NOVEL IMMUNOTHERAPEUTIC STRATEGIES FOR

CHRONIC LYMPHOCYTIC LEUKEMIA

DISSERTATION

Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University

By

Kyle Beckwith, B.S.

Graduate Program in Biomedical Sciences

The Ohio State University

2016

Dissertation Committee:

John Byrd MD (Co-Advisor)

Natarajan Muthusamy DVM, PhD (Co-Advisor)

Michael Caligiuri MD

Abhay Satoskar MD, PhD

Copyright by

Kyle A. Beckwith

2016

ABSTRACT

Chronic lymphocytic leukemia (CLL) is malignancy characterized by the progressive accumulation of neoplastic B-cells that are phenotypically mature, but functionally incompetent. It is the most prevalent adult leukemia in the United States, and has an incidence of approximately 18,000 new cases per year. The introduction of

CD20-targeted monoclonal antibody rituximab dramatically altered how this disease is managed, and immunotherapeutic strategies have become a mainstay of CLL therapy ever since. Newer immunotherapies approved by the FDA include anti-CD52 alemtuzumab and two anti-CD20 antibodies with improved activity. Due to expression of

CD52 on T-cells, alemtuzumab therapy puts an already immunocompromised population at greater risk for infection (which is already the most common cause of CLL patient death). Unfortunately current therapeutics are not curative, and resistance/relapse is inevitable. Given the limitations of these therapies, particularly when confronted with the challenge of treating relapsed/refractory disease, it is important to expand our immunotherapeutic arsenal beyond targeting CD20. Therefore, the overall aim of this thesis is to develop novel approaches to CLL immunotherapy that can be successfully translated to the clinic. One of the most promising alternative targets for immunotherapy is CD37. This is largely B-cell restricted and highly expressed in CLL and Non-Hodgkins Lymphoma. Furthermore, the single agent clinical activity of anti-

CD37 otlertuzumab in CLL validates this therapeutic target. A number of CD37-targeted therapies have been developed recently, but our ability to study them preclinically is significantly hampered by the lack of suitable animal models that can accurately

ii recapitulate CLL. Ideally we would use the TCL1 mouse model of CLL, which has been extensively characterized as a drug development tool, but the antibodies targeting human CD37 (hCD37) do not cross react with murine CD37. This problem was addressed by developing a model of murine leukemia expressing hCD37, crossing hCD37 transgenic mice developed by our laboratory with the TCL1 mouse. These hCD37xTCL1 mice facilitated the in vivo study of IMGN529, a CD37-targeting antibody- drug conjugate. Using our new model, I demonstrate that IMGN529 can not only improve overall survival, but is also capable of eliminating the proliferative subset of CLL B-cells

within lymphoid tissues. I go on to investigate combination strategies that can improve

the efficacy of anti-CD37 therapeutics, including addition of a phosphoinositide 3-kinase

(PI3K) inhibitor, which improves cytotoxicity against primary CLL B-cells (and is equally

effective against cells from high risk patients). I also investigate whether a CD37-

targeted bispecific antibody can effectively recruit T-cells to kill malignant B-cells, despite

the profound T-cell defects observed in CLL. Unfortunately, it appears to be inferior to

bispecific antibody formats and is only effective at high effector-to-target ratios. Finally, I

use a model of CD37-deficient CLL to explore the function of this tetraspanin in B-cell

malignancy. These mice exhibit decreased survival that suggests a tumor suppressor

role in this context. Overall, this thesis represents a comprehensive inquiry into both the

function of CD37, and a wide variety of approaches to targeting CD37 which provide

valuable information relevant to future clinical trials.

iii

DEDICATION

This dissertation is dedicated to my family and to my current and former mentors.

iv

ACKNOWLEDGMENTS

First, I would like to acknowledge Dr. Michael Uhler for his mentorship while I was at the University of Michigan. His trust and encouragement to seek out answers to my own questions fostered a love of research I did not discover until very late in college.

Without his mentorship and the (relative) freedom he afforded me, it is unlikely I would have continued to pursue laboratory research. This experience encouraged me to continue to seek out research opportunities in medical school, which led me to Dr. Phillip

Popovich’s laboratory. The short time I spent in this lab working closely with John

Gensel ultimately prompted me to delay entrance to Med 3 to pursue additional research opportunities.

I would like express my deepest thanks to my mentors Dr. John Byrd and Dr. Raj

Muthusamy for their guidance and support. Without the opportunity to pursue a

Pelotonia fellowship in the lab, I would not be seeking a PhD. Your neverending enthusiasm for science and mentorship inspires me. If I train future students half as well as you have, it would be considered a major success. To all members of the Byrd lab

(past and present) who have helped or provided insight along the way – thank you.

I am grateful to many faculty members at OSU for providing their expertise and support, but I would like to specifically extend thanks to Dr. Michael Caligiuri and Dr.

Abhay Satoskar for taking the time to be part of my committee. Thank you for your support and feedback throughout this entire process.

v

I would also like to thank my parents for their amazing support over the many years of my education. Finally, I am especially thankful to my brilliant wife Puneet for her endless support, insight, patience, and encouragement. You make me strive to be a better person and scientist.

vi

VITA

2009 ...... B.S. Biochemistry, with distinction

University of Michigan

2009 to present ...... Graduate/Medical Student

Ohio State College of Medicine

Biomedical Sciences Graduate Program

PUBLICATIONS

1. *Fraietta JA, *Beckwith KA, Patel PR, Ruella M, Zheng Z, Barrett DM, Lacey SF, Melenhorst JJ, McGettigan SE Cook DR, Zhang C, Xu J, Do P, Hulitt J, Kudchodkar SB, Cogdill AP, Gill S, Porter DL, Woyach JA, Long M, Johnson AJ, Maddocks K, Muthusamy N, Levine BL, June CH, *Byrd JC and *Maus MV. Ibrutinib Enhances Chimeric Antigen Receptor T-cell Engraftment and Efficacy in Leukemia. Blood. Pre-published online January 26, 2016. doi: http://dx.doi.org/10.1182/blood-2015-11-679134. Asterisk (*) indicates equal contribution by these authors. 2. Gensel JC, Wang Y, Guan Z, Beckwith KA, Braun KJ, Wei P, McTigue DM, Popovich PG. Toll-Like Receptors and Dectin-1, a C-Type Lectin Receptor, Trigger Divergent Functions in CNS Macrophages. J Neurosci. 35(27):9966-76, 2015. doi: 10.1523/JNEUROSCI.0337-15.2015 3. Beckwith KA, Byrd JC, and Muthusamy N. as therapeutic targets in hematological malignancy: a concise review. Front. Physiol. 6:91, 2015. doi: 10.3389/fphys.2015.00091 [Review article] 4. Hing ZA, Mantel R, Beckwith KA, Guinn D, Williams E, Smith L, Williams KE, Johnson AJ, Lehman A, Byrd JC, Woyach JA, Lapalombella R. Selinexor is effective in acquired resistance to ibrutinib and synergizes with ibrutinib in chronic lymphocytic leukemia. Blood. 125(20):3128-32, 2015. doi: 10.1182/blood- 2015-01-621391

vii

5. Liu T, Ling Y, Woyach JA, Beckwith K, Yeh Y, Hertlein E, Zhang X, Lehman A, Awan F, Jones JA, Andritsos LA, Maddocks K, MacMurray J, Salunke SB, Chen C, Phelps MA, Byrd JC, Johnson AJ. OSU-T315: a novel targeted therapeutic that antagonizes AKT membrane localization and activation of chronic lymphocytic leukemia cells. Blood. 125(2):284-95, 2015. doi: 10.1182/blood- 2014-06-583518 6. Beckwith KA, Frissora FW, Stefanovski MR, Towns WH, Cheney C, Mo X, Deckert J, Croce CM, Flynn JM, Andritsos LA, Jones JA, Maddocks KJ, Lozanski G, Byrd JC, and Muthusamy N. The CD37-targeting antibody-drug conjugate IMGN529 is highly active against human CLL and in a novel CD37 transgenic murine leukemia model. Leukemia. 28(7): 1501-10, 2014. 7. Zhong Y, El-Gamal D, Dubovsky JA, Beckwith KA, Harrington BK, Williams KE, Goettl VM, Jha S, Mo X, Jones JA, Flynn JM, Maddocks KJ, Andritsos LA, McCauley D, Shacham S, Kauffman M, Byrd JC, Lapalombella R. Selinexor suppresses downstream effectors of B-cell activation, proliferation and migration in chronic lymphocytic leukemia cells. Leukemia. 28(5): 1158-63, 2014. 8. Hertlein E, Beckwith KA, Lozanski G, Chen TL, Towns, WH, Johnson AJ, Lehman A, Ruppert AS, Bolon, B, Lozanski A, Rassenti L, Zhao W, Jarvinen T, Senter L, Croce CM, Symer DE, de la Chapelle A, Heerema N, Byrd JC. Characterization of a new chronic lymphocytic leukemia cell line for mechanistic in vitro and in vivo studies relevant to disease. PLoS One. 8(10), 2013. 9. Dubovsky JA, Beckwith KA, Natarajan G, Woyach JA, Jaglowski S, Zhong Y, Hessler JD, Liu T, Chang BY, Larkin KM, Stefanovski MR, Frissora FW, Smith LL, Smucker KA, Flynn JM, Jones JA, Andritsos LA, Maddocks KJ, Lehman AM, Furman R, Sharman J, Mishra A, Caligiuri MA, Satoskar AR, Buggy JJ, Muthusamy N, Johnson AJ, and Byrd JC. Ibrutinib is an irreversible molecular inhibitor of ITK driving a Th1 selective pressure in T-lymphocytes. Blood. 122(15): 2539-49, 2013.

FIELDS OF STUDY

Major Field: Biomedical Sciences

Specialization: Immunology

viii

TABLE OF CONTENTS

Abstract ...... ii

Dedication ...... iv

Acknowledgments ...... v

Vita ...... vii

List of Figures ...... x

Chapters:

1. Introduction ...... 1

2. A Mouse Model of Human CD37-Expressing Leukemia ...... 19

3. A CD37-Specific Antibody-Drug Conjugate for B-cell Malignancy ...... 30

4. Developing Strategies to Enhance Anti-CD37 Therapy ...... 60

5. Bispecific Antibodies: Bringing T-cells into the Fold ...... 73

6. Exploring the Function of CD37 in Leukemia ...... 89

7. Conclusions ...... 96

List of References ...... 107

ix

LIST OF FIGURES

Figure Page

1.1 Structural features of tetraspanins ...... 15

1.2 Signaling pathway associated with CD37 ligation by SMIP-016 ...... 16

1.3 CD37-targeted antibody therapeutics ...... 18

2.1 Generation and characterization of human CD37 transgenic mouse ...... 25

2.2 Human CD37 transgenic B-cells are sensitive to anti-CD37 therapy ...... 27

2.3 hCD37xTCL1 mice retain expression of both transgenes and develop CLL ...... 28

3.1 IMGN529 is directly cytotoxic to primary human CLL B-cells ...... 44

3.2 IMGN529 retains the Fc-mediated activities of its unconjugated precursor ...... 46

3.3 Gating strategy for measuring phagocytosis of CLL B-cells...... 48

3.4 Gating strategy for whole blood cytotoxicity assays ...... 50

3.5 IMGN529 demonstrates exceptional cytotoxicity in CLL whole blood ...... 52

3.6 DM1-conjugation improves killing and arrests proliferating B-cells ...... 54

3.7 IMGN529 demonstrates in vivo efficacy against hCD37+ murine leukemia ...... 55

3.8 IMGN529 eliminates splenomegaly and demonstrates CD37-specificity ...... 57

3.9 IMGN529 targets proliferating mouse leukemia cells within lymphoid tissues ..... 58

4.1 Combining TLR 7/8 agonist CL-075 with TRU-016 is not beneficial ...... 68

4.2 BI 836826 effector mediated killing ...... 70

4.3 Combining BI 836826 with idelalisib improves cytotoxicity ...... 71

4.4 Preliminary evaluation of BI 836826 in hCD37 leukemia model ...... 72

x

5.1 IgG-derived therapeutics constructed from scFV ...... 79

5.2 TSC-TRU recruits and activates T-cells ...... 81

5.3 Cytotoxicity of TSC-TRU with allogeneic or autologous T-cells ...... 82

5.4 T-cell activation by TSC-TRU in CLL PBMC cultures ...... 83

5.5 CLL B-cells evade killing by TSC-TRU in 48 hour PBMC cultures ...... 84

5.6 Longer incubation has minimal impact on TSC-TRU efficacy in PBMCs ...... 86

5.7 Gating strategy for PBMC co-culture experiments ...... 88

6.1 Confirmation of CD37 knockout ...... 93

6.2 CD37 deficiency reduces TCL1 mouse survival ...... 94

6.3 Measurement of leukemic burden in peripheral blood ...... 95

xi

CHAPTER 1

INTRODUCTION

Chronic Lymphocytic Leukemia (CLL)

Chronic lymphocytic leukemia (CLL) is a malignancy characterized by the progressive accumulation of neoplastic B-cells that are phenotypically mature, but functionally incompetent. It is the most prevalent adult leukemia in the United States, and has an incidence of approximately 18,000 new cases per year. Typically, CLL is diagnosed between the ages of 65-74 and is more common in men than women (white men being the most commonly afflicted group). (Siegel et al., 2013)

The highly heterogeneous nature of CLL has substantial impact on how this disease is managed clinically. While patients who are asymptomatic at diagnosis have a median survival of greater than 10 years (often permitting a “watch and wait” approach), the prognosis of those with symptomatic or advanced stage CLL is between 1.5 and 3 years without treatment (Rai et al., 1975). In the years that followed FDA approval of the anti-CD20 monoclonal antibody (mAb) rituximab, chemoimmunotherapy combining mAbs with traditional chemotherapeutics became the favored approach for initial treatment of patients with active disease (Woyach et al., 2012; Gribben, 2010). A regimen that adds rituximab to fludarabine and cyclophosphamide (FCR) improves both progression-free survival (PFS) and overall survival (OS) in previously untreated CLL patients (Hallek et al., 2008), although FCR still benefits patients with relapsed disease

(Wierda et al., 2005).

1

Despite the impact made by rituximab-based immunotherapy, it is not curative and resistance/relapse is inevitable (Woyach et al., 2011; Rezvani and Maloney, 2011).

In the years following rituximab’s success, several other mAb therapies have received

FDA approval for the treatment of CLL. These include the anti-CD52 mAb alemtuzumab and two CD20-targeting mAbs with improved activity (ofatumumab and obinatuzumab).

Alemtuzumab is a very potent mAb therapeutic, but unfortunately causes profound immunosuppression due to the elimination of CD52 expressing T-cells. While still used in certain cases, a delicate balance must be struck as infection is already the most common cause of death among CLL patients. Due to the limitations of these therapies, particularly when confronted with the challenge of treating relapsed/refractory disease, it is important to expand our immunotherapeutic arsenal beyond targeting CD20. This demand has been the motivation for the work presented herein, which focuses on the tetraspanin CD37 as an alternative target for immunotherapy.

Tetraspanin

The tetraspanin superfamily is comprised of at least 33 members in humans, and these transmembrane proteins are ubiquitous among metazoans (Maecker et al., 1997.

Many of the tetraspanins present on immune cells are also found in a variety of other tissues, but some display hematopoietic-restricted expression, including CD37, CD53,

Tssc6 (TSPAN32), and TSPAN33 (Tarrant et al., 2003; Heikens et al., 2007). The predominant view has been that tetraspanins are facilitators of signal transduction, providing organization to plasma membrane domains through lateral interaction with their numerous partners (Maecker et al., 1997; Hemler, 2005; Charrin et al., 2009).

However, there is recent evidence that certain tetraspanins also recruit signaling

2

proteins directly (Lapalombella et al., 2012). Tetraspanins have been reported to regulate diverse processes, including cellular migration, adhesion, activation, and apoptosis (Hemler, 2005). Furthermore, several tetraspanins influence cancer metastasis/progression and their functional roles in immune cells can impact anti-tumor immunity (Zoller, 2009; Veenbergen and van Spriel, 2011; Hemler, 2014).

Tetraspanins contain short N-terminal and C-terminal cytoplasmic tails, a small extracellular loop (EC1 domain), a large extracellular loop (EC2 domain) and four transmembrane domains (Figure 1.1). The EC2 domain contains a region conserved among tetraspanins, but also a highly variable region that is frequently involved in the specific interactions between tetraspanins and various non-tetraspanin partners (Yauch et al., 2000; Charrin et al., 2001; Shoham et al., 2006; Zevian et al., 2011). It is typical for tetraspanins to undergo extensive post-translational modification. Covalent attachment of palmitate to intracellular cysteine residues is implicated in mediating tetraspanin-tetraspanin interactions and assembly of tetraspanin-enriched domains that can support signaling (Berditchevski et al., 2002; Charrin et al., 2002; Yang et al., 2002;

Yang et al., 2004). Furthermore, nearly all tetraspanins display extensive N-linked glycosylation at extracellular sites (Maecker et al., 1997). This glycosylation is likely to have functional relevance, as shown with CD82 and CD9, which could only influence motility or apoptosis when glycosylated (Ono et al., 1999; 2000). A variety of glycosylation patterns are observed across cell lines, including those of the same lineage, but it remains unknown whether these differences have any impact on tetraspanin function (Schwartz-Albiez et al., 1988; White et al., 1998).

Numerous cis-interactions occur between tetraspanins and neighboring plasma membrane proteins within what are known as tetraspanin-enriched microdomains

3

(Hemler, 2005). These microdomains may function as signaling platforms, similar to lipid rafts but generally comprised of distinct components (Claas et al., 2001; Le Naour et al.,

2006a; Mattila et al., 2013; Zuidscherwoude et al., 2014). Many tetraspanin interactions depend on binding to the extracellular EC2 domain (Yauch et al., 2000; Charrin et al.,

2001; Shoham et al., 2006; Zevian et al., 2011), although transmembrane domains are also frequently involved (Charrin et al., 2001; Charrin et al., 2003; Shoham et al., 2006).

While numerous associations have been documented with other transmembrane proteins, there are fewer examples of tetraspanins interacting with cytoplasmic proteins.

This is not surprising, given their short cytoplasmic tails are generally less than 20 amino acids in length (Maecker et al., 1997). This has contributed to the thought that tetraspanins do not directly participate in signal transduction. However, several tetraspanins have been reported to associate with intracellular signaling proteins.

Common cytoplasmic partners include PI4K and PKC (Berditchevski et al., 1997; Zhang et al., 2001; Andre et al., 2006), but tetraspanins have also been shown to interact with several other signaling proteins (Clark et al., 2004; Little et al., 2004; Andre et al., 2006;

Le Naour et al., 2006b; Lapalombella et al., 2012). While tetraspanins clearly influence signaling, it remains possible that some of these interactions could be indirect as a result of association with adapter proteins.

Tetraspanins have been largely discounted as potential cell-surface receptors on the basis of their structure, which protrudes at most 5 nm into extracellular space

(Kitadokoro et al., 2001; Min et al., 2006). While their interactions do primarily occur in cis, recent publications have challenged the notion that tetraspanins cannot also function as receptors. CD9 is reported to have multiple soluble ligands, both acting as an alternative IL-16 receptor (Qi et al., 2006) and binding a placental protein released

4

during pregnancy (Waterhouse et al., 2002). CD81 has been identified as an essential receptor for Hepatitis C virus (Pileri et al., 1998). CD82 has been described as a receptor for an endothelial cell-surface protein, and this interaction was important for the suppression of tumor metastasis by this tetraspanin (Bandyopadhyay et al., 2006).

However, the capability of tetraspanins to bind endogenous ligands in a trans-fashion remains somewhat controversial in the field.

CD37

This protein is most highly expressed by mature B-cells, although other immune cells express CD37 to a lesser degree (Link et al., 1986; van Spriel et al., 2009; Deckert et al.,

2013). It is absent in the earliest stages of B-cell development and is lost again following differentiation into plasma cells; a pattern mirrored by B-cells malignancies originating from various developmental stages (Barrena et al., 2005). CD37 is highly expressed in mature B-cell malignancies, such as non-Hodgkin lymphoma and CLL, but is low or absent in acute lymphoblastic leukemia and multiple myeloma. The expression pattern of CD37 has led to considerable interest in targeting this tetraspanin therapeutically

(Zhao et al., 2007; Heider et al., 2011; Krause et al., 2012; Dahle et al., 2013; Deckert et al., 2013; Beckwith et al., 2014).

CD37-deficient mice exhibit defective IgG1 production in response to T-cell dependent antigens (Knobeloch et al., 2000), which is a consequence of decreased survival among IgG1-secreting B-cells in the days following antigen exposure (van Spriel et al., 2012). It was demonstrated that CD37 has an important role in clustering α4β1 integrin (also known as VLA-4) on the plasma membrane. Absence of CD37 impaired integrin-dependent Akt signaling that is typically activated through interaction with

5

follicular dendritic cells expressing a ligand of α4β1 integrin (van Spriel et al., 2012).

Similarly, ligation of CD37 by the antibody-derived peptide SMIP-016 also modulates the

Akt pathway in B-cells (Lapalombella et al., 2012). However, SMIP-016 induces both pro-apoptotic Akt inactivation and opposing pro-survival phosphoinositide 3-kinase δ

(PI3Kδ) activation (Figure 1.2). Analysis of its sequence suggested that the cytoplasmic tails of CD37 contained weak ITIM and ITAM-like motifs. Mutational studies support this function, providing evidence that the N-terminal ITIM can recruit SHP1 (which is capable of dephosphorylating/inactivating Akt) and the C-terminal ITAM can recruit PI3Kδ. While pro-survival and pro-apoptotic pathways are simultaneously induced by CD37 ligation, cellular death is favored. This may explain why several anti-CD37 therapeutics promote high degrees of apoptosis in leukemia cells (Zhao et al., 2007; Heider et al., 2011;

Krause et al., 2012; Lapalombella et al., 2012; Deckert et al., 2013; Beckwith et al.,

2014).

While the expression of CD37 is low in non-B cells (van Spriel et al., 2009;

Deckert et al., 2013), it still has important functions in T-cells, dendritic cells, and macrophages (van Spriel et al., 2004; Meyer-Wentrup et al., 2007; Sheng et al., 2009;

Gartlan et al., 2010; Gartlan et al., 2013). CD37 associates with Dectin-1 and appears to negatively regulate its activity in anti-fungal response, as IL-6 production is dramatically increased in CD37−/− macrophages following Dectin-1 stimulation (Meyer-Wentrup et al., 2007). It is possible that this regulation is accomplished through recruitment of phosphatases by CD37, which can associate with its N-terminal domain (Lapalombella et al., 2012). IL-6 production by CD37−/− cells likely supports the generation of IgA- secreting plasma cells, leading to excessive IgA secretion that ultimately provides these mice with resistance to fungal infections (van Spriel et al., 2009). CD37 plays a complex

6

role in T-cell responses, as made evident by the seemingly contradictory results of in vitro and in vivo studies. In vitro, CD37−/− dendritic cells are hyperstimulatory toward T- cells, and the data imply that CD37 negatively regulates peptide-MHC presentation

(Sheng et al., 2009; Gartlan et al., 2010). Furthermore, CD37 in T-cells may play a negative regulatory role in T-cell receptor (TCR) signaling. CD37-deficient T-cells proliferate more rapidly in response to TCR stimulation, which could be a result of decreased Lck phosphorylation (van Spriel et al., 2004). While the in vitro data imply a negative regulatory role for CD37 in T-cell responses, the opposite is observed using in vivo models. Mice deficient for CD37 are more susceptible to infection with murine malaria (Gartlan et al., 2010) and fail to reject syngeneic tumor cells transfected to express a foreign antigen (Gartlan et al., 2013). These discrepancies are explained by the observation that dendritic cells from CD37−/− mice have impaired migratory and adhesion capabilities, which clearly overshadows other potential contributions of CD37

(Gartlan et al., 2013). It remains unclear whether the hyperproliferative phenotype of

CD37-deficient T-cells is relevant beyond in vitro studies, but providing CD37−/− mice with wildtype dendritic cells did not appear to significantly increase the number of IFNγ producing T-cells relative to wildtype mice.

Anti-CD37 Therapy in Hematological Maligancy

Antibody-based strategies for treating cancer have rapidly increased in prevalence since anti-CD20 rituximab was introduced to the clinic. More than a dozen antibodies have been approved by the U.S. Food and Drug Administration (FDA) for cancer therapy and hundreds of ongoing human trials are registered at clinicaltrials.gov

(Scott et al., 2012). The first attempts to develop a CD37-targeted therapy predate the

7

approval of rituximab by nearly a decade, when 131I –labeled murine anti-CD37 antibody was tested in a small cohort of non-Hodgkin lymphoma (NHL) patients (Press et al.,

1989). While the early results were promising, targeting CD20 was quickly becoming the favored approach and CD37 was subsequently neglected for many years. Anti-CD37 therapy has experienced a recent resurgence, with five different targeting approaches being explored in B-cell malignancies. While several tetraspanins may be promising targets for cancer therapy, CD37 is by far the furthest in terms of clinical development.

CD37 is highly expressed on the surface of human B-cells (where its antigen density is at least 15 times greater than on non-B leukocytes) and it is present in the vast majority of CLL and NHL cases (Deckert et al., 2013), making it an attractive target for immunotherapy. Several CD37-targeting antibody-based therapeutics have been developed which are currently being evaluated in the clinic (Zhao et al., 2007; Heider et al., 2011; Deckert et al., 2013). Otlertuzumab was the first of these to begin clinical trials.

This therapeutic is a humanized, antibody-derived CD37-targeting peptide developed using the ADAPTIRTM platform. Mono-specific ADAPTIR molecules are built from a

single-chain variable fragment (scFv; a binding domain formed by linking the heavy and

light chain variable regions of an immunoglobulin), fused to the hinge region and Fc

domain of human IgG1 (Figure 1.3). These molecules form antibody-like dimers that are

smaller than IgG1 (intended to increase tissue penetration), but otherwise retain similar

pharmacokinetics and activity as traditional IgG1. Preclinical studies using SMIP-016, a

tool molecule not fully humanized (but containing human IgG1 Fc), demonstrated

superior NK cell-mediated antibody dependent cellular cytotoxicity (ADCC) compared to

anti-CD20 rituximab. This therapy also directly killed CLL tumor cells through induction

of caspase-independent apoptosis when in the presence of anti-Fc crosslinker (Zhao et

8

al., 2007). As discussed earlier, SMIP-016 was shown to induce both pro-apoptotic Akt inactivation and (to a lesser extent) pro-survival PI3Kδ activation (Lapalombella et al.,

2012). The simultaneous activation of these opposing signaling pathways provides an opportunity to utilize unique combination strategies for anti-CD37 therapies. Indeed,

Lapalombella et al. showed that SMIP-016 cytotoxicity against CLL B-cells was enhanced by the addition of either a pan-PI3K inhibitor (LY294002) or the PI3Kδ- selective CAL-101 (idelalisib; now FDA approved for CLL therapy). Further investigation of this potential combination is warranted, and should also be explored with newer anti-

CD37 therapies that can more efficiently induce apoptosis without dependence on additional crosslinking (Heider et al., 2011; Deckert et al., 2013).

Otlertuzumab (TRU-016) has been tested in CLL and NHL patients during recent clinical trials (Byrd et al., 2014; Gopal et al., 2014; Pagel et al., 2014). A phase I study in

CLL observed modest single-agent activity and found it to be well tolerated (Byrd et al.,

2014). Peripheral lymphocyte reduction was observed in 75.5% of patients with elevated initial lymphocyte counts, but overall response rate (ORR) was only 23% (19/83 patients) by NCI-96 criteria. Only partial responses (PR) were observed, which were more common among treatment-naïve CLL patients (6/7) or those who received 1 or 2 previous therapies (12/28). These results were encouraging, given that responses to single-agent rituximab are also limited (Byrd et al., 2001; Huhn et al., 2001), but can be dramatically improved by combination with chemotherapy (Keating et al., 2005).

Similarly, an early report from a randomized Phase II trial in relapsed CLL demonstrates

the improved efficacy of otlertuzumab when combined with bendamustine

(NCT01188681). Patients receiving otlertuzumab plus bendamustine had an ORR of

80% (16/20 patients) with 20% achieving a complete remission (CR), while those treated

9

with bendamustine alone had an ORR of only 42% (10/24) with a CR rate of 4% (Robak et al., 2013). A single-agent trial of otlertuzumab was also performed in NHL patients with follicular lymphoma, Waldenström's macroglobulinemia, or mantle cell lymphoma

(Pagel et al., 2014). Responses were limited to 2 of 16 patients (12%). However, another phase I study evaluated the combination of otlertuzumab, rituximab, and bendamustine in twelve patients with indolent NHL (follicular, mantle cell, and small lymphocytic) who had relapsed after receiving treatment regimens which included rituximab (Gopal et al.,

2014). Two doses of otlertuzumab were tested (10 or 20 mg/kg) with 6 patients per dose. This regimen was well tolerated and achieved an ORR of 83% (10/12), with four

CRs. All of the patients receiving the higher dose (6/6) responded, with 2 CRs and 4

PRs. Overall, these clinical studies highlight the promise of anti-CD37 therapies, particularly in combination with other agents.

Several newer CD37-targeted therapeutics have the potential to surpass the clinical benefits observed with otlertuzumab. To mediate ADCC, IgG1 requires covalent attachment of oligosaccharides at Asn297 within its Fc region, but eliminating fucose from

this carbohydrate structure is known to improve ADCC (Shinkawa et al., 2003). A non-

fucosylated variant of otlertuzumab has been generated which has enhanced binding to

FcγRIIIa, resulting in improved NK cell mediated ADCC and phagocytosis by

macrophages (Rafiq et al., 2013). Alternatively, ADCC can be enhanced by mutating

certain amino acid residues within the Fc region of IgG1 (Lazar et al., 2006). This

approach was taken in the generation of BI 836826 (Figure 1.3), an IgG1 with specific

mutations in the CH2 domain that augment ADCC (Heider et al., 2011; Krause et al.,

2012). This antibody has already entered clinical trials in Europe, with studies also

expected to begin in the United States.

10

This final broad category of therapies utilizes anti-CD37 antibodies to guide cytotoxic agents to tumor cells. CD37 internalizes moderately faster than CD20 when bound by antibody (Press et al., 1994), yet not so quickly that ADCC is prevented (Zhao et al., 2007; Heider et al., 2011; Krause et al., 2012; Deckert et al., 2013; Beckwith et al.,

2014). IMGN529 is a humanized anti-CD37 IgG1 conjugated to DM1 (Figure 1.3), a drug which inhibits microtubule assembly during mitosis (Deckert et al., 2013). Currently,

IMGN529 is being evaluated in NHL as part of an ongoing clinical trial (NCT01534715).

This trial has encountered some early difficulties, with several patients experiencing grade III/IV neutropenia that can be largely avoided with the addition of corticosteroids and G-CSF (Stathis et al., 2014). While possible that the low level of CD37 expression on neutrophils results in their direct elimination, evidence obtained from mouse models is suggestive of cell redistribution (Deckert et al., 2014). Only relatively low doses of

IMGN529 have been tested thus far, but 4 of 10 relapsed/refractory diffuse large B-cell lymphoma patients have responded to therapy (1 CR, 3 PR). This is expected to improve at higher doses, now that dose escalation is continuing with prophylaxis that addresses the neutropenia.

Similar to the above approach, anti-CD37 antibody can be used to deliver radioactive isotopes to tumor cells. This was first explored over 20 years ago (Press et al., 1989), but the renewed interest in targeting CD37 has led to the reemergence of

CD37-directed radioimmunotherapy (Dahle et al., 2013; Repetto-Llamazares et al.,

2014). There are currently two FDA-approved radiolabeled antibodies that target CD20

(Scott et al., 2012). However, the propensity of CD37 to internalize may make it a superior target, given that this occurs 10 times faster with 177Lu conjugated to anti-

CD37 tetulomab compared to anti-CD20 rituximab (Dahle et al., 2013). A phase I/II trial

11

recently initiated in Europe is exploring this therapeutic strategy in NHL using Betalutin, a 177Lu-conjugated anti-CD37 antibody. Thus far, 7 of 11 patients on this trial have

responded (ORR of 64%) with 4 CRs and 3 PRs (Kolstad et al., 2014).

A third approach to CD37-targeted drug delivery is the use of immunoliposomes

coated in antibody (Yu et al., 2013). CD37-coated immunoliposomes effectively

delivered cytotoxic drug to cell lines and CLL B-cells, and specificity could be altered by

using a dual targeting approach with an additional CD19 or CD20 antibody. Furthermore,

CD37 immunoliposomes that were not loaded with drug were capable of inducing

apoptosis in CD37+ cells, presumably due to a crosslinking effect. While this approach is

interesting experimentally, transitioning to full clinical development is quite complex due

to formulation related issues.

Eµ-TCL1 transgenic mouse model of CLL

A number of the studies described herein utilize the TCL1 mouse as a model of

CLL. These mice develops a leukemia closely resembling the more aggressive and

treatment-resistant subtype of CLL (Bichi et al., 2002; Yan et al., 2006). This leukemia

has the same CD5+CD19+ phenotype as human CLL in addition to displaying

immunodeficiencies and epigenetic changes that are observed in the human counterpart

(Ramsay et al., 2008; Chen et al., 2009). Overall, it is the best characterized drug

development tool for this malignancy (Johnson et al., 2006). Human CLL is a disease of

the elderly and TCL1 leukemia mimics this and requires a lengthy development period

that can exceed 1 year. However, the leukemia readily engrafts into normal mice on a

C57BL/6 background, which is advantageous from an experimental standpoint. Due to the lack of cross-reactivity of anti-human antibodies with equivalent mouse proteins, it is

12

challenging to study mAbs using this model (Macor et al., 2008). To address this for

CD37-targeted therapies we have developed a transgenic mouse expressing human

CD37 on its B-cells and crossed it with the TCL1 mouse to provide a model of spontaneous, human CD37-positive leukemia (hCD37xTCL1 mouse). This characterization of this model is the subject of Chapter 2 where it will be discussed in greater detail.

CLL microenvironment

Historically, CLL was viewed as a disease characterized by the slow accumulation of malignant B-cells with defects in apoptosis, but this has changed as the importance of a proliferative component has become increasingly apparent (Chiorazzi et al., 2005;

Messmer et al., 2005; Soma et al., 2006). Current evidence suggests that a subset of transformed B-cells form proliferative centers in lymphoid tissues, which are the source of the malignant cells that accumulate in the blood (Soma et al., 2006; Caligaris-Cappio et al., 2008; Vandewoestyne et al., 2011). In these tissues CLL cell survival, proliferation, and retention can be supported by accessory cells within the tumor microenvironment (Burger et al., 2000; Ghia et al., 2002; Burkle et al., 2007; Burger et al., 2013).

Kinase inhibitors

The tumor microenvironment in CLL is complex, and involves contribution from numerous cells types and signaling pathways. One of the most significant contributions of the lymphoid microenvironment is activation of B-cell receptor (BCR) signaling, which drives expansion of the malignant clone (Woyach et al., 2012). The importance of the

13

BCR pathway in CLL is highlighted by the clinical activity of drugs that inhibit various components, such as PI3K, Bruton’s tyrosine kinase (Btk), and spleen tyrosine kinase

(Woyach et al., 2012; Burger et al., 2013; Byrd et al., 2013). In particular, the Btk inhibitor ibrutinib has had a major impact on the management of CLL patients and even produces similar responses in high risk patients with deletion 17p (Byrd et al., 2013).

Idelalisib is another recently approved kinase inhibitor which has specificity for PI3Kδ.

These kinase inhibitors (and others still under clinical investigation) are rapidly altering how CLL is treated.

Thesis Objectives

The overall objective of this thesis is to develop novel approaches to CLL

immunotherapy that can be successfully translated to the clinic. In Chapter 2, I describe

a mouse model of human CD37-positive leukemia, providing a valuable preclinical tool

for evaluating anti-CD37 therapeutics. This mouse model enables the thorough study of

a CD37-specific antibody-drug conjugate in Chapter 3. The focus of Chapter 4 is the

development of combination strategies that enhance CD37-targeted therapy, which

includes immunomodulation and use of a kinase inhibitor. Chapter 5 investigates use of

a therapeutic that redirects T-cells to eliminate CD37+ tumor. Finally, Chapter 6

describes ongoing efforts to elucidate the function of CD37 in leukemia.

14

Figure 1.1. Structural features of tetraspanins.

Several common features of tetraspanins are depicted here. They possess 4 transmembrane domains (which are highly conserved), two short cytoplasmic tails, and two extracellular portions known as the EC1 domain (small extracellular loop) and EC2 domain (large extracellular loop).

Portions of the EC2 domain are conserved between various tetraspanins, but it also contains a highly variable region (shown in red). One of the features of this segment is the presence of 2-4 disulfide bonds (yellow lines) formed between cysteine residues (yellow circles), the number of which depend on the particular tetraspanin. The variable region of the EC2 domain contains binding sites for interactions with partner proteins and is frequently where epitopes for anti- tetraspanin antibodies are found. Many tetraspanins undergo palmitoylation at cysteine residues located near the intracellular border of the four transmembrane proteins. Additionally, most tetraspanins also experience N-linked glycosylation at extracellular asparagine residues (not depicted).

15

Figure 1.2. Signaling pathway associated with CD37 ligation by SMIP-016.

Lapalombella et al. described several cytoplasmic proteins which can associate with CD37, as depicted. Ligation by SMIP-016 leads to phosphorylation of Tyr13 within an ITIM-like motif in the

continued

16

Figure 1.2 continued

N-terminal cytoplasmic tail, which associates with a complex of proteins that includes Syk, Lyn,

and SHP1 (which likewise become phosphorylated). In addition, SMIP-016 induces

phosphorylation of an ITAM-like motif (containing Tyr274 and Tyr280) located in the C-terminal cytoplasmic tail that recruits PI3Kδ. Mutational studies suggest that the events requiring the N-

terminal ITIM drive apoptosis, while the C-terminal tail has a role in promoting cell survival. The

proposed mechanism of anti-CD37 induced cellular death involves a balance between these

signals, with preferential SHP1 activation driving apoptosis. SHP1 is capable of inactivating both

PI3K and Akt. SMIP-016 decreases the nuclear localization of Akt, preventing phosphorylation of

Fox O3a (and promoting retention in the nucleus) to allow transcription of pro-apoptotic BIM. An

opposing signal is transduced through PI3Kδ recruited to the C-terminal ITAM, activating Akt and

resulting in the downstream phosphorylation of GSK3β (which permits nuclear translocation of

pro-survival β-catenin). However, the contribution of PI3Kδ to survival can be eliminated by either

combination with a PI3K inhibitor or deletion of the ITAM-containing C-terminal domain of CD37.

While both pro-survival and pro-apoptotic signaling pathways are activated upon ligation by anti-

CD37 SMIP-016, those that promote cellular death predominate. Several other CD37-targeted

antibodies directly induce leukemia cell death, presumably in a similar fashion as SMIP-016/TRU-

016. However, they do not require additional receptor crosslinking (by use of anti-Fc antibody to

amplify the signal) as was observed with SMIP-016.

17

Figure 1.3. CD37 targeted antibody therapeutics.

Left: Otlertuzumab is an ADAPTIR™ molecule, constructed from an anti-CD37 single-chain variable fragment (scFv; a binding domain formed by linking the heavy and light chain variable regions of an immunoglobulin) which has been fused to the hinge region and Fc domain of human IgG1. Middle: mAb 37.1 is an Fc-engineered IgG1 with specific amino acid substitutions within the Fc region to increase ADCC mediated by effectors such as NK cells and macrophages.

Right: IMGN529 is a humanized anti-CD37 IgG1 (K7153A) conjugated to 3–4 molecules of cytotoxic drug (DM1) by stable thioether bonds. mAb, monoclonal antibody; CHO, Chinese hamster ovary; VH, heavy chain variable region; VL, light chain variable region; CH, heavy chain constant region (1, 2, or 3); CL, light chain constant region; D (orange circles), DM1; SMCC, N- succinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate.

18

CHAPTER 2

A MOUSE MODEL OF HUMAN CD37-EXPRESSING LEUKEMIA

As discussed in Chapter 1, the anti-CD20 mAb rituximab has made a

tremendous impact on the treatment of CLL. Despite the benefits it provides as part of

chemoimmunotherapy regimens, resistance is common and relapse essentially

inevitable (Rezvani and Maloney, 2011; Woyach et al., 2011). Expanding our arsenal

with novel immunotherapies that target alternative antigens could help circumvent

resistance to anti-CD20 therapy.

One of the most promising alternative targets is CD37, a tetraspanin protein

highly expressed on malignant B-cells in CLL and NHL (Link et al., 1986; Schwartz-

Albiez et al., 1988). While a natural ligand for CD37 has not been identified, signaling

induced by the anti-CD37 peptide SMIP-016 has implicated this tetraspanin as a

mediator of the PI3K/Akt survival pathway (Figure 1.2). The interest in this target has led

to the generation of several novel platforms of anti-CD37 therapeutics, including the

small modular immunopharmaceutical peptide otlertuzumab (TRU-016, the fully

humanized version of SMIP-016), an Fc-engineered IgG1 mAb, and an antibody-drug

conjugate (Zhao et al., 2007; Heider et al., 2011; Deckert et al., 2013). These therapies

are being explored for use in a variety of hematological malignancies, but my primary

focus has been their potential use in CLL. An ideal set of preclinical studies to evaluate

such therapies would include in vivo experiments in rodents, but suitable CLL mouse models have been lacking. The study of anti-human CD37 mAbs in vivo is impeded by

19

the lack of cross-reactivity with the murine protein. Xenograft of human cell lines into immunodeficient mice represents a common strategy in preclinical drug testing, although it is suboptimal as a tool for studying an immunotherapy and few CLL cell lines exist

(and they are rather poor representations of the disease). Therefore, a need exists for an improved mouse model that facilitates the preclinical evaluation of CD37-targeted therapies. To address this, our lab generated a human CD37 transgenic mouse. I have extensively characterized this mouse and used it to generate a spontaneous model of murine CLL expressing human CD37.

Materials and Methods

Mice

The human CD37 transgenic mouse (hCD37-Tg) was generated on a C57BL/6

background at the OSUCCC Transgenic Mouse Facility by conventional methodology as

previously described (Chen et al., 2006). B-cell restricted hCD37 expression is driven by

immunoglobulin (Ig) VH promoter and IgH-μ enhancer elements in the pBH expression

vector (Chen et al., 2006). The transgenic construct was generated by ligating the cDNA

sequence of human CD37 (with an additional FLAG sequence) into EcoRI and NotI sites

within the pBH vector. To generate our leukemia model, I crossed hemizygous hCD37-

Tg mice with homozygous Eμ-TCL1 mice (C57BL/6 background) that have been

previously described (Bichi et al., 2002; Johnson et al., 2006). Mice were housed in

microisolator cages under controlled temperature and humidity. All animal procedures

were performed in accordance with Federal and Institutional Animal Care and Use

Committee (IACUC) requirements.

20

Flow Cytometry

Flow experiments were performed using Beckman Coulter FC 500 flow cytometers (Brea, CA). Apoptosis assays were conducted on cells stained with fluorescein isothiocyanate (FITC) conjugated Annexin V and propidium iodide (PI) in 1X

Annexin V binding buffer (BD Biosciences, San Jose, CA). Other experiments used fluorochrome-labeled monoclonal antibodies against mouse CD3 (17A2), B220 (RA3-

6B2), CD19 (ID3), CD5 (53-7.3), CD4 (RM4-5), CD8 (53-6.7), CD45 (30-F11) (BD

Biosciences) and anti-human CD37 (K7153A) conjugated to R-Phycoerythrin (K7153A-

PE) that was provided by ImmunoGen, Inc. (Waltham, MA). Flow cytometric data was analyzed using Kaluza software (Beckman Coulter), with the exception of cell cycle experiments which were analyzed with Flowjo (Tree Star, Ashland, OR). Gating was verified with appropriate Fluorescence Minus One (FMO) controls. Quantification of

CD37 surface expression was performed using QuantiBRITE PE bead assay (BD

Biosciences) and 1:1 conjugated K7153A-PE.

Direct cytotoxicity assays

Mouse splenic B-cells were purified using EasySep kits (STEMCELL

Technologies). Cell viability was assessed by Annexin V-FITC and propidium iodide (BD

Biosciences) staining after 24 hour incubation at 37°C with 10 μg/ml anti-CD37 antibody

+/− 50 μg/ml goat anti-human Fc crosslinking antibody (Jackson ImmunoResearch,

West Grove, PA). Samples were analyzed by flow cytometry with at least 20,000 events

collected.

21

Results

Generation of a human CD37 transgenic mouse model of leukemia

With the ultimate goal of creating a mouse model accurately recapitulating the

characteristics of human CLL while expressing the human CD37 (hCD37) target protein,

the first steps were to characterize the hCD37 transgenic mouse (hCD37-Tg) generated

in our lab. The hCD37-Tg was created using an expression vector that contained IgVH

promoter and IgH-μ enhancer elements to achieve B-cell specific hCD37 expression

(Figure 2.1a). Presence of the transgene was confirmed by PCR using genomic DNA

(Figure 2.1b) and B-cell specific expression hCD37 expression was established using

flow cytometry performed on samples from spleen and peripheral blood (Figure 2.1c,d).

These data also demonstrated that hCD37 is not expressed on T-cells in the hCD37-Tg

mouse. However, quantification of CD37 antigen density of revealed that expression was

significantly lower than that observed on human B-cells (Figure 2.1e). CD37-specific

mAbs characteristically display high levels of direct cytotoxicity against leukemia cells in

vitro, particularly upon crosslinking with anti-Fc antibody (Zhao et al., 2007; Heider et al.,

2011; Deckert et al., 2013). To determine whether this also occurred with B-cells hCD37-

Tg mice, I purified splenic B-cells and incubated them with the anti-CD37 antibody

K7153A for 24 hours. This agent demonstrated in vitro cytotoxicity against hCD37-Tg B-

cells but did not decrease the viability of hCD37-negative B-cells from non-transgenic

littermates (Figure 2.2a,b). The observed cytotoxicity with K7153A prompted me to cross

the hCD37-Tg line with the Eμ-TCL1 mouse, an established model of IgVH unmutated

CLL which has been extensively characterized as a drug development tool (Bichi et al.,

2002; Johnson et al., 2006; Yan et al., 2006; Ramsay et al., 2008; Chen et al., 2009). B-

cells from these hCD37×TCL1 offspring retained expression of both transgenes (Figure

22

2.3a,b). As expected, elderly mice developed the CD5+CD19+ leukemia previously described in the original Eμ-TCL1 mouse (Bichi et al., 2002; Johnson et al., 2006). I then confirmed expression of hCD37 on this CD5+CD19+ cell population in the blood, spleen, and lymph nodes of a leukemic hCD37×TCL1 animal (Figure 2.3c).

Discussion

Herein I have described the generation of a human CD37 transgenic (hCD37-Tg) and its subsequent cross with the Eμ-TCL1 mouse to develop a model of spontaneous hCD37+ leukemia. This hCD37×TCL1 model addresses the need for improved platforms to preclinically evaluate anti-human CD37 therapeutics. Specifically, it provides the first model of a low-grade B-cell lymphoproliferative disorder that expresses human CD37.

This mouse is used extensively for key experiments described in the chapters to follow.

Existing models for evaluating anti-CD37 therapeutics are limited by their inability to reproduce the full spectrum of disease associated with B-cell malignancies. The most commonly used approach involves xenografts of human cells into immunodeficient mice, but these models fail to demonstrate the same tumor behavior or typical interactions with the tissue microenvironment (Macor et al., 2008). In CLL xenografts the injected cells largely remain in the peritoneal cavity, demonstrating a small presence of leukemic cells in the spleen and bone marrow while virtually no cells circulate in the peripheral blood

Shimoni et al., 1997; Durig et al., 2007). Models of spontaneous leukemia such as the

Eμ-TCL1 mouse produce a phenotype that more accurately portrays the compartmental distribution seen in human disease (Bichi et al., 2002; Macor et al., 2008). The leukemia developed by the Eμ-TCL1 model closely resembles the more aggressive subtype of

IgVH unmutated CLL, exhibiting similar behavior, immunodeficiencies, and epigenetic

23

changes as those observed in the human disease (Bichi et al., 2002; Johnson et al.,

2006; Yan et al., 2006; Ramsay et al., 2008; Chen e tal., 2009). The major disadvantage of this model, however, is that it cannot be utilized for preclinical evaluation of most therapeutic antibodies as they lack cross-reactivity with the mouse proteins. Expressing the human target protein in transgenic mice is an alternative, as when another group did so to evaluate their Fc-engineered CD37 antibody in vivo (Heider et al., 2011). However, this does not permit evaluation in the context of malignancy, where therapeutic efficacy may be dramatically altered. The hurdle for evaluating anti-CD37 therapies is overcome with the generation of our hCD37×TCL1 mouse model. In addition, the hCD37-Tg I used to generate this CLL model could be crossed with other models of spontaneous leukemia/lymphoma for testing CD37 targeted therapeutics in the context of different hematological malignancies. While the development of this mouse represents a major advance it should be noted that the expression of CD37 is lower than that observed in human CLL. This has the potential to influence response to anti-CD37 therapies tested in this model, and is further addressed in Chapter 3 where it is utilized for the preclinical evaluation of IMGN529, a CD37-specific antibody-drug conjugates.

24

Figure 2.1. Generation and characterization of human CD37 transgenic mouse.

A) Schematic representation of the construct used to generate the hCD37-Tg mouse. B) PCR

genotyping of founder lines with human CD37 specific primers. C) Surface expression of human

CD37 on transgenic (TG; top) and non-transgenic (NTG; bottom) splenocytes. Cells stained for

B220 and CD3 to analyze B and T lymphocytes, respectively. D) Surface expression of human

continued 25

Figure 2.1 continued

CD37 on transgenic (TG; top) and non-Ctransgenic (NTG; bottom) peripheral blood leukocytes.

Whole blood stained for CD19, CD4, CD8, and human CD37. After staining, RBCs were lysed and samples analyzed by flow cytometry. E) Quantification of human CD37 surface expression on CLL (n=10), hCD37-Tg (n=9), and NTG B-cells (n=8). ABC = Antibody Binding Capacity

26

Figure 2.2. Human CD37 transgenic B-cells are sensitive to anti-CD37 therapy

A) Example of flow cytometry performed on purified splenic B-cells stained with Annexin V/PI to assess viability after 24 hour treatment with 10 μg/ml anti-CD37 antibody (K7153A) and 50 μg/ml

goat anti-human Fc antibody (αFc) for crosslinking. B) Mean viability (+/- SEM) of purified splenic

B-cells from transgenic or non-transgenic spleens (n=7/group) 27

Figure 2.3. hCD37xTCL1 mice retain expression of both transgenes and develop CLL

continued 28

Figure 2.3 continued

A) Flow cytometry performed on peripheral blood from a CD37xTCL1 double transgenic mouse and a TCL1 littermate. Cells were stained with antibodies against mouse B220 to label B-cells

and human CD37. B) Western blot using protein lysates from purified B-cells obtained from

spleens of CD37xTCL1 mice or TCL1 littermates. Western blot is probed with anti-human TCL1 antibody. Lysates from C57BL/6 and leukemic TCL1 mice are included as controls in the first two lanes. C) Flow cytometry performed on samples obtained from peripheral blood, spleen, and lymph nodes of a leukemic CD37xTCL1 mouse. Cells were stained for CD5, CD19, and human

CD37. The right side of the panel demonstrates staining with K7153A-PE antibody and is gated on either CD5+CD19+ B-cells (dark gray histogram) or hCD37-negative T-cells (light gray histogram).

29

CHAPTER 3

A CD37-SPECIFIC ANTIBODY-DRUG CONJUGATE FOR B-CELL MALIGNANCY

IMGN529 is an antibody-drug conjugate (ADC) that consists of the cytotoxic

maytansine-derivative DM1 linked to a CD37-targeting humanized IgG1 antibody via a

stable SMCC linker (Deckert et al., 2013). This strategy seeks to combine the potent

cytotoxicity demonstrated by anti-CD37 therapeutics with the specific delivery of DM1,

which exerts anti-proliferative effects by disrupting microtubule dynamics during mitosis

(Oroudjev et al., 2010). The clinical viability of DM1-conjugated antibodies has been

demonstrated by ado-trastuzumab emtansine (T-DM1, KadcylaTM), now approved by the

U.S. Food and Drug Administration for treatment of HER2+ metastatic breast cancer

(Verma et al., 2012). Brentuximab vedotin, which is indicated for the treatment of refractory Hodgkins lymphoma and systemic anaplastic large cell lymphoma, is currently the only other ADC approved for marketing by the FDA (de Claro et al., 2012). However, given that over 20 ADCs are currently under evaluation in human trials for a wide variety of indications, this therapeutic strategy seems poised to become increasingly prominent.

While CLL was once viewed as a malignancy driven by apoptosis resistance, the importance of a proliferative component and interactions between tumor and the tissue microenvironment has become increasingly apparent (Caligaris-Cappio, 2003; Messmer et al., 2005; Soma et al., 2006; Chiorazzi et al., 2007). Current evidence suggests that a subset of transformed B-cells form proliferative centers in lymphoid tissues and are the source of the malignant cells which slowly accumulate in the peripheral blood (Soma et

30

al., 2006; Chiorazzi et al., 2007; Vandewoestyne et al., 2011). In vivo measurements indicate that birth rate of CLL B-cells can exceed 1% of the total malignant clone per day

(Caligaris-Cappio, 2003). However, it is still unknown whether ADCs carrying anti-mitotic payloads will have utility in CLL given that proliferation is lower than most subtypes of

NHL (Broyde et al., 2009).

Therefore, it is not surprising that the focus of IMGN529’s clinical development has been as a lymphoma therapy. My initial work with this ADC focused on its in vitro activity, but it is impossible to address the crucial question surrounding IMGN529 on the basis of these experiments because CLL B-cells do not proliferate ex vivo. Therefore, we

require an in vivo model of CLL in order to evaluate the potential utility of IMGN529. With the development of the hCD37xTCL1 model of CLL described in Chapter 3, the goal of characterizing IMGN529’s anti-proliferative effects became attainable.

Materials and Methods

Human Samples

Peripheral blood mononuclear cells (PBMCs) were obtained from normal donors or CLL patients in accordance with the Declaration of Helsinki. All subjects gave written, informed consent for their blood products to be used for research under an institutional review board approved protocol. Blood from CLL patients was collected at The Ohio

State University Wexner Medical Center (Columbus, OH). Normal cells were obtained from Red Cross partial leukocyte preparations or healthy donor blood. PBMCs were isolated by Ficoll density-gradient centrifugation (Ficoll-Paque Plus, GE Healthcare,

Uppsala, Sweden). Except when performing whole blood experiments, all CLL samples underwent negative selection of B-cells with RosetteSep (STEMCELL Technologies,

31

Vancouver, BC, CA) according to manufacturer’s protocol. Cells were cultured at 37°C and 5% CO2 in RPMI 1640 supplemented with 10% heat-inactivated fetal bovine serum

(Sigma, St. Louis, MO), 2 mM L-glutamine (Invitrogen, Carlsbad, CA), 56 U/mL penicillin and 56 μg/ml streptomycin (Invitrogen).

Monoclonal antibody therapeutics

Therapeutics used in our studies, which include alemtuzumab, rituximab, ofatumumab, and trastuzumab, were purchased from the pharmacy at The Ohio State

University Wexner Medical Center. ImmunoGen, Inc. (Waltham, MA) provided several

humanized IgG1 reagents: IMGN529, K7153A, and a non-binding chKTI-SMCC-DM1

(IgG-DM1) control ADC.

Mice

The generation of hCD37-Tg mice and subsequent breeding to create

hCD37xTCL1 mice are described in Chapter 2. Mice were housed in microisolator cages

under controlled temperature and humidity. All animal procedures were performed in

accordance with Federal and Institutional Animal Care and Use Committee (IACUC)

requirements.

Flow Cytometry

Flow experiments were performed using Beckman Coulter FC 500 flow

cytometers (Brea, CA). Apoptosis assays were conducted on cells stained with

fluorescein isothiocyanate (FITC) conjugated Annexin V and propidium iodide (PI) in 1X

Annexin V binding buffer (BD Biosciences, San Jose, CA). Other experiments used

32

fluorochrome-labeled monoclonal antibodies against mouse CD3 (17A2), CD19 (ID3),

CD5 (53-7.3), CD45 (30-F11) (BD Biosciences) and anti-human CD37 (K7153A) conjugated to R-Phycoerythrin (K7153A-PE) that was provided by ImmunoGen, Inc.

(Waltham, MA). Flow cytometric data was analyzed using Kaluza software (Beckman

Coulter), with the exception of cell cycle experiments which were analyzed with Flowjo

(Tree Star, Ashland, OR). Gating was verified with appropriate Fluorescence Minus One

(FMO) controls. Absolute cell concentrations were obtained by quantitative flow cytometry using Count Bright absolute counting beads (Invitrogen).

Mouse leukemia engraftment

Splenocytes from a moribund, leukemic hCD37×TCL1 donor were isolated by

Ficoll-Paque density gradient and re-suspended in sterile PBS for injection. Healthy female hCD37-Tg mice received an intravenous lateral tail vein injection of 200 μl containing 1×107 splenocytes. Age-matched mice were randomly assigned to the

following treatment groups (n=6-7 per group): the IMGN529 ADC, its K7153A antibody

component, an IgG-DM1 ADC control, or trastuzumab as an irrelevant humanized IgG1

antibody control. Mice were monitored for disease by flow cytometry on a weekly basis.

Upon leukemia diagnosis, a 10 mg/kg dose of the appropriate treatment was

administered intraperitoneally and repeat doses were given 2 times per week for 3

weeks (70 mg/kg total). Leukemia onset was defined as when greater than 20% of

CD45+ cells in the peripheral blood consisted of CD5+CD19+ leukemic B-cells or when

splenomegaly was evident upon light palpation. To avoid unnecessary suffering, mice

were euthanized with CO2 upon reaching standard IACUC criteria for early removal (e.g.

weight loss >20%, severe lethargy, labored breathing).

33

Direct cytotoxicity assays

Freshly isolated CLL B-cells from patient blood was enriched using RosetteSep

(STEMCELL Technologies). Cell viability was assessed by Annexin V-FITC and

propidium iodide (BD Biosciences) staining after 24 hour incubation at 37°C with 10

μg/ml antibody +/− 50 μg/ml goat anti-human Fc crosslinking antibody (Jackson

ImmunoResearch, West Grove, PA). For cell line experiments, Raji cells were incubated

with 1 μg/ml antibody for 72 hours. Data are reported as the percentage of remaining

viable cells (those that were both Annexin V and PI negative) normalized to untreated

control. Samples were analyzed by flow cytometry and at least 20,000 events collected.

Antibody dependent cellular cytotoxicity (ADCC)

The degree of ADCC was assessed using a standard 51Cr release assay, as

described previously.10 After 30 minutes of treatment with 10 μg/ml antibody, a total of

5×104 CLL target cells labeled with 51Cr were co-incubated with NK cells obtained from healthy donors for 4 hours at 37°C in 96-well plates at effector-to-target ratios of 25:1,

6.25:1, or without effectors. Following this incubation, supernatants were harvested and chromium release was measured with a Perkin Elmer Wizard 2 gamma counter

(Waltham, MA). Maximum chromium release was determined using targets lysed with sodium dodecyl sulfate (SDS). Cytotoxicity was calculated as follows: %Specific lysis =

(Experimental 51Cr release – Spontaneous 51Cr release) ÷ (Maximum 51Cr release –

Spontaneous 51Cr release) × 100. NK cells for this assay were enriched from Red Cross partial leukocyte preparations and healthy donor blood using RosetteSep kits

(STEMCELL Technologies).

34

Antibody dependent cellular phagocytosis (ADCP)

Monocytes were isolated from Red Cross partial leukocyte preparations using

MACS CD14+ selection kit (Miltenyi Biotec, Auburn, CA) according to manufacturer protocols. Monocytes were cultured in 10 cm2 dishes using RPMI 1640/10% FBS containing 20 ng/ml monocyte-colony stimulating factor (M-CSF; R&D Systems,

Minneapolis, MN) to promote differentiation into monocyte-derived macrophages

(MDMs). Fresh media containing M-CSF was provided every 2 days. After 7-10 days of

incubation, adherent macrophages were harvested and labeled with Claret dye (Sigma).

CLL cells were labeled with PKH67 fluorescent dye (Sigma), then treated with 10 μg/ml

antibody for 1 hour on ice and washed twice. Labeled cells were co-incubated at an

effector-to-target ratio of 1:5 (1×106 MDMs and 5×106 CLL cells) for 30 minutes at 37°C.

Samples were fixed with 1% paraformaldehyde prior to analysis by flow cytometry.

Relative phagocytosis = (%Claret-positive MDMs becoming PKH67+ in the treatment

condition) − (%Claret-positive MDMs becoming PKH67+ in the untreated control). At

least 10,000 Claret-positive (MDM) events were collected per sample.

Complement dependent cytotoxicity (CDC)

RosetteSep purified B-cells and plasma were isolated from the blood of CLL

patients. In a final volume of 500 μl, a total of 5×105 CLL B-cells were combined with 150

μl of autologous plasma (+/− heat-inactivation for 30 minutes at 57°C),10 μg/ml antibody, and incubated for 1 hour at 37°C. Following this incubation, samples were washed once and dead cells stained with LIVE/DEAD Fixable near-IR (Invitrogen). Cells were then fixed with 1% paraformaldahyde until analysis by flow cytometry. The percentage of

35

lysed cells was calculated by subtracting the percentage of dead cells in the corresponding untreated control. At least 20,000 events were collected per sample.

CLL B-cell depletion in whole blood

Whole blood from CLL patients (90 μl) was incubated with 10 μg/ml antibody for

1 hour at 37°C. 27 After this treatment, blood was stained with anti-CD3 FITC (UCHT1) and CD19 PE (HIB19) from BD Biosciences. Cal-lyse (Invitrogen) was used to simultaneously lyse RBCs while fixing the leukocytes. Count Bright absolute counting beads were added just prior to analysis by flow cytometry in order to calculate absolute concentrations of cells. To adjust for differences in initial leukemia counts between patients, data was normalized as follows: Percent depletion = 100 × (1 – [concentration of cells in treated sample] ÷ [concentration of cells in untreated control]).

Cell cycle analysis

Raji cells were synchronized with 2 μg/ml aphidicolin (Sigma) for 24 hours, transferred to fresh media, and treated with 1 μg/ml antibody for an additional 24 hours.

Following incubation, 1×106 cells (re-suspended in 1 ml PBS) were fixed by addition of 3

ml absolute ethanol. After fixation for at least 24 hours, these samples were stained in a

PBS solution of 0.1% (v/v) Triton X-100, 200 μg/ml DNase-free RNase, and 30 μg/ml

propidium iodide (Sigma). The proportion of singlet cells in G1, S, and G2 were obtained

with Flowjo analysis software (Tree Star) using the Dean-Jett-Fox model and the

assumption that the G2 coefficient of variance (CV) is equal to the G1 CV.

36

In vivo cell proliferation assay

To assess whether IMGN529 could inhibit in vivo proliferation, we engrafted

healthy hCD37-Tg mice with CD37xTCL1 leukemia. After detection of peripheral blood

leukemia (Day 0), mice were randomized to groups that would receive either IMGN529

or IgG-DM1 control in a blinded fashion. On Day 3 and Day 5 these mice received 10

mg/kg i.p. antibody, followed by 100 μg ethynyl-2’deoxyuridine (EdU) on Day 6. Tissues

were collected 24 hours later and incorporation of EdU was detected using Click-it EdU

Alexa Fluor 647 Flow Cytometery kit (Life Technologies) according to manufacturer

recommendations. Gating for EdU positivity was determined using a control mouse

which did not receive EdU.

Statistical analysis

For Raji cell line experiments, analysis of variance (ANOVA) was performed. For

patient sample data which involved repeated measures, mixed effect models were

utilized to account for dependencies across different treatment groups. For the in vivo

study, log-rank tests were used to compare the survival probabilities between mouse

groups. Holm’s method was used to adjust multiplicities. SAS 9.3 software was used for

data analysis (SAS, Inc; Cary, NC).

Results

The CD37-targeting IMGN529 directly induces apoptosis of CLL B-cells and maintains

Fc-dependent killing by innate immune cells in vitro

Addressing whether ADCs may be capable of targeting the proliferative

component of CLL is complex and ultimately requires a suitable in vivo model. However,

37

I initially characterized the antibody-derived activity of the IMGN529 ADC against primary human CLL, which does not proliferate ex vivo in the absence of stimulation.

Treatment with IMGN529 or its antibody component alone (K7153A) demonstrated significant in vitro cytotoxicity against peripheral blood CLL B-cells (Figure 3.1a,b). This effect was further augmented by the addition of anti-Fc crosslinking antibody, but did not require its presence to induce cellular apoptosis. The ability for these therapies to induce apoptosis of CLL B-cells was not dependent on IgVH mutational status (Figure 3.1c).

Interestingly, B-cells isolated from healthy donor blood were less susceptible to direct killing by these antibodies (Figure 3.1d). IMGN529 and its antibody component K7153A also mediated antibody-dependent cellular cytotoxicity (ADCC) against CLL by healthy donor NK cells (Figure 3.2a). No significant difference was observed between IMGN529 and K7153A with respect to their ability to mediate ADCC. Furthermore, both agents equally promoted phagocytosis of CLL by monocyte-derived macrophages (Figure 3.2b and Figure 3.3). Neither K7153A nor IMGN529 exhibited complement-dependent cytotoxicity when CLL B-cells were incubated with autologous plasma (Figure 3.2c).

While the above experiments are informative, I sought to further evaluate these reagents in a context where the collective impact of multiple potential mechanisms could be appreciated. Therefore, I performed B-cell depletion assays in CLL patient whole blood, where various innate immune components that may contribute to the therapeutic efficacy of antibodies (complement, NK cells, monocytes, and granulocytes) are present in physiologically relevant proportions to leukemia cells. One hour treatment of CLL patient whole blood with IMGN529 or K7153A resulted in significantly greater malignant

B-cell depletion than rituximab or alemtuzumab (Figure 3.4 and Figure 3.5a). In addition,

CD37-targeted therapeutics avoided the undesirable T-cell depletion that is observed

38

with anti-CD52 alemtuzumab (Figure 3.5b). A limited analysis suggests that activity in whole blood does not vary on the basis of cytogenetic abnormalities, although a much larger sample size would be required to achieve greater certainty (Figure 3.5c).

Likewise, IgVH mutational status did not significantly alter activity (Figure 3.5d).

Given the lack of in vitro proliferation associated with CLL B-cells, I sought to determine whether delivery of DM1 by IMGN529 would have an impact on actively dividing transformed B-cell lines. Raji cells were treated with K7153A, IMGN529, or control antibodies for 72 hours. In contrast to what was observed with non-dividing CLL

B-cells, IMGN529 demonstrated superior cytotoxicity against Raji cells compared to its

K7153A antibody component (Figure 3.6a). DM1 is known to kill cells through disruption of microtubules, resulting in G2/M arrest and mitotic catastrophe (Oroudjev et al., 2010).

Consistent with this mechanism, cell cycle analysis of IMGN529-treated Raji cells

revealed that a substantial proportion were arrested at the G2/M checkpoint (Figure

3.6b).

IMGN529 selectively depletes leukemic B-cells in vivo and improves overall survival of mice with hCD37×TCL1 leukemia

To evaluate the therapeutic potential of IMGN529 in vivo, I engrafted splenocytes

from a leukemic hCD37×TCL1 donor into healthy hCD37-Tg recipients and monitored

these mice for disease. Upon leukemia development, mice were treated with the

IMGN529 ADC, its antibody component K7153A, an IgG-DM1 ADC control, or

trastuzumab as a non-specific humanized IgG1 control (Figure 3.7a). In contrast with my

in vitro cytotoxicity studies using human CLL cells, which showed no significant

difference between IMGN529 and its antibody component K7153A alone (see Figure

39

3.1b), improved overall survival was only observed in mice treated with IMGN529

(Figure 3.7b). One week of IMGN529 treatment was sufficient to eliminate peripheral

blood leukemia, while the disease continued to progress in other groups (Figure 3.7c,d).

In addition, previously detected splenomegaly disappeared with IMGN529 treatment

(Figure 3.8a). Depletion of hCD37-negative T-cells did not occur with IMGN529 (Figure

3.8b). Only a transient reduction of peripheral CD19+CD5+ B-cells was observed on day

21 after treatment with the K7153A antibody (Figure 3.7d).

IMGN529 eliminates proliferating hCD37×TCL1 leukemia cells in vivo

To better characterize the in vivo activity of IMGN529, I sought to track the effects on the proliferating subset of leukemia cells within lymphoid tissues. Mice were engrafted with hCD37×TCL1 leukemia and randomly assigned to treatment groups upon development of disease (day 0). On days 3 and 5, mice received 10 mg/kg i.p. doses of

IMGN529 or IgG-DM1 control, followed by EdU injection on day 6 to monitor in vivo cell

proliferation. At the beginning of the study, the proportions of peripheral blood

CD5+CD19+ leukemia were similar, but IMGN529 therapy eliminated most leukemia 48

hours after the second dose (Figure 3.9a). Leukemia cells were present in the blood,

spleen, and bone marrow of mice injected with IgG-DM1, but those receiving IMGN529

demonstrated significantly less tumor burden in these tissues (Figure 3.9b,c). By visual

inspection, spleens from all mice treated with IMGN529 were much smaller than those

from control mice. Although it is an underestimate of actual size difference, this is also

evident when spleen length is quantified (Figure 3.9d). Moreover, incorporation of EdU

was significantly lower among CD5+CD19+ cells remaining in the spleens of IMGN529

treated mice (Figure 3.9e).

40

Discussion

This chapter describes the extensive characterization of IMGN529’s in vitro activity using primary human CLL samples. I also utilized the novel hCD37xTCL1 mouse from the previous chapter to demonstrate potent in vivo efficacy in the context of murine

CLL. To my knowledge, this is the first ADC shown to effectively target the proliferative component of CLL in vivo.

My in vitro studies of IMGN529 show that conjugation of the CD37-targeting

K7153A antibody to DM1 does not diminish its ability to promote effector-mediated killing of CLL by NK cells and macrophages. Furthermore, both IMGN529 and K7153A greatly surpass rituximab in their ability to deplete malignant B-cells in CLL patient whole blood.

I also observed direct cytotoxicity against CLL in the absence of crosslinker, similar to what has been reported with another recently developed anti-CD37 therapeutic (BI

836826) but unlike the CD37-directed peptide TRU-016 (Zhao et al., 2007; Heider et al.,

2011). Interestingly, healthy donor B-cells were less sensitive to anti-CD37 induced apoptosis than human CLL B-cells. This cannot be explained by decreased expression of the target protein, as normal B-cells do not have less surface CD37 (Barrena et al.,

2005). Both murine splenic B-cells and human CLL B-cells display high degrees of spontaneous apoptosis during in vitro culture. Therefore, observed differences in direct cytotoxicity could be a result of normal human B-cells exhibiting decreased priming to undergo apoptosis, thus reducing their sensitivity to anti-CD37 therapeutics (which are expected to induce expression of pro-apoptotic mitochondrial protein BIM following

CD37 ligation (Lapalombella et al., 2012).

IMGN529 and K7153A were able to mediate similar levels of cytotoxicity against human CLL cells in vitro. This is in sharp contrast to what was observed in vivo, where

41

K7153A produced only modest depletion of leukemic B-cells and was unable to significantly alter the disease course in the hCD37×TCL1 model. While its antibody alone was not effective, the full IMGN529 ADC rapidly eliminated peripheral blood leukemia, reversed splenomegaly and improved overall survival. The different in vitro

and in vivo activity I observed with these agents is not entirely surprising given that CLL

cells do not proliferate in culture, which would make the delivery of anti-mitotic DM1

largely irrelevant in these short-term in vitro studies. When tested against proliferating

Raji cells in vitro, IMGN529 possessed a distinct advantage over its antibody component

with enhanced cytotoxic activity resulting in G2/M cell cycle arrest. I hypothesized that

IMGN529 eliminated dividing leukemia cells within the proliferative centers via targeted

delivery of DM1 in vivo, thus resulting in a greater therapeutic benefit than the antibody

alone. This is supported by the observation that peripheral leukemia depletion with

IMGN529 was accompanied by a rapid decrease in splenomegaly. To further investigate

the in vivo activity of IMGN529, I monitored its effects on leukemia cells within various

compartments of disease and examined those from lymphoid tissues for signs of

proliferation using EdU. In doing so, I confirmed that IMGN529 is effective at targeting

leukemia in both peripheral blood and the lymphoid compartment. EdU incorporation

was largely absent among the few CD5+CD19+ cells that remained after 2 doses,

suggesting that IMGN529 was indeed eliminating this proliferative subset of cells.

Additionally, the splenomegaly observed with IgG-DM1 controls was not present in mice

receiving IMGN529. While a small number of leukemia cells remained after 2 doses,

these displayed very little EdU incorporation. It is evident that IMGN529 is highly active

across multiple compartments of disease in this CLL mouse model. Given that the

expression of hCD37 was suboptimal compared to human CLL (Figure 2.1e), it is

42

unlikely that antigen density has led to an exaggerated response in the mouse model.

For this reason, delivery of the cytotoxic DM1 payload is likely even more efficient in human CLL cells.

As clinical study of IMGN529 moves forward, it would be worthwhile to explore potential combination therapies that may take advantage of the unique therapeutic properties of this ADC. Given that IMGN529 maintains the Fc-mediated effector functions of the antibody from which it was generated, combination with immunomodulatory drugs (such as lenalidomide) could prove effective. The anti-CD37 peptide TRU-016 previously demonstrated in vitro synergy with PI3K inhibitors

(Lapalombella et al., 2012). Therefore, it may also be worthwhile to explore combinations with inhibitors of PI3Kδ such as idelalisib. The newly generated hCD37×TCL1 mouse model provides a unique tool that will enable the evaluation of combination strategies for IMGN529 and other CD37-targeted therapeutics in an immunocompetent model of spontaneous CLL.

To summarize, I have demonstrated the utility for evaluation of anti-CD37 therapeutics provided by the mouse model described in Chapter 2 (which develops a transplantable CD5+CD19+hCD37+ leukemia). This model facilitated improved preclinical studies of IMGN529, elucidating its robust anti-leukemic effects in vivo. This is impressive in the context of the Eμ-TCL1 leukemia that is somewhat resistant to treatment (Johnson et al., 2006; Lucas et al., 2009; Lapalombella et al., 2012). Given the significant benefits we observed with IMGN529, despite relatively low hCD37 expression, I propose that this therapeutic could exhibit efficacy in a wide range of

CD37-positive human B-cell malignancies.

43

Figure 3.1. IMGN529 is directly cytotoxic to primary human CLL B-cells

continued 44

Figure 3.1 continued

A) Example of 24-hour Annexin V/PI viability assay performed on RosetteSep purified B-cells from CLL patient blood.B) Viability of freshly isolated CLL patient B-cells (n=16) following 24 hour

treatment with 10 μg/ml IMGN529 or its antibody component K7153A +/− 50 μg/ml crosslinking

antibody (αFc). Anti-HER2/neu antibody trastuzumab included as an additional negative control

for n=11 patients. Data are normalized to untreated controls. Significance is indicated by

asterisks (*p<0.05, ***p<0.0001, or NS if p>0.05). C) CLL patient B-cells with either unmutated

(UM, n=9) or mutated (M, n=5) IgVH assayed for viability by Annexin V/PI staining

following 24 hour treatment with 10 μg/ml IMGN529 or its antibody component K7153A +/- 50

μg/ml anti-Fc crosslinking antibody (αFc). Data are normalized to untreated control. IgVH was

considered unmutated when there was >98% homology to germline sequence. D) Healthy donor

B-cells (n=4) isolated by negative selection with Rosette-Sep. Purity was confirmed to be greater

than 90% CD19+ B-cells for all samples. Viability was assessed by Annexin V/PI staining and

flow cytometry following 24 hour treatment with 10 μg/ml IMGN529, its antibody component

K7153A, or the anti-HER2 trastuzumab as a negative humanized IgG1 control. Error bars indicate

SEM.

45

Figure 3.2. IMGN529 retains the Fc-mediated activities of its unconjugated precursor

continued

46

Figure 3.2 continued

A) NK cell mediated antibody-dependent cytotoxicity as measured by 51Cr release assay. CLL target cells (n=7) were incubated with 10 μg/ml antibody for 30 minutes, followed by incubation with healthy donor NK cells (n=8) for 4 hours. Mean specific lysis displayed for n=18 NK/CLL combinations with error bars indicating standard error of the mean (SEM). B) Induced phagocytosis of CLL cells by monocyte-derived macrophages (MDMs) following 1 hour incubation with 10 μg/ml antibody. Relative phagocytosis displayed for a total of n=10 MDM/CLL combinations (7 MDMs, 5 CLL) with error bars indicating SEM. C) Complement-dependent cytotoxicity assay for B-CLL incubated with 10 μg/ml antibody and autologous plasma (+/− heat inactivation) for 1 hour. Mean values displayed for n=9 CLL patients with error bars indicating

SEM.

47

Figure 3.3. Gating strategy for measuring phagocytosis of CLL B-cells

continued 48

Figure 3.3 continued

Antibody-dependent cellular phagocytosis (ADCP) of CLL cells by monocyte-derived macrophages (MDMs). Gating scheme is displayed. Initially, a singlet gate is created using FS-

Area vs. FS-Peak plot. Gate for PKH67 (CLL) vs. Claret (MDM) was verified using a sample with only Claret stained MDMs. For determination of phagocytosis, a gate is created on the singlet

Claret-positive (MDM) events. The percentage of these MDM events that are PKH67-positive is considered the percent phagocytosis. For relative phagocytosis, the %Phagocytosis in the untreated is subtracted from treated samples. Examples of each treatment group are shown.

49

Figure 3.4. Gating strategy for whole blood cytotoxicity assays

continued 50

Figure 3.4 continued

Whole blood CLL B-cell depletion assay. First, a singlet gate is created from the FS-Area versus FS-Peak plot, which includes both the lymphocytes and the CountBright counting beads.

Subsequent FS vs. SS gates are on either the lymphocyte population or the bead population. The

CountBright bead gate is further fefined using the SS vs. FL4 gate, as they fluoresce brightly in

the FL4 channel. The number of events obtained for each cellular event of interest (CD19+ or

CD3+ within the lymphocyte gate) and number of bead events collected were used to calculate absolute concentrations of cells using the known concentration of beads, as outlined by the manufacturer protocol. Examples of each treatment condition are shown. Data acquisition stopped at approximately 1000 bead events for each sample.

51

Figure 3.5. IMGN529 demonstrates exceptional cytotoxicity in CLL whole blood

continued

52

Figure 3.5 continued

A-B) CLL patient whole blood was treated with 10 μg/ml antibody for 1 hour at 37°C. B-cells and

T-cells were stained with anti-CD19 and CD3 antibodies respectively. Counts were obtained

using CountBright absolute counting beads and normalized to untreated control. Mean percent

depletion of B-cells A) and T-cells B) are displayed for n=23 CLL patients with error bars

indicating SEM. Greater than 97% of the CD19+ cells in these patients were confirmed to be malignant CD5+ B-cells.C) Limited analysis examining the effects of cytogenetic status on efficacy in whole blood B-cell depletion assays. Patient groups are as follows: no cytogenetic abnormalities (n=5), del13q (n=8), del17p (n=3), Trisomy 12 (n=3). Several patients for which cytogenetic status was either unavailable or with abnormalities lacking replicates are excluded. D)

Analysis examining the effects of IgVH mutational status on B-cell depletion in whole blood

assays (n=11 patients with mutated IgVH and n=8 patients unmutated IgVH). For several patients

IgVH mutational status was unavailable, and thus they were excluded. No statistically significant differences were detected (p=0.8559 for IMGN529 and p=0.8848 for K7153A). Error bars indicate

SEM.

53

Figure 3.6. DM1-conjugation improves killing and arrests proliferating B-cells

A) Raji cells were treated with 1 μg/ml K7153A, IMGN529, or relevant controls for 72 hours and viability assayed by Annexin V and propidium iodide staining. Data are normalized to untreated control and reported as mean with error bars indicating SEM for n=5 replicate cultures from two independent experiments. B) Cell cycle analysis of Raji cells that were synchronized with the

DNA polymerase inhibitor aphidicolin for 24 hours, released from this inhibition, and then treated with 1 μg/ml antibody for 24 hours.

54

Figure 3.7. IMGN529 demonstrates in vivo efficacy against hCD37+ mouse leukemia.

A) Schematic outline of the hCD37×TCL1 engraftment experiment. Healthy hCD37-Tg recipients

were injected i.v. with 1×107 splenocytes from a leukemic hCD37×TCL1 donor. Mice were

continued 55

Figure 3.7 continued randomly assigned (n=6-7/group) for treatment with 10 mg/kg IMGN529 ADC, its K7153A antibody component, IgG-DM1 ADC control, or trastuzumab control. Treatment began following leukemia development, which was defined as when >20% of CD45+ events were CD5+CD19+ B- cells. Animals received a 10 mg/kg i.p. treatment on the day of leukemia diagnosis and repeat doses twice weekly for three weeks (70 mg/kg total). B) Kaplan-Meier plot comparing the overall survival of engrafted mice. IMGN529 significantly improved survival relative to its IgG-DM1 control (p=0.042), while parent antibody K7153A had no survival benefit compared with trastuzumab control (p=0.94). Log-rank tests were used for statistical analysis. Arrow indicates day 20, when mice received their last injection. C) Representative examples of flow cytometry performed on mice 24h after receiving their third dose. Whole blood was stained for

CD45/CD19/CD5 and plots are gated on singlet CD45+ events. D) Absolute concentration of the

CD5+CD19+ B-cell population to which the leukemia belongs. Data expressed as mean +/− SEM.

Arrow indicates day 20, when mice received their last injection.

56

Figure 3.8. IMGN529 eliminates splenomegaly and demonstrates specificity for CD37+ cells

A) Measurement of spleen sizes in the hCD37xTCL1 leukemia engraftment study. A score was assigned to each mouse based on spleen size. Scores were given according to the following criteria: 0 = spleen not palpable, 1 = felt upon deep palpation, 2 = palpable with light touch, 3 =

spleen palpable from the diaphragm to the lower abdomen, 4 = massive splenomegaly, where

vertical growth has reached its limit and the spleen begins to wrap around the opposite side of the

mouse. Data is represented as mean spleen score +/- SEM for each treatment group. B) Mean T-

cells/µl (+/- SEM) in peripheral blood of IMGN529 mice (n=6).

57

Figure 3.9. IMGN529 targets proliferating mouse leukemia cells within lymphoid tissues.

A-E) Healthy hCD37-Tg mice engrafted with hCD37×TCL1 leukemia. Upon leukemia

development (day 0), mice were randomized (n=5 mice/group) to receive 10 mg/kg i.p. doses of

continued

58

Figure 3.9 continued

IMGN529 or IgG-DM1 control on days 3 and 5. To detect in vivo proliferation 100 μg EdU was administered on day 6 and mice were euthanized on day 7. (Mean is displayed for n=5 mice/group with error bars indicating SEM. ***p<0.0001, **p<0.01, *p<0.05) A) Percentage

CD5+CD19+ leukemia cells (of total CD45+ events) in peripheral blood on day 0 and day 7.

(***p<0.0001 for change in cell percentage). B) Percentage CD5+CD19+ leukemia cells (of total

CD45+ events) in the blood, spleen, and bone marrow on day 7. C) Examples of flow cytometry performed on peripheral blood, spleen, and bone marrow from mice treated with IMGN529 or

IgG-DM1. Plots are gated on singlet, live, CD45+ cells. D) Examples of spleens removed from mice receiving IMGN529 or IgG-DM1 (left panel) and average spleen length of all mice in the study (n=5/group; right panel). E) Percentage of EdU+ cells among CD5+CD19+ events in the spleen and bone marrow.

59

CHAPTER 4

DEVELOPING STRATEGIES TO ENHANCE ANTI-CD37 THERAPY

Otlertuzumab (TRU-016) has clinically validated CD37 as a target for immunotherapy (Byrd et al., 2014; Gopal et al., 2014; Pagel et al., 2014), and human trials have now begun for several novel anti-CD37 therapeutics which displayed improved preclinical activity. However, the response to immunotherapy is typically impaired when it is administered as a single agent. One strategy for improving response is to enhance Fc-mediated killing, which can be accomplished by combination with an immunomodulatory drug (or by directly altering the mAb through modifications of its Fc region). A variety of combination therapies have been used to achieve this, for example: treatment with cytokines (Roda et al., 2006), lenalidomide (Wu et al., 2008), or toll-like receptor (TLR) 7/8 agonists (Wu et al., 2008; Butchar et al., 2010).

A second approach to combination therapy with anti-CD37 therapeutics could instead focus on the direct cytotoxicity they exhibit. We previously described a mechanism that provided a likely explaination for the the cytotoxicity displayed by SMIP-

016. Upon ligation of CD37 by SMIP-016, two cytoplasmic regulatory motifs with opposing function become phosphorylated. This leads to both pro-apoptotic inactivation of Akt and (to a lesser extent) pro-survival PI3Kδ activation (Figure 1.2). Unsurprisingly, combining SMIP-016 with PI3K inhibitors led to improved in vitro cytotoxicity

(Lapalombella et al., 2012). This combination has not been explored with other CD37-

targeting therapeutics, so it has been unknown whether this effect can be replicated with

60

newer mAbs (particularly those which kill without need of crosslinking). In this chapter, I investigate the use of two strategies that I hypothesized would improve CD37-targeted therapy: 1) combining otlertuzumab (TRU-016) with immunomodulatory toll-like receptor

7/8 agonist, and 2) addition of PI3Kδ inhibitor to the Fc-engineered BI 836826.

Materials and Methods

Reagents

The TLR 7/8 agonist CL-075 (Invivogen, San Diego, CA) was provided by Jon

Butchar and Susheela Tridandapani as part of an ongoing collaboration. The Fc- engineered anti-CD37 antibody BI 836826 was provided by Boehringer Ingelheim

(Ingelheim am Rhein, Germany), and TRU-016 was provided by Emergent Biosolutions

(Gaithersburg, MD).

Direct cytotoxicity assays

Freshly isolated CLL B-cells from patient blood was enriched using RosetteSep

(STEMCELL Technologies). Cell viability was assessed by Annexin V-FITC and propidium iodide (BD Biosciences) staining after 24 hour incubation at 37°C with 0.1

μg/ml antibody +/− 1 μM idelalisib. Data are reported as the percentage of remaining viable cells (those that were both Annexin V and PI negative) normalized to untreated control. Samples were analyzed by flow cytometry and at least 20,000 events collected.

Antibody dependent cellular phagocytosis (ADCP)

Monocytes were isolated from Red Cross partial leukocyte preparations using

MACS CD14+ selection kit (Miltenyi Biotec, Auburn, CA) according to manufacturer

61

protocols. Monocytes were cultured in 10 cm2 dishes using RPMI 1640/10% FBS containing 20 ng/ml monocyte-colony stimulating factor (M-CSF; R&D Systems,

Minneapolis, MN) to promote differentiation into monocyte-derived macrophages

(MDMs). Fresh media containing M-CSF was provided every 2 days. After 7-10 days of

incubation, adherent macrophages were harvested and labeled with Claret dye (Sigma).

CLL cells were labeled with PKH67 fluorescent dye (Sigma), then treated with 10 μg/ml

antibody for 1 hour on ice and washed twice. Labeled cells were co-incubated at an

effector-to-target ratio of 1:5 (1×106 MDMs and 5×106 CLL cells) for 30 minutes at 37°C.

Samples were fixed with 1% paraformaldehyde prior to analysis by flow cytometry.

Relative phagocytosis = (%Claret-positive MDMs becoming PKH67+ in the treatment

condition) − (%Claret-positive MDMs becoming PKH67+ in the untreated control). At

least 10,000 Claret-positive (MDM) events were collected per sample.

Antibody dependent cellular cytotoxicity (ADCC)

The degree of ADCC was assessed using a standard 51Cr release assay, as

described previously.10 After 30 minutes of treatment with 10 μg/ml antibody, a total of

5×104 CLL target cells labeled with 51Cr were co-incubated with NK cells obtained from healthy donors for 4 hours at 37°C in 96-well plates at effector-to-target ratios of 25:1,

6.25:1, or without effectors. Following this incubation, supernatants were harvested and chromium release was measured with a Perkin Elmer Wizard 2 gamma counter

(Waltham, MA). Maximum chromium release was determined using targets lysed with sodium dodecyl sulfate (SDS). Cytotoxicity was calculated as follows: %Specific lysis =

(Experimental 51Cr release – Spontaneous 51Cr release) ÷ (Maximum 51Cr release –

Spontaneous 51Cr release) × 100. NK cells for this assay were enriched from Red Cross

62

partial leukocyte preparations and healthy donor blood using RosetteSep kits

(STEMCELL Technologies).

Mouse engraftment studies

C57BL/6 mice were injected i.p. with 5x106 splenocytes from leukemic

hCD37xTCL1 donors. Peripheral blood leukemic cell counts were monitored on a weekly

basis using Countbrite beads using whole blood stained with anti-CD5/CD19/CD45

antibodies (BD Biosciences). For more detailed methodology, see Chapter 3. Upon

identification of leukemia (defined as CD5+ B-cell count exceeding 1200 cells/µl), mice

were randomized to treatment groups. The two components of the therapy, 2 mg/kg CL-

075 (or PBS control) and 10 mg/kg TRU-016 (or trastuzumab control), were co-

administered to mice i.p. on days 1, 5, 8, 12, 15, and 19 relative to the start of their

leukemia. Evaluation of mice was performed by a blinded technician who euthanized

mice reaching IACUC-approved early removal criteria.

Results

The initial strategy I pursued sought to augment Fc-mediated killing by effectors

cells of the innate immune system. It was previously shown that TLR7/8 agonist

improves mAb efficacy in a solid tumor mouse model (Butchar et al., 2010). However,

this had not been explored in hematological malignancy. Interested in whether an effect

could be observed in our hCD37xTCL1 model of CD37, I began a study investigating the

feasibility of combining the TLR7/8 agonist CL-075 with TRU-016. While peripheral

leukemia cell counts decreased in the first week following treatment, disease

progression was seen in the weeks that followed (Figure 4.1a, b). It quickly became

63

clear that combination with CL-075 was not beneficial. Normalization of the cell counts

(to account for differing initial tumor burden) suggest that if the combination had any effect, it performed even more poorly than TRU-016 alone (Figure 4.1a, b). As would be expected, there were no significant differences in overall survival (Figure 4.1c).

Rather than pursuing other immune modulating strategies further, I began to work with an Fc-engineered CD37-targeted IgG1 that had already displayed improved effector activity as a result of mutations in its CH2 domain. This therapeutic, BI 836826, exhibits substiantial direct cytotoxicity (as seen with other anti-CD37 mAbs) and does so in the absence of crosslinker (Heider et al., 2011). This latter trait is only shared with

K7153A/IMGN529 among the therapeutics being developed for clinical use (Figure 3.1a, b). Upon receipt of this mAb, I performed some initial characterization of its activity which included measurement of macrophage-mediated phagocytosis (Figure 4.2a) and ADCC by NK cells (Figure 4.2b). In both cases, it was superior to anti-CD20 rituximab.

Additional studies confirmed the reported crosslinker independent direct cytotoxicity and activity in CLL patient whole blood (not shown). At this point, I began collaborating with a hematology fellow in the lab (Deborah Stephens) on this project. We have been investigating combination of BI 836826 with the PI3Kδ inhibitor idelalisib. Dr. Stephens established suitable concentrations for this combination, characterized its impact on

ADCC (which was decreased, but not eliminated), and began exploring its effects in

high-risk cases of CLL. Continuing this work, I examined cytotoxicity against samples

from a larger cohort of high risk patients (and those with better prognosis). Not only did

combination of idelalisib with BI 836826 improve in vitro cytotoxicity, but it also achieved

a similar degree of killing in both high risk patients with deletion 17p and those with lower

risk cytotogenetics (Figure 4.3a, b). Furthermore, this combination was similarly effective

64

in ibrutinib-refractory patients who had developed mutations in Cysteine 481 of BTK

(Figure 4.3c). Ultimately, we would like to test this combination in vivo, although the poor pharmacokinetics of idelalisib in mice will require the use of a different inhibitor. As my previous studies with hCD37xTCL1 leukemia have shown, the effect of single-agent mAb therapy (unless delivering anti-mitotic toxin) is severely limited (Figure 3.7 and

Figure 4.1), However, a small pilot study indicated that BI 836826 was capable of depleting peripheral leukemia in this model (Figure 4.4).

Discussion

In this chapter I explore potential combination strategies to improve the effectiveness of anti-CD37 therapy. While initial studies with the TLR 7/8 agonist did not support its combination with anti-CD37 therapy (or at least, not with TRU-016), they still provided useful information. While these agonists can improve antibody-mediated killing in solid tumor models, there were no previous reports of their use in hematological malignancy. A potential problem with using this agonist is the provision of pro-survival signals to the leukemia cells, given that B-cells express TLR7/8. Further studies would be required to determine if this was occurring with this murine leukemia. In any case, we moved on to studying a CD37-targeted mAb therapeutic which already possessed improved Fc-mediated killing by effector cells: BI 836826.

Our lab previously demonstrated the benefit of combining SMIP-016 with PI3K inhibitors, although this has not yet been shown with other CD37-targeted therapies.

Given that BI 836826 directly induces apoptosis in leukemia cells without the need for crosslinking secondary antibody (which is a requirement by SMIP-016), this was an attractive candidate for further testing. Its performance against CLL in my studies was

65

similar to expectation based on published reports on other Fc-mediated activities. This therapy exhibited activity against CLL B-cells that exceeded that of anti-CD20 mAbs in assays measuring phagocytosis and ADCC by NK cells (Figure 4.2). My colleague Dr.

Stephens confirmed that as with SMIP-016, combination with idelalisib led to improved leukemia cell apoptosis. Furthermore, I observed similar degrees of cytotoxicity against cells from both patients with deletion 17p and those who were refractory to ibrutinib

(Figure 4.3). Given that these patient populations have poor prognosis and decreased response to many therapies, it is important to develop effective strategies for managing their disease. It is difficult to determine the extent to which mAb-induced apoptotic signaling contributes to in vivo efficacy, although it has been hypothesized that such killing could be augmented by effector cells that provide crosslinking of mAb-bound target. However, there is currently scant evidence to support such in vivo crosslinking. In

particular, the efficacy of rituximab was completely dependent on FcR ITAM signaling

(and thus killing by the effector cell itself) in a mouse model that expressed mutant FcR

incapable of signaling but still retaining surface expression and capacity to bind the Fc of

antibodies. (de Haij et al., 2010). Antibodies exhibiting crosslinker independent killing

may alter the equation. The anti-CD20 tositumomab is an example of this, as it retained

in vivo activity even when digested into F(ab)′2 fragments incapable of recruiting effector

cells (Cragg and Glennie, 2004). Because of its crosslinker independent killing, BI

836826 (and K7153A/IMGN529) may represent better options for combination strategies

that exploit their signaling characteristics (i.e. combination with PI3K inhibitor). This mAb

appears to have single agent activity based on initial testing in the hCD37xTCL1 model

(Figure 4.4). Ongoing studies will further explore its activity in vivo and determine

whether combining this mAb with a PI3Kδ-selective inhibitor improves survival beyond

66

monotherapy. Other questions will still remain, including to what degree direct induction of apoptosis contributes to efficacy in vivo, and whether a common mechanism is responsible for both the direct cytotoxicity of SMIP-016 and anti-CD37 antibodies that display crosslinker-independent killing.

67

Figure 4.1. Combining TLR 7/8 agonist CL-075 with TRU-016 is not beneficial.

continued 68

Figure 4.1 continued

C57BL/6 mice engrafted with hCD37 leukemia via lateral tail vein and randomized to treatment

groups that received 2 mg/kg CL-075 (or vehicle) and 10 mg/kg TRU-016 (or trastuzumab

control) following leukemia development (>1200 CD5+ B-cells/µl) in peripheral blood. Mean +/–

SEM for n=10-12 mice/group. A) Absolute count of leukemia cells in peripheral blood. B)

Leukemic B-cells normalized to the count prior to the first treatment. C) Overall survival. Mice

meeting IACUC approved early removal criteria were euthanized by a blinded technician.

69

Figure 4.2. BI 836826 effector-mediated killing

A) Phagocytosis of CLL B-cells by MDMs (n=8) measured by flow cytometry using fluorescently labeled cells B) ADCC with standard 4h 51Cr release assay. CLL B-cells and allogeneic NK cells from healthy donors (n=6). *p<0.05, ANOVA with Holm-SIdak post-hoc analysis.

70

Figure 4.3. Combining BI 836826 with idelalisib improves cytotoxicity

Direct cytotoxicity of BI 836826 (0.1 µg/ml) +/– 1 µM idelalisib after 24 hour incubation with primary human CLL B-cells from A) high risk patients with deletion of 17p (n=13), B) lower risk

cytogenetic groups: deletion 13q / trisomy 12 / normal cytogenetics (n=11), or C) ibrutinib-

refractory patients with cysteine 481 mutation (n=3). *p<0.05, ANOVA, Holm-Sidak test.

71

Figure 4.4. Preliminary evaluation of BI 836826 in hCD37 leukemia model

A) Example bivariate plots showing reduction in leukemia 48h after i.p. injection with 10 mg/kg BI

836826 (mAb 37.1), non-engineered control, or non-binding chimeric IgG1 control (rituximab) on days 0, 3, 7, 10, and 14. B) Absolute leukemic B-cell counts in peripheral blood of mice engrafted with hCD37 leukemia.

72

CHAPTER 5

BISPECIFIC ANTIBODIES: BRINGING T-CELLS INTO THE FOLD

The therapeutics which are the focus of the preceding chapters could be viewed as building upon traditional immunotherapy. Some simply improve upon existing function

(e.g. Fc-engineered BI836826), while others introduce new mechanisms of action while retaining those attributable to the original mAb (IMGN529). These anti-tumor mechanisms can include fixing complement proteins, blocking ligand binding, mediating cytotoxicity (or phagocytosis) by innate immune cells, or directly inducing apoptotic signaling. However, they have no means to recruit adaptive immune cells for this fight.

This chapter takes a detour from these more conventional immunotherapies, exploring a strategy that redirects T-cells to eliminate tumor.

CLL exhibits widespread signs of immune dysfunction, which can include: hypogammaglobinemia, autoimmunity, inhibition of NK cell function, and profound T-cell immunodeficiency (Riches and Gribben, 2014). CLL patients typically have a decreased

CD4:CD8 ratio and this inversion is correlated with decreased time to first treatment and progression-free survival (Nunes et al., 2012). Contact with malignant B-cells results in altered expression of genes responsible for proliferation, vesicle trafficking, cytoskeletal remodeling, and CD8 T-cell cytotoxicity (Gorgon et al. 2005). Unsurprisingly, T-cells from

CLL patients show decreased proliferation following stimulation and impaired immunological synapse formation (Kay and Kaplan 1986; Ayanlar-Batuman et al., 1986;

Ramsay et al., 2008).

73

The abundance of T-cell defects in CLL can present challenges that must be overcome for immunotherapeutic strategies relying on these cells. Bispecific antibodies capable of recruiting/activating T-cells (via CD3 binding) are one approach.

Blinatumomab, a CD19/CD3 targeting bispecific T-cell engager (BiTE), recently received

FDA approval for the treatment of relapsed/refractory acute lymphoblastic leukemia

(ALL). In a Phase II clinical trial 69% of patients with relapsed/refractory disease achieved CRs (Topp et al., 2011). This therapy has also been reported to be effective against primary CLL in vitro, despite the T-cell defects that plague CLL (Löffler et al.,

2003; Wong et al., 2013). While a number of B-cell antigens have been investigated as targets for bispecific antibodies, targeting CD37 has not been explored. TSC-TRU is a newly developed anti-CD37 × anti-CD3 bispecific ADAPTIR peptide (Figure 1.3) that was constructed by linking CD3-targeted scFv to TRU-016. In this chapter, I test whether the immunodeficiencies of CLL can be overcome by this therapy, and discuss potential reasons subpar performance.

Methods

Reagents

TSC-TRU and non-specific anti-HER2 × anti-CD3 control ADAPTIR therapeutics were provided by Emergent Biosolutions (Gaithersburg, MD).

Titration of TSC-TRU

CLL patient whole blood was incubated with TSC-TRU for 1 hour to identify concentrations at which this reagent caused T-cell association with B-cells. Association was measured by flow cytometry, without removing doublet cells from the analysis such 74

that CD19+CD3+ events were considered to be associated cells. The activation status at various concentrations was determined by CD69 staining after 20h co-culture of B/T- cells at a 1:1 ratio.

Cytotoxicity experiments

Healthy donor T-cells (n=3) or autologous T-cells from CLL patients (n=11) were cultured with PKH67-labeled CLL B-cells for 24 or 48h with antibody. Absolute cells counts of live CLL B-cells were obtained by flow cytometry. %Cytotoxicity = 100 x

[(experimental count − spontaneous death count) ÷ (100 − spontaneous death count)]

100 The CLL PBMC experiments were performed as above, but were treated for 48h

(n=10) or 6d (n=5). The gating strategy is outlined in Figure 5.1 for these studies in outlined in Figure 5.7.

Results

Engaging T-cells using a novel anti-CD37 × anti-CD3 bispecific ADAPTIR

As I reviewed above, T-cells from CLL patients are highly dysfunctional.

However, it may still be possible to overcome this immunodeficiency with a bispecific

antibody. The initial studies with TSC-TRU established a range of concentrations

capable of recruiting/activating T-cells in the presence of CLL B-cells (Figure 5.2). To

control for non-specific TCR stimulation by the anti-CD3 portion of the therapeutic, anti-

HER2 × anti-CD3 ADAPTIR (TSC-CTL) was used. Maximum B/T association was

achieved around 100 ng/ml (Figure 5.2a), but some non-specific activation of CD8 T-

cells also occurs at this concentration (Figure 5.2b). While non-specific activation by the

control was absent at both 1 ng/ml and 10 ng/ml, treatment with TSC-TRU at these

75

concentrations still achieved similar levels of activation (despite the rapid drop in cell association at 1 ng/ml).

In allogeneic cytotoxicity experiments (Figure 5.3a), the ability to eliminate CLL

B-cells at these concentrations correlated well with the trends in activation. At 10 ng/ml,

TSC-TRU displayed modest cytotoxicity while avoiding the non-specific killing that occurred at 100 ng/ml. Autologous killing assays produced a very similar result at a 5:1

T-cell to B-cell ratio (Figure 5.3b,c). Other bispecific antibodies have been reported to function at very low effector-to-target (E:T) ratios that may better reflect the endogenous cell proportions seen in patients (Löffler et al., 2003; Aigner et al., 2013; Wong et al.,

2013). With this in mind, I performed a new set of experiments using PBMCs from 11

CLL patients. Despite activating both CD4 and CD8 T-cells (Figure 5.4), TSC-TRU showed little evidence it could deplete CLL B-cells under these conditions (Figure 5.5a, b). T-cell numbers were unchanged by TSC-TRU, although treatment with the control led to decreased T-cell counts observed in the control condition (Figure 5.5c-f).

While some killing may be expected at 48 hours, a longer incubation period may be necessary when dealing with such low E:T ratios. To test this hypothesis, five PBMC samples from CLL patients were incubated with TSC-TRU or its control for 6 days

(Figure 5.6). The results were highly variable once again, resulting in no significant difference between the mean B-cell counts of TSC-TRU and its control. In fact, 2 of 5 patients appeared to experience improved B-cell survival (Figure 5.6a, b). The effect on

T-cells was similarly disappointing, the most notable being depletion of both CD4 and

CD8 populations by the anti-HER2 × anti-CD3 control (Fig. 5.6c-f).

76

Discussion

Initially, recruiting T-cells using TSC-TRU appeared to be a promising alternative strategy for targeting CD37 in CLL. Allogeneic T-cells were capable of eliminating CLL

B-cells at E:T ratios ranging from 20:1 to 1:1 (Figure 5.3a). Autologous T-cells from CLL patients were also capable of depleting B-cells at a 5:1 ratio (5.3b, c). However, other bispecific antibodies have been shown to retain activity at more physiologically relevant effector-to-target ratios, where malignant cell numbers are expected to far exceed those of T-cells. When tested under these conditions using CLL patient PBMCs, the results were far less favorable. Despite activating T-cells (Figure 5.4), the cytotoxicity of TSC-

TRU was largely unimpressive at these low E:T ratios (Figure 5.5a, b) and longer incubation times did little to improve upon this (Figure 5.5a, b). The lack of significant T- cell expansion is also not very encouraging (Figure 5.5c-f and Figure 5.6c-f).

The overall performance of TSC-TRU was relatively poor when compared with other bispecific antibodies that have been developed in recent years. For example, both the anti-CD33 BiTE (AMG 330) in AML and anti-CD19 BiTE (blinatumomab) in CLL were effective at eliminating malignant cells at very low E:T ratios (Löffler et al., 2003; Aigner et al., 2013; Wong et al., 2013). Possible explanations for the different outcomes observed with TSC-TRU include: 1) differences in experimental methodology, 2) BiTE design confers advantages not provided by a bispecific ADAPTIR, or 3) CD37 is a poor target for bispecific antibodies. The first possibility appears unlikely, given that my approach was nearly identical to those used in the BiTE publications. If the therapeutic was effective, activity should have been evident after 5+ days of culture. It may have been useful to include a positive control for T-cell proliferation (anti-CD3/CD28 stimulation), but the lack of consistent anti-tumor effect despite evidence of T-cell

77

activation would remain troubling. Furthermore, this ADAPTIR format appears to induce a high degree of non-specific TCR activation that may be promoting activation-induced cell death that ultimately decreases the T-cell counts (Figure 5.5c-f and Figure 5.6c-f).

The results from my experiments with TSC-TRU raise a question about the design of bispecific antibodies – is the BiTE design superior to the ADAPTIR format?

Early bispecific antibodies constructed from traditional mAbs showed poor activity compared to recent designs (Wolf et al., 2005). BiTEs consist of two scFv (one targeting

CD19 and the other CD3) linked together, leading to a very small therapeutic (54 kDa).

This is accompanied by some disadvantages, namely that the drug requires continuous infusion due to its rapid excretion (Topp et al., 2011). However, it also forces target cells into closer proximity with one another, which may play a major role in its activity (Wolf et al., 2005).

Finally, we must consider whether CD37 is even a suitable target for bispecific antibodies. T-cells express a very small amount CD37 that could be influencing the activity of TSC-TRU. Although there is no evidence that CD37-specific antibodies deplete this cell population, the expression threshold needed to induce target killing by

T-cell immunotherapies is significantly lower. Further studies would be required to determine if this T-cell fratricide was occurring. However, I currently suspect that difficulties primarily arise from the ADAPTIR design, as I will discuss further in the conclusions chapter.

78

Figure 5.1. IgG-derived therapeutics constructed from scFv

continued 79

Figure 5.1 continued

Top left: Otlertuzumab (TRU-016) is an ADAPTIR™ molecule, constructed from an anti-CD37 single-chain variable fragment (scFv; a binding domain formed by linking the heavy and light chain variable regions of an immunoglobulin) which has been fused to the hinge region and Fc domain of human IgG1. Top right: TSC-TRU is a bispecific ADAPTIR designed with anti-CD3 and anti-CD37 scFv linked to a hinge region and IgG4 backbone (CH2 and CH3) with N297A mutation to eliminate potential for ADCC. Bottom left: Structure of a normal immunoglobulin

(IgG). Bottom right: Blinatumomab, a BiTE (bispecific T-cell engager) constructed from anti-CD3 and anti-CD19 scFv. VH, heavy chain variable region; VL, light chain variable region; CH, heavy chain constant region (1, 2, or 3); CL, light chain constant region.

80

Figure 5.2. TSC-TRU recruits and activates T-cells

A) Association of CLL B-cells with T-cells in whole blood, measured as CD19+CD3+ doublets by flow cytometry with low flow rate B) Percentage of CD4 or CD8 T-cells expressing CD69 after

24h incubation at 1:1 ratio of B-cells to T-cells.

81

Figure 5.3. Cytotoxicity of TSC-TRU with allogeneic or autologous T-cells

Flow-based cytotoxicity assay, 48 hour incubation with various E:T ratios. A) Allogeneic T-cells cultured with CLL B-cells (Mean ± SEM; n=3) B) Autologous T-cells at 5:1 E:T ratio or without T- cells (Mean ± SEM; n=10) . %Cytotoxicity = [(experimental count − spontaneous death count) ⁄

(100 − spontaneous death count)] x 100. *p<0.05 relative to appropriate control. ANOVA. 82

Figure 5.4. T-cell activation by TSC-TRU in CLL PBMC cultures

Activation of T-cells in PBMC cultures after 48 hours, as measured by detection of CD69 with flow cytometry. Expressed as percentage of CD69+ events among A) CD4 and B) CD8 T-cells

83

Figure 5.5. CLL B-cells evade killing by TSC-TRU in 48 hour PBMC cultures

continued

84

Figure 5.5 continued

Two day antibody incubation with PBMCs from CLL patients (n=10) as measured by a flow-based cytotoxicity assay using count beads to calculate live cell concentrations. A, C, E) Absolute cell counts and B, D, F) normalized cytotoxicity. A-B) B-cells, C-D) CD4 T-cells, E-F) CD8 T-cells.

*p<0.05 by one-way ANOVA with Holm-Sidak test.

85

Figure 5.6. Longer incubation has minimal impact on TSC-TRU efficacy in PBMCs

continued 86

Figure 5.6 continued

Six day antibody incubation with PBMCs from CLL patients (n=5), as measured by a flow-based cytotoxicity assay using count beads to calculate live cell concentrations. A, C, E) Absolute cell counts and B, D, F) normalized cytotoxicity. A-B) B-cells, C-D) CD4 T-cells, E-F) CD8 T-cells.

*p<0.05 by one-way ANOVA with Holm-Sidak test.

87

Figure 5.7. Gating strategy for PBMC co-culture experiments

Cells stained with anti-CD3 and CD19 antibodies to identify T-cells and B-cells.

Activation determined using anti-CD69 antibody. Propidium iodide included to exclude

dead cells, and live cell counts were obtained using CountBright beads.

88

CHAPTER 6

EXPLORING THE FUNCTION OF CD37 IN LEUKEMIA

While CD37 has long been pursued as a target for immunotherapy, surprisingly little is known about its biological function. It is unknown whether a natural ligand for

CD37 exists, but ligation by anti-CD37 peptide simultaneously induces both pro- apoptotic inactivation of Akt and opposing pro-survival phosphoinositide 3-kinase (PI3K) activation. CD37 is also critical to normal plasma cell development, clustering VLA-4

(α4β1 integrin) on the surface of germinal center B-cells to receive pro-survival signals from follicular dendritic cells (van Spriel et al., 2012). In CLL, VLA-4 subunit expression is associated with more aggressive disease and promotes homing and adhesion to supportive cells in the tumor microenvironment (Vincent et al., 1996; Burger et al., 1999;

Gattei et al., 2008). Given its role in controlling VLA-4 function, CD37 could potentially influence the development or progression of CLL. This could occur through at least two mechanisms: 1) directly promoting integrin-mediated survival of CLL B-cells, or 2) altering VLA-4 dependent adhesion (which could alter cell trafficking). To explore this further, I sought to generate a model of CD37 deficient leukemia by crossing CD37 knockout mice with the TCL1 mouse model of CLL. My initial hypothesis was that loss of

CD37 expression would delay disease onset in TCL1 mice (and decrease leukemic B- cell homing to lymphoid tissue). This chapter describes one important aspect of an ongoing project to test this hypothesis.

89

Materials and Methods

Mice

CD37 knockout mice were graciously provided by Dr. Annemiek van Spriel and bred with TCL1 mice on a C57BL/7 background. The offspring were then bred with either homozygous knockout mice or C57BL/6 mice to produce the desired genotypes. Mice were housed in microisolator cages and euthanized upon reaching IACUC-approved early removal criteria.

Flow cytometry

Peripheral blood leukemia was monitored using anti-CD5, CD19, and CD45 antibodies

(BD Biosciences) and described in greater detail within Chapter 3.

Results

After obtaining CD37–/– mice, we immediately began to breed them with the TCL1

line with the goal of obtaining hemizygous TCL1 mice with varying CD37 genotypes,

which were determined by PCR (Figure 6.1a). Lack of CD37 expression was further

confirmed by flow cytometry (Figure 6.1b). These mice were followed longitudinally to

measure overall survival, with any decisions to euthanize made by a blinded technician.

Interestingly, loss of CD37 was associated with decreased survival among TCL1 mice

(Figure 6.2). Deceased mice displayed signs of leukemia consistent with expectations

for the TCL1 mouse. A cohort of mice were also monitored by flow cytometry to examine

whether leukemic burden or time to disease onset differed. No significant differences

were detected between the groups (Figure 6.3).

90

Discussion

CD37 is known to promote survival of germinal center B-cells due to its role in clustering the integrin VLA-4 (van Spriel et al., 2012). Given that VLA-4 is functionally relevant in human CLL, I suspected that CD37 may also have a pro-leukemic effect in murine CLL. Expression of the VLA-4 subunit CD49d is associated with more aggressive

CLL and it promotes homing and adhesion to supportive cells in the tumor microenvironment (Vincent et al., 1996; Burger et al., 1999; Gattei et al., 2008). I originally hypothesized that loss of CD37 would prolong the survival of TCL1 mice, but it appears the opposite is true based on my initial work (Figure 6.2). This is not entirely surprising, because CD37 is globally knocked out and it does have several reported functions in non-B leukocytes (despite its relatively low expression on these cells). In particular, CD37–/– mice exhibit defects in cellular immunity stemming from impaired dendritic cell migration and priming of T-cells. When murine RMA cell line expressing human MUC1 antigen was injected into CD37 knockout mice, they showed impaired ability to reject the tumor relative to wildtype mice (Gartlan et al., 2013). In addition to potential effects on anti-tumor immunity, it is also possible there are B-cell intrinsic effects which led to decreased survival. It was recently reported that approximately 50% of CD37–/– mice aged >12-18 months can develop an IL-6 dependent IgA+ lymphoma

with a phenotype resembling mature germinal B-cells. The authors provide evidence that

regulation of the IL-6 pathway by SOCS3 is disrupted upon loss of CD37, resulting in

constitutive activation and lymphoma development (de Winde et al., 2016). Elevated

serum IL-6 is correlated with more aggressive disease in human CLL (Fayad et al.,

2001), so this pathway could feasibly contribute to the decreased survival of CD37–/–

TCL1 mice. However, it should be noted that the lymphoma observed by Winde et al.

91

could not establish itself when adoptively transferred into wildtype mice, instead requiring either healthy CD37–/– mice or extremely immunocompromised NSG recipients for successful engraftment to occur. This lends support to the notion that B-cell extrinsic factors (e.g. dysfunctional cellular immunity) are major contributors to the development of B-cell malignancy in CD37–/– mice. Isolating the effects of intrinsic vs. extrinsic factors

in the CD37–/–TCL1 model will be important to understanding the role of CD37 in

leukemia/lymphoma, and will therefore be a focus of future studies.

92

Figure 6.1. Confirmation of CD37 knockout

A) Murine CD37 (mCD37) or the neomycin resistance (Neo; inserted during generation of knockout) amplified by PCR. Lanes from left to right: C57BL/6 control, CD37–/– founder, CD37–/+ offspring, #1-5 TCL1+ offspring. B) Expression of murine CD37 on B/T lymphocytes in peripheral blood from a wildtype control and TCL1+ mouse determined to be CD37–/– by PCR.

93

Figure 6.2. CD37 deficiency reduces TCL1 mouse survival.

Mice hemizygous for the TCL1 transgene with wildtype CD37 (n=28; median 413 days), heterozygous knockout (n=36; median 364 days), or homozygous knockout (n=20; median 305 days). Log-rank analysis of survival.

94

Figure 6.3. Measurement of leukemic burden in peripheral blood

Flow cytometric analysis of whole blood, displayed as percentage of CD45+ leukocytes that are leukemic CD5+CD19+ B-cells (+/− SEM) for n=8-20 mice per genotype.

95

CHAPTER 7

CONCLUSIONS

Summary of Key Findings

While recent advances have improved the care of CLL patients, disease progression is inevitable with currently available therapeutics. The overall objective of this thesis was to develop novel approaches to CLL immunotherapy that can be successfully translated to the clinic. One of the most promising targets for such therapy is CD37, due to its high expression in many B-cell malignancies (while it is very low or absent among non-B leukocytes) and its restriction to the hematopoietic system.

In Chapter 2, I attempt to address a major challenge in the preclinical study of

CD37-targeted therapy – the lack of suitable CLL animal models for evaluating these treatments. The TCL1 mouse represents the best characterized in vivo model for drug development in CLL, but anti-human CD37 mAbs do not bind to the murine protein. Our lab has generated a human CD37 transgenic mouse, which I have extensively characterized. In this mouse, human CD37 is specifically expressed on B-cells and they are sensitive to apoptosis induced by anti-CD37 mAb. Upon crossing these mice with the TCL1 line, the resulting hCD37xTCL1 develop the expected CLL-like leukemia that now expresses the desired human CD37. This provided a tool that would permit the in vivo evaluation of a novel anti-CD37 antibody-drug conjugate (IMGN529) in Chapter 3.

An extensive series of in vitro studies treating primary human CLL cells with IMGN529 demonstrate that conjugation to the anti-microtubule toxin DM1 does not diminish the

96

activity attributable to its IgG1 precursor (K7153A). This includes high levels of direct cytotoxicity, induction of phagocytosis by macrophages and killing mediated by NK cells.

This anti-CD37 therapy is also highly active in CLL patient whole blood, where a variety of potential effector cells are present at concentrations that are seen endogenously. In vivo studies with hCD37xTCL1 leukemia, demonstrate that IMGN529 quickly eliminates leukemic B-cells and improves overall survival of the mice. I hypothesized that this ADC could target the proliferative component of the leukemia through delivery of anti- microtubule toxin to neoplastic cells (in addition to its mAb-derived mechanisms of killing). IMGN529 proved very capable of eradicating this dividing cells within the lymphoid tissues of leukemic mice. Notably, this is the first evidence that antibody-drug conjugates carrying a microtubule-disrupting payload can successfully target this important subset of CLL B-cells.

In Chapter 4, I investigate potential strategies to improve the quality of anti-CD37 therapy. Initial attempts to augment effector-mediated killing by combining TRU-016 with a TLR7/8 agonist were unsuccessful. I moved on to study BI 836826, an Fc-engineered mAb where effector functions were already enhanced. We had been exploring addition of a PI3Kδ inhibitor to compensate for the activation that occurs concurrent with the induction of pro-apoptotic signaling triggered by CD37-targeted therapeutics. Notably, I provide evidence that not only supports use of idelalisib in combination with BI 836826, but also demonstrate this strategy retains its impact against B-cells from high risk patients with 17p deletion or resistance to ibrutinib therapy.

Chapter 5 shifts focus toward immunotherapy that re-directs T-cells to kill tumor

cells. The activity of bispecific (anti-CD37 × anti-CD3) TSC-TRU was initially promising,

demonstrating a capacity for recruiting and activating T-cells which killed CLL patient B-

97

cells. In contrast to reports with other bispecific mAbs, however, its benefits were lost when tested using the lower, endogenous effector-to-target ratios of PBMC cultures.

Chapter 6 moves away from targeting CD37, instead exploring the biological function of this tetraspanin in leukemia. This was examined using a CD37-deficient model of murine CLL that was developed by crossing the TCL1 line with CD37 knockout mice. The overall survival of CD37–/–TCL1 mice was decreased, although I did not observe significant differences in peripheral leukemia when monitoring mice for disease development. Further studies will be required to determine whether B-cell intrinsic or extrinsic effects are responsible for the altered survival.

Significance

As a whole, this thesis describes a variety of approaches to immunotherapy that can be built upon in the laboratory and the clinic. In particular, it represents a comprehensive inquiry into CD37-targeted therapeutic strategies. Use of CD37 as a target for immunotherapy is clinically validated by otlertuzumab (Byrd et al., 2014) and newer generations of anti-CD37 therapeutics are likely to further improve upon these results. Antibody-based immunotherapy and kinase inhibitors such as ibrutinib have significantly improved the management of CLL. However, the disease is still incurable without a hematopoietic cell transplant, which carries significant risk in the elderly population usually afflicted with CLL. Given the limitations of these therapies, particularly when confronted with the challenge of treating relapsed/refractory disease, it is important to expand our therapeutic arsenal. The work presented in this thesis builds toward accomplishing this goal.

98

The development of the hCD37xTCL1 mouse has already been an invaluable tool for the preclinical evaluation of anti-human CD37 therapeutics. Previously, we have been limited to engrafting severely immunodeficient mice with human cell lines in order to evaluate CD37-targeted therapies. These models do not accurately recapitulate the complexity of the multicompartmental disease observed in humans, where non-dividing

CLL B-cells accumulate in the blood, but those inhabiting bone marrow, spleen, and lymph nodes receive very different microenvironmental stimuli in these niches.

Responses to these immunotherapies could vary based on the location of the malignant

cells, thus it is critical to evaluate their activity in both blood and lymphoid tissues.

This importance of the hCD37xTCL1 model is particularly evident when examining the case of antibody-drug conjugate IMGN529. While conjugating drug to the

Fc fragment of a mAb can reduce its effector activities, I showed this did not occur with

IMGN529. In addition, it retains the potent direct (crosslinker-independent) cytotoxicity exhibited by its precursor mAb. This affords an opportunity to exploit the unique properties of CD37 with a therapeutic that: 1) maintains the Fc-mediated effector functions of IgG1, 2) delivers toxin into tumor cells through endocytosis, and 3) mediates potent antibody-induced apoptosis. This gives IMGN529 a unique repertoire of mechanisms among therapeutics for B-cell malignancy. With the hCD37xTCL1 model, I demonstrate that IMGN529 can eliminate the proliferative subset of leukemia cells found within lymphoid tissues. This is the first example of antibody-drug conjugate doing so within an in vivo CLL model. The impact of the anti-microtubule drug is clear, as the unconjugated antibody precursor could not control the disease, despite showing some signs of activity.

99

For many years CLL was viewed as a disease of slowly dividing neoplastic B- cells with defects in apoptosis, but this has changed as the importance of a proliferative component has become more apparent (Chiorazzi et al., 2005; Messmer et al., 2005;

Soma et al., 2006). A subset of transformed B-cells form proliferative centers in lymphoid tissues, which are the source of the malignant cells that accumulate in the blood (Soma et al., 2006; Caligaris-Cappio et al., 2008; Vandewoestyne et al., 2011). I suspect incomplete clearance of this population is partly responsible for why mAbs typically require combination with chemotherapy to achieve complete responses in patients (Byrd et al., 2001). While fludarabine, for example, can exhibit cytotoxicity against non-dividing lymphocytes in vitro, purine analogs are naturally more effective at killing proliferating

cells that are actively replicating their DNA (Sandoval et al., 1997; Genini et al., 2000).

Likewise, additional delivery of anti-microtubule drug by IMGN529 may help eliminate

the proliferative subset and provide the extra push needed to facilitate clearance of non-

dividing cells. It is unlikely that IMGN529 would be the treatment of choice for most CLL,

but as discussed earlier, the course of this disease is very heterogeneous. Some

patients will progress much faster and higher rates of proliferation can lead to a birth of

new CLL B-cells exceeding 1% of the total malignant clone per day (Caligaris-Cappio,

2003). A therapeutic like IMGN529 may be more suited to patients in the midst of a

progressive phase of their disease (especially if they are relapsed/refractory to other

treatment) or in the 5-10% of patients who experience Richter’s transformation, where

the disease changes into an aggressive lymphoma with very poor prognosis.

As mentioned previously, the number of CRs achieved by antibody monotherapy

is often underwhelming, but many of the partial responders can be converted to CRs

when a combination approach is taken. This was also true of otlertuzumab (TRU-016),

100

which performed much better with the addition of chemotherapy. Identifying combination strategies that can be logically integrated with anti-CD37 therapeutics will be an important next step to advancing these therapies. I first explored a strategy to improve

Fc-mediated killing by combining TRU-016 with a TLR 7/8 agonist (CL-075), which had appeared promising on the basis of in vitro studies performed by a collaborator. While the in vivo results did not support the further development of TLR 7/8 agonists in CLL, it was valuable information to have as these drugs had not been previously evaluated in hematological malignancy.

Signaling induced by SMIP-016 (the chimeric version of TRU-016) was

previously shown to induce both pro-apoptotic SHP1 and pro-survival PI3Kδ signaling

(Lapalombella et al., 2012). It remains unknown whether this mechanism is also shared

with Type II anti-CD37 antibodies (BI 836826, K7153A, and IMGN529) which

demonstrate crosslinker-independent direct cytotoxicity. However, we did observe improved cytotoxicity when combining idelalisib with BI 836826 (similar to what was observed with SMIP-016). It is notable that this killing was retained to an equivalent degree against cells from patients with 17p deletion. Identifying strategies that improve outcomes for this high risk population should be a very high priority, as their poor prognosis has persisted despite major treatment advances. The German CLL8 trial observed median progression-free survival of del(17p) patients on FCR was approximately 12 months; extremely poor compared to those with normal cytogenetics

(≈50 months), deletion 11q (≈50 months), trisomy 12 (≈70 months), or deletion 13q (≈70 months)(Stilgenbauer et al., 2014). In contrast, response to ibrutinib is similar across cytogenetic groups, although high risk patients with del(17p) or del(11q) still make up the vast majority of patients (10 out of 11) who saw progression (Byrd et al., 2013). Given

101

the persistent difficulty with treating del(17p), our observation that combination of BI

836826 and idelalisib does not have reduced activity against del(17p) cells ex vivo is encouraging and warrants additional investigation.

The eventual decision to abandon of study of anti-CD37 × anti-CD3 bispecific

TSC-TRU was disappointing after the initial promise it showed, but the data obtained does provide insight and raised important questions. In particular, what barriers prevent this therapy from functioning in CLL PBMC cultures with low E:T ratios, while the anti-

CD19 × anti-CD3 BiTE Blinatumomab remains active? Does the failure lie with TSC-

TRU itself, or is CD37 an unsuitable target for this class of therapeutic? There were several warning signs that suggest the design may be at fault. Even early studies where

TSC-TRU exhibited cytotoxicity suggest it is less potent than other bispecific antibodies.

While killing largely disappeared at lower E:T ratios using less than 0.01 µg/ml of TSC-

TRU, blinatumomab is reported to kill in CLL PBMC cultures at 0.005 µg/ml with E:T ratios of 1:50 (Löffler et al., 2003). The maximum killing seen with anti-CD19 × anti-CD3

DART bispecific was reached as low as 0.0001 µg/ml when tested against the Raji cell line (Moore et al., 2011). Furthermore, within concentration ranges where BiTE and

DART molecules kill tumor cells there was no activation T-cells when either a control antibody or CD19-negative cell lines were used. This is in stark contrast to what I observed with the bispecific ADAPTIR format, where even the anti-HER2 × anti-CD3 control activated T-cells and caused B-cell depletion at relevant concentrations. I believe the poor performance is more likely a result of the anti-CD3 used in the construction of the ADAPTIR. The binding affinity of the T-cell activating scFv could be too strong, as lower affinity formats tend to perform better in bispecific antibodies (Bortoletto et al.,

2002). It is also possible that fratricide contributes to poor efficacy, as CD37 is

102

expressed at very low levels on T-cells. However, there has been no evidence of this in any experiments performed by myself or the company. Ultimately, we lack evidence to conclude CD37 is not suitable for targeting by T-cell based immunotherapies.

The preliminary work with the CD37–/–TCL1 mice suggests that CD37 may be

acting as a tumor suppressor in this model. This may be related to the dysfunctional IL-6

signaling recently reported in very old CD37–/–- mice, which leads to lymphoma

development between the ages of 12-18 months (de Winde et al., 2016). However, it is

difficult to say whether the decreased survival results from B-cell intrinsic or extrinsic

effects of CD37 deficiency. Dendritic cell migration and priming of T-cells are defective in

CD37–/– mice, which impairs anti-tumor immunity (Gartlan et al., 2013). In light of my

observations and these publications, additional studies will be needed to further

elucidate the role of CD37 in leukemia.

Future Directions

Antibody-drug conjugates (ADCs)

There is considerable progress to be with strategies that utilize antibody-drug

conjugates, including (but is not limited to) work with IMGN529. Naturally, there are

unanswered questions in regards to its clinical efficacy that will be addressed in the near

future by ongoing trials. If the results in NHL are encouraging, it may be valuable to

pursue a trial which treats relapsed/refractory CLL patients who display signs of

accelerated disease, and/or patients with Richter’s transformation. Beyond the clinical

development of IMGN529, I believe there is significant work that could be done to

improve the efficacy of ADCs in general. One strategy to improve therapeutics which

retain strong ADCC activity post-conjugation is to augment Fc-mediated killing with

103

either combination therapy (e.g. lenalidomide) or by using an Fc- engineered/afucosylated antibody to construct them. This approach has been used for anti-BCMA antibody-drug conjugate constructed from an afucosylated IgG1 (Tai et al.,

2014). They do show evidence of macrophage recruitment to the tumor cells in vivo, but unfortunately neglect to compare the fucosylated and afucsoylated forms of the ADC in their mouse models (comparing only ADC vs. unconjugated afucosylated mAb). I think this approach merits further consideration, although its impact could vary significantly from one antigen to another due to the dependence on how quickly the antibody is internalized.

Targeting CD37+ B-cell leukemia with T-cells

Despite the disappointment with the TSC-TRU bispecific, I do not think this closes the door on CD37 as a target for T-cell dependent immunotherapy. Many bispecific antibody formats have failed in the past and as discussed previously, I would hypothesize this is a failure of design rather than the target. In addition to recruiting T- cells to CD37-expressing targets via bispecific antibody, a chimeric antigen receptor could also be utilized (and I have already designed an anti-CD37 CAR that could be used for this purpose). With that said, it may still be a suboptimal target for CAR T-cell

and bispecific antibody therapies, as the low expression required to initiate killing may

have unfortunate effects. Given the severe neutropenia observed in the IMGN529

clinical trial, CAR T-cells or bispecific antibodies might be expected to induce killing of

neutrophils. Rigorous preclinical testing would be required to evaluate this possibility, but

I would be cautious about using cellular immunotherapy against this target as it cannot

be easily withdrawn. For that reason, redirection of T-cells to CD37+ tumor might be

104

more appropriate with a bispecific antibody. With this approach, it may be possible to avoid neutropenia with suitable prophylaxis (as in the IMGN529 trial), yet there is still an opportunity to abort therapy if necessary (particularly if a BiTE format with low serum half-life was used).

Biological function of CD37

My work with the CD37-deficient TCL1 mice thus far has raised some interesting questions, but there is much we do not yet understand about this protein and tetraspanin biology in general. While there was no significant difference across genotypes in the peripheral leukemic burden of the cohort I followed more closely by flow, the disease may be reaching a terminal stage more quickly. It was not uncommon for leukemic

CD37–/– mice to die more rapidly than expected (based on past experience with TCL1

mice), such that mice that appeared to be weeks away from reaching early removal

criteria would be found dead several days later. Current data to support this is

insufficient, however, and a larger cohort of mice would need to be followed closely to

evaluate this possibility. Furthermore, with our current evidence it is not possible to

distinguish between the B-cell intrinsic vs. extrinsic effects of CD37 loss. It will be very

important to perform adoptive transfer experiments to better understand how this

tetraspanin influences murine CLL.

Extrinsic defects in cellular immunity could explain my preliminary findings. As

previously discussed, CD37–/– mice exhibit dendritic cell defects which impair clearance of adoptively transferred tumor (Gartlan et al., 2013). The described IL-6 related dysfunction and increased production of this cytokine is not evident until mice are over 1 year old (de Winde et al., 2016), a point at which less than 40% of CD37–/– TCL1 mice

105

remain alive in my studies. Therefore, I would hypothesize that B-cell extrinsic defects in anti-tumor immunity have a larger impact on the observations made in CD37–/– TCL1 mice. Despite what we have seen thus far, loss of CD37 may still be detrimental to the engraftment and progression of adoptively transferred CD37–/– leukemia. It is clear that

CD37 has a variety of complex roles and their relative contributions could be difficult to discern. Given the complexity of tetraspanin web, it is possible that different B-cell intrinsic functions of CD37 are more important in certain malignancies. While de Winde et al., reports a wide variety of diffuse large B-cell lymphoma cases that express low/absent CD37 (and exhibit a dysfunctional IL-6 signaling axis), I have never observed

CD37 negative CLL. The study of CD37 function is a challenging endeavor, but an improved understanding of its role in B-cell malignancy may not only provide valuable insight in these diseases, but could have broad relevance to the study of tetraspanins in general.

106

REFERENCES

Amiot M (1990) Identification and analysis of cDNA clones encoding CD53. A pan- leukocyte antigen related to membrane transport proteins. J Immunol 145: 4322- 4325.

Andre M, Le Caer JP, Greco C, Planchon S, El Nemer W, Boucheix C, Rubinstein E, Chamot-Rooke J, Le Naour F (2006) Proteomic analysis of the tetraspanin web using LC-ESI-MS/MS and MALDI-FTICR-MS. Proteomics 6: 1437-1449.

Badoux XC, Keating MJ, Wang X, O'Brien SM, Ferrajoli A, Faderl S, Burger J, Koller C, Lerner S, Kantarjian H, Wierda WG (2011) Fludarabine, cyclophosphamide, and rituximab chemoimmunotherapy is highly effective treatment for relapsed patients with CLL. Blood 117(11): 3016-24.

Bandyopadhyay S, Zhan R, Chaudhuri A, Watabe M, Pai SK, Hirota S, Hosobe S, Tsukada T, Miura K, Takano Y, Saito K, Pauza ME, Hayashi S, Wang Y, Mohinta S, Mashimo T, Iiizumi M, Furuta E, Watabe K (2006) Interaction of KAI1 on tumor cells with DARC on vascular endothelium leads to metastasis suppression. Nat Med 12: 933-938.

Barok M, Tanner M, Koninki K, Isola J (2011) Trastuzumab-DM1 causes tumour growth inhibition by mitotic catastrophe in trastuzumab-resistant breast cancer cells in vivo. Breast Cancer Res 13(2):R46.

Barreiro O, Yanez-Mo M, Sala-Valdes M, Gutierrez-Lopez MD, Ovalle S, Higginbottom A, Monk PN, Cabanas C, Sanchez-Madrid F (2005) Endothelial tetraspanin microdomains regulate leukocyte firm adhesion during extravasation. Blood 105: 2852-2861.

Barrena S, Almeida J, Yunta M, Lopez A, Fernandez-Mosteirin N, Giralt M, Romero M, Perdiguer L, Delgado M, Orfao A, Lazo, PA (2005) Aberrant expression of tetraspanin molecules in B-cell chronic lymphoproliferative disorders and its correlation with normal B-cell maturation. Leukemia 19:1376-1383.

Bauer P and Köhne K (1994) Evaluation of Experiments with Adaptive Interim Analyses. Biometrics. 50(4):1029-1041.

Beckmann J, Scheitza S, Wernet P, Fischer JC, Giebel B (2007) Asymmetric cell division within the human hematopoietic stem and progenitor cell compartment: identification of asymmetrically segregating proteins. Blood 109:5494-5501.

Beckwith KA, Frissora FW, Stefanovski MR, Towns WH, Cheney C, Mo X, Deckert J, Croce CM, Flynn JM, Andritsos LA, Jones JA, Maddocks KJ, Lozanski G, Byrd JC, Muthusamy N (2014) The CD37-targeting antibody-drug conjugate IMGN529 107

is highly active against human CLL and in a novel CD37 transgenic murine leukemia model. Leukemia 28: 1501-1510.

Beers SA, French RR, Chan HT, Lim SH, Jarrett TC, Vidal RM, Wijayaweera SS, Dixon SV, Kim H, Cox KL, Kerr JP, Johnston DA, Johnson PW, Verbeek JS, Glennie MJ, Cragg MS (2010) Antigenic modulation limits the efficacy of anti-CD20 antibodies: implications for antibody selection. Blood 115(25):5191-201.

Berditchevski F, Odintsova E, Sawada S, Gilbert E (2002) Expression of the palmitoylation-deficient CD151 weakens the association of alpha 3 beta 1 integrin with the tetraspanin-enriched microdomains and affects integrin- dependent signaling. J Biol Chem 277:36991-37000.

Berditchevski F, Tolias KF, Wong K, Carpenter CL, Helmer ME (1997) A novel link between integrins, transmembrane-4 superfamily proteins (CD63 and CD81), and phosphatidylinositol 4-kinase. J Biol Chem 272: 2595-2598.

Bichi R, Shinton SA, Martin ES, Koval A, Calin GA, Cesari R, Russo G, Hardy RR, Croce CM (2002) Human chronic lymphocytic leukemia modeled in mouse by targeted TCL1 expression. Proc Natl Acad Sci USA 99(10): 6955-60.

Bos SD, Lakenberg N, Van Der Breggen R, Houwing-Duistermaat JJ, Kloppenburg M, De Craen AJ, Beekman M, Meulenbelt I, Slagboom PE (2010) A genome-wide linkage scan reveals CD53 as an important regulator of innate TNF-alpha levels. Eur J Hum Genet 18: 953-959.

Bosca L, Lazo PA(1994) Induction of nitric oxide release by MRC OX-44 (anti-CD53) through a protein kinase C-dependent pathway in rat macrophages. J Exp Med 179, 1119-1126.

Broyde A, Boycov O, Strenov Y, Okon E, Shpilberg O, Bairey O (2009) Role and prognostic significance of the Ki-67 index in non-Hodgkin's lymphoma. Am J Hematol 84(6): 338-43.

Burger JA, Gribben JG (2013) The microenvironment in chronic lymphocytic leukemia (CLL) and other B cell malignancies: Insight into disease biology and new targeted therapies. Seminars in Cancer Biology.

Burger JA, Burger M, Kipps TJ (1999) Chronic lymphocytic leukemia B cells express functional CXCR4 chemokine receptors that mediate spontaneous migration beneath bone marrow stromal cells. Blood 94: 3658-67.

Burger JA, Tsukada N, Burger M, Zvaifler NJ, Dell’Aquila M, Kipps TJ (2000) Blood- derived nurse-like cells protect chronic lymphocytic leukemia B cells from spontaneous apoptosis through stromal cell-derived factor-1. Blood 96: 2655-63.

108

Burger JA, Zvaifler NJ, Tsukada N, Firestein GS, Kipps TJ (2001) Fibroblast-like synoviocytes support B-cell pseudoemperipolesis via a stromal cell-derived factor-1- and CD106 (VCAM-1)-dependent mechanism. J Clin Invest 107: 305- 315.

Burkle A, Niedermeier M, Schmitt-Graff A, Wierda WG, Keating MJ, Burger JA (2007) Overexpression of the CXCR5 chemokine receptor, and its ligand, CXCL13 in B- cell chronic lymphocytic leukemia. Blood 110: 3316-25.

Butchar JP, Mehta P, Justiniano SE, Guenterberg KD, Kondadasula SV, Mo X, Chemudupati M, Kanneganti TD, Amer A, Muthusamy N, Jarjoura D, Marsh CB, Carson WE, 3rd, Byrd JC, Tridandapani S (2010) Reciprocal regulation of activating and inhibitory Fc{gamma} receptors by TLR7/8 activation: implications for tumor immunotherapy. Clin Cancer Res. 16(7): 2065-75.

Byrd JC, Furman RR, Coutre SE, Flinn IW, Burger JA, Blum KA, Grant B, Sharman JP, Coleman M, Wierda WG, Jones JA, Zhao W, Heerema NA, Johnson AJ, Sukbuntherng J, Chang BY, Clow F, Hedrick E, Buggy JJ, James DF, O'Brien S (2013) Targeting BTK with Ibrutinib in Relapsed Chronic Lymphocytic Leukemia. N Engl J Med 369: 32-42.

Byrd JC, Pagel JM, Awan FT, Forero A, Flinn IW, Deauna-Limayo DP, Spurgeon SE, Andritsos LA, Gopal AK, Leonard JP, Eisenfeld AJ, Bannink JE, Stromatt SC, Furman, RR (2014) A Phase 1 Study Evaluating the Safety and Tolerability of Otlertuzumab (TRU-016), an Anti-CD37 Mono-specific ADAPTIRTM Therapeutic Protein in Chronic Lymphocytic Leukemia. Blood 123(9): 1302-8.

Byrd JC, Rai K, Peterson BL, Appelbaum FR, Morrison VA, Kolitz JE, Shepard L, Hines JD, Schiffer CA, Larson RA (2005) Addition of rituximab to fludarabine may prolong progression-free survival and overall survival in patients with previously untreated chronic lymphocytic leukemia: an updated retrospective comparative analysis of CALGB 9712 and CALGB 9011. Blood 105(1): 49-53.

Byrd JC, Murphy T, Howard RS, Lucas MS, Goodrich A, Park K, Pearson M, Waselenko JK, Ling G, Grever MR, Grillo-Lopez AJ, Rosenberg J, Kunkel L, Flinn IW (2001) Rituximab using a thrice weekly dosing schedule in B-cell chronic lymphocytic leukemia and small lymphocytic lymphoma demonstrates clinical activity and acceptable toxicity. J Clin Oncol 19: 2153-2164.

Caligaris-Cappio F, Ghia P (2008) Novel insights in chronic lymphocytic leukemia: are we getting closer to understanding the pathogenesis of the disease? J Clin Oncol 26(27): 4497-503.

Caligaris-Cappio F (2003) Role of the microenvironment in chronic lymphocytic leukaemia. Br J Haematol 123(3): 380-8.

109

Carrasco YR, Batista FD (2006) B-cell activation by membrane-bound antigens is facilitated by the interaction of VLA-4 with VCAM-1. EMBO 25: 889-899.

Charrin S, Le Naour F, Labas V, Billard M, Le Caer JP, Emile JF, Petit MA, Boucheix C, Rubinstein E (2003) EWI-2 is a new component of the tetraspanin web in hepatocytes and lymphoid cells. Biochem J 373: 409-421.

Charrin S, Le Naour F, Oualid M, Billard M, Faure G, Hanash SM, Boucheix C, Rubinstein E (2001) The major CD9 and CD81 molecular partner. Identification and characterization of the complexes. J Biol Chem 276: 14329-14337.

Charrin S, Le Naour F, Silvie O, Milhiet PE, Boucheix C, Rubinstein E (2009) Lateral organization of membrane proteins: tetraspanins spin their web. Biochem J 420: 133-154.

Charrin S, Manie S, Oualid M, Billard M, Boucheix C, and Rubinstein E (2002) Differential stability of tetraspanin/tetraspanin interactions: role of palmitoylation. FEBS Lett 516: 139-144.

Chen HC, Byrd JC, Muthusamy N (2006) Differential role for cyclic AMP response element binding protein-1 in multiple stages of B cell development, differentiation, and survival. J Immunol 176(4): 2208-18.

Chen SS, Raval A, Johnson AJ, Hertlein E, Liu TH, Jin VX, Sherman MH, Liu SJ, Dawson DW, Williams KE, Lanasa M, Liyanarachchi S, Lin TS, Marcucci G,Pekarsky Y, Davuluri R, Croce CM, Guttridge DC, Teitell MA, Byrd JC, Plass C (2009) Epigenetic changes during disease progression in a murine model of human chronic lymphocytic leukemia. Proc Natl Acad Sci USA 106(32): 13433-8.

Chen Z, Pasquini M, Hong B, Dehart S, Heikens M, Tsai S (2005) The human Penumbra gene is mapped to a region on 7 frequently deleted in myeloid malignancies. Cancer Genet Cytogenet 162: 95-98.

Chiorazzi N (2007) Cell proliferation and death: forgotten features of chronic lymphocytic leukemia B cells. Best Pract Res Clin Haematol 20(3): 399-413.

Chiorazzi N, Rai KR, Ferrarini M (2005) Chronic lymphocytic leukemia. N Engl J Med 352(8): 804-815.

Claas C, Stipp CS, Hemler ME (2001) Evaluation of prototype transmembrane 4 superfamily protein complexes and their relation to lipid rafts. J Biol Chem 276: 7974-7984.

Clark KL, Oelke A, Johnson ME, Eilert KD, Simpson PC, Todd SC (2004) CD81 associates with 14-3-3 in a redox-regulated palmitoylation-dependent manner. J Biol Chem 279: 19401-19406.

110

Clynes RA, Towers TL, Presta LG, Ravetch JV (2006) Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat Med 6(4): 443-6.

Cragg MS, Glennie MJ (2004) Antibody specificity controls in vivo effector mechanisms of anti-CD20 reagents. Blood 103(7):2738-43.

Dahle J, Repetto-Llamazares AH, Mollatt CS, Melhus KB, Bruland OS, Kolstad A, Larsen RH (2013) Evaluating antigen targeting and anti-tumor activity of a new anti-CD37 radioimmunoconjugate against non-Hodgkin's lymphoma. Anticancer Res 33: 85-95.

De Bruyne E, Bos TJ, Asosingh K, Vande Broek I, Menu E, Van Valckenborgh E, Atadja P, Coiteux V, Leleu X, Thielemans K, Van Camp B, Vanderkerken K, Van Riet I (2008) Epigenetic silencing of the tetraspanin CD9 during disease progression in multiple myeloma cells and correlation with survival. Clin Cancer Res 14: 2918- 2926.

Bortoletto N, Scotet E, Myamoto Y, D'Oro U, Lanzavecchia A (2002) Optimizing anti- CD3 affinity for effective T cell targeting against tumor cells. Eur J Immunol 32(11):3102-7. de Claro RA, McGinn K, Kwitkowski V, Bullock J, Khandelwal A, Habtemariam B, Ouyang Y, Saber H, Lee K, Koti K, Rothmann M, Shapiro M, Borrego F, Clouse K, Chen XH, Brown J, Akinsanya L, Kane R, Kaminskas E, Farrell A, Pazdur R (2012) U.S. Food and Drug Administration approval summary: brentuximab vedotin for the treatment of relapsed Hodgkin lymphoma or relapsed systemic anaplastic large-cell lymphoma. Clin Cancer Res 18(21): 5845-9.

de Haij S, Jansen JH, Boross P, Beurskens FJ, Bakema JE, Bos DL, Martens A, Verbeek JS, Parren PW, van de Winkel JG, Leusen JH (2010) In vivo cytotoxicity of type I CD20 antibodies critically depends on Fc receptor ITAM signaling. Cancer Res 70(8): 3209-17.

de Winde CM, Veenbergen S, Young KH, Xu-Monette ZY, Wang XX, Xia Y, Jabbar KJ, van den Brand M, van der Schaaf A, Elfrink S, van Houdt IS, Gijbels MJ, van de Loo FA, Bennink MB, Hebeda KM, Groenen PJ, van Krieken JH, Figdor CG, van Spriel AB (2016) Tetraspanin CD37 protects against the development of B cell lymphoma. J Clin Invest 126(2):653-66.

Deckert J, Park PU, Chicklas S, Yi Y, Li M, Lai KC, Mayo MF, Carrigan CN, Erickson HK, Pinkas J, Lutz RJ, Chittenden T, Lambert JM (2013) A novel anti-CD37 antibody-drug conjugate with multiple anti-tumor mechanisms for the treatment of B-cell malignancies. Blood 122(20): 3500-10.

Deckert J, Ponte JF, Coccia JA, Lanieri L, Chicklas S, Yi Y, Watkins W, Ruiz-Soto R, Romanelli A, Lutz RJ (2014) Preclinical Mechanistic Studies Investigating Neutrophil and Lymphoid Cell Depletion By IMGN529, a CD37-Targeting Antibody-Drug Conjugate (ADC). Abstract #3119, 56th annual meeting of the 111

American Society of Hematology, San Francisco, CA, USA.

Durig J, Ebeling P, Grabellus F, Sorg UR, Mollmann M, Schutt P, Göthert J, Sellmann L, Seeber S, Flasshove M, Dührsen U, Moritz T (2007) A novel nonobese diabetic/severe combined immunodeficient xenograft model for chronic lymphocytic leukemia reflects important clinical characteristics of the disease. Cancer Res 67(18): 8653-61.

Erickson HK, Park PU, Widdison WC, Kovtun YV, Garrett LM, Hoffman K, Lutz RJ, Goldmacher VS, Blättler WA (2006) Antibody-maytansinoid conjugates are activated in targeted cancer cells by lysosomal degredation and linker-dependent intracellular processing. Cancer Res 66(8): 4426-33.

Fayad L, Keating MJ, Reuben JM, O'Brien S, Lee BN, Lerner S, Kurzrock R (2001) Interleukin-6 and interleukin-10 levels in chronic lymphocytic leukemia: correlation with phenotypic characteristics and outcome. Blood 97(1):256-63.

Flygare JA, Pillow TH, Aristoff P (2013) Antibody-drug conjugates for the treatment of cancer. Chem Biol Drug Des 81(1):113-21.

Gartlan KH, Belz GT, Tarrant JM, Minigo G, Katsara M, Sheng KC, Sofi M, Van Spriel AB, Apostolopoulos V, Plebanski M, Robb L, Wright MD (2010) A complementary role for the tetraspanins CD37 and Tssc6 in cellular immunity. J Immunol 185: 3158-3166

Gartlan KH, Wee JL, Demaria MC, Nastovska R, Chang TM, Jones EL, Apostolopoulos V, Pietersz GA, Hickey MJ, Van Spriel AB, Wright MD (2013) Tetraspanin CD37 contributes to the initiation of cellular immunity by promoting dendritic cell migration. Eur J Immunol 43: 1208-1219.

Gattei V, Bulian P, Del Principe MI, Zucchetto A, Maurillo L, Buccisano F, Bomben R, Dal-Bo M, Luciano F, Rossi FM, Degan M, Amadori S, Del Poeta G (2008) Relevance of Cd49d protein expression as overall survival and progressive disease prognosticator in chronic lymphocytic leukemia. Blood 111: 865-873.

Genini D, Adachi S, Chao Q, Rose DW, Carrera CJ, Cottam HB, Carson DA, Leoni LM (2000) Deoxyadenosine analogs induce programmed cell death in chronic lymphocytic leukemia cells by damaging the DNA and by directly affecting the mitochondria. Blood 96(10):3537-43.

Ghia P, Strola G, Granziero L, Geuna M, Guida G, Sallusto F, Ruffing N, Montagna L, Piccoli P, Chilosi M, Caligaris-Cappio F (2002) Chronic lymphocytic leukemia B cells are endowed with the capacity to attract CD4+, CD40L+ T cells by producing CCL22. European Journal of Immunology 32: 1403-13.

Gopal AK, Tarantolo SR, Bellam N, Green DJ, Griffin M, Feldman T, Mato AR, Eisenfeld, AJ, Stromatt SC, Goy A (2014) Phase 1b study of otlertuzumab (TRU-016), an anti-CD37 monospecific ADAPTIR therapeutic protein, in combination with 112

rituximab and bendamustine in relapsed indolent lymphoma patients. Invest New Drugs.

Goschnick MW, Lau LM, Wee JL, Liu YS, Hogarth PM, Robb LM, Hickey MJ, Wright MD, Jackson DE (2006) Impaired "outside-in" integrin alphaIIbbeta3 signaling and thrombus stability in TSSC6-deficient mice. Blood 108: 1911-1918.

Gribben JG (2010) How I treat CLL up front. Blood 115(2):187-97.

Haining EJ, Yang J, Bailey RL, Khan K, Collier R, Tsai S, Watson SP, Frampton J, Garcia P, Tomlinson MG (2012) The TspanC8 subgroup of tetraspanins interacts with A disintegrin and metalloprotease 10 (ADAM10) and regulates its maturation and cell surface expression. J Biol Chem 287: 39753-39765.

Hallek M, Cheson BD, Catovsky D, Caligaris-Cappio F, Dighiero G, Döhner H, Hillmen P, Keating MJ, Montserrat E, Rai KR, Kipps TJ; International Workshop on Chronic Lymphocytic Leukemia (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):5446-56.

Heider KH, Kiefer K, Zenz T, Volden M, Stilgenbauer S, Ostermann E, Baum A, Lamche H, Kupcu Z, Jacobi A, Muller S, Hirt U, Adolf GR, Borges E (2011) A novel Fc- engineered monoclonal antibody to CD37 with enhanced ADCC and high proapoptotic activity for treatment of B-cell malignancies. Blood 118(15): 4159- 68.

Heikens MJ, Cao TM, Morita C, Dehart SL, Tsai S (2007) Penumbra encodes a novel tetraspanin that is highly expressed in erythroid progenitors and promotes effective erythropoiesis. Blood 109: 3244-3252.

Hemler ME (2005) Tetraspanin functions and associated microdomains. Nat Rev Mol Cell Biol 6: 801-811.

Hemler ME (2014) Tetraspanin proteins promote multiple cancer stages. Nat Rev Cancer 14: 49-60.

Horváth G, Serru V, Clay D, Billard M, Boucheix C, Rubinstein E (1998) CD19 Is Linked to the Integrin-associated Tetraspans CD9, CD81, and CD82. J Biol Chem 273: 30537-30543.

Hu X, Xuan H, Du H, Jiang H, Huang J (2014) Down-regulation of CD9 by methylation decreased bortezomib sensitivity in multiple myeloma. PLoS One 9, e95765.

Huhn D, Von Schilling C, Wilhelm M, Ho AD, Hallek M, Kuse R, Knauf W, Riedel U, Hinke A, Srock S, Serke S, Peschel C, Emmerich B (2001) Rituximab therapy of

113

patients with B-cell chronic lymphocytic leukemia. Blood 98: 1326-1331.

Imai T, Mayumi K, Nishimura M, Yoshie O (1995) Molecular analyses of the association of CD4 with two members of the transmembrane 4 superfamily, CD81 and CD82. J Immunol 155: 1229-1239.

Johnson AJ, Lucas DM, Muthusamy N, Smith LL, Edwards RB, De Lay MD, Croce CM, Grever MR, Byrd JC (2006) Characterization of the TCL-1 transgenic mouse as a preclinical drug development tool for human chronic lymphocytic leukemia. Blood 108(4): 1334-8.

Kay NE, Kaplan ME (1986) Defective T cell responsiveness in chronic lymphocytic leukemia: analysis of activation events. Blood 67(3):578-81.

Keating MJ, O'brien S, Albitar M, Lerner S, Plunkett W, Giles F, Andreeff M, Cortes J, Faderl S, Thomas D, Koller C, Wierda W, Detry MA, Lynn A, 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, 4079-4088.

Kinashi T (2005) Intracellular signaling controlling integrin activation in lymphocytes. Nature Reviews 5: 546-59.

Kitadokoro K, Bordo D, Galli G, Petracca R, Falugi F, Abrignani S, Grandi G, Bolognesi M (2001) CD81 extracellular domain 3D structure: insight into the tetraspanin superfamily structural motifs. EMBO J 20: 12-18.

Knobeloch, K.P., Wright, M.D., Ochsenbein, A.F., Liesenfeld, O., Lohler, J., Zinkernagel, R.M., Horak, I., and Orinska, Z. (2000). Targeted inactivation of the tetraspanin CD37 impairs T-cell-dependent B-cell response under suboptimal costimulatory conditions. Mol Cell Biol 20, 5363-5369.

Koebel CM, Vermi W, Swann JB, Zerafa N, Rodig SJ, Old LJ, Smyth MJ, Schreiber RD (2007) Adaptive immunity maintains occult cancer in an equilibrium state. Nature 450(7171):903-7.

Kolstad A, Madsbu U, Dahle J, Stokke C, Bach-Gansmo T, Løndalen AM, Holtedahl JE, Revheim ME, Bruland O, Bolstad B, Bolstad N, Tierens A, Larsen RH, Alfheim JA, Delabie J, Signe S, Erlanson M, Rudå SN, Holte H (2014) A Phase I Study of 177 lu-DOTA-HH1 (Betalutin) Radioimmunotherapy for Patients with Relapsed CD37+ Non-Hodgkin's B Cell Lymphoma. Abstract #3094, 56th annual meeting of the American Society of Hematology, San Francisco, CA, USA.

Konstandin MH, Sester U, Klemke M, Weschenfelder T, Wabnitz GH, Samstag Y (2006) A novel flow-cytometry-based assay for quantification of affinity and avidity changes of integrins. Journal of Immunological Methods 310: 67-77.

114

Krause G, Patz M, Isaeva P, Wigger M, Baki I, Vondey V, Kerwien S, Kuckertz M, Brinker R, Claasen J, Frenzel LP, Wendtner CM, Heider KH, Hallek M (2012) Action of novel CD37 antibodies on chronic lymphocytic leukemia cells. Leukemia 26: 546-549.

Lapalombella R, Sun Q, Williams K, Tangeman L, Jha S, Zhong Y, Goettl V, Mahoney E, Berglund C, Gupta S, Farmer A, Mani R, Johnson AJ, Lucas D, Mo X, Daelemans D, Sandanayaka V, Shechter S, McCauley D, Shacham S, Kauffman M, Chook YM, Byrd JC (2012) Selective inhibitors of nuclear export show that CRM1/XPO1 is a target in chronic lymphocytic leukemia. Blood 120(23): 4621- 34.

Lapalombella R, Yeh YY, Wang L, Ramanunni A, Rafiq S, Jha S, Staubli J, Lucas DM, Mani R, Herman SE, Johnson AJ, Lozanski A, Andritsos L, Jones J, Flynn JM, Lannutti B, Thompson P, Algate P, Stromatt S, Jarjoura D, Mo X, Wang D, Chen CS, Lozanski G, Heerema NA, Tridandapani S, Freitas MA, Muthusamy N, Byrd JC (2012) Tetraspanin CD37 directly mediates transduction of survival and apoptotic signals. Cancer Cell 21(5): 694-708.

Lazar GA, Dang W, Karki S, Vafa O, Peng JS, Hyun L, Chan C, Chung HS, Eivazi A, Yoder SC, Vielmetter J, Carmichael DF, Hayes RJ Dahiyat BI (2006) Engineered antibody Fc variants with enhanced effector function. Proc Natl Acad Sci USA 103: 4005-4010.

Le Naour R, Andre M, Boucheix C, Rubinstein E (2006) Membrane microdomains and proteomics: lessons from tetraspanin microdomains and comparison with lipid rafts. Proteomics 6: 6447-6454.

Le Naour R, Andre M, Greco C, Billard M, Sordat B, Emile JF, Lanza F, Boucheix C, Rubinstein E (2006) Profiling of the tetraspanin web of human colon cancer cells. Mol Cell Proteomics 5: 845-857.

Leung KT, Chan KY, Ng PK, Lau TK, Chiu WM, Tsang KS, Li CK, Kong CK, Li K (2011) The tetraspanin CD9 regulates migration, adhesion, and homing of human cord blood CD34+ hematopoietic stem and progenitor cells. Blood 117(6): 1840-50.

Levy S (2014) Function of the tetraspanin molecule CD81 in B and T cells. Immunol Res 58: 179-185.

Link MP, Bindl J, Meeker TC, Carswell C, Doss CA, Warnke RA, Levy R (1986) A unique antigen on mature B cells defined by a monoclonal antibody. J Immunol 137(9): 3013-8.

Little KD, Hemler ME, Stipp SC (2004) Dynamic regulation of a GPCR-tetraspanin-G protein complex on intact cells: central role of CD81 in facilitating GPR56-Galpha q/11 association. Mol Biol Cell 15: 2375-2387.

115

Löffler A, Gruen M, Wuchter C, Schriever F, Kufer P, Dreier T, Hanakam F, Baeuerle PA, Bommert K, Karawajew L, Dörken B, Bargou RC (2003) Efficient elimination of chronic lymphocytic leukaemia B cells by autologous T cells with a bispecific anti-CD19/anti-CD3 single-chain antibody construct. Leukemia 17(5):900-9.

Lucas DM, Edwards RB, Lozanski G, West DA, Shin JD, Vargo MA, Davis ME, Rozewski DM, Johnson AJ, Su BN, Goettl VM, Heerema NA, Lin TS, Lehman A, Zhang X, Jarjoura D, Newman DJ, Byrd JC, Kinghorn AD, Grever MR (2009) The novel plant-derived agent silvestrol has B-cell selective activity in chronic lymphocytic leukemia and acute lymphoblastic leukemia in vitro and in vivo. Blood 113(19): 4656-66.

Luu VP, Hevezi P, Vences-Catalan F, Maravillas-Montero JL, White CA, Casali P, Llorente L, Jakez-Ocampo J, Lima G, Vilches-Cisneros N, Flores-Gutierrez JP, Santos-Argumedo L, Zlotnik A (2013) TSPAN33 is a novel marker of activated and malignant B cells. Clin Immunol 149, 388-399.

Macor P, Secco E, Zorzet S, Tripodo C, Celeghini C, Tedesco F (2008) An update on the xenograft and mouse models suitable for investigating new therapeutic compounds for the treatment of B-cell malignancies. Curr Pharm Des 14(21): 2023-39.

Maecker HT, Levy S (1997) Normal lymphocyte development but delayed humoral immune response in CD81-null mice. J Exp Med 185: 1505-1510.

Maecker HT, Todd SC, Levy S (1997) The tetraspanin superfamily: molecular facilitators. FASEB J 11: 428-442.

Mattila PK, Feest C, Depoli D, Treanor B, Montaner B, Otipoby KL, Carter R, Justement LB, Bruckbauer A Batista FD (2013) The actin and tetraspanin networks organize receptor nanoclusters to regulate B cell receptor-mediated signaling. Immunity 38: 461-474.

Messmer BT, Messmer D, Allen SL, Kolitz JE, Kudalkar P, Cesar D, Murphy EJ, Koduru P, Ferrarini M, Zupo S, Cutrona G, Damle RN, Wasil T, Rai KR, Hellerstein MK, Chiorazzi N (2005) In vivo measurements document the dynamic cellular kinetics of chronic lymphocytic leukemia B cells. J Clin Invest 115(3): 755-64.

Meyer-Wentrup F, Figdor CG, Ansems M, Brossart P, Wright MD, Adema GJ, Van Spriel AB (2007) Dectin-1 interaction with tetraspanin CD37 inhibits IL-6 production. J Immunol 178: 154-162.

Min G, Wang H, Sun TT, Kong XP (2006) Structural basis for tetraspanin functions as revealed by the cryo-EM structure of uroplakin complexes at 6-A resolution. J Cell Biol 173: 975-983.

116

Mollinedo F, Fontan G, Barasoain I, Lazo PA (1997) Recurrent infectious diseases in human CD53 deficiency. Clin Diagn Lab Immunol 4: 229-231.

Mollinedo F, Martin-Martin B, Gajate C, Lazo PA (1998) Physiological activation of human neutrophils down-regulates CD53 cell surface antigen. J Leukoc Biol 63: 699-706.

Moore PA, Zhang W, Rainey GJ, Burke S, Li H, Huang L, Gorlatov S, Veri MC, Aggarwal S, Yang Y, Shah K, Jin L, Zhang S, He L, Zhang T, Ciccarone V, Koenig S, Bonvini E, Johnson S (2011) Application of dual affinity retargeting molecules to achieve optimal redirected T-cell killing of B-cell lymphoma. Blood 117(17):4542- 51.

Nakamoto T, Murayama Y, Oritani K, Boucheix C, Rubinstein E, Nishida M, Katsube F, Watabe K, Kiso S, Tsutsui S, Tamura S, Shinomura Y, Hayashi N (2009) A novel therapeutic strategy with anti-CD9 antibody in gastric cancers. J Gastroenterol 44: 889-896

Nicholson RH, Pantano S, Eliason JF, Galy A, Weiler S, Kaplan J, Hughes MR, Ko MS (2000) Phemx, a novel mouse gene expressed in hematopoietic cells maps to the imprinted cluster on distal chromosome 7. Genomics 68: 13-21.

Nunes C, Wong R, Mason M, Fegan C, Man S, Pepper C (2012) Expansion of a CD8(+)PD-1(+) replicative senescence phenotype in early stage CLL patients is associated with inverted CD4:CD8 ratios and disease progression. Clin Cancer Res 18(3):678-87.

Olweus J, Lund-Johansen F, Horejsi V (1993) CD53, a protein with four membrane- spanning domains, mediates signal transduction in human monocytes and B cells. J Immunol 151: 707-716.

Ono M, Handa K, Withers DA, Hakomori S (1999) Motility inhibition and apoptosis are induced by metastasis-suppressing gene product CD82 and its analogue CD9, with concurrent glycosylation. Cancer Res 59: 2335-2339.

Ono M, Handa K, Withers DA, Hakomori S (2000) Glycosylation effect on membrane domain (GEM) involved in cell adhesion and motility: a preliminary note on functional alpha3, alpha5-CD82 glycosylation complex in ldlD 14 cells. Biochem Biophys Res Commun 279: 744-750.

Oroudjev E, Lopus M, Wilson L, Audette C, Provenzano C, Erickson H, Kovtun Y, Chari R, Jordan MA (2010) Maytansinoid-antibody conjugates induce mitotic arrest by suppressing microtubule dynamic instability. Mol Cancer Ther 9(10): 2700-13.

Pagel JM, Spurgeon SE, Byrd JC, Awan FT, Flinn IW, Lanasa MC, Eisenfeld AJ, Stromatt SC, Gopal AK (2014) Otlertuzumab (TRU-016), an anti-CD37 monospecific ADAPTIR therapeutic protein, for relapsed or refractory NHL 117

patients. Br J Haematol 168(1):38-45.

Pileri P, Uematsu Y, Campagnoli S, Galli G, Falugi F, Petracca R, Weiner AJ, Houghton M, Rosa D, Grandi G, Abrignani S (1998) Binding of hepatitis C virus to CD81. Science 282: 938-941.

Plosker GL, Figgitt DP (2003) Rituximab: a review of its use in non-Hodgkin's lymphoma and chronic lymphocytic leukaemia. Drugs 63(8): 803-43.

Press OW, Eary JF, Badger CC, Martin PJ, Appelbaum FR, Levy R, Miller R, Brown S, Nelp WB, Krohn KA, Fisher D, DeSantes K, Porter B, Kidd P, Thomas D, Bernstein ID (1989) Treatment of refractory non-Hodgkin's lymphoma with radiolabeled MB-1 (anti-CD37) antibody. J Clin Oncol 7: 1027-1038.

Press OW, Howell-Clark J, Anderson S, Bernstein I (1994) Retention of B-cell-specific monoclonal antibodies by human lymphoma cells. Blood 83: 1390-1397.

Qi JC, Wang J, Mandadi S, Tanaka K, Roufogalis BD, Madigan MC, Lai K, Yan F, Chong BH, Stevens RL, Krilis SA (2006) Human and mouse mast cells use the tetraspanin CD9 as an alternate interleukin-16 receptor. Blood 107: 135-142.

Rafiq S, Siadak A, Butchar JP, Cheney C, Lozanski G, Jacob NK, Lapalombella R, Mcgourty J, Moledor M, Lowe R, Setter B, Jones J, Flynn JM, Andritsos L, Devine S, Mo X, Jarjoura D, Tridandapani S, Algate P, Byrd JC, Muthusamy N (2013) Glycovariant anti-CD37 monospecific protein therapeutic exhibits enhanced effector cell-mediated cytotoxicity against chronic and acute B cell malignancies. MAbs 5: 723-735.

Rai KR, Sawitsky A, Cronkite EP, Chanana AD, Levy RN, Pasternack BS (1975) Clinical staging of chronic lymphocytic leukemia. Blood 46(2):219-34.

Ramsay AG, Johnson AJ, Lee AM, Gorgun G, Le Dieu R, Blum W, Byrd JC, Gribben JG (2008) Chronic lymphocytic leukemia T cells show impaired immunological synapse formation that can be reversed with an immunomodulating drug. J Clin Invest 118(7): 2427-37.

Repetto-Llamazares AH, Larsen RH, Giusti AM, Riccardi E, Bruland OS, Selbo PK, Dahle J (2014) 177Lu-DOTA-HH1, a novel anti-CD37 radio-immunoconjugate: a study of toxicity in nude mice. PLoS One 9: e103070.

Rezvani AR, Maloney DG (2011) Rituximab resistance. Best Pract Res Clin Haematol 24(2): 203-16.

Robak T, Hellman A, Kloczko J, Loscertales J, Lech-Maranda E, JPagel JM, Mato A, Byrd JC, Awan FT, Hebart H, Garcia-Marco JA, Hill BT, Hallek M, Stromatt S, Jaeger, U (2013) Phase 2 Study Of Otlertuzumab (TRU-016), An Anti-CD37 ADAPTIRTM Protein, In Combination With Bendamustine vs Bendamustine Alone In Patients With Relapsed Chronic Lymphocytic Leukemia (CLL). Abstract 118

#2860, 55th annual meeting of the American Society of Hematology, New Orleans, LA, USA.

Robb L, Tarrant J, Groom J, Ibrahim M, Li R, Borobakas B, Wright MD (2001) Molecular characterisation of mouse and human TSSC6: evidence that TSSC6 is a genuine member of the tetraspanin superfamily and is expressed specifically in haematopoietic organs. Biochim Biophys Acta 1522: 31-41.

Roda JM, Joshi T, Butchar JP, McAlees JW, Lehman A, Tridandapani S, Carson WE, 3rd (2007) The activation of natural killer cell effector functions by cetuximab-coated, epidermal growth factor receptor positive tumor cells is enhanced by cytokines. Clin Cancer Res 13(21):6419-28.

Roda JM, Parihar R, Lehman A, Mani A, Tridandapani S, Carson WE, 3rd (2006) Interleukin-21 enhances NK cell activation in response to antibody-coated targets. J Immunol. 177(1): 120-9.

Romanska HM, Berditchevski F (2011) Tetraspanins in human epithelial malignancies. J Pathol 223: 4-14. Sandoval A, Consoli U, Plunkett W (1996) Fludarabine-mediated inhibition of nucleotide excision repair induces apoptosis in quiescent human lymphocytes. Clin Cancer Res 2(10):1731-41.

Schwartz-Albiez R, Dorken B, Hofmann W, Moldenhauer G (1988) The B cell-associated CD37 antigen (gp40-52). Structure and subcellular expression of an extensively glycosylated glycoprotein. J Immunol 140(3): 905-14.

Scott AM, Wolchok JD, Old LJ (2012) Antibody therapy of cancer. Nat Rev Cancer 12: 278-287.

Seita J, Sahoo D, Rossi DJ, Bhattacharya D, Serwold T, Inlay MA, Ehrlich LI, Fathman, JW, Dill DL, Weissman IL (2012) Gene Expression Commons: an open platform for absolute gene expression profiling. PLoS One 7: e40321.

Shallal S, Kornbluth, J (2000) CD9 expression enhances the susceptibility of myeloma cell lines to cell-mediated cytolysis. Blood 96: 224-233.

Sheng KC, Van Spriel AB, Gartlan KH, Sofi M, Apostolopoulos V, Ashman L, Wright MD (2009) Tetraspanins CD37 and CD151 differentially regulate Ag presentation and T-cell co-stimulation by DC. Eur J Immunol 39: 50-55.

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): 2210-8.

Shinkawa T, Nakamura K, Yamane N, Shoji-Hosaka E, Kanda Y, Sakurada M, Uchida K, Anazawa H, Satoh M, Yamasaki M, Hanai N, Shitara K (2003) The absence of 119

fucose but not the presence of galactose or bisecting N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical role of enhancing antibody-dependent cellular cytotoxicity. J Biol Chem 278: 3466-3473.

Shoham T, Rajapaksa R, Kuo CC, Haimovich J Levy S (2006) Building of the tetraspanin web: distinct structural domains of CD81 function in different cellular compartments. Mol Cell Biol 26: 1373-1385.

Siegel R, Naishadham D, Jemal A (2013) Cancer statistics, 2013. CA Cancer J Clin 63(1):11-30.

Smith P, DiLillo DJ, Bournazos S, Li F, Ravetch JV (2012) Mouse model recapitulating human Fcgamma receptor structural and functional diversity. Proc Natl Acad Sci USA 109(16): 6181-6.

Soma LA, Craig FE, Swerdlow SH (2006) The proliferation center microenvironment and prognostic markers in chronic lymphocytic leukemia/small lymphocytic lymphoma. Hum Pathol 37(2): 152-9.

Stathis A, Freedman AS, Flinn IW, Maddocks KJ, Weitman S, Berdeja JG, Mejia, AV, Zucca E, Green, R, Romanelli A, Zildjian SH, Ruiz-Soto R, Palomba M (2014) A Phase I Study of IMGN529, an Antibody-Drug Conjugate (ADC) Targeting CD37, in Adult Patients with Relapsed or Refractory B-Cell Non-Hodgkin’s Lymphoma (NHL). Abstract #1760, 56th annual meeting of the American Society of Hematology, San Francisco, CA, USA.

Stilgenbauer S, Schnaiter A, Paschka P, Zenz T, Rossi M, Döhner K, Bühler A, Böttcher S, Ritgen M, Kneba M, Winkler D, Tausch E, Hoth P, Edelmann J, Mertens D, Bullinger L, Bergmann M, Kless S, Mack S, Jäger U, Patten N, Wu L, Wenger MK, Fingerle-Rowson G, Lichter P, Cazzola M, Wendtner CM, Fink AM, Fischer K, Busch R, Hallek M, Döhner H (2014) Gene mutations and treatment outcome in chronic lymphocytic leukemia: results from the CLL8 trial. Blood 123(21):3247- 54.

Suzuki T, Coustan-Smith E, Mihara K Campana D (2002) Signals mediated by FcgammaRIIA suppress the growth of B-lineage acute lymphoblastic leukemia cells. Leukemia 16: 1276-1284.

Tai XG, Yashiro Y, Abe R, Toyooka K, Wood CR, Morris J, Long A, Ono S, Kobayashi M, Hamaoka T, Neben S, Fujiwara H (1996) A role for CD9 molecules in T cell activation. J Exp Med 184: 753-758.

Tai YT, Mayes PA, Acharya C, Zhong MY, Cea M, Cagnetta A, Craigen J, Yates J, Gliddon L, Fieles W, Hoang B, Tunstead J, Christie AL, Kung AL, Richardson P, Munshi NC, Anderson KC (2014) Novel anti-B-cell maturation antigen antibody- drug conjugate (GSK2857916) selectively induces killing of multiple myeloma. Blood 123(20):3128-38.

120

Takai T, Li M, Sylvestre D, Clynes R, Ravetch JV (1994) FcR gamma chain deletion results in pleiotrophic effector cell defects. Cell 76: 519-529.

Tarrant JM, Groom J, Metcalf D, Li R, Borobokas B, Wright MD, Tarlinton D, Robb L (2002) The absence of Tssc6, a member of the tetraspanin superfamily, does not affect lymphoid development but enhances in vitro T-cell proliferative responses. Mol Cell Biol 22: 5006-5018.

Tarrant JM, Robb L, Van Spriel AB, Wright MD (2003) Tetraspanins: molecular organisers of the leukocyte surface. Trends Immunol 24: 610-617.

Timmerman JM, Byrd JC, Andorsky DJ, Yamada RE, Kramer J, Muthusamy N, Hunder N, Pagel JM (2012) A phase I dose-finding trial of recombinant interleukin-21 and rituximab in relapsed and refractory low grade B-cell lymphoproliferative disorders. Clin Cancer Res 18(20): 5752-60.

Topp MS, Kufer P, Gökbuget N, Goebeler M, Klinger M, Neumann S, Horst HA, Raff T, Viardot A, Schmid M, Stelljes M, Schaich M, Degenhard E, Köhne-Volland R, Brüggemann M, Ottmann O, Pfeifer H, Burmeister T, Nagorsen D, Schmidt M, Lutterbuese R, Reinhardt C, Baeuerle PA, Kneba M, Einsele H, Riethmüller G, Hoelzer D, Zugmaier G, Bargou RC (2011) Targeted therapy with the T-cell- engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J Clin Oncol 29(18):2493-8.

Van Spriel AB, De Keijzer S, Van Der Schaaf A, Gartlan KH, Sofi M, Light A, Linssen PC, Boezeman JB, Zuidscherwoude M, Reinieren-Beeren I, Cambi A, Mackay F, Tarlinton DM, Figdor CG, Wright MD (2012) The tetraspanin CD37 orchestrates the alpha(4)beta(1) integrin-Akt signaling axis and supports long-lived plasma cell survival. Sci Signal 5: ra82.

Van Spriel AB, Puls KL, Sofi M, Pouniotis D, Hochrein H, Orinska Z, Knobeloch, KP, Plebanski M, Wright MD (2004) A regulatory role for CD37 in T cell proliferation. J Immunol 172: 2953-2961.

Van Spriel AB, Sofi M, Gartlan KH, Van Der Schaaf A, Verschueren I, Torensma R, Raymakers RA, Loveland BE, Netea MG, Adema GJ, Wright MD, Figdor CG (2009) The tetraspanin protein CD37 regulates IgA responses and anti-fungal immunity. PLoS Pathog 5: e1000338.

Vandewoestyne ML, Pede VC, Lambein KY, Dhaenens MF, Offner FC, Praet MM, Philippé JJ, Kipps TJ, Deforce DL (2011) Laser microdissection for the assessment of the clonal relationship between chronic lymphocytic leukemia/small lymphocytic lymphoma and proliferating B cells within lymph node pseudofollicles. Leukemia 25(5): 883-8.

Veenbergen S, Van Spriel AB (2011) Tetraspanins in the immune response against cancer. Immunol Lett 138: 129-136. 121

Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, Pegram M, Oh DY, Diéras V, Guardino E, Fang L, Lu MW, Olsen S, Blackwell K; EMILIA Study Group (2012) Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med 367(19): 1783-91. Vincent AM, Cawley JC, Burthem J (1996) Integrin function in chronic lymphocytic leukemia. Blood 87: 4780-4788.

Voehringer DW, Hirschberg DL, Xiao J, Lu Q, Roederer M, Lock CB, Herzenberg LA, Steinman L (2000) Gene microarray identification of redox and mitochondrial elements that control resistance or sensitivity to apoptosis. Proc Natl Acad Sci USA 97: 2680-2685.

Vugmeyster Y, Howell K, Bakshl A, Flores C, Canova-Davis E (2003) Effect of anti- CD20 monoclonal antibody, Rituxan, on cynomolgus monkey and human B cells in a whole blood matrix. Cytometry A 52(2): 101-9.

Waterhouse R, Ha C, Dveksler GS (2002) Murine CD9 is the receptor for pregnancy- specific glycoprotein 17. J Exp Med 195: 277-282.

White A, Lamb PW, Barrett JC (1998) Frequent downregulation of the KAI1(CD82) metastasis suppressor protein in human cancer cell lines. Oncogene 16: 3143- 3149.

Wierda W, O'Brien S, Wen S, Faderl S, Garcia-Manero G, Thomas D, Do KA, Cortes J, Koller C, Beran M, Ferrajoli A, Giles F, Lerner S, Albitar M, Kantarjian H, Keating M (2005) Chemoimmunotherapy with fludarabine, cyclophosphamide, and rituximab for relapsed and refractory chronic lymphocytic leukemia. J Clin Oncol 23(18):4070-8.

Witherden DA, Boismenu R, Havran WL (2000) CD81 and CD28 costimulate T cells through distinct pathways. J Immunol 165: 1902-1909.

Wolf E, Hofmeister R, Kufer P, Schlereth B, Baeuerle PA (2005) BiTEs: bispecific antibody constructs with unique anti-tumor activity. Drug Discov Today 10(18):1237-44.

Wong R, Pepper C, Brennan P, Nagorsen D, Man S, Fegan C (2013) Blinatumomab induces autologous T-cell killing of chronic lymphocytic leukemia cells. Haematologica 98(12):1930-8.

Worthington RE, Carroll RC, Boucheix C (1990) Platelet activation by CD9 monoclonal antibodies is mediated by the Fc gamma II receptor. Br J Haematol 74: 216-222.

Woyach JA, Ruppert AS, Heerema NA, Peterson BL, Gribben JG, Morrison VA, Rai KR, Larson RA, Byrd JC (2011) Chemoimmunotherapy with fludarabine and rituximab produces extended overall survival and progression-free survival in chronic lymphocytic leukemia: long-term follow-up of CALGB study 9712. J Clin Oncol 122

29(10): 1349-55.

Woyach JA, Johnson AJ, Byrd JC (2012) The B-cell receptor signaling pathway as a therapeutic target in CLL. Blood 120(6): 1175-84.

Wright MD, Geary SM, Fitter S, Moseley GW, Lau LM, Sheng KC, Apostolopoulos V, Stanley EG, Jackson DE, Ashman LK (2004) Characterization of mice lacking the tetraspanin superfamily member CD151. Mol Cell Biol 24: 5978-5988.

Yan XJ, Albesiano E, Zanesi N, Yancopoulos S, Sawyer A, Romano E, Petlickovski A, Efremov DG, Croce CM, Chiorazzi N (2006) B cell receptors in TCL1 transgenic mice resemble those of aggressive, treatment-resistant human chronic lymphocytic leukemia. Proc Natl Acad Sci USA 103(31): 11713-8.

Yang X, Claas C, Kraeft SK, Chen LB, Wang Z, Kreidberg JA, Hemler ME (2002) Palmitoylation of tetraspanin proteins: modulation of CD151 lateral interactions, subcellular distribution, and integrin-dependent cell morphology. Mol Biol Cell 13: 767-781.

Yang X, Kovalenko OV, Tang W, Claas C, Stipp SC, Hemler ME (2004) Palmitoylation supports assembly and function of integrin-tetraspanin complexes. J Cell Biol 167: 1231-1240.

Yauch RL, Berditchevski F, Harler MB, Reichner J, Hemler ME (1998) Highly stoichiometric, stable, and specific association of integrin alpha3beta1 with CD151 provides a major link to phosphatidylinositol 4-kinase, and may regulate cell migration. Mol Biol Cell 9: 2751-2765.

Yauch RL, Kazarov AR, Desai B, Lee RT, Hemler ME (2000) Direct extracellular contact between integrin alpha(3)beta(1) and TM4SF protein CD151. J Biol Chem 275: 9230-9238.

Yu B, Mao Y, Yuan Y, Yue C, Wang X, Mo X, Jarjoura D, Paulaitis ME, Lee RJ, Byrd JC, Lee LJ, Muthusamy N (2013) Targeted drug delivery and cross-linking induced apoptosis with anti-CD37 based dual-ligand immunoliposomes in B chronic lymphocytic leukemia cells. Biomaterials 34: 6185-6193.

Yunta M, Lazo PA (2003) Apoptosis protection and survival signal by the CD53 tetraspanin antigen. Oncogene 22: 1219-1224.

Zenz T, Mertens D, Kuppers R, Döhner H, Stilgenbauer S (2010) From pathogenesis to treatment of chronic lymphocytic leukemia. Nature Reviews Cancer 10: 37-50.

Zevian S, Winterwood NE, Stipp CS (2011) Structure-function analysis of tetraspanin CD151 reveals distinct requirements for tumor cell behaviors mediated by alpha3beta1 versus alpha6beta4 integrin. J Biol Chem 286: 7496-7506.

123

Zhang XA, Bontrager AL, Hemler ME (2001) Transmembrane-4 superfamily proteins associate with activated protein kinase C (PKC) and link PKC to specific beta(1) integrins. J Biol Chem 276: 25005-25013.

Zhao X, Lapalombella R, Joshi T, Cheney C, Gowda A, Hayden-Ledbetter MS, Baum PR, Lin TS, Jarjoura D, Lehman A, Kussewitt D, Lee RJ, Caligiuri MA, Tridandapani S, Muthusamy N, Byrd JC (2007) Targeting CD37-positive lymphoid malignancies with a novel engineered small modular immunopharmaceutical. Blood 110: 2569-2577.

Zijlstra A, Lewis J, Degryse B, Stuhlmann H, Quigley JP (2008) The inhibition of tumor cell intravasation and subsequent metastasis via regulation of in vivo tumor cell motility by the tetraspanin CD151. Cancer Cell 13: 221-234.

Zoller M (2009) Tetraspanins: push and pull in suppressing and promoting metastasis. Nat Rev Cancer 9: 40-55.

Zuidscherwoude M, De Winde CM, Cambi A, Van Spriel AB (2014) Microdomains in the membrane landscape shape antigen-presenting cell function. J Leukoc Biol 95: 251-263.

124