev.immunol.021908.132607

Weiss, L., Melchardt, T., Egle, A., Grabmer, C., Greil, R., & Tinhofer, I.

(2011). Regulatory T cells predict the time to initial treatment in early stage chronic lymphocytic leukemia. Cancer, 117(10), 2163-2169. doi: 10.1002/cncr.25752

Weinkove, R., Brooks, C. R., Carter, J. M., Hermans, I. F., & Ronchese, F.

(2013). Functional invariant natural killer T-cell and CD1d axis in chronic lymphocytic leukemia: implications for immunotherapy. Haematologica, 98(3), 376-384. doi: 10.3324/haematol.2012.072835

Wen, L., Brill-Dashoff, J., Shinton, S. A., Asano, M., Hardy, R. R., &

Hayakawa, K. (2005). Evidence of marginal-zone B cell-positive selection in spleen. Immunity, 23(3), 297-308. doi: 10.1016/j.immuni.2005.08.007

Wen, X., Zhang, D., Kikuchi, Y., Jiang, Y., Nakamura, K., Xiu, Y., . . . Hirose,

S. (2004). Transgene-mediated hyper-expression of IL-5 inhibits autoimmune disease but increases the risk of B cell chronic lymphocytic leukemia in a model of murine lupus. Eur J Immunol, 34(10), 2740-2749. doi: 10.1002/eji.200425267

Whiteside, T. L. (2008). The tumor microenvironment and its role in

promoting tumor growth. Oncogene, 27(45), 5904-5912. doi: 10.1038/onc.2008.271

Widhopf, G. F., Rassenti, L. Z., Toy, T. L., Gribben, J. G., Wierda, W. G., &

Kipps, T. J. (2004). Chronic lymphocytic leukemia B cells of more than 1% of patients express virtually identical immunoglobulins. Blood,

Page 215: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

196

104(8), 2499-2504. doi: 10.1182/blood-2004-03-0818 Wiemers, D. O., Shao, L. J., Ain, R., Dai, G., & Soares, M. J. (2003). The

mouse prolactin gene family locus. Endocrinology, 144(1), 313-325. Wilson, J. D., & Nossal, G. J. (1971). Identification of human T and B

lymphocytes in normal peripheral blood and in chronic lymphocytic leukaemia. Lancet, 2(7728), 788-791.

Wintrobe, M. M., & Hasenbush, L. L. (1939). Chronic leukemia: the early

phase of chronic leukemia, the results of treatment and effects of complicating infections; a study of eighty-six adults. Arch Intern Med, 64(4), 701-718.

Woyach, J. A., Johnson, A. J., & Byrd, J. C. (2012). The B-cell receptor

signaling pathway as a therapeutic target in CLL. Blood, 120(6), 1175-1184. doi: 10.1182/blood-2012-02-362624

Wu, D., Xing, G. W., Poles, M. A., Horowitz, A., Kinjo, Y., Sullivan, B., . . .

Wong, C. H. (2005). Bacterial glycolipids and analogs as antigens for CD1d-restricted NKT cells. Proc Natl Acad Sci U S A, 102(5), 1351-1356. doi: 10.1073/pnas.0408696102

Wu, Q. L., Zierold, C., & Ranheim, E. A. (2009). Dysregulation of Frizzled 6 is

a critical component of B-cell leukemogenesis in a mouse model of chronic lymphocytic leukemia. Blood, 113(13), 3031-3039. doi: 10.1182/blood-2008-06-163303

Xu, J. L., & Davis, M. M. (2000). Diversity in the CDR3 region of V(H) is

sufficient for most antibody specificities. Immunity, 13(1), 37-45. Xu, Y., Harder, K. W., Huntington, N. D., Hibbs, M. L., & Tarlinton, D. M.

(2005). Lyn tyrosine kinase: accentuating the positive and the negative. Immunity, 22(1), 9-18. doi: 10.1016/j.immuni.2004.12.004

Yan, X. J., Albesiano, E., Zanesi, N., Yancopoulos, S., Sawyer, A., Romano,

E., . . . Chiorazzi, N. (2006). B cell receptors in TCL1 transgenic mice resemble those of aggressive, treatment-resistant human chronic lymphocytic leukemia. Proc Natl Acad Sci U S A, 103(31), 11713-11718. doi: 10.1073/pnas.0604564103

Youinou, P., Hillion, S., Jamin, C., Pers, J. O., Saraux, A., & Renaudineau, Y.

(2006). B lymphocytes on the front line of autoimmunity. Autoimmun Rev, 5(3), 215-221. doi: 10.1016/j.autrev.2005.06.011

Youinou, P., Jamin, C., & Lydyard, P. M. (1999). CD5 expression in human

Page 216: ACCELERATED PROGRESSION OF CHRONIC LYMPHOCYTIC

197

B-cell populations. Immunol Today, 20(7), 312-316. Yuille, M. R., Matutes, E., Marossy, A., Hilditch, B., Catovsky, D., & Houlston,

R. S. (2000). Familial chronic lymphocytic leukaemia: a survey and review of published studies. Br J Haematol, 109(4), 794-799.

Zanesi, N., Aqeilan, R., Drusco, A., Kaou, M., Sevignani, C., Costinean, S., . .

. Croce, C. M. (2006). Effect of rapamycin on mouse chronic lymphocytic leukemia and the development of nonhematopoietic malignancies in Emu-TCL1 transgenic mice. Cancer Res, 66(2), 915-920. doi: 10.1158/0008-5472.CAN-05-3426

Zapata, J. M., Krajewska, M., Krajewski, S., Kitada, S., Welsh, K., Monks, A.,

. . . Reed, J. C. (2000). TNFR-associated factor family protein expression in normal tissues and lymphoid malignancies. J Immunol, 165(9), 5084-5096.

Zapata, J. M., Krajewska, M., Morse, H. C., Choi, Y., & Reed, J. C. (2004).

TNF receptor-associated factor (TRAF) domain and Bcl-2 cooperate to induce small B cell lymphoma/chronic lymphocytic leukemia in transgenic mice. Proc Natl Acad Sci U S A, 101(47), 16600-16605. doi: 10.1073/pnas.0407541101

Zent, C. S., & Kay, N. E. (2010). Autoimmune complications in chronic

lymphocytic leukaemia (CLL). Best Pract Res Clin Haematol, 23(1), 47-59. doi: 10.1016/j.beha.2010.01.004

Zenz, T., Eichhorst, B., Busch, R., Denzel, T., Häbe, S., Winkler, D., . . .

Stilgenbauer, S. (2010). TP53 mutation and survival in chronic lymphocytic leukemia. J Clin Oncol, 28(29), 4473-4479. doi: 10.1200/JCO.2009.27.8762

Zenz, T., Mertens, D., Döhner, H., & Stilgenbauer, S. (2011). Importance of

genetics in chronic lymphocytic leukemia. Blood Rev, 25(3), 131-137. doi: 10.1016/j.blre.2011.02.002

Zhang, Z. X., Yang, L., Young, K. J., DuTemple, B., & Zhang, L. (2000).

Identification of a previously unknown antigen-specific regulatory T cell and its mechanism of suppression. Nat Med, 6(7), 782-789. doi: 10.1038/77513

Zheng, S. G., Wang, J. H., Gray, J. D., Soucier, H., & Horwitz, D. A. (2004).

Natural and induced CD4+CD25+ cells educate CD4+CD25- cells to develop suppressive activity: the role of IL-2, TGF-beta, and IL-10. J Immunol, 172(9), 5213-5221.