<<

INTERFERON-GAMMA-MEDIATED IMMUNOEVASIVE STATEGIES IN MULTIPLE MYELOMA

DISSERTATION

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

By

Paul David Ciarlariello

Graduate Program in Molecular Cellular and Developmental Biology

The Ohio State University

2016

Dissertation Committee:

Don M. Benson, Jr., MD, PhD -Advisor

Michael A. Caligiuri, MD

Gregory Lesinski, PhD

Natarajan Muthusamy, DVM, PhD -Advisor

Flavia Pichiorri, PhD

Copyrighted by

Paul David Ciarlariello

2016

Abstract

Natural killer (NK) cells are a major source of -γ (IFN-γ) and may play a key role in innate immunity against multiple myeloma (MM). IFN-γ abrogates MM tumor in vitro

proliferation, however, clinical trials of recombinant IFN-γ in patients with MM showed no

benefit. MM cells exhibit strategies designed to evade NK cell surveillance and lysis. Herein,

we provide evidence of a novel means of MM immune evasion in which IFN-γ appears to

play a reciprocal relationship between MM and NK cells. Patients with MM exhibit higher

levels of serum IFN-γ levels than in the healthy setting. NK cells produce IFN-γ in response

to MM cells which express functional IFN-γ receptors. Stimulation with IFN-γ leads to

increased transcription and expression of the inhibitory ligands HLA-E and PD-L1 by MM

cells. This effect may be overcome by interruption of the NKG2A / HLA-E interaction.

Intriguingly, MM cells release extracellular vesicles (EV) which are capable of enhancing

IFN-γ production of NK cell, thus describing a potential cyclic mechanism of perpetual

immune detriment. Taken in total, these results suggest that MM cells subvert the favorable

effects of IFN-γ to attenuate NK cell tumor-directed cytotoxicity.

ii Dedication

To my wife, Nicole, and my baby, Gianluca.

To my parents, Paul and Sue.

I am a scientist - I seek to understand me all of my impurities and evils yet unknown

I am a scientist - I seek to understand me I am an incurable, and nothing else behaves like me

I am a scientist- I seek to understand me [...] the hole I dig is bottomless, but nothing else will set me free

-Guided By Voices

iii Acknowledgments

I would like to thank my advisor Dr. Don M. Benson, Jr. for taking the plunge and adopting me as his first and only graduate research student. Don, you have created a supporting environment in which I could grow as a scientist. If even an ounce of your knowledge, generosity, hospitality, and disposition have rubbed off on me, I will no doubt be a better researcher, father, and overall human being. Thank you, for lending your expertise and shepherding my training. I am forever grateful.

Of course, I would like to acknowledge my co-mentor Dr. Raj Muthusamy for the many constructive conversations which kept me on track to graduate. Raj, curiosities pour out of you and I have had such a rewarding experience discussing my data and its implications. I will always admire your method and approach to properly designed and controlled experiments.

A special thanks to Dr. Michael A. Caligiuri who brought me on to his team in 2007. Mike, you dispense positivity and influence with every word and action to those around you, and I am grateful for your support and guidance throughout the years.

To Dr. Flavia Pichiorri, thank you for the many collaborative efforts and willingness to meet and discuss our mutual findings. I have enjoyed our colorful conversations and your constant excitement for decoding life’s secrets.

To Dr. Greg Lesinski, thank you for your constructive and honest feedback throughout the years. I have always felt at ease in our exchanges, knowing that you were fully present and engaged in my scientific stories.

iv I extend my endless respect and gratitude to my science “mommy”, Dr. Rossana Trotta. Rox,

you make hypothesis-driven methods look deceivingly easy. I did not have a full appreciation of your expertise in experimental design until I was on my own. I think of you when considering experimental controls and especially when I am qualifying the conditions of my results. You have left such an impression on the scientist I have become. Thank you.

I must honor and recognize my lab mate, Dr. Tiffany Hughes for her many supportive roles during my tenure. You have offered your help every day for the past six years and have asked nearly nothing in return. We as a lab were truly lucky to recruit you, and I am confident your many contributions will help rid the world of myeloma, once and for all.

Some of the most rewarding experiences in the lab were moments while mentoring my undergraduates D. Maxwell Banaszak and Rohit Menon. I am glad to have been a part of your experience here at OSU. I know you both are ready to take on the world.

My parents Paul and Sue Ciarlariello have selflessly supported me my entire life. Through their efforts, I have been afforded the free reign of pursuing my passions. Mom and Dad, I am without words as to how incredible my life experience has been. Let this accomplishment symbolize your success as parents. I have been and will always be proud to be your son.

To my love, Nicole Danielle Ciarlariello. You are the most intuitive, thoughtful, and gentle soul I have ever come across and I am lucky to have you by my side. We now embark on new beginnings with our baby boy, Gianluca. I don’t care where we end up, as long as we are together. Thank you for believing in me, and trusting me with your heart.

v

Vita

November 4, 1983 ...... Born, Dayton, Ohio

June 2002 ...... West Carrollton High School

June 2006 ...... B.S. Biochemistry,

The Ohio State University

September 2010 - Present ...... Molecular, Cellular, and Developmental

Biology Graduate School Program,

The Ohio State University

Publications

Harshman, S. W., A. Canella, P. D. Ciarlariello, K. Agarwal, O. E. Branson, A. Rocci, H. Cordero, M. A. Phelps, E. M. Hade, J. A. Dubovsky, A. Palumbo, A. Rosko, J. C. Byrd, C. C. Hofmeister, D. M. Benson, Jr., M. E. Paulaitis, M. A. Freitas and F. Pichiorri (2016). "Proteomic characterization of circulating extracellular vesicles identifies novel serum myeloma associated markers." J Proteomics.

Briercheck EL, R. Trotta ,L. Chen , A. S. Hartlage, J. P. Cole, T. D. Cole, C. Mao , P. P. Banerjee, H. T. Hsu , E. M. Mace, D. Ciarlariello, B. L. Mundy-Bosse, I. Garcia-Cao, S. D. Scoville, L. Yu, R. Pilarski, W. E. Carson, G. Leone, P. P. Pandolfi, J.Yu, J. S. Orange and M. A. Caligiuri (2015). “PTEN Is a Negative Regulator of NK Cell Cytolytic Function” J Immunol 194(4):1832-40. doi: 10.4049/jimmunol.1401224. Epub 2015 Jan 16.

Collins, S. M., C. E. Bakan, G. D. Swartzel, C. C. Hofmeister, Y. A. Efebera, H. Kwon, G. C. Starling, D. Ciarlariello, S. Bhaskar, E. L. Briercheck, T. Hughes, J. Yu, A. Rice and D. M. Benson, Jr. (2013). "Elotuzumab directly enhances NK cell cytotoxicity against myeloma via CS1 ligation: evidence for augmented NK cell function complementing ADCC." Cancer Immunol Immunother 62(12): 1841-1849.

vi Harshman, S. W., A. Canella, P. D. Ciarlariello, A. Rocci, K. Agarwal, E. M. Smith, T. Talabere, Y. A. Efebera, C. C. Hofmeister, D. M. Benson, Jr., M. E. Paulaitis, M. A. Freitas and F. Pichiorri (2013). "Characterization of multiple myeloma vesicles by label-free relative quantitation." Proteomics 13(20): 3013-3029.

Trotta, R., L. Chen, S. Costinean, S. Josyula, B. L. Mundy-Bosse, D. Ciarlariello, C. Mao, E. L. Briercheck, K. K. McConnell, A. Mishra, L. Yu, C. M. Croce and M. A. Caligiuri (2013). "Overexpression of miR-155 causes expansion, arrest in terminal differentiation and functional activation of mouse natural killer cells." Blood 121(16): 3126-3134.

Trotta, R., L. Chen, D. Ciarlariello, S. Josyula, C. Mao, S. Costinean, L. Yu, J. P. Butchar, S. Tridandapani, C. M. Croce and M. A. Caligiuri (2012). "miR-155 regulates IFN-gamma production in natural killer cells." Blood 119(15): 3478-3485.

Trotta, R., D. Ciarlariello, J. Dal Col, H. Mao, L. Chen, E. Briercheck, J. Yu, J. Zhang, D. Perrotti and M. A. Caligiuri (2011). "The PP2A inhibitor SET regulates granzyme B expression in human natural killer cells." Blood 117(8): 2378-2384.

Costinean, S., S. K. Sandhu, I. M. Pedersen, E. Tili, R. Trotta, D. Perrotti, D. Ciarlariello, P. Neviani, J. Harb, L. R. Kauffman, A. Shidham and C. M. Croce (2009). "Src homology 2 domain-containing inositol-5-phosphatase and CCAAT enhancer-binding beta are targeted by miR-155 in B cells of Emicro-MiR-155 transgenic mice." Blood 114(7): 1374- 1382.

Trotta, R., J. Dal Col, J. Yu, D. Ciarlariello, B. Thomas, X. Zhang, J. Allard, 2nd, M. Wei, H. Mao, J. C. Byrd, D. Perrotti and M. A. Caligiuri (2008). "TGF-beta utilizes SMAD3 to inhibit CD16-mediated IFN-gamma production and antibody-dependent cellular cytotoxicity in human NK cells." J Immunol 181(6): 3784-3792.

Trotta, R., D. Ciarlariello, J. Dal Col, J. Allard, 2nd, P. Neviani, R. Santhanam, H. Mao, B. Becknell, J. Yu, A. K. Ferketich, B. Thomas, A. Modi, B. W. Blaser, D. Perrotti and M. A. Caligiuri (2007). "The PP2A inhibitor SET regulates IFN-gamma production." J Exp Med 204(10): 2397-2405.

Fields of Study

Major Field: Molecular, Cellular, and Developmental Biology

vii Table of Contents

Abstract ...... ii

Dedication...... iii

Acknowledgments ...... iv

Vita ...... vi

Publications ...... vi

Table of Contents ...... viii

List of Tables ...... xii

List of Figures...... xiii

List of Abbreviations ...... xv

CHAPTER 1: BACKGROUND...... 1

1.1Multiple Myeloma Biology ...... 1

1.1a Multiple myeloma overview and discovery ...... 1

1.1b Multiple myeloma diversity and progression ...... 3

1.1c Multiple myeloma and development ...... 10

1.1d Multiple myeloma emergence and the bone marrow niche ...... 11

1.1e Communication in multiple myeloma including extracellular vesicles ...... 13

viii 1.2 Natural Killer Cell Biology ...... 17

1.2a Natural killer cell effector function overview ...... 17

1.2b The immune synapse and natural killer cell receptor signaling ...... 18

1.2c Major histocompatibility complex class-I biology ...... 22

1.2d ...... 27

1.2e Interferon-gamma and STAT1 signaling ...... 28

1.3 STAT3 signaling in multiple myeloma ...... 29

CHAPTER 2: INTERFERON-GAMMA LOWERS THE SUSCEPTIBILITY OF

MULTIPLE MYELOMA TO NATURAL KILLER CELL-MEDIATED CYTOTOXICITY

...... 31

2.1 Introduction ...... 31

2.2 Results with Figures ...... 32

NK cells produce IFN-γ in response to MM ...... 32

NK cells produce IFN-γ in response to soluble MM factors ...... 33

IFN-γ increases MM cell surface expression of inhibitory ligands ...... 39

IFN-γ lowers the susceptibility of MM cells to NK cell-mediated cytotoxicity ...... 48

IFN-γ-mediated resistance is differentially recovered by IPH2201 (Anti-NKG2A) ...... 54

CD56Bright rather than CD56Dim NK cells exhibit greater killing against MM cells ...... 56

2.3 Discussion and Summary ...... 61

2.4 Experimental Methods ...... 63

ix Transwell co-cultures ...... 65

Flow cytometry and sorting ...... 65

Real time PCR ...... 66

Western blot analysis ...... 66

ELISA assays ...... 67

Cytotoxicity assays ...... 67

CHAPTER 3: MULTIPLE MYELOMA CELL-DERIVED EXTRACELLULAR VESICLES

ENHANCE IFN-GAMMA PRODUCTION BY NATURAL KILLER CELLS ...... 69

3.1 Introduction ...... 69

3.2 Results and Figures ...... 70

MM EV enhance NK cell-mediated IFN-γ production ...... 70

MM EV contain HLA-E and PD-L1 Inhibitory Ligands ...... 75

MM EV present ligands on their surface ...... 79

3.3 Discussion ...... 81

3.4 Methods ...... 83

Cell lines and NK cell preparations ...... 83

Treatments ...... 84

Extracellular vesicle isolation ...... 84

Flow cytometry ...... 85

EV surface staining and detection by Nanosight ...... 85

x ELISA assays ...... 86

Western blot analysis ...... 86

CHAPTER 4: FUTURE STUDIES AND EXTENDED DISCUSSION ...... 88

4.1 The mechanism for IFN-γ-mediated resistance of U266 to NK cell cytotoxicity ...... 88

4.2 IFN-γ treatment of MM cells to enhance efficacy of inhibitors ...... 89

4.3 Differential peptide cross-presentation in MM following IFN-γ treatment ...... 89

4.4 Concluding remarks ...... 90

Bibliography ...... 94

xi List of Tables

Table 1: IMWG Criteria for Diagnosing Multiple Myeloma ...... 4

Table 2: Common Chromosomal Abnormalities in Multiple Myeloma ...... 6

Table 3: Attempted Primary NK Cell Cytotoxicity Experiments Involving MM EV ...... 82

xii List of Figures

Figure 1: “Church spire” pattern in MM patient sera ...... 2

Figure 2: Immunoglobulin classes ...... 8

Figure 3: Extracellular vesicle release ...... 16

Figure 4: MHC-I Structure ...... 23

Figure 5: Healthy primary NK cells produce IFN-γ when cultured with MM ...... 33

Figure 6: IFN- γ production in MM-NK cell co-culture does not require cell-cell contact .... 34

Figure 7: MM cells constitutively express IFN-γRs on their surface ...... 36

Figure 8: IFN-γ activates STAT1 and STAT3 in MM cells ...... 37

Figure 9: Transcription of HLA-E and PD-L1 by MM cells after IFN-γ treatment ...... 38

Figure 10: Surface expression of HLA-ABC by MM after IFN-γ treatment ...... 41

Figure 11: Surface expression of HLA-E by MM after IFN-γ treatment ...... 41

Figure 12: Surface expression of PD-L1 by MM after IFN-γ treatment ...... 45

Figure 13: Transcription of HLA-E and PDL-1 by MM cells in contactless co-culture with

NK-92 ...... 45

Figure 14: Surface expression of HLA-E and PD-L1 by MM cells in contactless co-culture with NK92 ...... 47

Figure 15: Viability and susceptibility of MM cells to NK cell cytoxicity after IFN-γ treatment ...... 49

xiii Figure 16: IFN-γ confers resistance of MM cells to NK cell-mediated killing, no stimulation

...... 51

Figure 17: IFN-γ confers partial resistance of MM cells to NK cell-mediated killing, activating conditions ...... 52

Figure 18: IFN-γ dose-dependent resistance of MM cells to NK cell-mediated cytotoxicity . 53

Figure 19: IPH2201 (Anti-NKG2A) pretreatment of NK cells recovers IFN-γ-mediated resistance ...... 55

Figure 20: Comparing NK cell subsets in healthy and MM peripheral blood ...... 57

Figure 21: Sorted CD56Bright and CD56Dim NK cell cytotoxicity against MM cell line RPMI-

8226 ...... 58

Figure 22: Effect of IPH2201 (Anti-NKG2A) on CD56Bright and CD56Dim NK cytotoxicity against MM cell line RPMI-8226 ...... 60

Figure 23: Extracellular vesicle isolation and prep validation ...... 72

Figure 24: MM cell EV enhance NK cell IFN-γ production ...... 74

Figure 25: Protein profile of EV released from MM cell lines and patient sera ...... 76

Figure 26: HLA-E detected on mass spectrometry study ...... 77

Figure 27: HLA-E and PD-L1 in EV released from MM...... 78

Figure 28: NanoSight NS300 visualization of MM EV and HLA-ABC presentation ...... 80

Figure 29: Graphical Summary of Dissertation Work...... 92

xiv List of Abbreviations aa…………………………………………………………………………………... ADCC……………………………………...Antibody-Dependent Cell-mediated Cytotoxicity AML………………………………………………………………..Acute Myeloid Leukemia BCR…………………………………………………………………………. Receptor

β2M……………………………………………………………………..βeta-2-Microglobulin BLIMP1… ……………………………………B--Induced Maturation Protein 1 BM…………………………………………………………………..………….Bone Marrow BMSC…………………………………………………………….Bone Marrow C3…………………………………………………………………Complement Component 3 CD………………………………………………………………..…Cluster of Differentiation CLL………………………………………………………...Chronic Lymphoicytic Leukemia CLP…………..………………………………………………Common Lymphoid Progenitor CRAB………………………...hypercalcemia, renal insufficiency, anemia, lytic bone lesions CXCR5………………………………………………………….CXC- Receptor 5 EBF1……………………………………………………………...……..Early B Cell Factor 1 ECM……………………………………………………………..……….Extracellular Matrix ECs…………………………………………………………………………..Endothelial Cells ER…………………………………………………………………….Endoplasmic Reticulum EV………………………………………………………………………Extracellular Vesicles FasL………………………………………………………………………………..Fas FL………………...………………………………………………………………..Flt3 Ligand FLC…………………………………………………....Free--chain (of immunoglobulin) GAS…………………………………………………………….Gamma Activated Sequences HLA……………………..………………………………………..Human Leukocyte

xv HPC…………………………………………………………...Hematopoietic Progenitor Cell HSC………………………………………………………………....Hematopoietic Stem Cell IFN………..………………………………………………………………………...Interferon Ig..……………………………………………………………..…………….Immunoglobulin IGH……………………………………………………………Immunoglobulin Heavy Chain IL………..………………………………………………………………………… IMWG………………………..…………………………International Myeloma Work Group IRF1…………………………………………………………….Interferon Response Factor 1 IRF4…………………………………………………………...Interferon Regulatory Factor 4 IS…………………………………………………………………………….Immune Synapse ITAM…………………………………….Immunoreceptor Tyrosine-Based Activation Motif ITIM……………………………………...Immunoreceptor Tyrosine-Based Inhibitory Motif ITSM………………………………………...Immunoreceptor Tyrosine-Based Switch Motif JAK…………………………………………………………………...Janus Activated Kinase KIR…………………………………………………… Killer Immunoglobulin-like Receptor KL…………………………………………………………………...…………….KIT Ligand LFA-1…………………………………………..Lymphocyte Function-Associated Antigen 1 LPS……………………………………………………………………….. MGUS………..…………………...Monoclonal Gammopathy of Undetermined Significance MHC………………...……………………………………Major Histocompatibility Complex MM………………..………………………….……………………………Multiple Myeloma M-protein…………………………………………………………………Monoclonal protein MTOC……………………………………………………..Microtubule Organizing Complex MVB……………………………………………………………………Multivesicular Bodies MVD……………………………………………………………………..Microvessel Density NCR……………………………………………………………Natural Cytotoxicity Receptor NFκB…………………………………………………………………Nuclear Factor κappa B NK…..…………………………………………………………………..………Natural Killer NKG2…………………………………………………...……………..Natural Killer Group 2 PAX5…………………………………………………………………….Paired Box Protein 5

xvi

PC………..………………………………………………………………………..Plasma Cell PD-1……………………………………………………………………..Programmed Death 1 PD-L1…………………………………………………………...Programmed Death Ligand 1 RANK………………………………………..Receptor Activator of Nuclear Factor Kappa B SDF1………………………………………………………………..Stromal Derived Factor 1 SLAMF7……………………………..Signaling Lymphocytic Activation Molecule Family 7 SLO……………………………………………………………...Secondary Lymphoid Organ SLT……………………………………………………………...Secondary Lymphoid Tissue SMM…………………………………………………..…..….Smoldering Multiple Myeloma SOCS………………………………………………………Suppressor of Signaling STAT……………………………………...Signal Transducer and Activator of Transcription TAP………………………………………...Trasnporter Associated with Antigen Processing TBET……………………………………………………………..T-Box Expressed in T Cells TCR…………………………………………………………………………... Receptor TF………………………………………………….…………………….. TGF………………………………………………….………….Transforming TLR………………………………………………………………………..Toll-Like Receptor TNF……………………………………………………………………Tumor Factor TRAIL……………………………………………..TNF-Related -Inducing Ligand VEGF……………………………………………………Vascular Endothelial Growth Factor VLA-4……………………………………………………………………Very Late Antigen 4 XBP1………………………………………………………………..X-Box-Binding Protein 1

xvii

CHAPTER 1: BACKGROUND

1.1Multiple Myeloma Biology

1.1a Multiple myeloma overview and discovery

Multiple Myeloma (MM) is the uncontrolled proliferation of antibody-producing plasma cells

(PC) in the bone marrow (BM) for which there is essentially no cure. MM is the second most common hematological malignancy (after non-Hodgkin lymphoma) in the US and accounts for 1% of all malignant neoplasms and 13% of hematologic malignancies [1] [2]. In 2015, the lifetime risk of developing MM is 0.7% (1 in 143) with an average age range of 65-70 years at diagnosis.

In 1844, the first documented cases of MM in patients Sarah Newbury and Thomas

Alexander McBean were reported. Newbury suffered insufferable pain and multiple bone fractures leading up to her death. Upon McBean’s passing, an autopsy would reveal that his bones had softened and that their content, the marrow, was replaced with a “gelatiniform substance of blood-red colour and of unctuous (greasy) feel”[3]. While examining the urine of McBean, Henry Bence Jones would discover rich protein content, the identity of which would be determined nearly one hundred years later as monoclonal light chains. In 1873, pathologist Johann von Rustizky coined the term "multiple myelomas" upon observing eight separate bone tumors within a deceased patient [4]. With the development of X-ray imaging in the early 1900s, physicians were empowered to assess bone lesions; however therapies

1 were hardly effective. Further, as electrophoretic techniques in the 1940s-1950s were implemented, diagnosing myeloma became easier. MM patient sera have a distinct “church spire” signature attributed to elevated immunoglobulin when compared to healthy sera (Figure 1).To this day, sera protein electrophoresis remains an informative diagnostic for treatment assessment in MM.

Figure 1: “Church spire” pattern in MM patient sera

Figure 1: Cartoon of healthy(left) and MM patient(right) sera electrophoresis on

an agarose gel(top). Proteins separate into albumin α1, α2, β, and γ zones based on

size and charge. Densitometry produces the distinct “church spire” pattern in

(Continue on page 3) 2

(Continued from page 2) myeloma sera (middle, right). Immunofixation (bottom) allows for the Ig subclass

typing of the disease (in this example, IgG Myeloma with κ light chain).

1.1b Multiple myeloma diversity and progression

Despite MM’s prevalence and common symptomatic criteria, the disease is highly variable in

its manifestation. MM is clinically diagnosed at presentation of >10% PCs in the BM or BM

confirming plasmacytoma, the detection of serum or urinary Monoclonal (M-)protein

at any level, and end-organ damage attributed to plasma cell burden such as hypercalcemia, renal insufficiency, anemia, and/or lytic bone lesions (CRAB criteria, Table 1) [5].

Phenotypic markers of PCs include expression of cluster of differentiation (CD)138, CD38,

and more recently CD319 (SLAMF7, CS1) on their surface [6-8]. Aberrant expression of

CD56 and CD28, but lack of CD19 and CD27 are also commonly observed on MM PCs [9].

3 Table 1: IMWG Criteria for Diagnosing Multiple Myeloma

4 Beyond the surface of the cell, MM is a highly heterogeneous disease, presenting variable

antibody production, chromosomal aberrations, and karyotypic discordance. Like their

healthy PC counterparts, MM PCs clonally expand and selectively produce IgG, IgA, IgM,

IgD, or IgE antibodies (Figure 2), where one isotype dominates during active disease [10].

Although healthy and MM PCs incorporate either Kappa(κ) or Lambda(λ) Ig light chain

components, deviation from a healthy serum κ: λ range of 0.26 to 1.65, indicating clonal

expansion and/or incomplete immunoglobulin formation, is observed in MM [11]. The most common form of MM is IgG(52%) followed by IgA(21%), and free light chain (FLC, 16%)

[12]. Uncommon variants include non-secretory IgD(1%) and rarely IgE(0.01%) [13].

Frequently observed genetic abnormalities in most MM include hyperdiploid and nonhyperdiploid karyotypes. In hyperdiploid MM, a gain of odd 3, 5, 7, 9, 11,

15, 19, and 21 is observed, whereas in nonhyperdiploid MM chromosomal translocations of the IgH locus to chromosomes 4, 8, 11, and 16 occur [14-16]. Multivariate studies have refined both risk stratification and prognostic implications of abnormal karyotyping in MM, which are presented in Table 2 [17]. Further, new findings describe heterogeneity within an individual patient’s clonal population of malignant PCs, further complicating efforts to effectively treat MM [18].

5 Table 2: Common Chromosomal Abnormalities in Multiple Myeloma

6 MM is preceded by a premalignant phase known as Monoclonal Gammopathy of

Undetermined Significance (MGUS) [19, 20]. The term MGUS was coined in 1978 after an

extensive 5-year serum study would describe the ambiguous outcomes (some resulting in

death, some completely healthy) of 241 patients whom had all presented initially with

elevated serum M-protein but lacked evidence of advanced disease [21]. MGUS affects 2% and 3% of the population in the United States over the age of 50 and 70, respectively [22].

MGUS is often discovered incidentally during blood evaluations for a variety of clinical

symptoms, including fatigue and forgetfulness [23]. Recently, the International Myeloma

Working Group has refined MGUS criteria to include < 10% of PC in the BM, serum levels

of M-protein < 3 grams per deciliter, and lack of end-organ damage attributed to the plasma

cell burden including CRAB criteria [5]. Although MGUS is considered asymptomatic, some

patients describe neuropathy in their extremities, which is attributed to plasma cell-derived

FLC protein aggregates (amyloidosis) causing systemic microvasculitis [24]. The overall risk

of MGUS progressing to malignancy is ~1% per year, indefinitely [25]. Rather than

treatment, the standard of care for MGUS is active monitoring. Newly diagnosed patients are

recommended to check up every 3-4 months in the first year of MGUS diagnosis and subsequently every 6-12 months if no new symptoms develop [26].

7 Figure 2: Immunoglobulin classes

Figure 2: The prototypic immunoglobulin consists of two heavy chains bound by

disulfide bridges to two light chains (λ or κ). The C-termini of the heavy chains

(Continue on page 9) 8

(Continued from page 8)

make up the base region of the antibody, known as the Fc (Fragmentcrystallizable)

while the N-termini contain variable regions which distinguish the antibody’s

binding affinity, or Fab (Fragmentantigen-binding). The addition of the light chain,

which also contains a variable region, increases the possible binding

combinations the system can generate. Monomers IgG and IgE are free floating

whereas IgD is membrane bound. The Fc regions of both IgA, a dimer, and IgM,

a pentamer, are held together by a J-chain polypeptide.

Emerging from MGUS is an intermediary and indolent phase of MM termed Smoldering

MM (SMM). The smoldering was coined in a 1980 report describing prolonged (>5 years),

elevated, yet stable PC burden in asymptomatic and thus observation-only patients [27].

SMM clinically presents as serum M-protein > 3g/dL or urinary M-protein > 500mg/24hr,

and/or >10% plasma cells present in BM biopsy, but no end organ failure attributed to

CRAB criteria [5]. SMM carries a higher risk of progression to active disease than MGUS.

Indeed, SMM has a 10% per year chance of progressing to malignancy in the first 5 years,

followed by a 3% chance per year for the subsequent 5 years, and ~1% chance for every year

thereafter [28]. Interestingly, SMM patients which present with > 60% BM PC, serum or nonserum-associated FLC ratios > 100, and > 2 focal BM lesions by MRI are at a much greater risk [29, 30]. Such patients carry a 40% chance of progression per year, and

malignancy is nearly inevitable [31].

9 1.1c Multiple myeloma and plasma cell development

The early development of both healthy and MM PCs begin as progenitor hematopoietic stem

cells (HSCs) producing CD34+ common lymphoid progenitor (CLP) cells in the BM. The

commitment to a B cell lineage is driven by released by stromal cells in the BM

microenvironment, inducing a number of transcription factors (TF) including but not limited

to early B cell factor 1(EBF1), PU.1, E2A, and paired box protein 5 (PAX5) [32].

Development continues as each pro-B cell rearranges its immunoglobulin heavy chain (IgH)

segments, whose products pair with a universal surrogate light chain to assemble a

transmembrane IgM capable of binding antigen, the pre-B cell receptor (pre-BCR) [33]. The

pre-B cells phenotypically express CD45, major histocompatibility complex class-II (MHC-

II), CD19, and CD40 on their surface [34, 35]. Recombination-activating RAG1 and

RAG2 facilitate combination of 1 of 51 variable(V), 1 of 27 diversity(D) and 1 of 6 joining

(J) gene segments, known as V(D)J recombination, can collectively produce a repertoire of

IgH recognizing > 107 unique antigen specificities [36, 37].

The Pre-B cell, while in the BM, undergoes a crucial test of autoreactivity, by IgM recognition of either cell membrane proteins or soluble factors. Self-reactive events result in clonal deletion (apoptosis) after unsuccessful receptor editing, or anergy. The tolerant naïve

B cell, representing < 3% of all B cells generated, will then exit the BM and home to

CXCL13-rich splenic follicles via CXC- 5 (CXCR5), CC-chemokine receptor CCR7, and lymph nodes via adhesion molecule JAM-C to await [38-40]. The secondary lymphoid organs (SLO), such as the spleen and lymph nodes, provide a constant supply of circulating antigen.

10

Mature B cell activation occurs when the BCR engages an antigen, either free-floating or facilitated via an antigen-presenting cell ( and ). B cell activation can also occur by the T-cell-dependent mechanism of MHC-II peptide presentation and T- cell-independent mechanisms via toll-like receptor (TLR) recognition of lipopolysaccharide

(LPS) and hypomethylated CpG-DNA, or Complement component 3 (C3)-coated antigen which has been extensively reviewed elsewhere [41].

Activated B cells then undergo affinity maturation of the BCR, through further somatic hypermutation, class-switch recombination, and clonal selection allows the B cell with the highest affinity towards the specific antigen to predominate by proliferation. Most clonally expanded B cells will become short-lived effector plasmablasts which immediately secrete antibody against the invading pathogen. Days later, the differentiation of plasmablast to plasma cell initiates through IL-21/STAT3 and CD40L mediated repression of PAX5 [42,

43]. The plasma cell is now committed through the expression of transcription factors B- lymphocyte-induced maturation protein 1 (BLIMP1), interferon regulatory factor 4 (IRF4),

X-box-binding protein 1 (XBP1), and will express cell-binding molecule CD138/Syndecan-1 on its surface [44-47].

1.1d Multiple myeloma emergence and the bone marrow niche

B cells develop in the bone marrow, are affinity-selected in the GCs of SLT and, as plasma cells, egress back to the BM. The cause of PC malignant transformation in MM is currently unknown, but oncogenic events may occur during PC activation in the GC. An early event

11 described in both MGUS and MM is the dysregulation/over expression of the cyclin D gene, a key factor in cell cycle regulation [48]. Chromosomal abnormalities including translocations of the IgH locus during PC class switching in the GC have also been described

[49]. These observations likely serve as primary initiation events to malignancy. Subsequent mutations allow for constitutive activation of the nuclear factor kappa B (NFκB) pathway and somatic activating-mutations of NRAS and KRAS signaling oncoproteins (~15% of MM)

[50]. Plasma cells, malignant or otherwise, are strongly dependent on the BM microenvironment for survival. The homing of PCs to the BM is dependent on the release of chemokine stromal cell-derived factor 1 (SDF1) by BM stromal cells (BMSC) and its interaction with plasma cell-bound receptor CXCR4 [51, 52]. PC adhesion and extravasation to the BM microenvironment is initiated when CD138 binds to fibronectin and type I collagen-rich extracellular matrix (ECM) proteins [53-55]. Further, surface expression and activation of PC integrins, namely lymphocyte function-associated antigen 1 (LFA-1) and very late antigen 4 (VLA-4), enable the PC to directly interact with osteoblast, , stromal, and endothelial cells of the niche [56]. An adhesion event of a MM PC to a BMSC is sufficient to initiate both the MM PC activation of the anti-apoptotic NFκB pathway and

BMSC secretion of key MM survival and growth factor IL-6. In response to IL-6 activation, the MM PCs produce and secrete proangiogenic vascular endothelial growth factor (VEGF) which aids in the restructuring and optimization of the bone marrow niche for long term MM cell growth [57, 58].

12 1.1e Communication in multiple myeloma including extracellular vesicles

The bone marrow niche is a dynamic environment, constantly self-renewing, while

restructuring ECM components and providing hematopoietic support for developing

erythrocytes and immune cells. As MM progresses and tumor burden increases, PCs

outcompete and squelch normal hematopoietic cell processes, thereby altering the makeup

and function of cells, cytokines, and the ECM in the BM.

As discussed in 1.1d, PCs binding to BMSCs, induces proangiogenic factor VEGF and bFGF

[59]. BM and neovascularization, provided by endothelial cells (ECs) and measured by microvessel density (MVD), is increased in MM, but not MGUS [60]. Further, bone resorption and reformation is disrupted in MM, resulting in systemic hypercalcemia and

focused lytic bone lesions [61]. Bone destruction occurs proximal to MM PCs whose surface expression of receptor activator of NF-κB (RANK) ligand binds to RANK on neighboring osteoclast precursor cells, driving their survival, differentiation, and activation (bone

dissolution) [62, 63]. Conversely, osteoblasts, cells responsible for bone reformation, are

developmentally inhibited by MM PC-secreted wnt-signaling antagonist DDK1[64].

MM also influences the cytokine profile, both in the BM and systemically. Age-related changes in systemic cytokine milieu, termed “inflamm-aging”, skews the system to a pro- inflammatory state, whereas patients with end-stage myeloma have a profile that resembles immune senescence [65]. A recent study describes a trend of markedly elevated levels of proinflammatory factors IL-6, IL-8, alpha (TNFα),

(IFN-γ), and IL-10 in the peripheral blood (PB) and IL-6, IL-8, tumor necrosis factor alpha

TNFa, and IL-2 in the BM of MM patients [66]. Pleiotropic transforming growth factor beta

(TGFβ) is released by both PCs and BMSC in MM, which inhibits maturation and effector

13 functions of both the innate and adaptive , namely natural killer (NK) cells, , T-cells and antigen-presenting dendritic cells [67-73] . Of note, as MM progresses and immunodeficiency develops, ineffective immune surveillance and disease clearance will also leave the body susceptible to infection [74]. Indeed, myeloma patients when compared to healthy age-matched individuals are 7.1-fold and 10-fold more likely to develop a bacterial and viral infections, respectively[75].

Eventually, MM-driven remodeling of the BM will result in PC overcrowding in an unsustainable microenvironment, and subsequent malignant infiltration to a new bone.

Recent findings are beginning to recognize the influential role of extracellular vesicles in facilitating the spread of cancers to the bone. In a healthy setting, the BM is not naturally adapted for metastasis and requires preparation by the invading cancer. In , the BM premetastatic niche is re-educated by EV released by cancer cells, thereby reorganizing ECM components to facilitate attachment and establishment of a secondary site of malignancy

[76].

Extracellular vesicles (EV) are a collective term for non-organelle-containing, plasma membrane-enveloped packets of subcellular material. EV were originally believed to be cellular debris with little biological significance, until one study described B cell-derived

MHC-II-bearing EVs that were able to induce T-cell response [77]. Two types of EV commonly recognized are exosomes and microvesicles (Figure 3). Exosomes, which range from 50-100nm in diameter, are packaged internally and are released by the Trans-Golgi network from the cell as multivesicular bodies (MVB) [78, 79]. The larger of the EV,

14 microvesicles, which range from 100nm-1000nm in diameter, are generated by the outward blebbing of the cell membrane, and thus contain cargo and surface markers proximal to the membrane [80]. Capable of intercellular communication, EV contain DNA, mRNA, miRNA, and proteins, and can present surface ligands for functional delivery to specific recipient cell types [78, 81]. The collective term of EV is apropos in vesicle studies due to the limitations of successfully separating exosomes and microvesicles during preparation –contemporarily differential ultracentrifugation [82] .

15

Figure 3: Extracellular vesicle release

Figure 3: Exosomes(A) are packaged within multivesicular bodies and released

to the extracellular space. Microvesicles(B) form from outward blebbing of the

cell membrane.

16

1.2 Natural Killer Cell Biology

1.2a Natural killer cell effector function overview

Natural Killer cells are a unique subset of which function at the interface of the

innate and . Classically, natural killer (NK) cells are defined as large

granular lymphocytes that express surface cluster of differentiation (CD) marker CD56 but

lack CD3, a prototypic T cell marker. Natural killer cells make up 1-6% of total leukocytes

and have a healthy range of 0.08-0.43x106 cells per ml of peripheral blood [83]. NK cells have dual effector cell functions, namely to release recruitment factors and cytokines in response to inflammatory stimuli, and to exhibit cytotoxicity against virally infected and tumor-transformed cells [84]. Differential CD56 expression separates NK cells into two functionally distinct subpopulations, CD56Dim and CD56 Bright [85]. CD56Dim NK cells, considered to be more functionally mature, constitute ~90% of the NK cells circulating in the peripheral blood and are highly cytotoxic. Further, CD56Dim NK cells express an active

CD16 receptor, also known as FCγRIIIA, which recognizes the IgG1 (fragment crystallizable region) Fc region of antibodies, enabling CD56Dim to engage and attack targets coated by

IgG1-isotype antibodies [86]. CD56Bright NK cells constitute ~10% of the peripheral blood

NK and generally lack CD16 [87]. CD56Bright NK cells marshal an immune response by releasing a number of proinflammatory factors.

NK cells develop from bone marrow-derived CD34+ hematopoietic progenitor cell (HPC) to common lymphoid progenitor (CLP) cells that able to give rise to all lymphocyte subsets. NK cells differ from B and T cells due to lack of somatic gene rearrangement in establishing a

17

diverse repertoire of clones. NK cells develop in the bone marrow where BM stromal cells

produce the necessary cytokine/factors including but not limited to FL, KL, IL-7, and IL-15 to drive maturation. NK cell maturation and acquisition of functional competency appears to be site-specific and includes the secondary lymphoid tissues (SLT, i.e. tonsil and lymph nodes) as well as the spleen [84]. The alternative fate and function of tissue-specific NK cells is appreciated in the diverse NK cell populations found in the setting of pregnancy, SLT, BM, and cancer.

NK cells possess a variety of methods to exhibit cytotoxicity against a target. These include natural and antibody-dependent cell-mediated cytotoxicity (ADCC) through engagement of

Fc receptor CD16 and release of lytic granules. NK cells may also utilize members of the tumor necrosis factor (TNF) family TNF-related apoptosis-inducing ligand (TRAIL) and (FasL) to induce -mediated apoptosis on TRAIL receptor and - bearing target cells. Natural and ADCC killing can be measured within 4 hours in vitro while internal apoptotic dispatching by TRAIL and FasL requires at least 6 hours [88]. An NK cell’s preference for killing is established by signals received from its environment and potential target cells discussed in 1.2b.

1.2b The immune synapse and natural killer cell receptor signaling

When an NK cell engages a potential target, the rearrangement of its molecules at the cell surface interface forms the immune synapse (IS). The formation of the IS is not exclusive to

18 modulating cell death, but rather shares an adhesion mechanism for delivering proteins and signaling intermediates for activation and inhibition of recipient cells . In NK cell cytotoxicity, the IS provides a cleft between the NK cell and its target cell for specific and direct delivery of an apoptotic message via cytolytic granules [89]. However, the IS also provides a platform for target cells to present inhibitory ligands upon their surface. The NK cell is required to assess the balance of activating and inhibitory signals in the IS before inducing apoptosis. Interestingly, a single inhibitory event overrides activation, and thus multiple activation events are required to deliver a death message. This balancing mechanic serves two purposes –it has evolved to prevent a trigger-happy immune system while simultaneously promoting both cellular and systemic energy conservation.

The elegant formation of the IS is divided into three discrete stages, which is extensively described and reviewed elsewhere [90]. Briefly, the initiation stage begins as the NK cell, through chemotactic signaling or random surveying, makes early contact and tethers a potential target via members of the selectin family and CD2 [91, 92]. Early commitment to cytotoxicity and firm adhesion to the target is then established by integrins LFA-1 and MAC-

1, and activation of the NK cell through natural cytotoxicity receptor (NCR) members may support further action. In the absence of inhibitory signals, the effector phase describes the F- actin/WASP-dependent polarization and migration of the microtubule organizing complex

(MTOC), docked with many lytic granules containing perforin, granzyme A and granzyme B, towards the IS [93]. As granule contents are released into the synaptic cleft, CD107A- studded granule membrane incorporates into the NK cell surface and provides protection against its own weaponry [94]. In order to keep the neighboring bystander cells from being

19 exposed to lytic factors, the termination phase involves a period of inactivity followed by

disengagement of the NK cell from the target. Intriguingly, NK cells are serial killers and can

re-exhibit IS formation and cytotoxic potential in excess of 5 killings in 60 minutes [95].

These observations showcase the efficacy of the ever-ready NK cell when killing criteria is

properly corroborated.

Activation signals promote an NK cell to mobilize against a target. Multiple overlapping

activation signal intermediates and functional redundancy within SRC-family kinase

members Lck, Fyn, Src, Yes, and Fgr provide an obstacle for determining which case-

specific member is responsible [96]. Engagement of activating receptors bearing cytosolic

immunoreceptor tyrosine-based activation motifs (ITAMs) induces by SRC-

family kinases, pairing of affiliated adapter proteins (i.e. ZAP70, DAP10, DAP12, etc…) and

eventual downstream activation/phosphorylation of VAV1 and PLC-γ2 [97]. Vav1 and PLC-

γ2 signaling are required for calcium mobilization and release of cytolytic granules [98].

ITAM-bearing receptors include the natural cytotoxicity receptor (NCR) family members

NKp30, NKp44, and NKp46. FcγRIIIa(CD16, binds IgG1), SLAM family member

2B4(CD244, binds CD48) as well as lectin-like natural killer cell group 2 (NKG2)family members NKG2C-CD94(binds HLA-E under cellular stress) and NKG2D (binds

MICA/MICB) receptors also potentiate their signals through ITAM [99].

Inhibitory signals abrogate activation signals and serve to spare target cells from NK cell- mediated death. Engagement of inhibitory receptors results in activation of immunoreceptor tyrosine-based inhibitory motifs (ITIMS) which initiate the reciprocation of activating

20 signals. ITIMS signal through intermediates SHIP-1, SHP1and SHP2 which serve to reverse early activation of concomitant ITAMS and prevent calcium flux by dephosphorylating

VAV1, respectively [100, 101]. ITIM-bearing receptors include killer Ig-like receptors (KIR, bind MHC-I), lectin like NKG2 family member NKG2A-CD94 (Binds HLA-E), and programmed death 1(PD-1) which binds programmed death ligand 1(PD-L1) [102]. KIR and

NKG2A activation phosphorylate Crk, which promotes the disassembly of Crk, CBL-C and

C3G, neutralizing actin reorganization required for cytolytic potential [103]. PD-1 engagement upregulates CBL-b and CBL-C, which in T cells, promote ubiquitin-mediated proteasome dispatching of activating TCR surface receptors [104]. Signaling lymphocytic activation molecule family 7 (SLAMF7 or CS1, self-ligating) contains an immunoreceptor tyrosine-based switch motif (ITSM), which has potential to activate or inhibit downstream signaling. In NK cells, SLAMF7 interacts with CD45 and assembles inhibitory signaling microclusters on the cell surface, which restricts the formation of activating complex assembly. SHIP-1 and its downstream intermediates enable SLAMF7 to inactivate VAV1 and PLC-γ2 [105].

Many therapeutic approaches in MM have been designed to clinically target/neutralize the inhibitory messaging of NK cells, thereby enabling proper clearance of aberrantly presenting

MM cells. Indeed, monoclonal antibodies against CS1(Elotuzumab), inhibitory

KIR(lirilumab), PD-1/PD-L1(BMS-936558/936559), and NKG2A(IPH2201,

AstraZeneca/InnatePharma) are currently on the market or in clinical trials. However, it should be noted that careful consideration for treatment strategies should be made, as the

21 possibility of selectively promoting the rise of more aggressive and resistant MM PC clones

may occur.

1.2c Major histocompatibility complex class-I biology

All jawed vertebrates commonly express major histocompatibility complex (MHC) gene products and their cognate innate and adaptive immune receptors, killer immunoglobulin receptors (KIR) and T-cell receptors (TCR) respectively. Thus the and

NK-like cells have been functioning to protect organisms against pathogens and genetic maladies for > 450 million years [106, 107]. MHC genes, known as human leukocyte antigen

(HLA) genes in humans, are clustered on the short arm of 6 and fall into subclasses MHC-I (classical), MHC-Ib (non-classical), MHC-II based on function. The

general function of all MHC products is to provide a sampling of endogenous and exogenous

peptides, a process termed cross-presentation, to members of the immune system.

MHC-I proteins are either expressed or inducible in all nucleated cells. A functional MHC-I

complex is on the surface of the cell as a heterodimer, consisting of one MHC heavy chain

bound with antigenic peptide and associated with a light chain Beta-2-Microglobulin (β2M)

[108]. The polymorphic 45 kDa MHC-I heavy chain consists of 3 αlpha domains (Figure 4).

The α3 domain comprises a transmembrane domain that non-covalently binds the 12kDa β2M during complex formation, whereas the α1 and α2 domains form the binding cleft for an 8-9 amino acid (aa) long peptide [109, 110].

22

Figure 4: MHC-I Structure

Figure 4: The basic structure of MHC-I, featuring the non-covalent joining of the

alpha chain(blue) with β2M (yellow), and a peptide octamer (red) loaded within

the binding groove. The polymorphic regions within the α1 and α2 domains form

binding grooves of varying specificities towards peptides of 8-10 amino acids in

length. The α3 domain, which contains the membrane-spanning portion, also

serves as a recognition domain to T cells during antigen/peptide presentation.

23

In order to maintain the economy of the ever-changing cell, protein turnover via

constantly helps repurpose amino acids to new protein synthesis, all-the-while providing peptides for the process of antigen presentation [111]. Proteins whose function are no longer needed, or are improperly folded/translated are often tagged for degradation by

(poly)ubiquitination [112]. The ubiquitin tag, recognized by the proteasome’s 19s regulatory cap, serves as entry criteria for degradation [113]. Within the giant multicatalytic proteasome

core, multiple rounds of cleavage events fragment the protein and provide an ample library of

peptides for potential MHC loading. To expand the potential antigen pool, longer precursor

peptides are trimmed to 8-9aa in length by peptidase ERAP1 and ERAP2 [114]. The peptides

released from the proteasome are free-floating in the cytosol, only have a half-life of ~10

seconds, and will require the function of the transporter associated with antigen presentation

(TAP-1 and TAP-2) pump to gain access to the endoplasmic reticulum(ER) for incorporation

into the MHC-I heterodimers [115].

The ER lumen is the site of MHC-I peptide loading. Within minutes of translation, the

MHC-I heavy chain associates with β2M, enhancing the binding groove’s affinity for peptides [116]. The MHC-I heterodimers are partially folded and are stabilized by chaperone proteins tapasin, , and PDIA3 [117, 118]. The peptide loading complex is completed when tapasin associates with TAP which creates a close junction for efficient peptide loading during peptide translocation to the ER [119]. Bound MHC-I will undergo a proper folding quality control program in the ER prior to transport to the Golgi-apparatus and eventual incorporation into the plasma membrane –reviewed elsewhere [120].

24

The MHC-I (HLA-A, HLA-B, HLA-C) genes are highly polymorphic in distinct regions of the α1 and α2 domains. Although the general structure of the MHC-I is conserved, peptide preference and binding capacity of the 10 aa residue binding groove can differ considerably

[121]. To date, 3356 HLA-A, 4179 HLA-B and 2902 HLA-C alleles have been sequenced and identified [122]. However, an individual genome will only have 2 alleles of each MHC gene represented, one allele inherited from each parent. NK cells, through KIRs, assess the quantitative and qualitative MHC-I self-antigen surface expression of neighboring cells. Lack of MHC-I, or misfolded MHC-I due to cellular or viral stress, will support the activation of the NK cells, and likely induce apoptosis of the target cell. Indeed, the chronic myeloid leukemia-derived cell line K562 completely lacks surface expression of MHC-I and is highly susceptible to natural NK-mediated cytotoxicity [123]. Interestingly, primary MM PCs and some MM cell lines constitutively express MHC-I in abundance (data not shown) [124].

MHC-Ib (HLA-E, HLA-F, and HLA-G) are highly conserved, less polymorphic and provide

a qualitative signal during peptide presentation. To date, 21 HLA-E, 22 HLA-F, and 52

HLA-G alleles have been reported [122]. HLA-E and HLA-G contain 8 and 9 aa binding

groove residues, whereas HLA-F retains only 5aa residues [125, 126]. MHC-Ib members

generally present cell-survival ligands to inhibitory receptors on innate and adaptive immune

cells.

HLA-F remains the least studied and understood member of the MHC-Ib family. Evidence

suggests the interaction of HLA-F tetramers with inhibitory receptor LIR-1 of NK cells, thus

altering the activation threshold of contemporary signals [127]. Shown to associate with free

25 HLA-I heavy chains, HLA-F is proposed to aid in cross-presentation of antigen among activated immune cells [128]. HLA-F expression is tissue restricted to the thymus, spleen, and tonsil where it can be observed on/in NK cells, B cells, T Cells (but not Tregs), and [129].

In a healthy setting, HLA-G expression is highly restricted to the invading trophoblast in the setting of pregnancy [130]. The embryo requires protection in the as a hemi- allograft, only sharing half of its MHC identity with its mother. HLA-G, on the developing fetus or surrounding it in soluble form, will engage inhibitory receptors LIR and KIR2DL4 on tissue-specific decidual NK and initiate a phase of potent immune tolerance at the fetal- maternal interface that will last until birth [131-133]. Of interest, dysregulation and aberrant expression of HLA-G-bearing cells has been reported in blood cancers, including acute myeloid leukemia(AML) and chronic lymphocytic leukemia (CLL)[134, 135]. Further, the release of soluble HLA-G from cancer cells serves as another dimensional setback to overcome in targeted therapies, as systemic inhibitory signaling may restrict proper immune trafficking to the source.

Unlike other nonclassical MHC-Ib members HLA-G and HLA-F, HLA-E transcriptional expression is not tissue-restricted, and is likely transcribed in all MHC-I-expressing cells

[136]. HLA-E shares the same structure and functional domain distribution with classical

MHC-I. The peptide binding groove of HLA-E has high affinity for its cognate peptides, the

5’-leading strand signal peptides of classical MHC-1 (HLA/B/C) and HLA-G [137, 138].

Upon binding a peptide HLA-E, which is otherwise restricted to ER lumen, is trafficked to

26

the cell surface to present an inhibitory signal to its receptor on NK cells, NKG2A-

CD94(1.2b) [139]. Of note, the affinity for HLA-E to NKG2A is highest when HLA-E

binds/presents the leading peptide of HLA-G [140, 141]. HLA-E’s potent inhibitory

influence was first demonstrated after a xenogenic transfection of human HLA-E to porcine endothelial cells was sufficient in ablating human NK cell xenoreactivity, in vitro [142, 143].

Further, one study describes the protective potential of HLAE on the surface of various cancers including cervical cancer, breast cancer, melanoma, and some leukemias [144].

1.2d Interferons

Interferons were discovered for their anti-proliferative effect on virally infected cells, which in turn, interferes with viral propagation [145, 146]. Interferons are a family of small (<146 aa) naturally occurring yet pleiotropic proteins divided into three classes- Type I-III. Type I interferons include IFN-α and IFN-β, who share some , are produced in

response to viral dsDNA detection by DNA sensors RIG-1 and DAI, as well as TLR3 and

TLR4 stimulation via LPS and dsRNA, respectively[147]. IFN-γ, originally dubbed immune

interferon, is the exclusive member of Type II interferon. IFN-γ is produced by NK cells and

T cells commonly after IL-2, IL-15, IL-12/IL18, and Type I IFN stimulation [148]. The least

studied family, Type III interferon members include IFN-λ1 (IL-29) IFN-λ2(IL-28a) and

IFN-λ3(IL-28b), signal through paired IFNλR1/IL10R2 receptor, can augment other IFN

family member responses, and can potently inhibit replication of specific viral families

(HSV-2, ECMV, HBV, HCV) [149-151].

27 In NK cells, IFN-γ is under the transcriptional control of TBET and EOMES where the observed expression of each TF is higher in the cytokine-producing CD56Bright than the cytotoxic CD56Dim cell subset [152]. Some human NK cells constitutively express IFN-γ mRNA transcript in order to expedite their response, and can actively secrete IFN-γ within 30 minutes of activation [153]. IFN-γ can promote TH1 cell differentiation, a subset who produce IFN-γ upon stimulation [154]. IFN-γ can also support inducible Treg differentiation, an immunosuppressive T cell subset that is linked to immune tolerance and suppression in

MM [155]. Unlike perforin and lytic granules who associate with MTOC, IFN-γ secretion facilitated through the endosome recycling [156].

1.2e Interferon-gamma and STAT1 signaling

IFN-γ orchestrates transcriptional control of many gene families and cellular processes.

Extracellular IFN-γ binds surface-expressed IFN-γR1 and/or IFN-γR2 which homodimerize and thereby autophosphorylate their associated tyrosine kinase members janus activated kinase 1 (JAK1) and/or JAK2, respectively [157]. Activated JAKs provide a docking site for signal transducer and activator of transcription family members (STATs). JAK1 phosphorylates the c-terminal tyrosine residue Y701 of STAT1 while JAK2 phosphorylates

Y690 of STAT2 [158, 159]. Both pSTAT1 and pSTAT2 heterodimerize with other STAT proteins and transmigrate to the nucleus where they bind to gamma activated sequences

(GAS) and activate or suppress transcription of >200 downstream target genes such as interferon response factor 1 (IRF1) and suppressor of cytokine signaling (SOCS) genes.

Interestingly, STAT1-mediated activation of T-box Expressed in T cells (TBET) completes a loop [160]. Importantly, STAT1 activation induces TAP1/2, the antigen

28 peptide pump required for peptide loading and increased surface expression of MHC-I (1.2c)

[161]. Like MHC-I, IFN-γR1 is expressed or inducible on all nucleated cells. However, IFN-

γR2 expression is restricted to immune cells such as NK cells, CD4+ T helper cells, and dendritic cells after antigen endocytosis during microbial infection [162].

However, JAK-STAT signaling alone is not sufficient to describe the totality of the IFN-γ response. A recent ChIP-Seq based study reports 1441 STAT1-regulated genes, of which only 212 were known IFN-γ regulated genes, and only 194 genes (13.5%) and 42 genes

(2.5%) were upregulated and downregulated in the mRNA transcriptome [163]. The ChIP-

Seq study only describes the influence of IFN-γ as a single agent, underlying the complexity

of STAT1-mediated processes, involving many cooperating initiation factors of specific gene

programs in situ and in vivo.

1.3 STAT3 signaling in multiple myeloma

Recent mathematical modeling describes the crosstalk mechanism between IFN-γ signaling

and the IL-6/STAT3 pathway [164]. IL-6, a key growth factor for MM PC (1.1d), provides a

crucial MM survival signal via STAT3 activation. STAT3 is constitutively activated in some

MM cell lines (U266) and is correlated with resistance to apoptosis through a Bcl-xL-

dependent mechanism [165, 166]. Further, studies have shown in a healthy STAT1-deficient

mouse, IFN-γ can alternatively activate STAT3, which is likely a last-resort act of

resourcefulness to preserve some of the original message through redundant/common STAT

target genes [167]. In human , STAT3 activation by IFN-γ has also been reported

[168]. Further, transforming growth factor beta (TGFβ) produced by BMSC in MM can

29 perpetuate the IL-6/STAT3 survival mechanism for MM PC, while simultaneously enacting potent inhibitory effects upon NK cells [169].

30 CHAPTER 2: INTERFERON-GAMMA LOWERS THE SUSCEPTIBILITY OF MULTIPLE MYELOMA TO NATURAL KILLER CELL-MEDIATED CYTOTOXICITY

2.1 Introduction

Natural killer (NK) cells are large granular lymphocytes that comprise a key cellular subset of the innate immune response. Capable of lysing virally-infected and malignant targets independent of co-stimulatory signals and gene rearrangement events, NK cells are also a major source of potent, immunomodulatory cytokines which can marshal and guide the immune response. Among these, NK cells are a principal source of interferon-γ (IFN-γ) which serves to augment immune function, induce Th1 polarization and recruit immune cells to inflammatory sites [170].

NK cells appear to play an important role in the immune response to multiple myeloma

(MM). However, MM cells utilize strategies to evade detection and lysis by immune cells.

The specific mechanisms responsible for these immunoevasive events are incompletely understood. Herein, we provide new evidence supporting a role for IFN-γ in mediating MM resistance to NK cell surveillance and killing. NK cells produce IFN-γ when exposed to MM cells; however, MM cells express functional IFN-γ receptors and, via STAT1 and STAT3 signaling, mediate transcription of HLA-E, an inhibitory ligand of the NK receptor NKG2A

31 [171]. HLA-E in turn suppresses NK cell-mediated cytotoxicity against MM targets through

interaction with the inhibitory receptor NKG2A, which appears to be overexpressed on NK

cells in MM (unpublished observations). This immunosuppressive axis may be overcome

with the use of an anti-NKG2A antibody to interrupt this receptor/ligand interaction.

Collectively, these data provide novel insight into the means by which MM cells may subvert

signaling in the immune system to achieve NK-cell directed immunoevasion; furthermore, these findings highlight new considerations for MM treatment strategies. In combination with the observation of NKG2A overexpression on NK cells in MM, these findings implicate this antigen as a potential novel target for checkpoint inhibitor in MM, as well.

2.2 Results with Figures

NK cells produce IFN-γ in response to MM

We have previously observed that MM patient-derived NK cells secrete IFN-γ in response to autologous, primary MM tumor cells [172]. Numerous studies have indicated abnormal NK cell effector functions in the setting of MM, so we verified this effect utilizing healthy donor

NK cells against U266 MM cell line targets (Figure 5, A) as well as primary MM tumor cells

(Figure 5, B).

32 Figure 5: Healthy primary NK cells produce IFN-γ when cultured with MM

Figure 5: ELISpot assays describe spontaneous IFN-γ production by primary

enriched NK cells co-cultured 10 NK cells: 1MM cell for 24 hrs. Spontaneous

IFN- γ production by primary NK cells was observed when co-cultured with

either the MM cell line U266 (A) or primary CD138+ purified MM PC (B).

NK cells produce IFN-γ in response to soluble MM factors

Interestingly, NK cell IFN-γ production does not require direct NK cell: MM cell contact. To

assess the contribution of soluble factors to IFN-γ induction, a porous membrane which

restricts cell migration, thus preventing NK cell:MM cell contact, was used (Figure 13, A). In

this contactless co-culture system, when challenged with the RPMI-8226 MM cell line

(Figure 6, A) as well as primary, CD138+ MM tumor cells (Figure 6, B) at 24hrs, IFN-γ production by the NK cell line, NK-92, increased as a function of the quantity of MM target cells present.

33 Figure 6: IFN- γ production in MM-NK cell co-culture does not require cell-cell contact

* *

**

Figure 6: Either MM cell line RPMI-8226(A) or primary CD138+ MM PCs(B)

were co-cultured with NK cell line NK92 for 24hrs in contact-restrictive

conditions, while allowing the passing and exchange of soluble factors under

400nm in diameter. Supernatants from each co-culture were harvested and

relative IFN-γ production was assessed by -linked Immunosorbent Assay

(ELISA). NK92 cells made significantly more IFN-γ when contactless co-

cultured with MM cell line RPMI-8226 (A, P-value <0.001) and primary plasma

cells (B, both P-values < 0.003) at MM:NK cell ratios of 4:1 and 2:1(B only). Of

note, as the MM:NK cell ratio increased, IFN-γ output was amplified.

34 MM cells express functional IFN-γ receptors

Elevated IFN-γ levels are a systemic factor in MM, and our novel finding of MM-induced

IFN-γ production led us to assess the sensitivity of MM cells to IFN-γ. In normal culture conditions (i.e. RPMI media supplemented with 10% serum), MM cell lines RPMI-8, A) and

U266 (, B) constitutively expressed both IFN-γR1 and IFN-γR2 on their surface and expression was not altered after treatment with IFN-γ (data not shown, 48hr). In accordance with the canonical IFN-γ signaling pathway, a time course treatment of RPMI-8226 (Figure

8, A) and U266 (Figure 8, B) cells with IFN-γ revealed early and sustained activation/phosphorylation of signal transducer and activator of transcription 1

(STAT1,Y701) protein, confirming the prior observation by liu, et al [173]. With the recent discovery that IFN-γ can alternatively activate/phosphorylate STAT3, we assessed and observed early, though unsustained, activation/phosphorylation of STAT3(Y705) by IFN-γ

(Figure 8A/B) [167]. STAT1 regulates many genes (>1400 known) and robustly drives gene transcription and surface expression of MHC-Ia molecules. However, STAT1 has also been reported to upregulate the inhibitory ligand PD-L1 on MM cells [173]. The regions of PD-L1 and the inhibitory MHC-Ib ligand HLA-E are studded with STAT1 and STAT1/3/5 binding sites (Figure 9A), respectively. We next tested whether IFN-γ treatment was sufficient to drive of the inhibitory ligand HLA-E while verifying PD-L1 gene upregulation. Indeed, both HLA-E (Figure 9, B left) and PD-L1(Figure 9, B right) gene transcription is enhanced after (48hr) treatment with IFN-γ, but not by the MM cell proliferation factor IL-10 in both normal and serum-starved culture conditions.

35 Figure 7: MM cells constitutively express IFN-γRs on their surface

RPMI-8226 A.

U266

B.

IFN-γ R1 IFN-γ R1

Figure 7: MM cell lines RPMI-8226 and U266 were maintained in normal

culture conditions of RPMI media supplemented with 10% fetal bovine serum, 2

mM Glutamax (glutamine) and antibiotic/antimycotic added. Flow cytometric

analysis of IFN-γR1 and IFN-γR2 on MM cell lines RPMI-8226 (Top) and U266

(Bottom) shows constitutive expression of both IFN-γR1 and IFN-γR2 in both

cell lines. Of note, 48hr IFN-γ treatment of either cell line showed no change in

IFN-γR1 and IFN-γR2 surface expression (data not shown).

36 Figure 8: IFN-γ activates STAT1 and STAT3 in MM cells

Figure 8: A two hour time course and assessment of IFN-γ early activation of

STAT1 and STAT3 on MM cell lines RPMI-8226 (A) and U266 (B). pSTAT3

depicts the assessment of tyrosine 705 (Y705). GAPDH serves as a loading

control and neither total STAT1 nor total STAT3 protein levels were affected by

IFN-γ treatment. Due to labeling and label-stripping restrictions, both A and B

represent two separate western blots of the same protein lysates with loading

equality verified by uniform GAPDH signal.

37 Figure 9: Transcription of HLA-E and PD-L1 by MM cells after IFN-γ treatment

Figure 9: A database search(SABiosciences) provides a profile of the promoter

regions of both PD-L1 and HLA-E (CD274 and HLAE genes) which contain

multiple STAT binding sites (A). Treatment of MM cell line RPMI-8226 with

IFN-γ for 48hrs induced both HLA-E (B, left, P< 0.002) and PD-L1(B, right,

P<0.02) gene transcripts. HLA-E and PD-L1 transcription was not altered by

serum-starvation (light grey) or treatment with the MM cell proliferation factor

IL-10.

38 IFN-γ increases MM cell surface expression of inhibitory ligands

The biological readout for IFN-γ signaling is the cross presentation of potentially antigenic peptides through MHC-I. We assessed dose response effects of IFN-γ treatment on surface expression of HLA-ABC(Figure 10), HLA-E(Figure 11) and PD-L1(Figure 12) in MM cell lines, as well as K562 cells, which lack MHC-I. Interestingly, although the MM cell lines

U266 and OPM2 also lacked MHC-I on their surface, both cell lines still showed a dose- dependent increase in expression of all markers tested. Further, the RPMI-8226 cell line had the most robust measured response with regards to upregulating HLA-E and PD-L1, thus was chosen to serve as a model cell line for future functional assays. When RPMI-8226 cells were co-cultured with NK-92 cells in a contactless system for 48 hours in IFN-γ-secreting conditions, the RPMI-8226 showed a marked increase in the inhibitory ligands HLA-E and

PD-L1 gene transcripts (Figure 13). As expected, in the same contactless co-culture setup, increased surface expression of both HLA-E and PD-L1 was observed (Figure 14).

.

39

(Continue on page 41) on (Continue

γ treatment - ABC by MM after IFN after MM by ABC - Surface expression of HLA Figure 10:

40 (Continued from page 40)

Figure 10: Surface expression of HLA-ABC by MM after IFN-γ treatment

Flow cytometric assessment of MHC-I (HLA-ABC) on MM cell lines after

culture in normal conditions (No IFN-γ, grey line), or after low dose (1ng/ml ,

black line) and high dose (10ng/ml, filled in) IFN-γ treatment. Note that K562,

U266, and OPM2 cells lack MHC-I surface expression following culture in

normal conditions.

41

(Continue on page 43) on (Continue t γ treatmen γ - E by MM after IFN MM after E by - Surface expression of HLA Figure 11:

42 (Continued from page 42)

Figure 11: Surface expression of HLA-E by MM after IFN-γ treatment

Flow cytometric assessment of inhibitory ligand HLA-E (MHC-1b) on MM cell lines in normal culture conditions (No IFN-γ, grey line), or after low dose

(1ng/ml , black line) and high dose (10ng/ml, filled in) IFN-γ treatment. A stepwise, dose-dependent increase in surface expression of HLA-E was observed for all cell lines.

.

43

(Continue on page 45 ) on (Continue t γ treatmen - L1 by MM after IFN MM after by L1 - Surface expression of PD Figure 12:

44 (Continued from page 44)

Figure 12: Surface expression of PD-L1 by MM after IFN-γ treatment

Flow cytometric assessment of inhibitory ligand PD-L1(CD274) on MM cell lines in normal culture conditions (No IFN-γ, grey line), or after low dose

(1ng/ml , black line) and high dose (10ng/ml, filled in) IFN-γ treatment. The induction of PD-L1 surface expression was detected in all cell lines following treatment with IFN-γ(10ng/ml); the marginal shift in positive PD-L1 expression may be attributed to inefficient epitope binding.

45 Figure 13: Transcription of HLA-E and PDL-1 by MM cells in contactless co-culture

with NK-92

A. RPMI-8226 400nm porous membrane NK-92 Soluble Factors

B. RPMI-8226 RPMI-8226 * *

Figure 13: Contactless co-culture (schematic, A) of NK-like cell line NK92 with

MM cell line with RPMI-8226 in IFN-γ-producing conditions (24 hrs) increases

RPMI-8226 cells’ transcription of both HLA-E (B, left, P <0.001) and PD-L1 (B,

right, P < 0.001) genes. HLA-E shows an increased trend of transcription as the

MM:NK cell ratio also increased. The opposite can be said of PD-L1, but in the

higher populated conditions, the observed effect may be at least partially

attributed to approaching conditions of nutrient depletion.

46 Figure 14: Surface expression of HLA-E and PD-L1 by MM cells in contactless co- culture with NK92

IL-2 Stimulation

Figure 14: Flow cytometric analysis of 48hr contactless co-cultured MM cell line

RPMI-8226 with the NK-like cell line NK92 + IFN-γ-producing(IL-2)

conditions. Although a slight increase in surface expression of HLA-E on RPMI-

8226 cells was observed in the unstimulated control sample(2nd row, right,

23.21%, MFI 2549 vs 1419), both PD-L1(3rd row, middle, 26.09%, MFI 2767 vs

994) and HLA-E(3rd row, right, 59.79%, MFI 4361 vs 1419) expression was

greatest at 1:1 NK:MM cell ratio in IFN-γ-producing(IL-2) conditions.

47 IFN-γ lowers the susceptibility of MM cells to NK cell-mediated cytotoxicity

Next we assessed the direct effects of IFN-γ treatment on MM cell viability and

proliferation. The proliferation rate of all MM cell lines tested at 48hrs by MTS

assay (Figure 15, A), as well as viability (data not shown), remained unchanged.

With no observed direct toxicity, we proceeded to assess the effect of IFN-γ on

the MM cell’s susceptibility to NK cell-mediated lysis. MM cell lines U266

(Figure 15, B and C) and RPMI-8226 (Figure 15, D and E) treated with IFN-γ

were less susceptible to primary enriched NK cell-mediated cytotoxicity. IFN-γ

treatment conferred resistance of MM cell lines to enriched primary NK-cell

mediated natural (Figure 16) and IL-2-activated cytotoxicity (Figure 17). Further,

differential sensitivity to IFN-γ and resistance to NK cell-mediated lysis was

observed in RPMI-8226 and U266 cell lines (Figure 18).

48 Figure 15: Viability and susceptibility of MM cells to NK cell cytoxicity after IFN-γ treatment A.

B. U266 C. U266

* *

D. E. RPMI-8226 RPMI-8226

*

*

(Continue on page 50) 49 (Continued from page 49) Figure 15: High dose IFN-γ (10ng/ml) treatment of MM cell lines did not affect viability or proliferation as measured by MTS assay at 48hrs (A). IFN-γ

treatment (10ng/ml, 48hrs) of U266 and RPMI-8226 confers their resistance to

primary enriched NK cell-mediated natural cytotoxicity in both NK-unstimulated

(B, P-value < 0.001 and D, P-value < 0.026 respectively) and NK-activated

conditions (IL-2, 24hrs, C, P-value <0.01 and E, P-value <0.001, respectively).

Data depicts a standard 51Cr release assay and error bars depict the standard deviation of 3 technical replicates.

50 Figure 16: IFN-γ lowers the susceptibility of MM cells to NK cell-mediated killing, no stimulation

* *

* *

Figure 16: IFN-γ treatment (10ng/ml, 48hrs) of MM cell lines confers their

resistance to primary enriched NK cell-mediated natural cytotoxicity in NK-

unstimulated conditions. NK cells killed cell lines L363, MM1.S, OPM2 and

U266 significantly less when the MM cell lines were pretreated with IFN-γ for

48hrs (All P-values < 0.01). Note, the values and scale for H929 and RPMI-8226

data were low. Data represents 1 of 3 donors/experiments with similar trends.

Data depicts a standard 51Cr release assay and error bars depict standard deviation

of technical triplicates.

51 Figure 17: IFN-γ confers partial resistance of MM cells to NK cell-mediated

killing, activating conditions

* *

*

Figure 17: IFN-γ treatment (10ng/ml, 48hrs) of MM cell lines confers their resistance to primary enriched NK cell-mediated natural cytotoxicity in NK- activated (IL-2, 24hrs) conditions. NK cells activated with IL-2 killed cell lines

H929,MM1.S and RPMI-8226 significantly less when the MM cell lines were pretreated with IFN-γ for 48hrs (All P-values < 0.005). Also, the observed resistance of MM cell lines OPM2, U266, and L363 to NK cell-mediated killing

(Figure 16 data) was overcome in NK cell-activated conditions. Data represents 1

of 3 donors/experiments with similar trends. Data depicts a standard 51Cr release

assay and error bars depict standard deviation of technical triplicates

52 Figure 18: IFN-γ dose-dependent resistance of MM cells to NK cell-mediated cytotoxicity

Figure 18: MM cell lines were treated with a low dose of IFN-γ (1ng/ml) or a high dose of IFN-γ (10ng/ml) for 48hrs. MM cell lines U266 and RPMI-8226 exhibit differential sensitivity to IFN-γ treatment doses 1ng/ml and 10ng/ml. The data suggests that U266(Left) cells require higher amounts of IFN-γ in order to gain protection from NK cell-mediated lysis. The RPMI-8226(Right) cells show a measurable resistance to NK cell-mediated lysis at the lower dose(1ng/ml), and resistance is increased as the dose escalates(10ng/ml). Data depicts a standard

51Chromium release assay and error bars depict the standard deviation of technical triplicates.

53 IFN-γ-mediated resistance is differentially recovered by IPH2201 (Anti-NKG2A)

IPH2201 is a humanized IgG4 (non-depleting) anti-NKG2A antibody which sterically blocks the potential for inhibitory ITIM signaling in the NK cell. Thus IPH2201 serves as an apropos tool to assess HLA-E’s part in mediating MM cell resistance to NK-mediated cytotoxicity. Primary enriched NK cells pretreated with IPH2201 were able to overcome

RPMI-8226 (Figure 19, A and B) cells’ resistance to NK-mediated cytotoxicity, but oddly, the same was not true for the U266 cell line (Figure 19, C and D). Further, IFN-γ treated

U266 remained resistant to NK-mediated killing after blocking TRAIL (data not shown) or

MHC-I entirely (data not shown).

54 Figure 19: IPH2201 (Anti-NKG2A) pretreatment of NK cells recovers IFN-

γ-mediated resistance

A. B. * *

C. D. n.s. n.s. n.s.

n.s.

Figure 19: Primary enriched NK cells pretreated + IL-2 or + IL-15 for 24hrs

were given a two hour pretreatment of anti-NKG2A antibody IPH2201(2ug/ml).

IPH2201 reverses IFN-γ-mediated resistance of MM cell line RPMI-8226 to

primary NK cell-mediated cytotoxicity in IL-2(A, P-value <0.025) and IL-15(B,

P-value<0.025)-activated conditions. Interestingly, reversal of IFN-γ-mediated resistance of MM cell line U266(B) to NK cell killing by IPH2201 was not (Continue on page 56) 55 (Continued from page 55) observed in unstimulated(C) and IL-2-activated(D) conditions. Data represents 1

of 3 donors/experiments with similar trends. Data depicts a standard 51Cr release

assay and error bars depict the standard deviation of technical triplicates.

CD56Bright rather than CD56Dim NK cells exhibit greater killing against MM cells

When compared to healthy PB, NK subpopulations in the PB of MM patients

display an over-representation of CD56BrightCD94+ cells (Figure 20). Our group

has previously observed an abundance of NKG2A/CD94 on the surface of NK

cells from MM patients. As nearly all healthy CD56Bright but only ~50% of the

CD56Dim NK cells express CD94/NKG2A, we separated the two subpopulations

to see if they have a differential response to IPH2201. Impressively, CD56Bright

NK cells (Figure 21, left) exhibit greater cytotoxicity than do CD56Dim NK cells

(Figure 21, Right) against RPMI-8226 cells. Further, IPH2201 treatment of

CD56Bright NK cells (Figure 22) reestablished killing potential against an IFN-γ-

responding MM target.

56 Figure 20: Comparing NK cell subsets in healthy and MM peripheral blood

Figure 20: Peripheral blood mononuclear cells were isolated from three healthy

donors (Leukopacks, Red Cross) and three random MM patients. Surface

expression of prototypic NK cell markers CD56 and CD94 of each was assessed.

+ + Flow cytometric analysis excluding CD3 and CD138 populations in the

PB(excludes T cells and MM PCs , respectively) from three healthy donors (top)

and three MM patients(bottom). Graphs depict NK populations via markers CD56

(Y-axis) and CD94(X-axis). The CD56/CD94 profile which appears uniform in

the healthy donor samples is not represented in the patient samples. Both CD56+

and CD94+ markers are highly expressed and variably over-represented in the

MM samples.

57 Figure 21: Sorted CD56Bright and CD56Dim NK cell cytotoxicity against MM cell line RPMI-8226

*

Figure 21: Healthy primary NK cells were enriched, labeled for CD56

expression and FACS sorted/separated into CD56Bright and CD56Dim subset

populations. Both CD56Bright and CD56Dim NK cells were pretreated for 24 hrs

with IL-2 and their cytotoxic potential was assessed on the target MM cell line

RPMI-8226 + 48hr pretreatment with IFN-γ (10ng/ml). In IL-2 activating

conditions, CD56Bright NK cells exhibit greater cytotoxicity than CD56Dim NK cells towards MM cell line RPMI-8226 ( P-value < 0.001). Of note, both

(Continue on page 59) 58 (Continued from page 58) CD56Bright and CD56Dim NK cells exhibit undetectable amounts of killing toward

RPMI-8226 without stimulation/activation with IL-2 (data not shown). Graph depicts 1 of 3 experiments with similar trends. Data depicts a standard

51Chromium release assay and error bars depict the standard deviation of

technical triplicates.

59 Figure 22: Effect of IPH2201 (Anti-NKG2A) on CD56Bright and CD56Dim NK cytotoxicity against MM cell line RPMI-8226

* **

Figure 22: Two hour pretreatment of activated(IL-2, 24hrs) CD56Bright NK cells

with IPH2201(2ug/ml) significantly enhances cytotoxic potential against

untreated RPMI-8226 (P-value <0.003). In the same activating conditions,

IPH2201 significantly enhances CD56Bright NK cell-mediated cytotoxicity against

IFN-γ-treated RPMI-8226 target (P-value <0.025). The killing capacity of

IPH2201-pretreated CD56Bright NK cells is comparable regardless of target cell

pretreatment. Of interest, killing was successfully measured at E:T ratios less than

1. Killing for the CD56Dim for all conditions was below the threshold of

detection(data not shown).

60 2.3 Discussion and Summary

The body’s systemic cytokine profile skews towards proinflammatory (i.e. IFN-γ) conditions

with aging, a paradox termed inflamm-aging [174]. However, as MM manifests, the cytokine

milieu changes and resembles an immunosuppressive profile [65]. IFN-γ contributes to

differentiation of regulatory T cells, which are an immunosuppressive population often found

in elevated quantities during MM [175]. NK cells have altered and often muted effector

response in MM, and thus we tested if healthy NK cells would similarly respond in a MM co-

culture. Our data extends the observed phenomenon of spontaneous NK cell-mediated IFN-γ

production in the presence of MM cells to a healthy setting. IFN-γ production did not require

cell:cell contact, implying that soluble factors and extracellular vesicles may play a role

(Chapter 3). Although mathematical models have implicated the association and crosstalk of

IFN-γ with the IL-6/STAT3 networks, we are the first to report that IFN-γ alternatively

activates STAT3 in MM cells [164]. These observations taken together imply a mechanism in

early MM disease development that may perpetuate STAT3 activation and aid in the survival

of MM PCs.

We have also demonstrated that MM cells respond to IFN-γ by upregulating inhibitory

ligands on their surface. Gao, et al describe the mechanism of MM resistance to NK-

mediated lysis to be HLA-I-dependent, which may be true; however, it should be noted that

the authors of this study used a method of acid treatment that dissociated the MHC-I:β2M, and likely rendered all MHC-I unrecognizable[124]. Others have proposed that STAT1 is involved in MM cell immune evasion through the PD-1/PD-L1 axis [176]. HLA-E

61 expression has also been implied; however a mechanism for its expression on MM cells was

not determined [177].

HLA-E is classically recognized as an inhibitory ligand, however, a cellular stress

mechanism has also been described that blocks HLA-E’s affinity for NKG2A. This

subversion can occur when heat shock protein 60 (HSP60)-derived peptides compete with

MHC-I leading strand peptides for the HLA-E binding groove [178]. These ambiguities in

HLA-E function, especially under control of pro-inflammatory response element IFN-γ,

required further study. Our data indicate that both basal and induced HLA-E expression and

its inhibitory influence on NK-mediated lysis of RPMI-8226 can be overcome with the novel

anti-NKG2A antibody, IPH2201. A previous report also describes RPMI-8226’s natural resistance to NK-mediated lysis is in part dependent on NKp30 activation [179]. Further, the overexpression of IFN-γ/STAT-affiliated genes contributes to RPMI-8226’s resistance to

both IR and doxorubicin therapies [180]. These collective observations provide a rationale

for combined therapeutic approaches. However, our functional blocking data also implicates

IFN-γ in affording U266 cells an elusive and opportunistic resistance to NK cell

mobilization, independent of HLA-E, TRAIL, or MHC-I expression.

The accepted understanding of NK cell biology and maturation traditionally designates the

CD56Bright NK subset as a less cytotoxic effector to its CD56Dim counterpart. However, we present a scenario in which CD56Bright NK cells exhibit greater killing potential than CD56Dim

NK cells against the MM cell line RPMI-8226. Further, CD56Bright NK cells, even at low

NK:MM ratios, are capable of producing measurable killing while CD56Dim NK cells did not

62 (Figure 22 subtext). Perhaps the over-representation of CD56Bright NK cells in MM is not a consequence of dysregulated NK cell maturation, but rather describes a regulatory albeit incomplete response of NK cells towards malignancy. Our data may describe a previously unrecognized role for CD56Bright NK cells in the natural detection and clearance of MM

tumor cells and their precursors.

2.4 Experimental Methods

Cell lines and NK cell preparations

The multiple myeloma cell lines RPMI 8226 and U266 (ATCC, Manassas, Virginia, USA)

were maintained in normal culturing conditions; in RPMI 1640 media, supplemented with

10% Fetal Bovine Serum, 2mM GLUTIMAX®, and 2mM antibiotic/antimycotic (Life

Technologies, Carlsbad, California, USA). The human IL-2-dependent natural killer cell line

NK-92 (ATCC) was maintained in culture in RPMI 1640 media supplemented with 20%

FBS, 2 mM GLUTIMAX®, 2mM antibiotic/antimycotic, and 150 IU/ml recombinant human

IL-2 (catalog Ro 23-6019, Hoffmann-La Roche, Basel, Switzerland). Primary natural killer

cells were enriched from healthy donor sourced leukocytes (American Red Cross, Columbus,

Ohio, USA) by addition of RosetteSep™ Human NK Enrichment Cocktail (Catalog 15065,

StemCell Technologies, Vancouver, British Columbia, Canada) and density centrifugation on

Ficoll-Paque™PLUS gradient (Catalog 17-440-03, GE Healthcare Life Sciences). Residual

red blood cells were lysed using Ammonium Chloride Solution (Catalog 07850, StemCell

Technologies). In all instances, an enrichment of >75% CD56+ cells was achieved.

63 Treatments

When indicated, primary NK or NK-92 cells were treated with either 150 IU/ml of recombinant human IL-2 or recombinant human IL-15 (Catalog 130-093-955, Miltenyi

Biotec, San Diego, California, USA). MM cell lines were treated with 1ng/ml recombinant human IFN-γ (Catalog RIFNG50, Thermo Scientific) for at least 24 hours. Primary enriched

NK cells were treated with humanized IgG4 Anti-NKG2A antibody (IPH-2201, Innate

Pharma S.A., Marseille, France) or isotype control at a final concentration of 2ug/ml 4 hours prior to and throughout 51Cr release assays.

ELISPOT assays

ELISPOT experiments were conducted using MultiScreen 96-well plates (Millipore) as described previously [181]to measure NK-cell IFN-γ production. Spots were counted using an Immunospot Imaging Analyzer (Cellular Technology).

MTS assay

20,000 cells in 200ul (100,000/ml) were plated in a 96-well round bottom plate at day 0 in

10% FBS supplemented RPMI + 10ng/ml IFN-γ. At 24 and 48 hrs, cells were resuspended by pipette and 100ul transferred to a 96-well flat bottom plate. Subsequently 25ul of MTS agent

(Promega) was added and absorbance readings at 490nm were taken at 30’, 60’ 120’ and

240’. Values reflect the Abs490nm of sample minus that of a media-only control.

64 Transwell co-cultures

Contactless co-culture of MM and NK cells was achieved by placing 0.4 μm pore size

transwell inserts (Catalogs 3472 and 3450, Corning, Tewksbury, Massachusetts, USA) into

the wells of a culture plate. When used, NK-92 cells were washed 2x with PBS and starved

from IL-2 for 24 hours prior to co-culture.

Flow cytometry and sorting

For analysis of multiple myeloma cell surface expression, we used the following antibodies:

HLA-E-PE (Catalog 12-9953, clone 3D12HLA-E, eBioscience, Inc., San Diego, California,

USA), PD-L1-PE (Catalog 12-5983, clone MIH1, eBioscience, Inc.) IFNγR1-PE (Catalog

FAB673P, clone 92101, R&D Systems) IFNγR2-APC (Catalog FAB773, R&D Systems).

Appropriately labeled isotypes were used as controls. All samples were run on an LSRII flow

cytometer (BD Biosciences) and further analyses and graphics were generated using the

FlowJo v7.5.5 Software (FlowJo, LLC, Ashland, Oregon, USA). FACS sorting of CD3-

CD56bright and CD3-CD56dim NK cell populations were performed on a BD FACS ARIA II

system (BD Biosciences). Briefly, primary enriched NK cell preparations were labeled with

CD3-FITC (Catalog 555339, clone HIT3a, BD Biosciences, San Jose, California, USA) and

CD56-APC (Catalog IM2474, clone N901, Beckman Coulter, Pasadena, California, USA) antibodies and Sytox Blue viability dye (Catalog S34587, Life Technologies). Gates were strictly set on distinct target populations, and purity of viable sorted populations was > 98% before proceeding.

65 Real time PCR

Cells from each condition were washed 2x with PBS before lysis in P1 Buffer from the

Qiagen RNeasy (Qiagen, Valencia, California, USA) and RNA processing was carried out

according to manufacturer’s instructions. RNA was quantified using a NanoDrop 2000

(Thermo Fisher Scientific, Waltham, Massachusetts, USA) instrument. Samples with

acceptable Abs260/280 and Abs260/230 values > 1.9 were used for further analysis. For each sample, cDNA was generated from 1ug RNA template with M-MLV Reverse Transcriptase reagents (Catalog 28025, Thermo Fisher Scientific) and 10ng cDNA template was loaded uniformly for each real-time PCR reaction. Both HLA-E and PD-L1 primer/probes were

purchased as TaqMan® gene expression Assays (Catalog Hs03045171_m1 and

Hs01125301_m1, Life Technologies, Grand Island, New York, USA) and ribosomal 18s was

used as a housekeeping gene to normalize results. Real-Time PCR was carried out on an

ABI7900HT system with fast cycling conditions standard for the TaqMan® FAST Advanced

Reaction mix (Catalog 4444556, Life Technologies) and after threshold and baseline was

established using SDS 3.0.1 Software (Life Technologies), expression was determined by the

2-ΔΔCT method of calculation. Values represent the mean of technical triplicates + SEM.

Western blot analysis

Cells were harvested, washed once with ice-cold PBS, and lysed (108 cells/ml) in RIPA

buffer: 0.15 M NaCl, 1% Nonidet P-40, 0.1% SDS, 50 mM Tris (pH 8.0), supplemented with

Halt™ Protease and Phosphatase Inhibitor Cocktail (Catalog 78440, Life Technologies) as

described [182].Western blotting was performed according to previously published protocols

[183] and Ab-reactive proteins were detected with HRP-labeled donkey anti-rabbit (Catalog

66 NA934V, GE Healthcare Life Sciences, Pittsburgh Pennsylvania, USA) and sheep anti-

mouse antibodies (Catalog NA931V, GE Healthcare Life Sciences), and visualized using

ECL western blotting substrate (Catalog RPN2106, GE Healthcare Life Sciences). Proteins

were analyzed in 4–15% SDS-PAGE or any kD TGX ready-gels (Catalogs 456-1084 and

456-9034, Bio-Rad Laboratories, Hercules California, USA) using reducing conditions.

Primary antibodies used include Phospho-Stat1Tyr701 (Catalog #9171), Phospho-Stat2Tyr690

(Catalog #4441), Phospho-Stat3Tyr705 (Catalog #9145, Clone D3A7), Stat1 (Catalog #9172), and Stat3 (Catalog #9132, all Technologies, Danvers Massachusetts, USA).

ELISA assays

To quantify the amount of human IFN-γ secreted by NK cells, an ELISA using capture mAb

(Catalog M700a, clone 2G1, Thermo Fisher Scientific) and detection biotin-conjugated mAb

(Catalog M701B, clone B133.5, Thermo Fisher Scientific) was performed as previously described in [184]. Graphed values represent the mean of technical triplicates + standard

deviation.

Cytotoxicity assays

Natural cytotoxicity was assessed by standard 51Chromium release assay as previously described [69]. Briefly, effector cells treated or stimulated for 4 hours, 12 hours, or overnight, as indicated, were plated and serially diluted. Target cells were labeled with 50uCi 51Cr for

75 minutes, washed repeatedly and plated. Cell-free supernatants were harvested after a 4-

hour incubation and transferred to scintillation vials and radioactivity was counted (CPM) on

an LS6500 Scintillation Counter (Beckman Coulter). A positive control of 5% SDS detergent

67 (+ 51Cr-labeled target cells) for maximum 51Cr release and a negative control of media-alone

(+ 51Cr-labeled target cells) for spontaneous 51Cr release were included. The %51Cr release in

each sample was directly proportional to the % of target cells lysed. Percentage of targets

lysed was calculated by (CPMsample – CPMspontaneous)/(CPMmaximum -CPMspontaneous) x 100.

Graphed values represent the mean of technical triplicates + Standard Deviation.

68 CHAPTER 3: MULTIPLE MYELOMA CELL-DERIVED EXTRACELLULAR VESICLES ENHANCE IFN-GAMMA PRODUCTION BY NATURAL KILLER CELLS

3.1 Introduction

Multiple myeloma (MM) is an incurable hematologic manifestation of the bone marrow

(BM). After homing and adhesion to bone marrow stromal cells (BMSC), malignant plasma

cell (PC) survival is largely dependent on and growth factors (i.e. IL-6, IL-10) provided by

the bone marrow microenvironment (BMME). Although MM cell communication in the

BMME has been demonstrated to include cell:cell contact and the exchange of soluble

factors, contemporary reports are beginning to describe the various roles of extracellular

vesicles (EV) in MM progression.

EV are membrane-encapsulated organelle-free subcellular material containing mRNA, DNA,

protein, and are capable of presenting antigen and delivery of contents to cell-specific targets

[78, 81]. Upon their discovery in the 1940s, EV were regarded as cellular debris. Although

detected in a variety of settings, including viral infection and tumor release, EV functionality

remained elusive until 1996 when Raposo, et al discovered that EV isolated from EBV-

transformed B cells were antigenic [77]. The report that figuratively opened the flood gates to current EV studies in cancer described how a mutant oncogenic receptor (EGFRVIII) on EV

69 could be transferred to recipient cells and initiate their malignant transformation [185]. With

regard to MM, new studies implicate EV functionality in promoting angiogenesis, driving

osteoclast differentiation, and activating myeloid-derived suppressor cells in the BMME

[186-188]. Interestingly, BMSC EV have been reported to promote MM progression while providing drug resistance to the protease inhibitor bortezomib [189]. The complexity and elegance of EV biology is only beginning to be understood and appreciated.

Our findings implicate MM EV as potential mediators of the natural killer (NK) cell pro- inflammatory response. Upon treatment with MM EV, NK cells show enhanced IFN-γ secretion. In reciprocation, IFN-γ induces the expression of inhibitory ligands HLA-E and

PD-L1 on the surface of MM cells (2.2 , Figure 11, Figure 12). IFN-γ-treated MM cells

release EV bearing MHC-I, HLA-E and PD-L1, which may possess inhibitory potential to

their cognate ligands on both NK cells and T cells of the innate and adaptive immune

systems, respectively. Altogether, our findings may describe a previously unrecognized

mechanism of immune evasion both initiated and maintained by MM EV.

3.2 Results and Figures

MM EV enhance NK cell-mediated IFN-γ production

Previously, our lab observed that spontaneous release of IFN-γ by NK cells in co-culture with

MM cells does not require contact (Figure 6, Chapter 2). The transwell membrane used in

that assay had a pore size of 400nm, which is roughly the average diameter of reported EV

sizes and includes all exosomes (50-100nm) but restricts some microvesicles (100-1000nm).

In lieu of the many soluble factors present in the co-culture, we focused our efforts on the

70 potential contribution of MM EV to the observed effect. We adopted a differential centrifugation protocol originally described by Thery, et al (Figure 23, A) and validated the

MM EV prep fractions by flow cytometry to ensure optimal yield (Figure 23, B) [82].

Functionally, MM cell-derived EV were sufficient to enhance NK cell-mediated IFN-γ production in IL-2-activated conditions (Figure 24).

71 Figure 23: Extracellular vesicle isolation and prep validation A.

B.

Figure 23: The protocol for isolating EV by differential ultracentrifugation (A)

including the spin forces and pellet contents. Briefly, cells are spun out and

discarded with a 300 x g (x gravitational force). Subsequently, apoptotic bodies

and cellular debris including organelle-containing vesicles is precipitated and

discarded in the 2000 x g and 10,000 x g spins. Isolation of EV occurs with

100,000 x g spin, when pellets are resuspended, washed, and pooled into a single

prep tube to precipitate and uniformly distribute for multiple readouts/treatments.

Visualization of EV preparation (B) from the MM cell line U266 on an LSRII (Continue on page 73) 72 (Continued from page 72) flow cytometer using sizing gates generated from standardized polystyrene beads of known sizes (100 nm, 200nm, 1000nm, and 2000nm in diameter). Note that

PBS, EV-Free Media, and EV Wash Step serve as control samples to validate prep purity and yield. Of note, our diluted EV prep generated 108,940 events in

30 seconds whereas the other fractions (PBS alone, EV-Free Media, and EV wash step) all generated <9000 events.

73 Figure 24: MM cell EV enhance NK cell IFN-γ production

A. * B. *

Figure 24: NK-92 cells were starved for 24 hours from growth factor IL-2 prior

to a 24hr re-stimulation of IL-2 + EV isolated from MM cell lines U266(A) and

RPMI-8226(B). In IL-2 NK cell-activated conditions, both U266 (A, P-value

=0.044) and RPMI-8226 (B, P-value<0.01) cell line-derived EV significantly

enhanced NK-92 cell IFN-γ production vs EV-free media control. Cell free

supernatants were harvested and used in an ELISA for hIFN-γ detection. Error

bars represent the standard deviation of technical replicates. Ratios represent the

EV produced by X number of MM cells (calculated by known number of cells

from which the EV were derived and the known volume of EV prep).

74 MM EV contain HLA-E and PD-L1 Inhibitory Ligands

We were the first to characterize the protein content of EV from MM cell lines and reported

an abundance of MHC-I within the EV [190]. We isolated EV from the PB and BM of MM

patients and healthy donors (PB only) and observed that their EV contained a full range of

detectable proteins ranging from 12~200 kDa (Figure 25, A). Further, MHC- I and β2M were elevated in the EV from MM but not from healthy PB (Figure 25, B). The mass spectrometry profile of identified MHC-1/HLA-I spectral hits in Harshman, et al. (Figure 26, A) showed a single identified unique peptide signature for the inhibitory ligand HLA-E (Figure 26, B)

[190]. Paired with our data regarding the upregulation of HLA-E and PD-L1 expression by

IFN-γ, we assessed if either inhibitory ligand was present in the EV and if increased cellular

expression of either would be translated to the vesicles. Indeed, EV derived from the MM

cell line RPMI-8226 contained both HLA-E and PD-L1 proteins (Figure 27). Further,

treatment of RPMI-8226 cells with IFN-γ increased the amount of detectable HLA-E and

PD-L1 in the EV fraction (Figure 27, right).

75 Figure 25: Protein profile of EV released from MM cell lines and patient

sera

Figure 25: PonceauS staining of EV proteins on a nitrocellulose membrane (A) reveals proteins of many sizes in healthy PB (lanes 2-4), MM PB (5-10) and MM

BM (Lane 11) when compared to MW ladder (lane 1). Western blot analysis of

MHC-I and β2M (B) describes increased expression in the MM samples, but not in the healthy PB. Impressive expression of both MHC-I and β2M was observed

in the MM BM sample, but the study lacks a healthy BM sample for baseline

comparison. Due to biological variability, the uniform signal from PonceauS

(total protein) staining serves as the loading control.

76 Figure 26: HLA-E detected on mass spectrometry study

Figure 26: All HLA unique spectral identities and heat map of expression in MM

cell lines and their EV (A) imply enrichment of HLA in the EV. A single

spectrum count for MHC-Ib member and inhibitory ligand HLA-E was also

detected (B).

Data was adapted from [190] with permission from the authors: Harshman, S. W., Canella, A., Ciarlariello, P. D., Rocci, A., Agarwal, K., Smith, E. M., Talabere, T., Efebera, Y. A., Hofmeister, C. C., Benson, D. M., Paulaitis, M. E., Freitas, M. A. and Pichiorri, F. (2013), Characterization of multiple myeloma vesicles by label-free relative quantitation. Proteomics, 13: 3013–3029. doi:10.1002/pmic.201300142 77 Figure 27: HLA-E and PD-L1 in EV released from MM

Figure 27: MM cell line RPMI-8226 was either placed in normal culture

conditions or starved from serum for 48hrs + IFN-γ (10ng/ml). Serum-starvation

is required due to the possibility of cross contamination of vesicles naturally

present in the serum. Whole cell lysates and EV isolated from the serum-starved

cells were assessed for inhibitory ligands HLA-E and PD-L1, as well as Lamin

B1 (nuclear protein absent in EV) and GAPDH (loading control). Both HLA-E

and PD-L1 proteins are detected on the EV fractions, and enhancement of their

signals are observed when RPMI-8226 were pretreated with IFN-γ) Note the

enrichment of PD-L1 in the EV when compared to the global cell lysates.

78 MM EV present ligands on their surface

To determine the orientation of membrane proteins of the EV, we chose to fluorescently

label RPMI-8226 EV preps with a pan-HLA-I antibody and visualized the EV using the

NanoSight NS300 particle tracking system (Figure 28). Detected EV particles in the fluorescent channel for both untreated(Figure 28, top right) and IFN-γ-treated conditions(Figure 28, bottom right) imply orientation of surface ligands HLA-ABC, PD-L1

and HLA-E on MM EV are likely presenting outward, and have the capacity to engage their cognate ligands on recipient cells.

79 Figure 28: NanoSight NS300 visualization of MM EV and HLA-ABC presentation

Figure 28 Simultaneous visualization, quantification and size distribution of

RPMI-8226 EV in untreated (top left) and IFN-γ-treated (10 ng/ml, bottom left) conditions. Fluorescently labeled MHC-I (HLA-ABC)-bearing EV are detected in both in untreated (top right) and IFN-γ-treated (10 ng/ml, bottom right) conditions.

80 3.3 Discussion

In this study, we demonstrated that we can reproducibly isolate EV from MM cell lines and patient sera and further study their contents, properties of extracellular-presentation and biological functionality. Further, our data implicate a previously unrecognized form of communication between MM PC and the proximal BMME and may extend systemically. Of note, PD-L1 appears to be selectively loaded on the EV of MM for release (Figure 21). These data indicate that circulating EV may saturate the PB with inhibitory messaging specific to

NK and T cell responses. We attempted, extensively, to capture the potential inhibitory effect of HLA-E and PD-L1-bearing vesicles on NK cell cytoxicity but could not reproducibly confirm either inhibitory or activatory influence (Data not shown). Conditions attempted are summarized in Table 3. We speculate that the over-abundance of HLA-ABC represented in the EV may out-compete the conveyance of potential inhibitory signaling provided by HLA-E and PD-L1. Further, paracrine and autrocrine factors in vivo may contribute to the specific cargo and surface landscape of the MM EV, thus an assessment of

MM EV and NK cell cytotoxic influence in an autologous setting would best resolve these inquiries.

81 Table 3: Attempted Primary NK Cell Cytotoxicity Experiments Involving MM EV 82

Our data also present a counter-intuitive observation in that EV laden with known NK and T cell inhibitory receptors were directly able to enhance the NK cell cytokine response.

Although demonstrated in an allogeneic system with EV from cell lines, our previous data showing the spontaneous release of IFN-γ by autologous NK cells in culture with MM PC suggests the phenomenon may be physiologically relevant. Limited quantities of EV source material (i.e. patient blood and bone marrow) and availability of autologously paired samples will remain the biggest barriers to overcome for progress in future studies.

Interestingly, our data supports a multifunctional role for EV in MM. Serum starvation of

RPMI-8226 increases both the output and angiogenic potential (via delivery of miR-135b) of its EV [186, 191]. We may be describing a phenomenon whereby MM cells release EV to simultaneously subvert NK cell recognition and induce IFN-γ release for beneficial ECM remodeling in the BMME.

3.4 Methods

Cell lines and NK cell preparations

The multiple myeloma cell lines RPMI 8226 and U266 (ATCC, Manassas, Virginia, USA) were maintained in RPMI 1640 media, supplemented with 10% Fetal Bovine Serum,

2mM GLUTIMAX®, and 2mM antibiotic/antimycotic (All Life Technologies). The human

IL-2-dependent natural killer cell line NK-92 (ATCC) was maintained in culture in RPMI

1640 media supplemented with 20% FBS 2 mM GLUTIMAX®, 2mM antibiotic/antimycotic,

83 and 150 IU/ml recombinant human IL-2 (catalog Ro 23-6019, Hoffmann-La Roche, Basel,

Switzerland). Primary natural killer cells were enriched from healthy donor sourced

leukocytes (American Red Cross, Columbus, Ohio, USA) by addition of RosetteSep™

Human NK Enrichment Cocktail (Catalog 15065, StemCell Technologies, Vancouver,

British Columbia, Canada) and density centrifugation on Ficoll-Paque™PLUS gradient

(Catalog 17-440-03, GE Healthcare Life Sciences). Residual red blood cells were lysed using

Ammonium Chloride Solution (Catalog 07850, StemCell Technologies). In all instances, an

enrichment of >75% CD56+ cells was achieved.

Treatments

NK-92 cells were treated with 150 IU/ml of recombinant human IL-2 in the presence of EV derived from MM cell lines U266 and RPMI-8226 for 24hrs. Extracellular vesicle dose ratios represent the amount of EV generated by the number of MM cells to the number of NK cells plated. A cell’s amount of EV is directly proportional to the known concentration of cells plated and the harvested volume. EV treatments ranged from 4 to 24 hours as indicated for each assay.

Extracellular vesicle isolation

Isolation of cell line-derived extracellular vesicles has been previously described by Théry, et al [82]. and performed in Harshman, et al [190]. Concisely, differential centrifugation was used to isolate cells (300 x g), cellular debris (2000 x g), and apoptotic bodies (10,000 x g) for removal, and the remaining supernatant was used to pellet EVs (100,000 x g). EV pellets

84 were washed/pooled with PBS and centrifuged one more time (100,000 x g), after which

supernatants were discarded and EV pellets resuspended in application-dependent buffer.

Flow cytometry

Visualization of purified EV preps was carried out on an LSR II flow Cytometer (BD

Biosciences, San Jose, California, USA). Logarithmic scaling for both FSC and SSC

channels was required for proper separation of microparticles. To compare the contents of

each fraction of EV prep, uniform acquisition of all events for a set 30” period was used. To

determine the size distribution of EV present, EV acquisition events were compared to

standardized polystyrene beads of known 0.1 um, 0.2 um, 1.0 um and 2.0 um in diameters

(Spherotech, Lake forest, Illinois, USA).

EV surface staining and detection by Nanosight

EV surface labeling with antibodies of HLA-ABC (Catalog 561349, clone G46-2.6, BD

Pharmingen) and isotype control (Catalog 557872, clone MOPC-21, BD Pharmingen)

conjugated to PE-Cy7 fluorochrome. For each sample, EVs were prepared from a starting

volume of 107 cells’ worth of vesicles. EV pellets were resuspended in a small volume of

PBS before equal distribution for antibody labeling and placed on ice for 30 mins. PBS was

added to block the antibody to a dilution of 200x the labeling volume (10 ml wash added to

50ul labeling volume), mixed thoroughly by inversion and vortexing before being loaded into the Nanosight Syringe Pump for automated injection and analysis with the NS300 system

(Malvern Instruments Ltd, Malvern, UK) fitted with a laser diode of 532 nm. To detect PE-

Cy7 fluorescently labeled EVs, the camera channel was switched to detection of long pass

85 570nm filter. Both total EV and fluorescent EV populations were quantified and size distribution profile determined within the same tube by consecutive 5 x 60” video series recordings at 60 frames per second throughout an automated and continuous sample flow stream. All graphics and analyses were processed using Nanoparticle Tracking Analysis

Software 3.0 (Malvern Instruments Ltd).

ELISA assays

To quantify the amount of human IFN-γ released by NK cells, an ELISA using capture mAb

(Catalog M700a, clone 2G1, Thermo Fisher Scientific) and a biotin-conjugated detection mAb (Catalog M701B, clone B133.5, Thermo Fisher Scientific) previously described in

(source) was used. Graphed values represent the means of technical triplicates + standard deviation. Extracellular vesicle dose ratios represent the amount of EV generated by the number of MM cells to the number of NK cells plated. A cell’s amount of EV is directly proportional to the known concentration of cells plated and the harvested volume. EV treatments ranged from 4 to 24 hours as indicated for each assay.

Western blot analysis

Cells or EV were harvested, washed once with ice-cold PBS, and lysed (108 cells/ml or 25ul total per EV pellet) in RIPA buffer: 0.15 M NaCl, 1% Nonidet P-40, 0.1% SDS, 50 mM Tris

(pH 8.0), supplemented with Halt™ Protease and Phosphatase Inhibitor Cocktail (Catalog

78440, Life Technologies) as described [182]. Western blotting was performed according to previously published protocols [183], and Ab-reactive proteins were detected with HRP- labeled donkey anti-rabbit (Catalog NA934V, GE Healthcare Life Sciences, Pittsburgh

86 Pennsylvania, USA) and sheep anti-mouse (Catalog NA931V, GE Healthcare Life Sciences)

and ECL (Catalog RPN2106, GE Healthcare Life Sciences). Proteins were analyzed in 4–

15% SDS-PAGE or any kD TGX ready-gels (Catalogs 456-1084 and 456-9034, Bio-Rad

Laboratories, Hercules California, USA) using reducing conditions. Primary antibodies used includeGAPDH (Catalog ab8245, Abcam, Cambridge Massachusetts, USA), PD-L1 (Catalog

ab174838, Abcam), and HLA-E (Catalog ab2216, Abcam), and Lamin B1(Abcam).

87 CHAPTER 4: FUTURE STUDIES AND EXTENDED DISCUSSION

4.1 The mechanism for IFN-γ-mediated resistance of U266 to NK cell cytotoxicity

We have shown that the MM cell line U266 cells resist NK cell-mediated natural cytotoxicity upon treatment with IFN-γ (Figure 15 B and C, Figure 16) and that the mechanism does not involve HLA-E (Figure 19, C and D), MHC-I (data not shown) over-expression, and

CD107A (data not shown). Further, healthy primary NK cells express little, if any, PD-1 on their surface, supporting that PD-L1 is likely not involved in the mechanism of action [192,

193]. We hypothesize that the mechanism of resistance is mediated by cell surface ligands which are up-regulated and present in response to IFN-γ. To refine the candidates mediating this phenomenon, I plan to prepare surface-protein extracts from U266 cells + IFN-γ and perform a 2D Western blot, first separating the proteins by isoelectric focusing, followed by a second separation by size. Our hope is that the differential protein profile will provide three observable groups when blots are overlaid – unchanged with treatment, up-regulated with

IFN-γ treatment and down-regulated with IFN-γ treatment. Candidate surface protein signals up-regulated by IFN-γ will be identified using mass spectrometry, and their contribution to

IFN-γ-mediated resistance will be validated using neutralizing antibodies (if available).

Moreover, our proposed method may lead to the discovery of a novel surface protein, or implicate a novel role for an already identified protein in MM cell resistance to NK-mediated

88 cytoxicity. Of course, we would likely extend this study to EV lysates to see if any candidate

inhibitory ligands are released either spontaneously or in response to IFN-γ.

4.2 IFN-γ treatment of MM cells to enhance efficacy of proteasome inhibitors

Long term exposure of MM cells to IFN-γ decreases their proliferation [194, 195]. Cell cycling is halted while the immune system assesses MM cells for stress and potentially antigenic endogenous peptides. We observed that our MM cells robustly respond to short term IFN-γ treatment by newly synthesizing proteins with little effect on proliferation capacity. Further, treatment of MM cells with the irreversible proteasome inhibitor bortezomib down-regulates their MHC-I expression and increases their susceptibility to NK cell-mediated lysis [196]. We hypothesize that the pretreatment of MM cells with IFN-γ and subsequent treatment with either proteasome inhibitor bortezomib or carfilzomib may enhance their susceptibility to direct and indirect (NK-cell mediated) cytotoxicity. These data may indicate that we can abrogate the otherwise advantageous response of MM cells to IFN-

γ; however, the therapeutic translation for this data may be moot in that IFN-γ is not a tolerable treatment in the clinical setting, and is associated with a variety of flu-like symptoms including , headache, and diarrhea.

4.3 Differential peptide cross-presentation in MM following IFN-γ treatment

Our understanding of HLA-E peptide presentation is still evolving. Classically, HLA-E presents leading strand peptides of MHC-1a. Recently, a newly discovered peptide repertoire of which all members are capable of presentation via HLA-E has been reported [197]. This peptidome, consisting of 36 peptides ranging from 7-16 aa in length, represents a paradigm

89 shift in HLA-E functionality. Even more bizarre is the discovery that the proteasome during

protein degradation can splice peptides whose product sequences do not seem to reflect

genomically possible combinations [198]. Although this process was not studied in the

myeloma cell per se, it hints toward the possibility of identifying specific and novel peptide

sequences within the MM peptidome. These peptides may serve as biomarkers, or better yet,

novel therapeutic targets. I propose to characterize the peptides presented on MM cells +

IFN-γ treatment. The method uses an acid wash which detaches β2M from all MHC proteins,

and therefore the pool of peptides will represent all MHC (Ia, Ib, and II)-bound peptides.

Peptides will be separated by size via HPLC and sequenced using mass spectrometry.

Perhaps the most informative data from this proposed study would be to assess if the peptides

loaded upon MHC molecules within the vesicles, show a difference in distribution and

character. This may highlight a selective mechanism in MM cells in order to release

unfavorable peptides in EV while maintaining a less suspect cell surface.

4.4 Concluding remarks

In this thesis, for the first time MM, we have provided evidence of an alternative role of IFN-

γ in activating STAT1 and STAT3 which facilitates the increased surface expression of PD-

L1 and HLA-E, and their eventual incorporation and release on extracellular vesicles. If anything, the phenomenon described herein should provide considerations towards cancer therapies and their targets. To elaborate, if an MM patient’s system is saturated with HLA-E and PD-L1-bearing vesicles, then therapeutic dosing would likely need reconsideration to account for the non-active binding of these agents. Further, this report provides evidence that perhaps targeting the inhibitory receptors towards these ligands may be more effective.

90 We also present a potential and previously unrecognized role for CD56Bright NK cells as potent cytotoxic effector cells towards an MM target, perhaps alluding to a regulatory mechanism of NK cells in detecting early malignant transformation.

Lastly, we describe a reciprocating communication between NK cells and Myeloma cells summarized in Figure 29, which is previously unreported, and requires more investigation to determine the extent of the biological functionality of MM EV on the innate immune system.

91 Figure 29: Graphical Summary of Dissertation Work

Figure 29 (Continue on page 93) 92 (Continued from page 92)

Figure 29: Graphical Summary of Dissertation Work

We observed spontaneous IFN-γ production by NK cells when co-cultured with

MM PC (Figure 5) , the effect does not require cell:cell contact (Figure 6), and

later discover that MM-derived EV were sufficient to enhance NK cell IFN-γ

release (Figure 24). These observations may describe a perpetuating cycle in

which MM cells benefit from the constant release of IFN-γ and other associated

factors. Further, MM Cells have functional IFN- γRs (Figure 7) which signal

through STAT1 and STAT3 (Figure 8) to increase expression of many

genes/proteins including MHC-I (Figure 10), and inhibitory ligands HLA-E

(Figure 11,Figure 13) and PD-L1 (Figure 12,Figure 13). Treatment of MM cells

with IFN-γ allows measurable protection from NK cell mediated lysis (Figure 16,

Figure 17, Figure 18), which is partially dependent by HLA-E:NKG2A

interaction in the RPMI-8226 cell line (Figure 19,Figure 22), but not for U266

(Figure 19). Myeloma cells also respond to IFN-γ releasing vesicles with

membrane bound HLA-E and PD-L1 (Figure 26, Figure 27, Figure 28) whose

biological functionality are subject to future studies.

93 Bibliography

1. Raab, M.S., et al., Multiple myeloma. Lancet, 2009. 374(9686): p. 324-39. 2. Palumbo, A. and K. Anderson, Multiple myeloma. N Engl J Med, 2011. 364(11): p. 1046-60. 3. Macintyre, W., Case of Mollities and Fragilitas Ossium, accompanied with urine strongly charged with animal matter. Med Chir Trans, 1850. 33: p. 211-32. 4. Von Rustizky, J., Multiples Myelom. Deutsche Zeitschrift Chir (Berlin), 1873. 3: p. 162-72. 5. Kyle, R.A., et al., Monoclonal gammopathy of undetermined significance (MGUS) and smoldering (asymptomatic) multiple myeloma: IMWG consensus perspectives risk factors for progression and guidelines for monitoring and management. Leukemia, 2010. 24(6): p. 1121-7. 6. Ridley, R.C., et al., Expression of syndecan regulates human myeloma plasma cell adhesion to type I collagen. Blood, 1993. 81(3): p. 767-74. 7. Jackson, N., et al., An analysis of myeloma plasma cell phenotype using antibodies defined at the IIIrd International Workshop on Human Leucocyte Differentiation . Clin Exp Immunol, 1988. 72(3): p. 351-6. 8. Hsi, E.D., et al., CS1, a potential new therapeutic antibody target for the treatment of multiple myeloma. Clin Cancer Res, 2008. 14(9): p. 2775-84. 9. Bataille, R., et al., The phenotype of normal, reactive and malignant plasma cells. Identification of "many and multiple myelomas" and of new targets for myeloma therapy. Haematologica, 2006. 91(9): p. 1234-40. 10. Attaelmannan, M. and S.S. Levinson, Understanding and identifying monoclonal gammopathies. Clin Chem, 2000. 46(8 Pt 2): p. 1230-8. 11. Katzmann, J.A., et al., Serum reference intervals and diagnostic ranges for free kappa and free lambda immunoglobulin light chains: relative sensitivity for detection of monoclonal light chains. Clin Chem, 2002. 48(9): p. 1437-44. 12. Kyle, R.A., et al., Review of 1027 patients with newly diagnosed multiple myeloma. Mayo Clin Proc, 2003. 78(1): p. 21-33. 13. Pandey, S. and R.A. Kyle, Unusual myelomas: a review of IgD and IgE variants. Oncology (Williston Park), 2013. 27(8): p. 798-803. 14. Smadja, N.V., et al., Chromosomal analysis in multiple myeloma: cytogenetic evidence of two different diseases. Leukemia, 1998. 12(6): p. 960-9. 15. Debes-Marun, C.S., et al., Chromosome abnormalities clustering and its implications for pathogenesis and prognosis in myeloma. Leukemia, 2003. 17(2): p. 427-36. 16. Fonseca, R., et al., The recurrent IgH translocations are highly associated with nonhyperdiploid variant multiple myeloma. Blood, 2003. 102(7): p. 2562-7. 17. Segges, P. and E. Braggio, Genetic markers used for risk stratification in multiple myeloma. Genet Res Int, 2011. 2011: p. 798089.

94 18. Corre, J., N. Munshi, and H. Avet-Loiseau, Genetics of multiple myeloma: another heterogeneity level? Blood, 2015. 125(12): p. 1870-6. 19. Landgren, O., et al., Monoclonal gammopathy of undetermined significance (MGUS) consistently precedes multiple myeloma: a prospective study. Blood, 2009. 113(22): p. 5412-7. 20. Weiss, B.M., et al., A monoclonal gammopathy precedes multiple myeloma in most patients. Blood, 2009. 113(22): p. 5418-22. 21. Kyle, R.A., Monoclonal gammopathy of undetermined significance. Natural history in 241 cases. Am J Med, 1978. 64(5): p. 814-26. 22. Berenson, J.R., et al., A retrospective study to evaluate the work-up and follow-up of patients with monoclonal gammopathy of undetermined significance. Clin Lymphoma Myeloma Leuk, 2011. 11(4): p. 336-41. 23. Bianchi, G. and I.M. Ghobrial, Does my patient with a serum monoclonal spike have multiple myeloma? Hematol Oncol Clin North Am, 2012. 26(2): p. 383-93, ix. 24. Sanchorawala, V., Light-chain (AL) amyloidosis: diagnosis and treatment. Clin J Am Soc Nephrol, 2006. 1(6): p. 1331-41. 25. Kyle, R.A., et al., A long-term study of prognosis in monoclonal gammopathy of undetermined significance. N Engl J Med, 2002. 346(8): p. 564-9. 26. Korde, N., S.Y. Kristinsson, and O. Landgren, Monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM): novel biological insights and development of early treatment strategies. Blood, 2011. 117(21): p. 5573-81. 27. Kyle, R.A. and P.R. Greipp, Smoldering multiple myeloma. N Engl J Med, 1980. 302(24): p. 1347-9. 28. Kyle, R.A., et al., Clinical course and prognosis of smoldering (asymptomatic) multiple myeloma. N Engl J Med, 2007. 356(25): p. 2582-90. 29. Rajkumar, S.V., et al., International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol, 2014. 15(12): p. e538-48. 30. Kastritis, E., et al., Extensive bone marrow infiltration and abnormal free light chain ratio identifies patients with asymptomatic myeloma at high risk for progression to symptomatic disease. Leukemia, 2013. 27(4): p. 947-53. 31. Rajkumar, S.V., O. Landgren, and M.V. Mateos, Smoldering multiple myeloma. Blood, 2015. 125(20): p. 3069-75. 32. Busslinger, M., Transcriptional control of early B cell development. Annu Rev Immunol, 2004. 22: p. 55-79. 33. Mårtensson, I.L. and R. Ceredig, Review article: role of the surrogate light chain and the pre-B-cell receptor in mouse B-cell development. Immunology, 2000. 101(4): p. 435-41. 34. Hardy, R.R. and K. Hayakawa, B cell development pathways. Annu Rev Immunol, 2001. 19: p. 595-621. 35. Anolik, J., et al., Down-regulation of CD20 on B cells upon CD40 activation. Eur J Immunol, 2003. 33(9): p. 2398-409. 36. Oettinger, M.A., et al., RAG-1 and RAG-2, adjacent genes that synergistically activate V(D)J recombination. Science, 1990. 248(4962): p. 1517-23. 37. Volpe, J.M. and T.B. Kepler, Large-scale analysis of human heavy chain V(D)J recombination patterns. Immunome Res, 2008. 4: p. 3.

95 38. Doñate, C., et al., Homing of human B cells to lymphoid organs and B-cell lymphoma engraftment are controlled by cell adhesion molecule JAM-C. Cancer Res, 2013. 73(2): p. 640-51. 39. Hargreaves, D.C., et al., A coordinated change in chemokine responsiveness guides plasma cell movements. J Exp Med, 2001. 194(1): p. 45-56. 40. Gunn, M.D., et al., A B-cell-homing chemokine made in lymphoid follicles activates Burkitt's lymphoma receptor-1. Nature, 1998. 391(6669): p. 799-803. 41. Crotty, S., A brief history of T cell help to B cells. Nat Rev Immunol, 2015. 15(3): p. 185-9. 42. Ding, B.B., et al., IL-21 and CD40L synergistically promote plasma cell differentiation through upregulation of Blimp-1 in human B cells. J Immunol, 2013. 190(4): p. 1827-36. 43. Nera, K.P., et al., Loss of Pax5 promotes plasma cell differentiation. Immunity, 2006. 24(3): p. 283-93. 44. Shapiro-Shelef, M., et al., Blimp-1 is required for the formation of immunoglobulin secreting plasma cells and pre-plasma memory B cells. Immunity, 2003. 19(4): p. 607-20. 45. Klein, U., et al., Transcription factor IRF4 controls plasma cell differentiation and class-switch recombination. Nat Immunol, 2006. 7(7): p. 773-82. 46. Shaffer, A.L., et al., XBP1, downstream of Blimp-1, expands the secretory apparatus and other organelles, and increases protein synthesis in plasma cell differentiation. Immunity, 2004. 21(1): p. 81-93. 47. Reimold, A.M., et al., Plasma cell differentiation requires the transcription factor XBP-1. Nature, 2001. 412(6844): p. 300-7. 48. Bergsagel, P.L., et al., Cyclin D dysregulation: an early and unifying pathogenic event in multiple myeloma. Blood, 2005. 106(1): p. 296-303. 49. Bergsagel, P.L. and W.M. Kuehl, Chromosome translocations in multiple myeloma. Oncogene, 2001. 20(40): p. 5611-22. 50. Manier, S., et al., Bone marrow microenvironment in multiple myeloma progression. J Biomed Biotechnol, 2012. 2012: p. 157496. 51. Bleul, C.C., et al., A highly efficacious lymphocyte chemoattractant, stromal cell- derived factor 1 (SDF-1). J Exp Med, 1996. 184(3): p. 1101-9. 52. Nie, Y., et al., The role of CXCR4 in maintaining peripheral B cell compartments and humoral immunity. J Exp Med, 2004. 200(9): p. 1145-56. 53. Koda, J.E., A. Rapraeger, and M. Bernfield, Heparan sulfate proteoglycans from mouse mammary epithelial cells. Cell surface proteoglycan as a receptor for interstitial collagens. J Biol Chem, 1985. 260(13): p. 8157-62. 54. Saunders, S. and M. Bernfield, Cell surface proteoglycan binds mouse mammary epithelial cells to fibronectin and behaves as a receptor for interstitial matrix. J Cell Biol, 1988. 106(2): p. 423-30. 55. Stepp, M.A., et al., Syndecan-1 regulates cell migration and fibronectin fibril assembly. Exp Cell Res, 2010. 316(14): p. 2322-39. 56. Sanz-Rodríguez, F., A. Hidalgo, and J. Teixidó, Chemokine stromal cell-derived factor-1alpha modulates VLA-4 integrin-mediated multiple myeloma cell adhesion to CS-1/fibronectin and VCAM-1. Blood, 2001. 97(2): p. 346-51.

96 57. Dankbar, B., et al., Vascular endothelial growth factor and interleukin-6 in paracrine tumor-stromal cell interactions in multiple myeloma. Blood, 2000. 95(8): p. 2630-6. 58. Gupta, D., et al., Adherence of multiple myeloma cells to bone marrow stromal cells upregulates vascular endothelial growth factor secretion: therapeutic applications. Leukemia, 2001. 15(12): p. 1950-61. 59. Podar, K. and K.C. Anderson, The pathophysiologic role of VEGF in hematologic malignancies: therapeutic implications. Blood, 2005. 105(4): p. 1383-95. 60. Kumar, S., et al., Prognostic value of bone marrow angiogenesis in patients with multiple myeloma undergoing high-dose therapy. Bone Marrow Transplant, 2004. 34(3): p. 235-9. 61. Bataille, R., et al., Mechanisms of bone destruction in multiple myeloma: the importance of an unbalanced process in determining the severity of lytic bone disease. J Clin Oncol, 1989. 7(12): p. 1909-14. 62. Roodman, G.D., Pathogenesis of myeloma bone disease. Leukemia, 2009. 23(3): p. 435-41. 63. Ehrlich, L.A. and G.D. Roodman, The role of immune cells and inflammatory cytokines in Paget's disease and multiple myeloma. Immunol Rev, 2005. 208: p. 252- 66. 64. Tian, E., et al., The role of the Wnt-signaling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma. N Engl J Med, 2003. 349(26): p. 2483-94. 65. Zheng, M.M., et al., The systemic cytokine environment is permanently altered in multiple myeloma. PLoS One, 2013. 8(3): p. e58504. 66. Zingone, A., et al., Altered cytokine and chemokine profiles in multiple myeloma and its precursor disease. Cytokine, 2014. 69(2): p. 294-7. 67. Urashima, M., et al., Transforming growth factor-beta1: differential effects on multiple myeloma versus normal B cells. Blood, 1996. 87(5): p. 1928-38. 68. Allan, D.S., et al., TGF-β affects development and differentiation of human natural killer cell subsets. Eur J Immunol, 2010. 40(8): p. 2289-95. 69. Trotta, R., et al., TGF-beta utilizes SMAD3 to inhibit CD16-mediated IFN-gamma production and antibody-dependent cellular cytotoxicity in human NK cells. J Immunol, 2008. 181(6): p. 3784-92. 70. Brabletz, T., et al., Transforming growth factor beta and cyclosporin A inhibit the inducible activity of the interleukin-2 gene in T cells through a noncanonical octamer-binding site. Mol Cell Biol, 1993. 13(2): p. 1155-62. 71. Swain, S.L., et al., Transforming growth factor-beta and IL-4 cause helper T cell precursors to develop into distinct effector helper cells that differ in secretion pattern and cell surface phenotype. J Immunol, 1991. 147(9): p. 2991-3000. 72. Bogdan, C. and C. Nathan, Modulation of macrophage function by transforming growth factor beta, interleukin-4, and interleukin-10. Ann N Y Acad Sci, 1993. 685: p. 713-39. 73. Geissmann, F., et al., TGF-beta 1 prevents the noncognate maturation of human dendritic Langerhans cells. J Immunol, 1999. 162(8): p. 4567-75. 74. Pratt, G., O. Goodyear, and P. Moss, Immunodeficiency and immunotherapy in multiple myeloma. Br J Haematol, 2007. 138(5): p. 563-79. 75. Blimark, C., et al., Multiple myeloma and infections: a population-based study on 9253 multiple myeloma patients. Haematologica, 2015. 100(1): p. 107-13.

97 76. Peinado, H., et al., Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med, 2012. 18(6): p. 883-91. 77. Raposo, G., et al., B lymphocytes secrete antigen-presenting vesicles. J Exp Med, 1996. 183(3): p. 1161-72. 78. Théry, C., L. Zitvogel, and S. Amigorena, Exosomes: composition, biogenesis and function. Nat Rev Immunol, 2002. 2(8): p. 569-79. 79. Beach, A., et al., Exosomes: an overview of biogenesis, composition and role in . J Ovarian Res, 2014. 7: p. 14. 80. Borges, F.T., L.A. Reis, and N. Schor, Extracellular vesicles: structure, function, and potential clinical uses in renal diseases. Braz J Med Biol Res, 2013. 46(10): p. 824- 30. 81. Théry, C., Exosomes: secreted vesicles and intercellular communications. F1000 Biol Rep, 2011. 3: p. 15. 82. Théry, C., et al., Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol, 2006. Chapter 3: p. Unit 3.22. 83. Technologies, S. Frequencies of Cell Types in Human Peripheral Blood. July 17, 2015; 1]. Available from: http://www.stemcell.com/~/media/Files/wallchart_CellTypes_WEB.pdf?la=en. 84. Caligiuri, M.A., Human natural killer cells. Blood, 2008. 112(3): p. 461-9. 85. Nagler, A., et al., Comparative studies of human FcRIII-positive and negative natural killer cells. J Immunol, 1989. 143(10): p. 3183-91. 86. Perussia, B., Fc receptors on natural killer cells. Curr Top Microbiol Immunol, 1998. 230: p. 63-88. 87. Cooper, M.A., T.A. Fehniger, and M.A. Caligiuri, The biology of human natural killer-cell subsets. Trends Immunol, 2001. 22(11): p. 633-40. 88. Zamai, L., et al., Natural killer (NK) cell-mediated cytotoxicity: differential use of TRAIL and Fas ligand by immature and mature primary human NK cells. J Exp Med, 1998. 188(12): p. 2375-80. 89. McCann, F.E., et al., The size of the synaptic cleft and distinct distributions of filamentous actin, ezrin, CD43, and CD45 at activating and inhibitory human NK cell immune synapses. J Immunol, 2003. 170(6): p. 2862-70. 90. Orange, J.S., Formation and function of the lytic NK-cell immunological synapse. Nat Rev Immunol, 2008. 8(9): p. 713-25. 91. Chen, S., et al., Suppression of tumor formation in lymph nodes by L-selectin- mediated natural killer cell recruitment. J Exp Med, 2005. 202(12): p. 1679-89. 92. Orange, J.S., et al., The mature activating natural killer cell immunologic synapse is formed in distinct stages. Proc Natl Acad Sci U S A, 2003. 100(24): p. 14151-6. 93. Orange, J.S., et al., Wiskott-Aldrich syndrome protein is required for NK cell cytotoxicity and colocalizes with actin to NK cell-activating immunologic synapses. Proc Natl Acad Sci U S A, 2002. 99(17): p. 11351-6. 94. Cohnen, A., et al., Surface CD107a/LAMP-1 protects natural killer cells from degranulation-associated damage. Blood, 2013. 122(8): p. 1411-8. 95. Vanherberghen, B., et al., Classification of human natural killer cells based on migration behavior and cytotoxic response. Blood, 2013. 121(8): p. 1326-34.

98 96. Watzl, C. and E.O. Long, Signal transduction during activation and inhibition of natural killer cells. Curr Protoc Immunol, 2010. Chapter 11: p. Unit 11.9B. 97. Cella, M., et al., Differential requirements for Vav proteins in DAP10- and ITAM- mediated NK cell cytotoxicity. J Exp Med, 2004. 200(6): p. 817-23. 98. Caraux, A., et al., Phospholipase C-gamma2 is essential for NK cell cytotoxicity and innate immunity to malignant and virally infected cells. Blood, 2006. 107(3): p. 994- 1002. 99. Lanier, L.L., Up on the tightrope: natural killer cell activation and inhibition. Nat Immunol, 2008. 9(5): p. 495-502. 100. Bolland, S., et al., SHIP modulates responses by regulating membrane association of Btk. Immunity, 1998. 8(4): p. 509-16. 101. Stebbins, C.C., et al., Vav1 dephosphorylation by the tyrosine phosphatase SHP-1 as a mechanism for inhibition of cellular cytotoxicity. Mol Cell Biol, 2003. 23(17): p. 6291-9. 102. Long, E.O., Negative signaling by inhibitory receptors: the NK cell paradigm. Immunol Rev, 2008. 224: p. 70-84. 103. Peterson, M.E. and E.O. Long, Inhibitory receptor signaling via tyrosine phosphorylation of the adaptor Crk. Immunity, 2008. 29(4): p. 578-88. 104. Karwacz, K., et al., PD-L1 co-stimulation, ligand-induced TCR down-modulation and anti-tumor immunotherapy. Oncoimmunology, 2012. 1(1): p. 86-88. 105. Guo, H., et al., Immune cell inhibition by SLAMF7 is mediated by a mechanism requiring src kinases, CD45, and SHIP-1 that is defective in multiple myeloma cells. Mol Cell Biol, 2015. 35(1): p. 41-51. 106. Cooper, M.D. and B.R. Herrin, How did our complex immune system evolve? Nat Rev Immunol, 2010. 10(1): p. 2-3. 107. Parham, P. and A. Moffett, Variable NK cell receptors and their MHC class I ligands in immunity, reproduction and human evolution. Nat Rev Immunol, 2013. 13(2): p. 133-44. 108. Jones, E.Y., MHC class I and class II structures. Curr Opin Immunol, 1997. 9(1): p. 75-9. 109. Tysoe-Calnon, V.A., J.E. Grundy, and S.J. Perkins, Molecular comparisons of the beta 2-microglobulin-binding site in class I major-histocompatibility-complex alpha- chains and proteins of related sequences. Biochem J, 1991. 277 ( Pt 2): p. 359-69. 110. Zacharias, M. and S. Springer, Conformational flexibility of the MHC class I alpha1- alpha2 domain in peptide bound and free states: a molecular dynamics simulation study. Biophys J, 2004. 87(4): p. 2203-14. 111. Yewdell, J.W., Not such a dismal science: the economics of protein synthesis, folding, degradation and antigen processing. Trends Cell Biol, 2001. 11(7): p. 294-7. 112. Glickman, M.H. and A. Ciechanover, The ubiquitin-proteasome proteolytic pathway: destruction for the sake of construction. Physiol Rev, 2002. 82(2): p. 373-428. 113. Finley, D., Recognition and processing of ubiquitin-protein conjugates by the proteasome. Annu Rev Biochem, 2009. 78: p. 477-513. 114. Saveanu, L., et al., Concerted peptide trimming by human ERAP1 and ERAP2 aminopeptidase complexes in the endoplasmic reticulum. Nat Immunol, 2005. 6(7): p. 689-97.

99 115. Reits, E., et al., Peptide diffusion, protection, and degradation in nuclear and cytoplasmic compartments before antigen presentation by MHC class I. Immunity, 2003. 18(1): p. 97-108. 116. Momburg, F. and P. Tan, Tapasin-the keystone of the loading complex optimizing peptide binding by MHC class I molecules in the endoplasmic reticulum. Mol Immunol, 2002. 39(3-4): p. 217-33. 117. Wearsch, P.A. and P. Cresswell, The quality control of MHC class I peptide loading. Curr Opin Cell Biol, 2008. 20(6): p. 624-31. 118. Neefjes, J., et al., Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nat Rev Immunol, 2011. 11(12): p. 823-36. 119. Tan, P., et al., Recruitment of MHC class I molecules by tapasin into the transporter associated with antigen processing-associated complex is essential for optimal peptide loading. J Immunol, 2002. 168(4): p. 1950-60. 120. Chapman, D.C. and D.B. Williams, ER quality control in the biogenesis of MHC class I molecules. Semin Cell Dev Biol, 2010. 21(5): p. 512-9. 121. Parham, P., Function and polymorphism of human leukocyte antigen-A,B,C molecules. Am J Med, 1988. 85(6A): p. 2-5. 122. Robinson, J., et al., The IPD and IMGT/HLA database: allele variant databases. Nucleic Acids Res, 2015. 43(Database issue): p. D423-31. 123. Lozzio, C.B. and B.B. Lozzio, Human chronic myelogenous leukemia cell-line with positive Philadelphia chromosome. Blood, 1975. 45(3): p. 321-34. 124. Gao, M., et al., Myeloma cells resistance to NK cell lysis mainly involves an HLA class I-dependent mechanism. Acta Biochim Biophys Sin (Shanghai), 2014. 46(7): p. 597-604. 125. Lury, D., H. Epstein, and N. Holmes, The human class I MHC gene HLA-F is expressed in lymphocytes. Int Immunol, 1990. 2(6): p. 531-7. 126. Geraghty, D.E., et al., Human leukocyte antigen F (HLA-F). An expressed HLA gene composed of a class I coding sequence linked to a novel transcribed repetitive element. J Exp Med, 1990. 171(1): p. 1-18. 127. Lepin, E.J., et al., Functional characterization of HLA-F and binding of HLA-F tetramers to ILT2 and ILT4 receptors. Eur J Immunol, 2000. 30(12): p. 3552-61. 128. Goodridge, J.P., et al., HLA-F and MHC-I open conformers cooperate in a MHC-I antigen cross-presentation pathway. J Immunol, 2013. 191(4): p. 1567-77. 129. Lee, N., A. Ishitani, and D.E. Geraghty, HLA-F is a surface marker on activated lymphocytes. Eur J Immunol, 2010. 40(8): p. 2308-18. 130. van Wijk, I.J., et al., HLA-G expression in trophoblast cells circulating in maternal peripheral blood during early pregnancy. Am J Obstet Gynecol, 2001. 184(5): p. 991-7. 131. Goodridge, J.P., et al., KIR2DL4 (CD158d) genotype influences expression and function in NK cells. J Immunol, 2003. 171(4): p. 1768-74. 132. Gonen-Gross, T., et al., Complexes of HLA-G protein on the cell surface are important for leukocyte Ig-like receptor-1 function. J Immunol, 2003. 171(3): p. 1343-51. 133. Gonen-Gross, T., et al., The CD85J/leukocyte inhibitory receptor-1 distinguishes between conformed and beta 2-microglobulin-free HLA-G molecules. J Immunol, 2005. 175(8): p. 4866-74.

100 134. Yan, W.H., et al., Unfavourable clinical implications for HLA-G expression in acute myeloid leukaemia. J Cell Mol Med, 2008. 12(3): p. 889-98. 135. Nückel, H., et al., HLA-G expression is associated with an unfavorable outcome and immunodeficiency in chronic lymphocytic leukemia. Blood, 2005. 105(4): p. 1694-8. 136. Wei, X.H. and H.T. Orr, Differential expression of HLA-E, HLA-F, and HLA-G transcripts in human tissue. Hum Immunol, 1990. 29(2): p. 131-42. 137. Braud, V., E.Y. Jones, and A. McMichael, The human major histocompatibility complex class Ib molecule HLA-E binds signal sequence-derived peptides with primary anchor residues at positions 2 and 9. Eur J Immunol, 1997. 27(5): p. 1164-9. 138. Braud, V.M., et al., TAP- and tapasin-dependent HLA-E surface expression correlates with the binding of an MHC class I leader peptide. Curr Biol, 1998. 8(1): p. 1-10. 139. Lee, N., et al., HLA-E is a major ligand for the natural killer inhibitory receptor CD94/NKG2A. Proc Natl Acad Sci U S A, 1998. 95(9): p. 5199-204. 140. Kaiser, B.K., et al., Structural basis for NKG2A/CD94 recognition of HLA-E. Proc Natl Acad Sci U S A, 2008. 105(18): p. 6696-701. 141. Llano, M., et al., HLA-E-bound peptides influence recognition by inhibitory and triggering CD94/NKG2 receptors: preferential response to an HLA-G-derived nonamer. Eur J Immunol, 1998. 28(9): p. 2854-63. 142. Forte, P., et al., HLA-E expression on porcine cells: protection from human NK cytotoxicity depends on peptide loading. Am J Transplant, 2005. 5(9): p. 2085-93. 143. Lilienfeld, B.G., et al., Transgenic expression of HLA-E single chain trimer protects porcine endothelial cells against human natural killer cell-mediated cytotoxicity. Xenotransplantation, 2007. 14(2): p. 126-34. 144. Marín, R., et al., Analysis of HLA-E expression in human tumors. Immunogenetics, 2003. 54(11): p. 767-75. 145. ISAACS, A. and J. LINDENMANN, interference. I. The interferon. Proc R Soc Lond B Biol Sci, 1957. 147(927): p. 258-67. 146. ISAACS, A., J. LINDENMANN, and R.C. VALENTINE, Virus interference. II. Some properties of interferon. Proc R Soc Lond B Biol Sci, 1957. 147(927): p. 268- 73. 147. Kawai, T. and S. Akira, Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity, 2011. 34(5): p. 637-50. 148. Schoenborn, J.R. and C.B. Wilson, Regulation of interferon-gamma during innate and adaptive immune responses. Adv Immunol, 2007. 96: p. 41-101. 149. Ank, N., et al., Lambda interferon (IFN-lambda), a type III IFN, is induced by and IFNs and displays potent antiviral activity against select virus infections in vivo. J Virol, 2006. 80(9): p. 4501-9. 150. Kotenko, S.V., et al., IFN-lambdas mediate antiviral protection through a distinct class II complex. Nat Immunol, 2003. 4(1): p. 69-77. 151. Robek, M.D., B.S. Boyd, and F.V. Chisari, Lambda interferon inhibits B and C virus replication. J Virol, 2005. 79(6): p. 3851-4. 152. Knox, J.J., et al., Characterization of T-bet and eomes in peripheral human immune cells. Front Immunol, 2014. 5: p. 217. 153. Keppel, M.P., et al., Activation-specific metabolic requirements for NK Cell IFN-γ production. J Immunol, 2015. 194(4): p. 1954-62.

101 154. Bradley, L.M., et al., Characterization of antigen-specific CD4+ effector T cells in vivo: immunization results in a transient population of MEL-14-, CD45RB- helper cells that secretes (IL-2), IL-3, IL-4, and interferon gamma. J Exp Med, 1991. 174(3): p. 547-59. 155. Chang, J.H., et al., IFN-gamma-STAT1 signal regulates the differentiation of inducible Treg: potential role for ROS-mediated apoptosis. Eur J Immunol, 2009. 39(5): p. 1241-51. 156. Reefman, E., et al., Cytokine secretion is distinct from secretion of cytotoxic granules in NK cells. J Immunol, 2010. 184(9): p. 4852-62. 157. Murray, P.J., The JAK-STAT signaling pathway: input and output integration. J Immunol, 2007. 178(5): p. 2623-9. 158. Shuai, K., et al., A single phosphotyrosine residue of Stat91 required for gene activation by interferon-gamma. Science, 1993. 261(5129): p. 1744-6. 159. Stark, G.R., et al., How cells respond to interferons. Annu Rev Biochem, 1998. 67: p. 227-64. 160. Lighvani, A.A., et al., T-bet is rapidly induced by interferon-gamma in lymphoid and myeloid cells. Proc Natl Acad Sci U S A, 2001. 98(26): p. 15137-42. 161. Min, W., J.S. Pober, and D.R. Johnson, Kinetically coordinated induction of TAP1 and HLA class I by IFN-gamma: the rapid induction of TAP1 by IFN-gamma is mediated by Stat1 alpha. J Immunol, 1996. 156(9): p. 3174-83. 162. Marchetti, M., et al., Stat-mediated signaling induced by type I and type II interferons (IFNs) is differentially controlled through lipid microdomain association and clathrin-dependent endocytosis of IFN receptors. Mol Biol Cell, 2006. 17(7): p. 2896-909. 163. Satoh, J. and H. Tabunoki, A Comprehensive Profile of ChIP-Seq-Based STAT1 Target Genes Suggests the Complexity of STAT1-Mediated Gene Regulatory Mechanisms. Gene Regul Syst Bio, 2013. 7: p. 41-56. 164. Qi, Y.F., et al., Elucidating the crosstalk mechanism between IFN-gamma and IL-6 via mathematical modelling. BMC Bioinformatics, 2013. 14: p. 41. 165. Catlett-Falcone, R., et al., Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity, 1999. 10(1): p. 105-15. 166. Bharti, A.C., N. Donato, and B.B. Aggarwal, Curcumin (diferuloylmethane) inhibits constitutive and IL-6-inducible STAT3 phosphorylation in human multiple myeloma cells. J Immunol, 2003. 171(7): p. 3863-71. 167. Qing, Y. and G.R. Stark, Alternative activation of STAT1 and STAT3 in response to interferon-gamma. J Biol Chem, 2004. 279(40): p. 41679-85. 168. Caldenhoven, E., et al., Lineage-specific activation of STAT3 by interferon-gamma in human neutrophils. J Leukoc Biol, 1999. 65(3): p. 391-6. 169. Kubiczkova, L., et al., TGF-β - an excellent servant but a bad master. J Transl Med, 2012. 10: p. 183. 170. Schroder, K., et al., Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol, 2004. 75(2): p. 163-89. 171. Braud, V.M., et al., HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. Nature, 1998. 391(6669): p. 795-9.

102 172. Benson, D.M., et al., IPH2101, a novel anti-inhibitory KIR antibody, and combine to enhance the natural killer cell versus multiple myeloma effect. Blood, 2011. 118(24): p. 6387-91. 173. Liu, J., et al., Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-{gamma} and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood, 2007. 110(1): p. 296-304. 174. Ostan, R., et al., Inflammaging and cancer: a challenge for the Mediterranean diet. Nutrients, 2015. 7(4): p. 2589-621. 175. Wang, Z., et al., Role of IFN-gamma in induction of Foxp3 and conversion of CD4+ CD25- T cells to CD4+ Tregs. J Clin Invest, 2006. 116(9): p. 2434-41. 176. Bellucci, R., et al., Interferon-γ-induced activation of JAK1 and JAK2 suppresses tumor cell susceptibility to NK cells through upregulation of PD-L1 expression. Oncoimmunology, 2015. 4(6): p. e1008824. 177. Sarkar, S., et al., Optimal selection of natural killer cells to kill myeloma: the role of HLA-E and NKG2A. Cancer Immunol Immunother, 2015. 64(8): p. 951-63. 178. Michaëlsson, J., et al., A signal peptide derived from hsp60 binds HLA-E and interferes with CD94/NKG2A recognition. J Exp Med, 2002. 196(11): p. 1403-14. 179. Swift, B.E., et al., Natural killer cell lines preferentially kill clonogenic multiple myeloma cells and decrease myeloma engraftment in a bioluminescent xenograft mouse model. Haematologica, 2012. 97(7): p. 1020-8. 180. Fryknäs, M., et al., STAT1 signaling is associated with acquired crossresistance to doxorubicin and radiation in myeloma cell lines. Int J Cancer, 2007. 120(1): p. 189- 95. 181. Shafer-Weaver, K.A., et al., Evaluating the cytotoxicity of innate immune effector cells using the GrB ELISPOT assay. J Transl Med, 2004. 2(1): p. 31. 182. Trotta, R., et al., BCR/ABL activates mdm2 mRNA translation via the La antigen. Cancer Cell, 2003. 3(2): p. 145-60. 183. Trotta, R., P. Kanakaraj, and B. Perussia, Fc gamma R-dependent -activated activation in leukocytes: a common signal transduction event necessary for expression of TNF-alpha and early activation genes. J Exp Med, 1996. 184(3): p. 1027-35. 184. Cooper, M.A., et al., Human natural killer cells: a unique innate immunoregulatory role for the CD56(bright) subset. Blood, 2001. 97(10): p. 3146-51. 185. Al-Nedawi, K., et al., Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol, 2008. 10(5): p. 619-24. 186. Umezu, T., et al., Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood, 2014. 124(25): p. 3748-57. 187. Raimondi, L., et al., Involvement of multiple myeloma cell-derived exosomes in osteoclast differentiation. Oncotarget, 2015. 6(15): p. 13772-89. 188. Wang, J., et al., The bone marrow microenvironment enhances multiple myeloma progression by exosome-mediated activation of myeloid-derived suppressor cells. Oncotarget, 2015. 6(41): p. 43992-4004. 189. Wang, J., et al., Bone marrow stromal cell-derived exosomes as communicators in drug resistance in multiple myeloma cells. Blood, 2014. 124(4): p. 555-66.

103 190. Harshman, S.W., et al., Characterization of multiple myeloma vesicles by label-free relative quantitation. Proteomics, 2013. 13(20): p. 3013-29. 191. Sun, L., et al., Serum deprivation elevates the levels of microvesicles with different size distributions and selectively enriched proteins in human myeloma cells in vitro. Acta Pharmacol Sin, 2014. 35(3): p. 381-93. 192. Hallett, W.H., et al., Immunosuppressive effects of multiple myeloma are overcome by PD-L1 blockade. Biol Blood Marrow Transplant, 2011. 17(8): p. 1133-45. 193. Benson, D.M., et al., The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti- PD-1 antibody. Blood, 2010. 116(13): p. 2286-94. 194. Portier, M., et al., gamma-Interferon in multiple myeloma: inhibition of interleukin-6 (IL-6)-dependent myeloma cell growth and downregulation of IL-6-receptor expression in vitro. Blood, 1993. 81(11): p. 3076-82. 195. Palumbo, A., et al., Interferon-gamma in multiple myeloma. Leuk Lymphoma, 1995. 18(3-4): p. 215-9. 196. Shi, J., et al., Bortezomib down-regulates the cell-surface expression of HLA class I and enhances natural killer cell-mediated lysis of myeloma. Blood, 2008. 111(3): p. 1309-17. 197. Kraemer, T., et al., HLA-E: Presentation of a Broader Peptide Repertoire Impacts the Cellular Immune Response-Implications on HSCT Outcome. Stem Cells Int, 2015. 2015: p. 346714. 198. Berkers, C.R., et al., Peptide Splicing in the Proteasome Creates a Novel Type of Antigen with an Isopeptide Linkage. J Immunol, 2015. 195(9): p. 4075-84.

104