+ All Categories
Home > Documents > SOCS3 DEFICIENCY IN MYELOID CELLS … · socs3 deficiency in myeloid cells promotes tumor...

SOCS3 DEFICIENCY IN MYELOID CELLS … · socs3 deficiency in myeloid cells promotes tumor...

Date post: 12-Sep-2018
Category:
Upload: dinhquynh
View: 223 times
Download: 0 times
Share this document with a friend
154
SOCS3 DEFICIENCY IN MYELOID CELLS PROMOTES TUMOR DEVELOPMENT by HAO YU ETTY N. BENVENISTE, COMMITTEE CHAIR JESSY DESHANE DOUGLAS R HURST SELVARANGAN PONNAZHAGAN HONGWEI QIN A DISSERTATION Submitted to the graduate faculty of The University of Alabama at Birmingham, In partial fulfillment of the requirements for the degree of Doctor of Philosophy BIRMINGHAM, ALABAMA 2015
Transcript

SOCS3 DEFICIENCY IN MYELOID CELLS PROMOTES TUMOR DEVELOPMENT

by

HAO YU

ETTY N. BENVENISTE, COMMITTEE CHAIR

JESSY DESHANE

DOUGLAS R HURST

SELVARANGAN PONNAZHAGAN

HONGWEI QIN

A DISSERTATION

Submitted to the graduate faculty of The University of Alabama at Birmingham,

In partial fulfillment of the requirements for the degree of

Doctor of Philosophy

BIRMINGHAM, ALABAMA

2015

ii

Copyright by

Hao Yu

2015

iii

SOCS3 DEFICIENCY IN MYELOID CELLS PROMOTES TUMOR DEVELOPMENT

HAO YU

CELL, MOLECULAR, AND DEVELOPMENTAL BIOLOGY

ABSTRACT

STAT3 signaling is a major intrinsic pathway for cancer inflammation owing to its

frequent activation in malignant cells, and key role in regulating many genes crucial for

inflammation in the tumor microenvironment. Persistently activated STAT3 increases

tumor cell proliferation, survival, and invasion while suppressing anti-tumor immunity.

Suppressor Of Cytokine Signaling (SOCS) proteins are negative regulators of the

JAK/STAT pathway, and generally function as tumor suppressors. The absence of

SOCS3 in particular leads to heightened activation of the STAT3 transcription factor. In

the present study, we demonstrate that genetic deletion of SOCS3 specifically in myeloid

cells significantly enhances tumor growth, which correlates with elevated levels of

myeloid-derived suppressor cells (MDSC) in the tumor microenvironment, and

diminished CD8+ T cell infiltration in tumors. The importance of MDSC in promoting

tumor growth is documented by reduced tumor growth upon depletion of MDSC.

Furthermore, SOCS3-deficient bone-marrow-derived cells exhibit heightened STAT3

activation and preferentially differentiate into the Gr-1+CD11b

+Ly6G

+ MDSC phenotype.

Importantly, we identify granulocyte-colony stimulating factor (G-CSF) as a critical

factor secreted by the tumor microenvironment that promotes development of MDSC via

a STAT3-dependent pathway. Abrogation of tumor-derived G-CSF reduces the

iv

proliferation and accumulation of Gr-1+CD11b

+ MDSC and inhibits tumor growth. These

findings highlight the critical function of SOCS3 as a negative regulator of MDSC

development and function, via inhibition of STAT3 activation. Utilizing SOCS3/STAT3

expression to predict prostate cancer progression and immune therapy responses (anti

PD-L1) will also be discussed.

Keywords: myeloid-derived suppressor cells (MDSC), Gr-1+CD11b

+ Cells, JAK/STAT

pathway, suppressor of cytokine signaling (SOCS), myeloid cells, tumor microenvironment,

granulocyte-colony stimulating factor (G-CSF)

v

DEDICATION

I would like to dedicate this dissertation to my family.

vi

ACKNOWLEDGEMENTS

I am greatly indebted to the many people who made this dissertation possible.

First and foremost, I would like to thank Dr. Etty (Tika) Benveniste, my most dedicated

mentor, and Dr. Hongwei Qin, for their support and guidance on my research projects

and training. They inspired me to find my own scientific niche and to think independently.

I am very fortunate to learn a wealth of scientific knowledge and laboratory techniques in

the cancer-related field under this valuable scientific environment which led me to the

completion of the Ph.D study. I would also like to thank my committee members: Drs.

Jessy Deshane, Douglas R Hurst, and Selvarangan Ponnazhagan. They have provided

extremely important contributions and guidance to this research. I am inspired by their

motivation, immense knowledge, and enthusiasm in mentoring and research. They have

been supportive and helpful every step of the way, and I am extremely grateful for their

patience and guidance. I would also like to acknowledge all the members in Dr.

Benveniste’s laboratory and Drs. Richard Lopez, Bradley Yoder, Dongqi Xing, Xiaosi

Han, and other faculty from UAB for their insightful comments and encouragement.

I would like to thank my family. Without their constant support and unconditional

love, I could not have completed my graduate studies. In addition, I would like to

acknowledge the support from my friends. I will always be grateful to have all of you in

my life.

vii

TABLE OF CONTENTS

Page

ABSTRACT ..................................................................................................................iii

DEDICATION ..............................................................................................................v

ACKNOWLEDGEMENTS ..........................................................................................vi

LIST OF FIGURES ......................................................................................................x

LIST OF ABBREVIATIONS .......................................................................................xii

INTRODUCTION ........................................................................................................1

The Janus Kinase (JAK)/Signal Transducers and

Activators of Transcription (STAT) Pathway...................................................1

Dysregulation of the JAK/STAT Pathway in Cancer .......................................2

Constitutive activation of STAT3 in human cancer .................................2

The importance of STAT3 in tumorigenesis and metastasis ....................4

The importance of STAT3 in inflammation and cancer ...........................5

Role of SOCS3 in Regulating the JAK/STAT Pathway ...................................8

Role of SOCS3 in Regulating Immune Cell Functions and Responses ...........10

viii

Immune Contexture in the Tumor Micro-environment ....................................13

Immune cells shape tumor immunogenicity .............................................14

Understanding immune contexture in

cancer and predicting patient outcome....................................................15

Role of Myeloid Cells in Tumor Development and Progression ......................17

Dendritic cells ..........................................................................................17

Macrophages............................................................................................18

Neutrophils/granulocytes .........................................................................19

Origin of Myeloid-Derived Suppressor Cells (MDSCs) .................................22

Myeloid cell mobilization and clearance .................................................24

Role of MDSCs in Regulating Anti-tumor Immunity ......................................25

Using the TRAMP Model to Investigate

Prostate Tumor Development ...........................................................................27

Prostate cancer ........................................................................................28

TRAMP prostate model ............................................................................29

Rationale of Dissertation Study ........................................................................30

ix

SOCS3 Deficiency in Myeloid Cells Promotes Tumor Development: Involvement of

STAT3 Activation and Myeloid-Derived Suppressor Cells .........................................40

Abstract .............................................................................................................42

Introduction .......................................................................................................43

Materials and Methods ......................................................................................46

Results ...............................................................................................................51

Discussion .........................................................................................................59

References .........................................................................................................65

Figures...............................................................................................................69

CONCLUSIONS...........................................................................................................94

REFEENCES ................................................................................................................101

APPENDIX ...................................................................................................................138

x

LIST OF FIGURES

Figure Page

INTRODUCTION

1. JAK/STAT Signal Pathway in Cytokine Signaling. ............................................ 33

2. Suppressor Of Cytokine Signaling 3 (SOCS3) Protein Negatively Regulates the

JAK/STAT Pathway ............................................................................................ 35

3. Suppressive Mechanisms Mediated by MDSCs .................................................. 37

4. TRAMP Tumor Grading Scale ............................................................................ 39

SOCS3 Deficiency in Myeloid Cells Promotes Tumor Development: Involvement of

STAT3 Activation and Myeloid-Derived Suppressor Cells

1. Myeloid-specific SOCS3 Loss Promotes Tumor Growth ................................... 69

2. Gr-1+CD11b

+ Cells from SOCS3

MyeKO Tumor-Bearing Mice Suppress Antigen-

specific T-cell Responses .............................................................................................. 71

xi

3. Orthotopic Prostate Tumor Growth is Enhanced in SOCS3MyeKO

mice .............. 73

4. Loss of SOCS3 Promotes Proliferation of Gr-1+CD11b

+ Cells ........................... 75

5. Absence of SOCS3 Enhances STAT3 Activation and

BM Differentiation into Gr-1+CD11b

+ MDSC .................................................... 77

6. G-CSF Induces the Differentiation of BM Precursors into Functional MDSC in a

STAT3-dependent Manner ........................................................................................... 80

7. G-CSF Neutralization Limits Gr-1+CD11b

+ Cell Proliferation In Vitro and Tumor

Growth In Vivo .............................................................................................................. 83

S1. Characterization of Immune Responses in TRAMP Tumor-bearing Mice ....... 85

S2. Gr-1+CD11b

+ Cells Isolated from SOCS3

MyeKO Mice

Exhibit Enhanced MDSC Suppressive Function ............................................... 87

S3. Effect of SOCS3 on Gr-1+CD11b

+ Cell Proliferation........................................ 89

S4. G-CSF Expression by TRAMP Tumor-Bearing Mice ...................................... 91

S5. Effects of G-CSF Neutralizing Antibody .......................................................... 92

xii

LIST OF ABBREVIATIONS

APC Antigen-presenting cell

BBB Blood-brain barrier

CCL Chemokine (C-C motif) ligand

CCR C-C chemokine receptor

CXCL Chemokine (C-X-C motif) ligand

CXCR CXC chemokine receptor

CFA Complete Freund’s adjuvant

CNS Central nervous system

DC Dendritic cell

dLNs Draining lymph nodes

EAE Experimental autoimmune encephalomyelitis

ELISA Enzyme-linked immunosorbent assay

FDA Food and Drug Administration

GA Glatiramer acetate

G-CSF Granulocyte colony-stimulating factor

GM-CSF Granulocyte-macrophage colony-stimulating factor

GWAS Genome-wide association study

xiii

IFN Interferon

IL Interleukin

JAK Janus kinase

LPS Lipopolysaccharide

MBP Myelin basic protein

MCP-1 Monocyte chemotactic protein-1

M-CSF Macrophage colony-stimulating factor

MFI Mean fluorescent intensity

MHC Major histocompatibility complex

MOG Myelin oligodendrocyte glycoprotein

MS Multiple sclerosis

NF-κB Nuclear factor kappa-light-chain-enhancer of activated B cells

NO Nitric oxide

OVA Ovalbumin

PLP Proteolipid protein

RAG Recombination activating gene

SC Spinal cord

STAT Signal transducers and activators of transcription

xiv

TCR T cell receptor

TH T helper

TGF Transforming growth factor

TLR Toll-like receptor

TNF Tumor necrosis factor

Treg Regulatory T cell

WT Wild Type

1

INTRODUCTION

The Janus Kinase (JAK)/Signal Transducers and Activators of Transcription (STAT)

Pathway

The JAK/STAT signaling pathway is the predominant signal transduction cascade

utilized by numerous cytokines, and is critical for initiating innate immunity,

orchestrating adaptive immune mechanisms, and ultimately constraining inflammatory

and immune responses (1-3). The JAK/STAT pathway is utilized by over 60 cytokines

and growth factors including the interleukin-6 (IL-6) family, interferon (IFN) superfamily,

epidermal growth factor (EGF) and fibroblast growth factor (FGF) (4, 5). Cytokines and

growth factors activate receptor-associated JAKs, which phosphorylate the receptor

cytoplasmic domain on tyrosine residues, leading to recruitment of STATs (5-7). The

JAKs then tyrosine phosphorylate STATs, promoting their activation (5). Once activated,

STATs dimerize as either homodimers or heterodimers, and translocate to the nucleus,

then bind to regulatory elements to induce transcription of target genes (Fig. 1) (8, 9).

There are four JAKs (JAK1, JAK2, JAK3 and TYK2), and a total of seven STATs

(STAT 1, 2, 3, 4, 5a, 5b and 6) (9, 10). Various combinations of JAK/STAT usage result

in differential gene expression, particularly depending on the STAT transcription factor(s)

that is activated (11-14). STAT proteins can be divided into two groups. The first group

consists of STAT2, STAT4, and STAT6, which are predominantly involved in regulating

2

lymphocyte development (15). The second group includes STAT1, STAT3, and STAT5,

which are activated in different tissues through various ligands and are involved in

cytokine signaling, development of mammary glands, embryogenesis as well as

malignancy (16). STAT-mediated signaling is typically transmitted through the formation

of phosphorylated homodimers. However, complex cytokine signaling required for

generating a robust and specialized immune response is mediated through the use of a

limited number of STAT molecules, which involves the heterodimerization of STAT

proteins (17, 18). Cytokines, through activation of the JAK/STAT pathway, are of

paramount importance in regulating the development, differentiation and function of

immune cells (19, 20). JAK1/2 and STAT1 activation mediate the effects of IFN-γ on

macrophage function (21), JAK1/2 and STAT3 are involved in IL-6 family signaling (22),

and granulocyte macrophage colony-stimulating factor (GM-CSF) signals through JAK2

and STAT5 to affect myeloid development (22-24). STATs are also involved in

regulating tumor cell survival (12, 25) and maintaining a tumor-promoting

microenvironment (1, 26-28).

Dysregulation of the JAK/STAT Pathway in Cancer

Constitutive activation of STAT3 in human cancer.

Inflammation plays an important role in cancer initiation and malignant progression.

Inflammatory conditions can facilitate oncogenic transformation, and genetic and

epigenetic changes in malignant cells can also generate an inflammatory

3

microenvironment that further supports tumor development (29). Cancer-associated

inflammation is marked by the presence of specific inflammatory cells and inflammatory

mediators, including cytokines and chemokines (29). The critical link between chronic

inflammation and cancer is clearly illustrated by the fact that the major pro-inflammatory

transcription factor, STAT3, can be activated by several of these important cancer risk

factors (30). STAT3 is activated in response to many cytokines such as IL-6 family

members, growth factors (e.g., EGF, FGF, TGF-α, G-CSF, and GM-CSF) and oncogenic

proteins through phosphorylation of tyrosine 705 (17). Other classes of non-receptor

protein tyrosine kinases have also been reported to stimulate STAT3 activation. The Src

family of kinases (SFKs) can either activate STAT3 directly or may function downstream

of the activation of receptor tyrosine kinase (RTKs)/G protein-coupled receptors (GPCRs)

(31). Phosphorylation of a single serine (Ser727) in the C-terminal transactivation domain

of STAT3 by numerous serine kinases including mTOR and protein kinase C delta (PKC)

allows for maximal activation of transcription of responsive genes (32). Transcriptional

activation by STAT3 requires the recruitment of co-activators such as CREB-binding

Protein (CBP)/p300 and apurinic/apyrimidinic endonuclease (APE)/Ref-1 (33, 34).

The potential oncogenic role of STAT3 was established by the evidences that

constitutively activated STAT3 in Src-transformed cell lines and that interrupting STAT3

signaling blocks the transformation of mouse fibroblasts by the Src oncoprotein (31). The

first direct links between STATs and human cancer came from findings that constitutive

STAT3 activity is required for the growth of head and neck cancer and of acute

myelocytic leukemia and chronic myelocytic leukemia cells (35). In contrast to the

transient nature of STAT3 activation in normal cells, persistent activation of STAT3 has

4

been reported in a variety of human tumor cell lines and primary human tumors,

including leukemia, lymphomas, multiple myeloma, glioma, breast, ovarian, cervical,

colon, lung, and prostate cancers (36-40).

The importance of STAT3 in tumorigenesis and metastasis.

Cancer cells expressing constitutively activated STAT3 are more resistant to apoptosis

and chemotherapies aimed at initiating apoptosis (41). Numerous groups have shown that

in vivo administration of STAT3 inhibitors have antitumor effects in human cancer

xenograft mouse models (42, 43). The first evidence towards the role of STAT3 in

survival was that STAT3 activation is essential for gp130-induced proliferation of the IL-

3-dependent pro-B hematopoietic cell line, BAF/B03 (44). In gastric, glioblastoma and

colorectal cancer cells, the active form of STAT3 was found to promote the G1/S phase

transition of the cell cycle through the expression of cyclin D1, which can associate with

cdk4 or cdk6 and control progression from G1 to S phase (45). Constitutively

phosphorylated STAT3 has been found to induce over-expression of target genes such as

cdc2, cyclin B1, m-ras, and E2F-1 in colon and breast carcinomas (46). Gp130-mediated

STAT3 signaling has been shown to up-regulate the expression of several growth-

promoting genes, such as c-Myc, Pim-1 and Pim-2, and promote emergent tumor cells to

escape terminal differentiation (47-50). Constitutive STAT3 signaling contributes to

malignancy by preventing the apoptosis pathway in multiple myeloma cells through

increased expression of Bcl-xL (51). Inhibition of STAT3 with the JAK inhibitor,

AZD1480, was found to decrease tumor growth through down regulation of cell cycle

regulators, anti-apoptotic genes Bcl-2 and survivin, the metastasis-related factor TIMP-1,

and c-Myc (52).

5

STAT3 is involved in regulating cell movement, mainly by cytoskeleton reorganization

and controlling cell adhesion properties. Recent studies demonstrate enhanced STAT3

activation with increased cell–cell contact or increased confluence, indicating that

STAT3 involved in tumor cell contact and able to up-regulate genes necessary for cell

invasion and migration (30, 53, 54). The introduction of constitutively-activated mutated

STAT3-C in epithelial cells decreased E-cadherin levels, increased numbers of

lamellipodia and stress fibers, and enhanced migratory capacity and tumor formation by

inducing the expression of fibronectin (55). STAT3 target genes include several members

of the MMP family, which are known to contribute to tumor invasion, angiogenesis and

metastasis (56, 57). High levels of phosphorylated STAT3 are a prominent feature in

colon and gastric cancers associated with adverse outcomes (58). Experimentally induced

STAT3 activity enhanced both the level of MMP-1 mRNA and secreted MMP-1

enzymatic activity in colorectal carcinomas (59). Activation of STAT3 by IL-6 was also

found to induce Twist expression in human breast cancer cells by binding to the second

proximal STAT3-binding site on the Twist promoter and activating its transcription (60,

61). STAT3-deficient keratinocytes demonstrated increased cell adhesiveness and

compromised growth factor-induced cell migration (62).

The importance of STAT3 in inflammation and cancer.

STAT3 signaling is a major intrinsic pathway for cancer inflammation because it is

frequently activated in malignant cells and capable of inducing a large number of genes

that are crucial for inflammation. STAT3 is crucial for regulating cytokines, chemokines

and other mediators that induce and maintain a cancer-promoting inflammatory

environment (63). Within the tumor microenvironment, the persistent activation of

6

STAT3 in tumor cells leads to the over-production of cytokines, chemokines and growth

factors and the associated receptors which in turn activate STAT3 in stromal

inflammatory cells and release inflammatory mediators to the microenvironment (64, 65).

The IL-6/JAK/STAT3 pathway is also crucial for inflammatory conditions caused by

environmental and other factors that are associated with increased cancer risk (1, 8).

Many tumor viruses are also known to activate STAT3 by various distinct mechanisms,

including hepatitis B virus (66), human papillomavirus (67), human T-lymphotropic virus

type 1 (68) and Epstein-Barr virus (69). Both lipopolysaccharide and live bacteria are

able to activate STAT3, resulting in the production of IL-1β and IL-6, which are major

mediators of inflammation-induced cancer (70). Importantly, a crucial role of STAT3

signaling in mediating ultraviolet light-induced skin cancer has been demonstrated in a

transgenic mouse model (71). Cigarette smoke-associated cancer development may also

be linked to STAT3 activation through the nicotinic receptor (59). Chronic stress is

another contributor to cancer pathogenesis and progression, and a recent study has

suggested the potential importance of STAT3 activation in mediating tumorigenicity by

the stress mediators noradrenaline and adrenaline (53, 72).

Several recent studies have identified Toll-like receptors, such as TLR9 and TLR4, as

important activators of the JAK/STAT3 pathway (73, 74). STAT3 upregulates the

expression of certain TLRs in malignant cells and promotes tumor progression (8).

STAT3 can directly interact with the NF-κB family member RelA (also called p65),

through acetyltransferase p300-mediated acetylation, trapping it in the nucleus and

thereby contributing to constitutive NF-κB activation in tumor-associated hematopoietic

cells and various malignancies (75-77). A major role of microRNAs (miRNAs) in cancer

7

has also emerged, and several of these miRNAs, including miR-17 (78), miR-19a (79),

miR-20 (80), miR-24 (81), and miR-135 (81), have been shown to be crucial for

regulating the JAK/STAT3 pathway. Additionally, miRNAs released by tumor cells are

packaged into microvesicles and delivered to neighboring stromal cells in the tumor

microenvironment (82). For example, tumor cell-derived miR-9 strongly induces

endothelial cell migration and tumor angiogenesis by inhibiting SOCS5 in endothelial

cells, which leads to JAK/STAT3 activation in macrophages (8). Cancer-promoting

functions of STAT3, such as its role in mitochondria, epigenetic regulation, and the

tumor microenvironment further highlight the importance of targeting STAT3 for cancer

therapy. Consequently, inhibiting the STAT3 pathway abrogates formation of and

suppresses established pre-metastatic niches, effectively reducing tumor metastasis (83,

84).

Myeloid cells display increased proliferation and survival capacity through upregulated

expression of genes underlying growth in a STAT3‑dependent manner (85). Survival,

proliferation and differentiation of myeloid cells are facilitated by triggering a cascade

through the JAK/STAT3 signaling pathway which is the main transcription factor

involved in myeloid cell expansion (86, 87). STAT3 not only prevents apoptosis and

promotes cell proliferation by upregulating the anti-apoptotic or pro-proliferative factors

BCL-XL, MYC, cyclin-D1 and survivin, but also regulates the expression of multiple

proteins that are crucial for the differentiation of myeloid cells (84). In addition, STAT3

regulates the transcription factor CCAAT/enhancer-binding protein-β (C/EBPβ) (88)

which regulates myelopoiesis in healthy individuals and has a crucial role in controlling

the differentiation of myeloid progenitors to functional myeloid cells (89). STAT3 also

8

cooperates with NF-κB signaling regulating the mobilization of myeloid cells to sites of

infection, injury or tumor growth. The pro-inflammatory mediators COX2 and PGE2,

which enhance myeloid cell accumulation and suppressive activity, are also potential

targets for STAT3 and NF-κB (90).

Role of SOCS3 in Regulating the JAK/STAT Pathway

JAKs and STATs are essential mediators of almost all biological signaling events

initiated by cytokines. As such, unrestrained activation of the JAK/STAT pathway is

detrimental and has been associated with inflammation and cancer (91). Activating

mutations in STAT proteins are rare, thus STAT hyper-activation is usually due to an

over-abundance of cytokines, and/or dysregulation of endogenous negative regulators of

JAKs or STATs. The JAK/STAT pathway is

tightly regulated at many steps through distinct mechanisms, including phosphotyrosine

phosphatases (PTPs), protein inhibitor of activated STATs (PIAS), and suppressors of

cytokine signaling (SOCS) proteins (Fig. 1) (92). PTPs participate in the regulation of the

JAK/STAT signaling pathway through dephosphorylation and have important

implications in physiology and diseases (93). PIAS proteins regulate the activity of many

transcription factors through the sumoylation machinery (94). Different PIAS bind

different STATs and probably act by inhibiting their DNA binding or by recruiting

histone deacetylases and promoting protein degradation (95).

9

Most notably, SOCS proteins are cytokine-inducible proteins, acting as potent negative

regulators of the JAK/STAT signaling pathway. Besides their obligate intracellular role,

SOCS proteins can serve as vectors mediating macrophage to epithelial cell cross-talk

through exosome trafficking (96). The SOCS family is composed of eight members; CIS

and SOCS1-7, and serve to restrict the duration of activation of cytokine-induced

signaling by inhibiting JAK kinase activity after it has been turned on (97, 98).

Experiments in different genetically manipulated mice have demonstrated a crucial role

of SOCS proteins in pathophysiology. For example, SOCS1-deficient mice die within 3

weeks of birth due to severe systemic inflammation resulting from uncontrolled

interferon-γ (IFN-γ) signaling (99). SOCS2-deficient mice develop gigantism due to

enhanced responses to growth hormone (100). Mice lacking SOCS3 die perinatally due to

defective placental formation (101, 102).

SOCS proteins contain an N-terminal variable region, a classical SH2 domain, and a C-

terminal SOCS box (Fig. 2A) (98). SOCS proteins are not constitutively expressed, rather,

are induced after cytokine binding to its receptor (103). The SOCS proteins create a

negative feedback loop to prevent excessive activation of cytokine-induced JAK/STAT

signaling (104). SOCS proteins are able to bind JAKs and certain cytokine receptors via

their SH2 domains, thereby suppressing further signaling events (Fig. 2B) (105, 106).

The SOCS box recruits a complex containing elongin B and C (102, 107, 108), and this

complex then interacts with the Cullin-5 ubiquitin machinery, leading to proteasome

mediated degradation of SOCS-associated targets (109). Any tyrosine phosphorylated

signaling intermediate (phospho-JAK, phospho-STAT, phosphorylated receptors) is

conceivable a SOCS protein substrate (110). Thus, the SH2 domain of SOCS proteins

10

function as an adapter bringing ubiquitin ligases close to kinase-activated signaling

proteins and mediate their degradation (111). SOCS1 and SOCS3, but not the other

members of the SOCS family, bind directly to JAK proteins, conferring inhibition of JAK

kinase activity (98). Studies using truncated or chimeric forms of SOCS proteins showed

that SOCS1 and SOCS3 contain a 12 amino acid N-terminal kinase inhibitory region

(KIR) resembling a JAK substrate, which allows them to suppress signaling by direct

inhibition of JAK’s catalytic activity (Fig. 2B) (112). The KIR region of SOCS1 binds

directly to the auto-phosphorylation site of JAK2 and a peptide mimic corresponding to

this site can function as an inhibitor of the JAK/STAT pathway (113). SOCS1 and

SOCS3 in particular have critical functions in repressing innate and adaptive immunity,

in part by inhibiting STAT activation induced by IFN-γ, IL-6, IL-12, IL-23 and GM-CSF,

which are all implicated in inflammation and cancer (97, 114).

Role of SOCS3 in Regulating Immune Cell Functions and Responses

SOCS3, as a negative regulator of the JAK/STAT3 pathway, is regulated primarily by

activation and STAT3, although its expression can be mediated through other signaling

cascades, including the mitogen activated protein kinase (MAPK) and NF-κB pathways

(115). Reduced SOCS3 expression levels are detected in cancerous lesions infected with

HCV compared with non-cancerous lesions (116-121). Hyperactivation of STAT3 by

reduced SOCS3 expression may contribute to malignancies and carcinogenesis by

inducing multiple tumor-promoting genes (119). Suppression of SOCS3 expression

11

causes constitutive STAT3 activation, which is considered to be important for the linkage

between inflammation and cancer.

Genetic deletion of SOCS3 leads to mid-gestational embryonic lethality due to increased

STAT3 and MAPK activation (115). Mice with a deletion of SOCS3 in hematopoietic

cells (SOCS3fl/fl vav-cre) have been shown to develop a severe inflammatory disease

during adult life (122). SOCS3fl/fl vav-cre mice developed a spectrum of inflammatory

pathologies with inflammatory neutrophil infiltration into multiple tissues and consequent

hind-leg paresis (123). Enhanced IL-6 responses accounted for the enhanced

susceptibility of SOCS3fl/fl vav-cre or SOCS3fl/fl LysM-cre (deficient in SOCS3 in

myeloid cells) mice to induced inflammatory diseases like rheumatoid arthritis (RA) or

experimental autoimmune encephalomyelitis (EAE) (124). Gp130-deficient mice

spontaneously develop a RA-like disease that is accelerated by IL-6 administration (104).

Accordingly, adenoviral-delivered SOCS3 reduced joint inflammation in mice with

arthritis via inhibition of IL-6 signaling (125).

SOCS3 is expressed in the double negative (early) stage of thymocyte differentiation and

regulates T cell development in the thymus (126). Thymic T cells lacking SOCS3 have

an increased frequency of γδ+ T cells compared to wild type controls. SOCS3-deficient

CD8+ T cells showed higher proliferation in response to TCR ligation than wild-type

cells despite normal activation of signaling pathways downstream of TCR and CD28

receptors (127). Suppression of IL-27 signaling was found to substantially reduce the

activation of CD8+ T cells after deletion of SOCS3 (128). TH2 cells contain higher

amounts of SOCS3 compared to TH1 cells. Accordingly, over-expression of SOCS3 in T

cells inhibits TH1 and promotes TH2 development, suggesting that SOCS3 stimulates

12

allergic responses (129). SOCS3 has been also suggested to inhibit IL-12-induced STAT4

activation by direct binding to the IL-12Rβ2. Inhibition of SOCS3 expression in T cells

exhibited markedly suppressed airway hyper-responsiveness and eosinophilia (130).

Mice with T cells over-expressing SOCS3 also showed a delayed onset of EAE and

restricted TH17 differentiation (131). SOCS3 expression is relatively low in progenitor

BM B cells and high in peripheral blood and spleen B cells. SOCS3 regulates CXCL12-

induced FAK phosphorylation through the ubiquitin-proteosome pathway (132). SOCS3

is an important negative regulator of immature B cells by affecting their survival and pro-

adhesive responses in the bone marrow microenvironment.

The majority of macrophages activated within an in vivo pro-inflammatory conditioning

environment show strong upregulation of SOCS3 expression and this cell population co-

express the M1 marker, iNOS (133). Without SOCS3, both human and rodent

macrophages have a reduced ability to develop pro-inflammatory features but instead

display immunoregulatory characteristics. In SOCS3-deficient macrophages, IL-6 signals

in a similar manner to the immunosuppressive cytokine IL-10, through prolonged STAT3

activation (134). Studies of SOCS3-deficient macrophages confirm that SOCS3

positively regulates TLR4 signaling and M1 activation by inhibition of IL-6R-mediated

STAT3 activation, as well as TGF-β-mediated SMAD3 activation (135). Forced

activation of Notch signaling enhances both M1 polarization and anti-tumor activity via

SOCS3 induction (136). However, myeloid-specific SOCS3-deficient mice are

vulnerable to a neuroinflammation model, which is characterized by enhanced STAT3

signaling, expression of M1-related genes, and an immune response dominated by TH1

and TH17 cells (137, 138). SOCS3-deficient bone marrow-derived macrophages

13

(BMDMs) express higher levels of genes related to M1 polarization, such as IL-1β, IL-6,

IL-12, IL-23, iNOS, CCL2, and CXCL10, compared with BMDMs from WT mice upon

stimulation with M1 inducers (137). Furthermore, SOCS3 deletion enhanced LPS, IFN-γ,

and GM-CSF-induced STAT activation, but it had no significant effect on LPS-induced

NF-κB and MAPK activation (139).

SOCS3 deficient DCs exhibited constitutive activation of STAT3 and expressed low

levels of MHC class II molecules, co-stimulatory molecules, and IL-12 (140, 141).

Adoptive transfer of SOCS3 deficient DCs suppressed EAE. SOCS3 deficient DCs

produced a higher amount of TGF-β than WT DCs, resulting in a selective expansion of

Treg cells (142). Thus, in the absence of SOCS3, DCs tend to become tolerogenic DCs.

In addition, SOCS3 is an important negative regulator of DCs maturation because SOCS3

negatively regulates GM-CSF receptor signaling (143).

Myeloid cells in future metastatic sites, under the influence of tumor-produced factors,

exhibit increased proliferation and survival. Of particular importance, both the production

of tumor-derived factors that promote metastasis and the outgrowth of myeloid cells in

distant sites are dependent on the JAK/STAT signaling axis. Therapeutic trials using

SOCS3-specific anti-sense oligonucleotides, small hairpin RNAs, or cell-penetrating

SOCS3 proteins, have been performed (144).

Immune Contexture in the Tumor Microenvironment

14

Immune cells shape tumor immunogenicity.

Tumors grow within an intricate network of malignant cells, vascular and lymphatic

vessels, cytokines and chemokines, and infiltrating immune cells (145). Different types of

infiltrating immune cells have different effects on tumor progression, which can vary

according to cancer types (146). Mouse studies by R. D. Schreiber and colleagues

demonstrate that infiltrating T cells have a major effect on the clinical attributes of human

cancers (147, 148). Cancer immune-editing can be divided in three different phases:

“elimination,” “equilibrium” and “escape” (149, 150). In the “elimination” stage both

innate and adaptive immunity act to identify the formation of tumor cells and to destroy

them, resembling the immunosurveillance theory (151). The “equilibrium” stage is

described as a period of tumor latency. In almost every case, once tumors survive

elimination, the innate and adaptive immune responses contribute to tumor cell growth

and shape its immunogenicity (152). The “escape” phase normally occurs when tumor

cells have developed the ability, through a number of mechanisms, to evade the

recognition of the immune system and/or their elimination (148, 152).

The progress of cancer disease results from several mechanisms developed by tumors to

evade antitumor immune responses. Rather than the ignorance and defects of anti-tumor

T cells, the suppressive tumor microenvironment and infiltrating immune cells contribute

mostly to cancer development (153). The presence of different cells and their dynamic

interaction with malignant cells have a profound effect on tumor progression (27). It has

been reported that there is a reduction or even loss of MHC I molecules, mostly

associated to gene mutations or impairment of MHCI-dependent antigen processing (154).

Similarly, the lack or reduction of the expression of co-stimulatory patterns by tumor

15

cells direct T lymphocytes to an anergic state (155). The ability of tumor cells to avoid

immune destruction is emerging as a hallmark of cancer, in addition to the previously

established hallmark of tumor-promoting inflammation (156).

Understanding immune contexture in cancer and predicting patient outcome.

Conventional clinical and pathological risk prediction in cancer patients is usually

achieved by evaluating tissue samples obtained during surgical removal of the primary

tumor, mostly focusing on their histopathological characteristics, including the size of the

tumor, tissue integrity, aberrant expression of proteins and recently, genetic markers, and

various characteristics of the invasive margin (IM) (157, 158). Still, it is well known that

histopathological characteristics provide limited information for prognosis since cancer

outcome can significantly vary among patients within the same histological tumor stage

(159). Furthermore, histopathological analysis has revealed that tumors are often

infiltrated by a variable degree of inflammatory and lymphocytic cells (160). Studies

reveal that these immune cells are not distributed randomly, but seem to be organized in

more or less dense infiltrates in the center of the tumoral zone (CT), at the IM of tumoral

nests and in lymphoid islets adjacent to the tumor (161, 162). A very important clinical

translation is the establishment of an immune score based on the density of cytotoxic and

memory T cells (CD8/CD45RO), both in the CT and the IM of tumors, to establish

prognosis of clinical outcome in patients, even when there is no cancer associated

prognostic marker such as in early tumor stage (I/II) patients (163, 164). In human

cancers, a high density of TH1/cytotoxic memory T lymphocytes located both in the CT

and the IM of the primary tumor is associated with long disease free time, better overall

16

survival and low risk of relapse and metastasis (165). Immune score classification may

help identify high-risk patients who would benefit the most from adjuvant therapy (166).

All immune cell types may be found in a tumor, including macrophages, dendritic cells,

mast cells, natural killer (NK) cells, naive and memory lymphocytes, B cells and effector

T cells, including various subsets of T cells: T helper cells, TH1 cells, TH2 cells, TH17

cells, regulatory T (Treg) cells, T follicular helper (TFH) cells and cytotoxic T cells (167-

169). It is now well established that in general, tumors with M2-phenotype macrophages

and myeloid derived suppressor cells (MDSCs) favor tumor growth and spread, whereas

infiltration of memory cytotoxic and TH1 T cells and M1-phenotype macrophages are

often associated with good clinical outcome and good response to immunotherapy (170).

NK cells are found in the stroma and are not in contact with tumor cells (171). B cells are

mostly found in the invasive margin of growing tumors and in stroma that are adjacent to

tumor beds (159, 170). T cells, particularly CD8+ T cells, may be located in the IM but

can also be in the tumor core (29, 172). More importantly, bone marrow-derived myeloid

cells, such as macrophages, neutrophils, eosinophils, mast cells and dendritic cells are

present in large numbers and have a crucial role in regulating the formation and

maintenance of the tumor microenvironment (173, 174). Myeloid cells present in the

tumor microenvironment mediate crosstalk between tumor cells and adaptive immune

cells and are correlated with patient disease progression (161, 175-177). With the

knowledge of the evolution of the immune contexture in the primary tumor, in the

periphery and in metastatic sites as disease progresses, therapeutic tools could be

designed to positively influence anti-tumor immunity.

17

Role of Myeloid Cells in Tumor Development and Progression

Pathological changes in solid cancers include recruitment and modifying of various types

of dysregulated immune cells and endothelial cells to form the tumor microenvironment

(153). A variety of chemokines and cytokines are produced by cancer cells and

surrounding stromal cells and recruit leukocytes from the circulation to local sites

according to their chemokine gradient (176). Although tumor cells were first thought to

drive the cellular events underpinning tumor angiogenesis, evidence has now emerged for

a central role of tumor-infiltrating myeloid cells such as monocytes/macrophages,

neutrophils, and dendritic cells in this process (175, 178-180). Myeloid cells are a major

component of the inflammatory infiltrate frequently seen in primary tumors (181).

Classically, these cells protect organisms from pathogens, and eliminate dying cells.

However, only recently, mounting evidence indicates that the tumor microenvironment

alters myeloid cells by converting them into potent immunosuppressive cells.

Dendritic cells.

DCs are terminally differentiated myeloid cells that specialize in antigen processing and

presentation. Two major subsets of DCs are currently recognized: conventional DCs and

plasmacytoid DCs (182). Differentiated DCs reside in tissues as immature cells that

actively take up tissue antigens but are poor antigen presenters and do not promote

effector T cell differentiation (183). Only functionally activated DCs can effectively

process and present tumor antigen, migrate to lymph node and then stimulate effective

immune responses (184). It is well established that DCs in tumor bearing hosts do not

18

adequately stimulate an immune response, and this potentially contributes to tumor

evasion of immune recognition (170). Multiple clinical studies have indicated that there

is a decreased presence and defective functionality of mature DCs in patients with breast,

non-small cell lung, pancreatic, cervical, hepatocellular or prostate cancer, or glioma

(185). DC migration and function are severely impaired by hypoxia and adenosine (186).

The transcription factor hypoxia-inducible factor 1α (HIF1α) is upregulated by DCs in

the hypoxic tumor environment and was shown to induce the expression of the adenosine

receptor, causing these DCs to drive the development of TH2 cells rather than that of

TH1 cells (187). Both phenotypically immature and phenotypically mature DCs may be

conditioned by the environment to support immune tolerance or immunosuppression

(188). MHC-II+CD11b

+CD11c

+ tumor infiltrating mouse DCs have been shown to

suppress CD8+ T cells and antitumor immune responses through arginase 1 (ARG1)

(188-190). The accumulation of indoleamine 2, 3-dioxygenase (IDO) expressing DCs

(most of which are pDCs) in tumor-bearing mice and in some patients with cancer

provides another mechanism of immune suppression, as IDO activity limits T cell growth

by depleting L-tryptophan (188, 191).

Macrophages.

Macrophages are a group of terminally differentiated myeloid cells that are closely

related to DCs. They are tissue-resident cells derived from monocytes circulating in the

peripheral blood. Their function in healthy individuals is to eliminate infectious agents,

promote wound healing and regulate adaptive immunity (180, 192). Activated

macrophages can be divided into M1 (classical activated) and M2 (alternative activated)

phenotypes. M1 or classically activated macrophages are activated by IFN-γ and bacterial

19

products, express high levels of IL-12, and low levels of IL-10, and are tumoricidal (193-

195). By contrast, M2 or alternatively activated macrophages are activated by IL-4, IL-10,

IL-13 and glucocorticoid hormones, express high levels of IL-10 and ARG1, and low

levels of IL-12, and facilitate tumor progression (180, 196-198). Macrophages in tumors,

usually termed tumor-associated macrophages (TAMs), often resemble the M2 phenotype.

However, recent evidence suggested that the phenotype of TAMs varies with the stage of

tumor progression (199). M1 macrophages are often abundant in chronic inflammatory

sites, where tumors are initiated and start to develop. Then macrophages switch to an M2-

like phenotype as the tumor begins to invade, vascularize, and develop (199-201).

TAMs are ineffective antigen-presenting cells, and they produce CC-chemokine

ligand 22 (CCL22), which attracts Treg cells that inhibit T cell activation (202). Secretion

of prostaglandin E2 (PGE2) and TGF-β by TAMs further contributes to immune

suppression (203). TAMs can also cause T cell apoptosis through their expression of PD1

ligand 1 (PD-L1), which binds to its receptor programmed cell death protein 1 (PD-1) on

activated T cells (204). Mouse M2 macrophages produce ARG1, which deprives T cells

of the L-arginine that is necessary for their growth (201). Pro-angiogenic TAMs may

express the angiopoietin receptor TIE2 and/or have low expression levels of MHC

class II molecules, and they localize to hypoxic regions (205). TAMs that promote early

tumor cell invasion are enriched for WNT7B, a protein that is involved in normal

developmental and repair responses (206).

Neutrophils/granulocytes.

20

Granulocytes are myeloid cells that are characterized by the presence of cytoplasmic

granules and a specific nuclear morphology. The most abundant type of granulocytes in

the body are neutrophils, which are also commonly referred to as polymorphonuclear

leukocytes owing to their polylobed nuclei (207). Granulocyte differentiation is regulated

by the coordinated expression of key myeloid transcription factors, with granulocytes and

macrophages differentiating from a common committed progenitor cell (208).

Neutrophils are produced within haematopoietic cords interspersed within the venous

sinuses of the bone marrow. Transcriptional profiling studies suggest that macrophages

represent the default myeloid cell, and that granulocytes arise through the selective

expression of a subset of transcription factors (e.g. Egr1, HoxB7 and STAT3), proteins

(e.g. S100A8, S100A9 and neutrophil elastase) and cytokines including G-CSF and GM-

CSF (86, 209, 210). Neutrophils are the most abundant circulating leukocyte in humans

and play a fundamental role in the innate immune response (211). The primary role of

neutrophils is to kill invading bacteria and certain fungal species through phagocytosis by

release of preformed granular enzymes and proteins, and by the production of a range of

oxygen species (206, 211, 212). However, the highly destructive capacity of these cells

also raises the potential for neutrophils to damage healthy tissues, which occurs in many

inflammatory diseases such as acute respiratory distress syndrome, inflammatory bowel

disease and rheumatoid arthritis (211, 213).

Myeloid cells are not released from the bone marrow until they reach full maturity, but

during inflammation, neutrophil precursors can be released (206). Human tumors can be

infiltrated by mature granulocytes, the numbers of which can be independent prognostic

factors for tumor recurrence (211, 213). Recent evidence has linked granulocytes, and

21

particularly neutrophils, with tumor angiogenesis and metastasis, and has provided initial

clues about the immunoregulatory roles of these cells in cancer.

Tumor cells and tumor associated stromal cells produce neutrophil-attracting CXC-

chemokines and prokineticin 2 (the mammalian orthologue of BV8) (214). In the lungs,

tumor-derived G-CSF also mobilizes granulocytes to pre-metastatic niches and supports

subsequent metastasis formation. It is likely that granulocytes facilitate the angiogenic

switch by expressing MMP9, which promotes tumor angiogenesis by inducing VEGF

expression in neoplastic tissue (215). Mobilized granulocytes also release elastase, which

then enters endosomal compartments of neoplastic cells and degrades insulin receptor

substrate 1 (IRS1). Degradation of IRS1 facilitates interactions between phosphoinositide

3 kinase (PI3K) and the receptor for the mitogen platelet-derived growth factor (PDGF),

thus promoting tumor cell proliferation (216, 217).

In mice bearing 4T1 breast tumors, neutrophils inhibited the formation of tumor

metastases through direct antitumor effects mediated by reactive oxygen species (ROS)

(218). Similarly to macrophages, neutrophils have been shown to shift from an

antitumoral ‘N1’ phenotype to a pro-tumoral ‘N2’ phenotype in the cancer

microenvironment (219). TGF-β drives the N2 phenotype, and TGF-β blockade promotes

an N1 phenotype with antitumor activity (219). In lung adenocarcinoma and

mesothelioma models, TGF-β induces tumor-infiltrating neutrophils to develop a pro-

tumoral phenotype, which is characterized by ARG1 expression and low levels of TNF,

CCL3 and intercellular adhesion molecule 1 (ICAM1) (215, 220). In tumor-bearing

animals, depletion of N2 neutrophils led to an increase in CD8+ T cell activity (221).

Similar results were observed in human melanoma: serum amyloid A1 protein induced

22

the expansion of IL-10 secreting neutrophil populations that were able to suppress the

antigen-specific proliferation of CD8+ T cells (222).

Origin of Myeloid-Derived Suppressor Cells (MDSCs)

Tumor-induced granulocytic hyperplasia is associated with tumor vasculogenesis and

escape from immunity via T cell suppression (211). Initially, these myeloid cells were

identified as granulocytes or monocytes; however, recent studies have revealed that this

hyperplasia is associated with populations of multipotent progenitor cells that have been

identified as MDSCs (223). MDSCs were originally identified in tumor-bearing mice as

cells that co-express integrin-αM (CD11b) and Myeloid Differentiation Antigen (GR-1,

also known as Ly6G/C); however, their phenotype in cancer is rather diverse (209, 224).

Mouse MDSCs have recently been identified on the basis of expression of lymphocyte

antigens Ly-6C and Ly-6G (209). CD11b+Ly-6G

lowLy-6C

hi cells have a monocytic-like

morphology, preferentially express iNOS, have increased T cell suppressive activity and

have been identified as monocytic-MDSCs (225). This contrasts with CD11b+Ly-6G

+Ly-

6Clow

cells that have a granulocyte-like morphology, express high levels of ARG1, and

are identified as granulocytic-MDSCs (G-MDSCs) which have a polymorphonuclear

(PMN) morphology (225) (Fig. 3). In addition to their specific markers, monocytic

MDSCs express varying levels of classic monocyte markers, such as F4/80 (also known

as EMR1), CD115 (also known as M-CSFR), 7/4 (also known as LY6B) and CCR2 (226).

They suppress CD8+ T cells predominantly via expression of the enzymes ARG1 and

23

inducible nitric oxide synthase (iNOS) and through the production of reactive nitrogen

species (227). Monocytic MDSCs may also include progenitors that give rise to a subset

of CD11b+GR-1

lowLY6G

-F4/80

hiMHC-II

+ macrophages with potent immunosuppressive

properties (228).

The equivalent MDSCs in humans is defined as the CD14-CD11b

+CD33

+CD15

+

phenotype or cells that express the CD33 marker but lack the expression of markers of

mature myeloid and lymphoid cells and HLA-DR (207, 229). In human patients with

renal cell carcinoma and colorectal carcinoma, MDSCs account for a 10-fold higher level

in the circulation compared with ~0.5% of peripheral blood mononuclear cells in healthy

individuals (225). MDSCs with the phenotype LIN-HLA-DR

-CD33

+CD11b

+ have been

isolated from the blood of patients with glioblastoma, breast cancer, colon cancer, lung

cancer or kidney cancer (230, 231). The frequency of this immature cell population may

reflect the tumor burden, and a high frequency correlates with poor prognosis and

radiographic progression in patients with breast or colorectal cancer (179, 225).

Polymorphonuclear MDSCs and neutrophils are functionally and phenotypically different.

First, polymorphonuclear MDSCs are immunosuppressive. Second, polymorphonuclear

MDSCs express higher levels of CD115 and CD244 than neutrophils but lower levels of

CXCR1/2 (228). Third, compared with neutrophils, polymorphonuclear MDSCs are less

phagocytic, express higher levels of ARG1 and myeloperoxidase, have increased ROS

production and show reduced chemotaxis (207, 232). Similarly, although monocytic

MDSCs and inflammatory monocytes share a common phenotype and morphology, these

cell populations are functionally distinct. Monocytic MDSCs are highly

immunosuppressive, as they express, among other factors, high levels of both iNOS and

24

ARG1. By contrast, iNOS and ARG1 are not coordinately upregulated in monocytes (221,

226) (Fig. 3). Furthermore, MDSCs include direct progenitors of DCs, macrophages and

granulocytes and are able to undergo terminal differentiation depending on the

microenvironment (227). It is clear that the myeloid lineage is globally altered in cancer

as a single, closely integrated system involving all terminally differentiated myeloid cells

and their pathologically activated immature progenitors (225, 230, 233). MDSCs are

generally defined as a heterogenous population of immune cells from the myeloid lineage

to which dendritic cells, macrophages and neutrophils also belong.

Myeloid cell mobilization and clearance.

The principal regulator of physiological granulopoiesis is G-CSF whose effects include

commitment of progenitor cells to the myeloid lineage, proliferation of granulocytic

precursors, reduction of transit time through the granulocytic compartment, and release of

mature cells from bone marrow (234). G-CSF exerts its effects through the G-CSF

receptor, which is a member of the class I cytokine receptor family. Mice that lack the G-

CSF receptor and humans who express a dominant negative receptor mutation are

profoundly neutropenic (235). IL-6, GM-CSF and IL-3 also stimulate granulopoiesis in

vivo, but in all three cases, single knock-out mice exhibit normal basal levels of

granulopoiesis, which suggests significant redundancy or reserve (236, 237).

To exit the bone marrow, myeloid cells must migrate across the sinusoidal endothelium

that separates the haematopoietic compartment from the circulation. Myeloid cells

migrate across the bone marrow endothelium through tight-fitting pores by a unique

process of trans-cellular migration, and pass through the cell body of the endothelium,

25

rather than at cell-cell junctions (238, 239). Myeloid cells maintain G-CSF receptors at

high levels on their surface from early in their development; CXCR4, a G-protein

coupled receptor, is also expressed at low levels on the cell surface of mature myeloid

cells (240-242). The major ligand for CXCR4 is stromal-derived factor 1 (SDF-1), a

CXC chemokine that is produced constitutively by bone marrow stromal cells (243). The

role of CXCR4-SDF-1 interaction in regulating myeloid cells egress from the bone

marrow is further supported by the finding that Cxcr4 deletion in murine myeloid cells

results in increased neutrophil release, and by the observation that treatment with a

CXCR4 antagonist or blocking antibodies leads to rapid mobilization of neutrophils from

both human and mouse bone marrow (243-245).

Role of MDSCs in Regulating Anti-tumor Immunity

MDSCs exploit a variety of redundant mechanisms to influence both innate and adaptive

immune responses. MDSCs deplete nutrients which are required by lymphocytes,

specifically, L-arginine depletion through iNOS, ARG1-dependent consumption; and L-

tryptophan and L-cysteine deprivation via its consumption and sequestration (224) (Fig.

3). The depletion of these amino acids causes down-regulation of the ζ chain in the T cell

receptor (TCR) complex and proliferative arrest of antigen-activated T cells (246).

MDSCs generate oxidative stress, which is caused by the production of ROS and reactive

nitrogen species by MDSCs. Peroxynitrite and hydrogen peroxide are produced by

MDSCs, and these reactive species drive several molecular blocks in T cells, ranging

26

from the loss of TCR ζ chain expression and interference with IL-2 receptor signaling to

the nitration and subsequent desensitization of the TCR (224, 233) (Fig. 3). MDSCs

interfere with lymphocyte trafficking and viability. MDSCs decrease the expression of

CD62L on naive T cells through expression of ADAM17 (disintegrin and

metalloproteinase domain containing protein 17) (247). MDSCs also decrease the number

and inhibit the function of mouse and human NK cells, mostly through membrane

contact-dependent mechanisms (248). MDSCs are involved in the activation and

expansion of Treg cell populations. MDSCs promote the clonal expansion of antigen-

specific natural Treg cells and also induce the conversion of naive CD4+ T cells into

induced Treg cells (249). Provided that MDSCs and T cells are in close proximity, the

factors that mediate MDSCs suppressive function can inhibit T-cell proliferation

regardless of the antigen specificity of the T cells. However evidence suggests that

MDSCs-mediated immunosuppression in peripheral lymphoid organs is mainly antigen-

specific (84). Stable contacts of antigen-specific interactions between antigen-presenting

cells and T cells are necessary for MDSCs-derived ROS and peroxynitrite to mediate

effects on the molecules on the surface of T cells that render the T cells unresponsive to

specific antigen (249, 250).

Accumulating evidence from tumor-bearing mice and human cancers indicates that the

induction and expansion of MDSCs in the tumor microenvironment is mediated by a

combined effect of multiple factors including cytokines, growth factors and pro-

inflammatory mediators. Neoplastic cells and tumor-associated stromal cells release

multiple tumor-derived soluble factors that perturb the myeloid compartment (89, 251).

Cytokines such as GM-CSF, G-CSF, M-CSF, stem cell factor (SCF; also known as KIT

27

ligand), VEGF and IL-3 promote myelopoiesis and contribute, in part, to a blockade of

myeloid cell maturation (229). Tumor-derived soluble factors that are pro-inflammatory

(such as IL-1β, IL-6, S100A8 and S100A9), as well as cytokines released by activated T

cells (such as IFN-γ, IL-4, IL-10 and IL-13), initiate the immunosuppressive pathways

that commit immature myeloid cells to become MDSCs and then further promote the

differentiation of MDSCs towards immunosuppressive macrophages and DCs (251, 252).

The tumor-derived factors CCL2, CCL12, CXCL5, prokineticin 2, S100A8 and S100A9

recruit immature myeloid cells to the tumor stroma (87, 253, 254). Immature myeloid

cells are also attracted by CCL2 that is nitrated or nitrosylated in the tumor environment.

By contrast, effector CD8+ T cells are not recruited by modified CCL2, which may

explain the selective enrichment of myelomonocytic cells within mouse and human

tumors (255). Continuous stimulation of myelopoiesis and activation of immature

myeloid cells by tumor-derived soluble factors may drive the subsequent accumulation of

immunosuppressive MDSCs that support tumor promotion and form the metastatic niche.

Accordingly, the oncogenic program may have a greater influence on the functional

immunosuppressive activities of MDSCs than on their accumulation (179, 225).

Using the TRAMP Model to Investigate Prostate Tumor Development

The benefit of preclinical tumor models for evaluating cancer therapeutics depends

largely on whether the tumors growing in animals closely mimic the characteristics of the

human counterpart (256). The most frequently used preclinical models are those from

28

tumor cell implantation, as they are easily controlled as compared with chemically

induced tumors or those developed from genetic alterations (146). Depending on the

source of the tumor cells, xenograft or syngeneic tumor cells/tissues are transplanted into

animals. While xenografted tumors must be grown in immune deficient animals,

syngeneic tumors are grown in animals whose immune systems are intact. This is an

especially desirable characteristic since the efficacy of cancer therapeutics can only be

truly evaluated in the presence of an intact host immune system (257).

Prostate cancer.

Prostate cancer is a leading cause of illness and death among men in the United States

and Western Europe. Autopsy studies have revealed small prostatic carcinomas in up to

29% of men 30 to 40 years of age and 64% of men 60 to 70 years of age (258). Initially,

hormone therapy or androgen-deprivation therapy is used for prostate cancer to lower the

levels of androgens through surgically removing the testicles or with drugs that stop the

testicles from making androgens or blocking androgen receptor (AR) signaling (259-261).

Overall, most men who develop prostate cancer (99%) are expected to live at least five

years after diagnosis. However, for men diagnosed with prostate cancer that has spread to

other parts of the body, the five-year survival rate drops to 28% (259). Eventually, many

men with metastatic prostate cancer develop castration-resistant disease (CRPC). This

means that the cancer is able to grow and continue to spread despite using hormone

therapy (262). At this stage of disease, effective treatment options are very limited. Until

recently, chemotherapeutic agents and immune therapy are able to prolong the mean life

span of patients (263). Sipuleucel-T (Provenge, Dendreon, Seattle, WA, USA) was

approved by the US FDA in April 2010 and is indicated for the treatment of

29

asymptomatic or minimally symptomatic mCRPC (264). Sipuleucel-T is an autologous

cellular cancer vaccine that is designed to stimulate an immune response to prostate

cancer, consisting of peripheral blood mononuclear cells (PBMCs) obtained through

leukapheresis from each patient and cultured in vitro for 2–3 days with prostate cancer

antigen and with the cytokine GM-CSF. The myeloid cells isolated from patients undergo

maturation during the stimulation and present prostate cancer antigen in cells surface.

Once the cells are reinfused back to patients, they are able to activate adaptive immune

cells and induce an immune response to prostate cancer cells. Treatment with Sipuleucel-

T was able to improve the median overall survival for 4.1 months compared with control

arm (25.8 versus 21.7 months, p=0.03), after a median follow up of 34.1 months (265).

Chronic or recurrent inflammation has a role in the development of many types of cancer

in humans, including prostate cancer (39). Symptomatic prostatitis occurs in 9% or more

of men between 40 and 79 years of age; about half of these men have more than one

episode of prostatitis by 80 years of age (157, 266). Intake of antioxidants or nonsteroidal

anti-inflammatory drugs is correlated with a decreased risk of prostate cancer (39). Focal

atrophic lesions associated with chronic inflammation are often directly adjacent to

lesions of prostatic intraepithelial neoplasia, prostate cancers, or both (267-269).

Mounting clinical evidence suggests that the immune system plays an important role in

regulating prostate cancer development. As such, understanding the role of immune cells

within prostate cancer would provide invaluable insights about utilizing anti-tumor

immunity to fight this cancer.

TRAMP prostate model.

30

Mouse and human prostate anatomy is dissimilar. The mouse prostate has a lobular

structure with four lobes—anterior, ventral, dorsal, and lateral. Alternatively, the human

prostate has one “lobe” divided into three zones: central, transitional, and peripheral (270,

271). The majority of human prostate cancer is found in the peripheral zone, which

comprises about 75% of the tissue in the prostate. The mouse dorsolateral lobe has been

described as the most similar to the human peripheral zone (257, 270, 272, 273).

The TRAMP (transgenic adenocarcinoma of the mouse prostate) model for prostate

cancer was generated and characterized in 1995–1997 (257, 270). In this model,

expression of both the large and small SV40 tumor antigens (T/tag) is regulated by the

prostate-specific rat probasin promoter (PB) (270). TRAMP mice developed epithelial

hyperplasia by 8 weeks of age (corresponding to sexual maturity), progressed to prostatic

intraepithelial neoplasia (PIN) by 18 weeks of age, and after 28 weeks of age, 100% of

the mice displayed lymphatic metastases, and approximately two-thirds displayed

pulmonary metastases (270). As disease develops in TRAMP mice, tumor tissue

histologically and biochemically resembles human disease with pathology ranging from

noncancerous PIN to aggressive adenocarcinoma of the prostate (Fig. 4) (274). TRAMP

mice exhibit progressive stages of prostate cancer ranging from PIN, cribriform structures,

and focal adenocarcinoma to extracapular extension, and seminal vesicle invasion with

metastasis to lymph nodes, adrenal glands, lung and bone (257, 275, 276). The versatility

of the TRAMP model has been extended further by the establishment of several TRAMP-

derived prostate tumor cells lines (including TRAMP-C1 and TRAMP-C2) that can be

injected into syngeneic male nontransgenic C57BL/6 mice to induce ectopic prostate

31

tumorigenesis (257). The TRAMP model has been widely adopted for use in a variety of

studies designed to assess novel therapies directed against prostate cancer.

Rationale of Dissertation Study

STAT3 signaling is a major intrinsic pathway for cancer inflammation owing to its

frequent activation in malignant cells, and key role in regulating many genes crucial for

inflammation in the tumor microenvironment. Persistently activated STAT3 increases

tumor cell proliferation, survival, and invasion while suppressing anti-tumor immunity

(1). Many of the key immunoregulatory cytokines involved in cancer, including IL-6, IL-

12, IL-23, IFN-γ and GM-CSF, require activation of JAK1, JAK2 or both for subsequent

activation of STATs, and ultimate biological responses. SOCS proteins, CIS and SOCS1-

7, inhibit JAK-STAT signaling through a variety of mechanisms. SOCS proteins are

induced by cytokines, creating a negative feedback loop to prevent excessive activation

of cytokine-induced pathways. The predominant function of SOCS3 is inhibition of

signaling by the IL-6 family of cytokines, leading to inhibition of STAT3 activation.

SOCS3, as a negative regulator of STAT3, is a key pathological regulator of

inflammation as well as tumor immunity.

We generated mice with conditional knockout of SOCS3 in cells of the myeloid lineage

(LysMCre-SOCS3fl/fl mice) to investigate the function of myeloid SOCS3 in prostate

cancer development (139). We recently demonstrated that deletion of SOCS3 in myeloid

cells (neutrophils, DCs, monocytes/macrophages) leads to heightened activation of

32

STAT3, and enhanced expression of proinflammatory genes including IL-1, TNF-α, IL-6

and iNOS (137). However, the role of SOCS3 in myeloid cells within the prostate tumor

microenvironment has not been explored. Therefore, the hypothesis of my thesis work

was to determine if SOCS3 is protective in prostate cancer by regulating antitumor

immunity through myeloid cells. To test this hypothesis, we evaluated the effect of

myeloid specific SOCS3 deletion on prostate cancer growth. In the present study, we

provide evidence that the loss of SOCS3 in myeloid cells enhances prostate tumor growth,

and is associated with elevated levels of Gr-1+CD11b

+ MDSC in tumors of SOCS3-

deficient mice. We identify G-CSF as a critical factor secreted by the tumor

microenvironment that promotes MDSC expansion via a STAT3/SOCS3-dependent

pathway. Abrogation of tumor-derived G-CSF inhibits tumor growth by reducing the

accumulation of MDSC (277).

Collectively, contained within is evidence highlighting the important role of myeloid

SOCS3 in regulating the development of myeloid cells and maintaining an

immunosuppressive milieu.

33

Adopted from Shuai K, and Liu B, Nat. Rev. Immunol. 2003;3: 909-917.

Figure 1. JAK/STAT Signal Pathway in Cytokine Signaling. Ligand-induced receptor

oligomerization activates JAKs that subsequently phosphorylate tyrosine residues on the

cytoplasmic portion of the receptor. This allows the recruitment of STAT proteins to the

phosphorylated receptor by their Src-homology 2 (SH2) domains. STATs are

phosphorylated by the JAKs on a conserved tyrosine residue in the c-terminal domain to

form STAT homodimers or heterodimers. STATs dissociate from the receptor after

dimerization and translocate into the nucleus. In the nucleus, STATs bind to specific

response elements and induce gene transcription. The negative regulators of this pathway

34

are phosphotyrosine phosphatases (PTPs), protein inhibitor of activated STAT (PIAS),

and suppressor of cytokine signaling (SOCS) proteins.

35

Adopted from Baker BJ, et.al., Trends Immunol 2009;30: 392-400.; Kershaw NJ, et.al.,

Nat Struct Mol Biol 2013;20:469-76.

Figure 2. Suppressor Of Cytokine Signaling 3 (SOCS3) Protein Negatively

Regulates the JAK/STAT Pathway. (A). SOCS proteins contain an N-terminal variable

region, a classical SH2 domain, and a C-terminal SOCS box. The SOCS box interacts

with components of the ubiquitin ligase machinery and mediates proteosomal degradation

of associated proteins, most commonly, JAKs. SOCS1 and 3 contain a kinase inhibitory

region (KIR) which acts as a pseudosubstrate for JAKs, conferring inhibition of JAK

kinase activity. (B). Ribbon diagram of the JAK2 (beige)–SOCS3 (green)–gp130 (black)

complex. The gp130 peptide is located in the canonical phosphotyrosine-binding groove

on the SH2 domain of SOCS3, while the opposing face of the SH2 domain contacts

JAK2. SOCS3 binds to gp130 through the canonical phospho-tyrosine-binding groove on

A.

B.

36

the SH2 domain with the BC loop (Ser73-Phe79). The SOCS3 KIR (Leu22-Ser29),

which is unstructured in isolation, folds back underneath the BC loop and sits in a groove

formed by the JAK2 activation loop.

37

Adopted from Gabrilovich D, et.al., Nature Reviews Immunology 9, 162-174.

Figure 3. Suppressive Mechanisms Mediated by MDSCs. MDSCs consist of two main

subsets: monocytic MDSCs, which have a CD11b+LY6G

-LY6C

hi phenotype, and

granulocytic MDSCs, which have a CD11b+LY6G

+LY6C

low phenotype. The granulocytic

subset of MDSCs has increased activity of STAT3 and NADPH, which results in high

levels of reactive oxygen species (ROS). ROS and, in particular, peroxynitrite induce

post-translational modification of T-cell receptors and may cause antigen-specific T-cell

unresponsiveness. The monocytic MDSC subset has upregulated expression of iNOS, and

increased levels of NO which suppresses T-cell function through the inhibition of MHC

38

class II expression and the induction of T-cell apoptosis. Both subsets have increased

levels of arginase 1, which causes T-cell suppression through depletion of L-arginine.

39

Adopted from Liu Z, et.al., J Immunol 2008;180:6044-53.

Figure 4. TRAMP Tumor Grading Scale. Prostate lesions are scored using a 1–6 scale

that has been established for TRAMP mice. Noncancerous lesions are graded as 1

(normal tissue); 2 (low prostatic intraepithelial neoplasia); or 3 (high prostatic

intraepithelial neoplasia). Cancerous lesions (adenocarcinomas) are graded as 4 (well-

differentiated); 5 (moderately differentiated); or 6 (poorly differentiated).

40

SOCS3 Deficiency in Myeloid Cells Promotes Tumor Development:

Involvement of STAT3 Activation and Myeloid-Derived Suppressor Cells

HAO YU1, YUDONG LIU

1, BRADEN C. MCFARLAND

1, JESSY S. DESHANE

2,

DOUGLAS R. HURST3, SELVARANGAN PONNAZHAGAN

3, ETTY N.

BENVENISTE1* AND HONGWEI QIN

1*

Running Title: Role of STAT3/SOCS3 in MDSC

Departments of Cell, Developmental and Integrative Biology1, Medicine

2 and Pathology

3,

University of Alabama at Birmingham, Birmingham, Alabama 35294

Cancer Immunol Res; 3(7)

Copyright

2015

Used by permission

Format adapted for dissertation

41

SOCS3 Deficiency in Myeloid Cells Promotes Tumor Development: Involvement of

STAT3 Activation and Myeloid-Derived Suppressor Cells

Hao Yu1, Yudong Liu

1, Braden C. McFarland

1, Jessy S. Deshane

2, Douglas R. Hurst

3,

Selvarangan Ponnazhagan3, Etty N. Benveniste

1* and Hongwei Qin

1*

Running Title: Role of STAT3/SOCS3 in MDSC

Departments of Cell, Developmental and Integrative Biology1, Medicine

2 and Pathology

3,

University of Alabama at Birmingham, Birmingham, Alabama 35294

*Co-Corresponding Authors:

Dr. Hongwei Qin, Department of Cell, Developmental and Integrative Biology,

University of Alabama at Birmingham, 1918 University Boulevard, MCLM 390,

Birmingham, AL 35294. Phone: 205-934-2573. E-mail address: [email protected]

Dr. Etty (Tika) Benveniste, Department of Cell, Developmental and Integrative Biology,

University of Alabama at Birmingham, 1900 University Boulevard, THT 926A,

Birmingham, AL 35294. Phone: 205-934-7667. E-mail address: [email protected]

Financial Support: This work was supported in part by National Institutes of Health

grants CA158534 (ENB), CA132077 (SP) and CA133737 (SP), American Cancer

Society grants RSG-11-259-01-CSM (DRH) and IRG-60-001-53 (JSD),

METAvivorResearch and Support, Inc. (DRH) and a grant from the American Brain

Tumor Association in honor of Paul Fabbri (BCM).

42

The authors disclose no potential conflicts of interest.

ABSTRACT

Suppressor Of Cytokine Signaling (SOCS) proteins are negative regulators of the

JAK/STAT pathway, and generally function as tumor suppressors. The absence of

SOCS3 in particular leads to heightened activation of the STAT3 transcription factor,

which has a striking ability to promote tumor survival while suppressing anti-tumor

immunity. We report for the first time that genetic deletion of SOCS3 specifically in

myeloid cells significantly enhances tumor growth, which correlates with elevated levels

of myeloid-derived suppressor cells (MDSC) in the tumor microenvironment, and

diminished CD8+ T-cell infiltration in tumors. The importance of MDSC in promoting

tumor growth is documented by reduced tumor growth upon depletion of MDSC.

Furthermore, SOCS3-deficient bone-marrow-derived cells exhibit heightened STAT3

activation and preferentially differentiate into the Gr-1+CD11b

+Ly6G

+ MDSC phenotype.

Importantly, we identify granulocyte-colony stimulating factor (G-CSF) as a critical

factor secreted by the tumor microenvironment that promotes development of MDSC via

a STAT3-dependent pathway. Abrogation of tumor-derived G-CSF reduces the

proliferation and accumulation of Gr-1+CD11b

+ MDSC and inhibits tumor growth. These

findings highlight the critical function of SOCS3 as a negative regulator of MDSC

development and function, via inhibition of STAT3 activation.

PRECIS

The loss of SOCS3, a negative regulator of STAT3, in myeloid cells, leads to the

development of MDSC and immunosuppressive activity within the tumor

43

microenvironment, via a G-CSF/STAT3 axis. Thus, SOCS3 in myeloid cells may serve

as a therapeutic target to regulate anti-tumor immunity.

INTRODUCTION

The Janus Kinase/Signal Transducers and Activators of Transcription

(JAK/STAT) signaling pathway is utilized by numerous cytokines, and is critical for

induction of innate and adaptive immunity, and ultimately suppressing inflammatory and

immune responses (1). Of the seven STAT proteins, STAT3 has been implicated in

inducing and maintaining an immunosuppressive tumor microenvironment (2, 3). The

persistent activation of STAT3 mediates tumor-promoting inflammation, tumor survival

and invasion, and suppression of anti-tumor immunity (3). Hyperactivation of STAT3 is

implicated in tumor progression and poor patient prognosis in a large number of cancers,

including breast, prostate, melanoma, pancreatic cancer and brain tumors (3). Activating

mutations in STAT3 are rare, thus STAT3 hyperactivation is usually caused by an over-

abundance of cytokines such as IL-6 and/or dysregulation of endogenous negative

regulators, most notably, Suppressors Of Cytokine Signaling (SOCS) proteins (4-6).

There are eight SOCS proteins: SOCS1-7 and CIS, which inhibit the duration of

cytokine-induced JAK/STAT signaling. The predominant function of SOCS3 is

inhibition of STAT3 activation by inhibiting JAK kinase activity (5, 7). As such, loss of

SOCS3 expression leads to hyperactivation of JAKs and downstream STAT3, and

expression of STAT3-mediated genes.

SOCS3 is tightly linked to cancer cell proliferation, as well as cancer-associated

inflammation (8). Yet, the role of SOCS3 in various types of cancer is controversial;

44

there are reports of either increased or reduced SOCS3 expression in breast and prostate

cancer (9-12). In other cancers, including gastric cancer, hepatocellular carcinoma, head

and neck squamous cell carcinoma and colon cancer, SOCS3 functions as a tumor

suppressor (8). The loss of SOCS3 expression by hypermethylation of the SOCS3

promoter is generally associated with poor clinical outcome, metastasis and aggressive

phenotype (9). In pre-clinical models, conditional knock-down of SOCS3 results in

accelerated tumorigenesis, which is associated with hyper-activation of various signaling

pathways, including STAT3 (8).

The inflammatory milieu within the microenvironment of cancers supports tumor

cell survival and angiogenesis. In tumor models and human cancers, innate leukocytes are

predominantly of myeloid origin, and are composed of tumor associated macrophages,

dendritic cells (DCs), and myeloid-derived suppressor cells (MDSC) (13, 14). MDSC,

characterized by expression of CD11b and Gr-1, are a heterogeneous population of

activated immature myeloid cells found within tumors that exert immunosuppressive

properties (13-15). MDSC have the capacity to suppress the cytotoxic activities of natural

killer (NK) and NKT cells, and adaptive immune responses elicited by CD4+ and CD8

+

T-cells (15, 16). Under normal conditions, Gr-1+CD11b

+ cells are maintained at very low

levels, but in patients with tumors, those cells can make up to 50% of total CD45+

hematopoietic cells in the tumor mass (17). Numbers of MDSC in tumors are negatively

associated with overall survival and treatment efficacy in patients with colorectal,

pancreatic and prostate cancer (18). In a tumor promoting environment, MDSC expand

and migrate from the bone marrow (BM) into the blood, spleen and tumors by numerous

cytokines and soluble mediators including M-CSF, G-CSF, GM-CSF, IL-6, IL-1, TNF-α

45

and S100A8/S100A9 (14, 19-23). The expansion and functional activation of MDSC

involves numerous transcription factors, with STAT3 being the most crucial (24).

We recently demonstrated that deletion of SOCS3 in myeloid cells (neutrophils,

DCs, monocytes/macrophages) leads to heightened activation of STAT3, and enhanced

expression of proinflammatory genes including IL-1, TNF-α, IL-6 and iNOS (25, 26). To

investigate the function of myeloid SOCS3 in tumor growth, the TRAMP model of

prostate cancer was examined in SOCS3 floxed (SOCS3fl/fl

) and SOCS3 myeloid specific

deletion (SOCS3MyeKO

) mice. Our results demonstrate that prostate tumor growth is

significantly enhanced in SOCS3MyeKO

mice, and is associated with elevated levels of Gr-

1+CD11b

+ MDSC in tumors of SOCS3-deficient mice. We identify G-CSF as a critical

factor secreted by the tumor microenvironment that promotes MDSC expansion via a

STAT3/SOCS3-dependent pathway. Abrogation of tumor-derived G-CSF inhibits tumor

growth by reducing the accumulation of MDSC. Our results highlight the important role

of myeloid SOCS3 in regulating the development of MDSC, and demonstrate that in the

absence of SOCS3, an immunosuppressive milieu is established in the tumor

microenvironment that promotes tumor growth.

46

MATERIALS AND METHODS

Mice. Mice 6-8 weeks of age were used. C57BL/6 and transgenic OT-1 mice (specific to

OVA peptide257-264) (27) were bred at the University of Alabama at Birmingham (UAB)

(Birmingham, AL). SOCS3 conditional knockout (SOCS3MyeKO

) mice were generated by

breeding of SOCS3fl/fl

mice (28) with mice expressing Cre recombinase under the control

of the LysM promoter, in which the conditional SOCS3 allele is excised in myeloid cells

(25). All experiments were approved by the IACUC of UAB.

Cell Lines and Primary Cells. Murine epithelial prostate cancer cells TRAMP-C1 and

TRAMP-C2 were cultured in DMEM medium with 10% FBS (Sigma, St. Louis, MO).

Conditioned media (CM) was generated by incubation of TRAMP-C1 or C2 cells for 48

h. Primary BM cells were flushed from the femur and tibia of mice (25, 26), and cultured

under conditions to generate MDCSs, including TRAMP CM, 10 ng/ml of murine GM-

CSF or 20 ng/ml of murine G-CSF for 3-4 days.

Peptides, Antibodies, Cytokines and Lentiviral Vector. OVA257-264 (SIINFEKL) was

obtained from AnaSpec (Fremont, CA). Antibodies (Abs) against phospho-STAT3

(Tyr705) and STAT3 were from Cell Signaling Technology (Beverly, MA), and Ab

against GAPDH was from Abcam (Cambridge, MA). Neutralizing Ab to G-CSF

(MAB414) and isotype control (IgG1) were from R&D Systems (San Diego, CA).

Neutralizing Ab to Gr-1 (RB6-8C5) and isotype control (IgG2b) were from BioXcell

(West Lebanon, NH). Recombinant murine G-CSF and GM-CSF were from R&D

Systems (San Diego, CA). Lentiviral expression of SOCS3 was generated as described

(29).

47

Tumor Models and Ab Treatment. TRAMP-C1 or C2 cells (3.0 × 106) in 100 μl of

PBS were s.c. inoculated in the flank of SOCS3fl/fl

or SOCS3MyeKO

mice. Tumor volumes

were calculated using the formula (0.5 × l × w2, where l is length and w is width). Three

weeks after s.c. inoculation of TRAMP-C1 cells, anti-mouse G-CSF Ab or isotype

control Ab (10 μg per mouse) was administered intraperitoneally every two days for a

total of 9 treatments (19, 20). For Gr-1+ cell depletion, mice were treated with anti-Gr-1

or control Ab (100 μg per mouse) every two days for a total of 9 treatments (21). In an

orthotopic model, 5.0 × 105 TRAMP-C2 cells in 50 μl of PBS were injected into the

ventrolateral prostate gland, and analyzed after 30 days (30).

Flow Cytometry. Subcutaneous TRAMP tumors, prostate containing tumor, and normal

prostate tissue were minced into fragments, and incubated in collagenase solution in the

presence of DNase I (1 mg/ml) at 37°C for 1 h. Dissociated cells were passed through a

100 μm cell strainer. Spleens or BM extracted from mouse femurs were homogenized

and passed through a 100 μm cell strainer. Cells were resuspended and stained with direct

labeled Abs against: CD45 (30-F11), CD11b (M1/70), Gr-1 (RB6-8C5), Ly6G (1A8) and

Ly6C (HK1.4) (BioLegend); and B220 (RA3-6B2), F4/80 (BM8) and CD11c (N418)

(eBioscience). For T cell analysis, Abs from BioLegend were used: CD3 (145-2C11),

CD4 (GK1.5), CD8 (53-6.7), IFN-γ (XMG1.2) and Foxp3 (MF-14). Mouse G-CSF

R/CD114 Ab was from R&D Systems. For intracellular staining, Abs against phospho-

STAT3 (Y705) and phospho STAT5 (Y694) (BioLegend) were used. Staining was

performed as previously described (25, 31). All samples were analyzed on an LSRIIB

FACS instrument and were further analyzed with FlowJo software.

48

Isolation of Gr-1+CD11b

+ Cells and Functional T-cell Suppression Assay. Single-cell

suspensions were prepared from tumors or spleens by digestion with Collagenase/Dispase

and DNase for 1 h at 37°C. Gr-1+CD11b

+ myeloid cells were sorted by FACSAria

(>90%). Splenocytes isolated from OT-1 mice were resuspended at 10 × 106 cells/ml and

incubated with CFSE (0.5 μM) at room temperature for 10 min. Suppression of T cells

was evaluated in a co-culture system with OT-1 splenocytes (1 × 106 per well) with

increasing ratios of Gr-1+CD11b

+ cells in the presence of OVA peptide (3 μg/ml). OT-1

CD8+

T-cells were evaluated on day 3 by flow cytometry for CFSE dilution. The percent

of dividing cells was determined by drawing a gate outside a reference CFSE peak from

unstimulated CFSE+ cells.

Nitric Oxide Detection, IFN-γ Production and Arginase Activity Assay. Nitric oxide

production was evaluated in supernatants from co-cultured Gr-1+CD11b

+ cells and OT-1

T-cells using the Griess Reagent System (Promega) (25). The same culture supernatants

were evaluated for IFN-γ production using an IFN-γ ELISA kit (BioLegend) (25).

Arginase activity was measured in cell lysates using the QuantichromTM Arginase Assay

Kit (BioAssay Systems, Hayward, CA) following the manufacturer’s instructions.

Cytology of Gr-1+CD11b

+ Cells. CD45

+Gr-1

+CD11b

+ cells were sorted from spleens or

tumors of TRAMP-C1 tumor-bearing mice using FACSAria. Cytospin preparations were

stained with Diff-Quick modified Giemsa reagent (Polysciences) according to the

manufacturer's protocols.

In Vivo Cell Proliferation Assay. Naïve or TRAMP-C1 tumor-bearing mice were

injected intraperitoneally with 100 μg EdU (Invitrogen). After 20 h, mice were sacrificed

49

and single-cell suspensions prepared from BM, spleens and tumors. Cells were stained

with Alexa Fluor 647 Azide according to the Click-iT® EdU Flow Cytometry Assay Kits

(Invitrogen) (31).

Histopathology and Immunofluorescence. Slides were stained with nuclear dye

(hematoxylin), and then stained with counterstain (eosin). Immunofluorescence was

performed with Abs to CD45 (1:50) or Gr-1 (1:100). Sections were fixed in 3%

formaldehyde, blocked with 10% donkey serum for 30 min, labeled with primary

antibody overnight at 4°C, then incubated with Alexa Fluor 568 secondary antibody

(Invitrogen; 1:200) along with DAPI (Invitrogen; 1:1000) for 1 h at room temperature.

Remaining steps were performed as described (25, 26).

RNA Isolation, RT-PCR, and Gene Expression Assays. Total RNA was isolated from

untreated or cytokine-stimulated MDSC (26). Five hundred ng of RNA was used to

reverse transcribe into cDNA and subjected to qRT-PCR. The abundance of mRNA was

normalized to that of 18S or HPRT (hypoxanthine guanine phosphoribosyl transferase)

and the data analyzed using the comparative Ct method to obtain relative quantitation

values (26).

Immunoblotting. Thirty μg of cell lysate was separated by electrophoresis on 10% SDS-

polyacrylamide gels and probed with specific Abs (26).

ELISA and Multiplex Analysis of Cytokine Expression. Tumor supernatants, TRAMP

tumor cell CM, and serum were collected and analyzed using a G-CSF ELISA kit (R&D

Systems). Millipore mouse cytokine/chemokine panel LI (MPXMCYTO-70K) (Millipore,

50

Billerica, MA) was used for detection of cytokines and chemokines. Expression levels of

cytokines/chemokines were normalized to total protein levels.

Statistical Analyses. Graphpad Prism v6.01 was used for the graphs and for statistics.

ANOVA test was performed. All results are shown as mean ± SD. A p value of < 0.05

was considered to be statistically significant.

51

RESULTS

Myeloid-specific SOCS3 Loss Promotes Tumor Growth. We utilized a syngeneic

tumor model of murine TRAMP C1 and C2 prostate cancer cells (32) inoculated into the

flank of C57BL/6 SOCS3fl/fl

and SOCS3MyeKO

mice. Tumor growth was significantly

increased in SOCS3MyeKO

mice compared with SOCS3fl/fl

mice (Fig. 1A).

Immunofluorescence staining demonstrates enhanced tumor infiltration of CD45+

hematopoietic-derived cells and Gr-1+ myeloid cells in SOCS3

MyeKO mice at day 45 (Fig.

1B). The Gr-1+CD11b

+ cell population was substantially increased in SOCS3

MyeKO mice

compared to SOCS3fl/fl

mice (Fig. 1C), whereas the percentages of other cells such as

macrophages (F4/80high

, CD11b+), DCs (CD11b

+, CD11c

+), B-cells (B220

+) and T-cells

(CD3+) were largely unchanged (Fig. 1D). Associated with the increased frequency of

Gr-1+CD11b

+ cells in the tumor, there was a significant reduction in infiltrating CD8

+ T-

cells in the tumors of SOCS3MyeKO

mice (Fig. 1E). The percentage of Gr-1+CD11b

+ cells

was elevated in the spleen of SOCS3MyeKO

mice compared to SOCS3fl/fl

mice (Fig. S1A),

although no changes in CD8+ T-cells were observed in the spleen (Fig. S1B). Similar

percentages of CD4+Foxp3

+CD25

+ regulatory T-cells (Treg) were detected in tumors

from SOCS3MyeKO

and SOCS3fl/fl

mice (Fig. S1C). To determine if the increased tumor

growth observed in SOCS3MyeKO

mice is due to enhanced MDSC function, depletion of

Gr-1+ MDSC with anti-Gr-1 neutralizing antibody in tumor-bearing mice was conducted.

TRAMP-C1 cells were injected into the flanks of SOCS3fl/fl

and SOCS3MyeKO

mice, and

anti-Gr-1 mAb or isotype control was administrated at day 24 (Fig. S1D). We observed

reduced levels of MDSC in the spleen of SOCS3MyeKO

mice after treatment with

52

neutralizing Gr-1 mAb (Fig. S1E). More importantly, tumor growth was significantly

reduced after treatment with anti-Gr-1 mAb in SOCS3MyeKO

mice (Fig. 1F). These results

suggest a critical role of Gr-1+ MDSC in contributing to tumor growth in SOCS3

MyeKO

mice.

Gr-1+CD11b

+ Cells from Tumor-bearing Mice Suppress Antigen-specific T-cell

Responses. Gr-1+CD11b

+ cells from the tumors of TRAMP-C1 mice displayed multi-

lobed nuclei (Fig. 2A). Gr-1 identifies the monocyte and neutrophil markers Ly6C and

Ly6G, respectively. MDSC consist of two major subsets of Ly6G+Ly6C

low granulocytic

and Ly6G-Ly6C

hi monocytic cells (33). Gr-1

+CD11b

+ cells from TRAMP-C1 tumors are

a mixture of granulocytic and monocytic cells, with the majority of cells expressing

Ly6G (Fig. 2B). Further, higher percentages of both granulocytic and monocytic MDSC

were detected in SOCS3MyeKO

mice (Fig. 2B). We next examined the influence of SOCS3

on various MDSC effector functions, first testing the ability of Gr-1+CD11b

+ cells

isolated from tumors to suppress in vitro proliferation of OVA-specific CD8+ T-cells.

The immunosuppressive function of tumor-associated MDSC isolated from SOCS3MyeKO

mice was more potent than MDSC from SOCS3fl/fl

mice (Fig. 2C). MDSC from the

tumors of SOCS3MyeKO

mice decreased IFN-γ production by T-cells more potently (Fig.

S2A) and produced higher levels of nitrite upon co-culture with OT-1 splenocytes (Fig.

S2B) than MDSC from SOCS3fl/fl

mice. Gene expression analysis of MDSC isolated from

tumors was performed. Loss of SOCS3 increased expression of mediators of immune

suppression, including Arginase 1 and S100A8, with a trend of increased iNOS and

S100A9 (Fig. 2D). MDSC from the spleen of SOCS3MyeKO

mice inhibited T-cell

53

proliferation more potently (Fig. S2C) and had significantly increased arginase activity

compared to MDSC from SOCS3fl/fl

mice (Fig. S2D). Furthermore, Gr-1+CD11b

+ cells

isolated from the BM, spleen and tumor of SOCS3MyeKO

mice had elevated STAT3

activation compared to cells from SOCS3fl/fl

mice (Fig. 2E).

Orthotopic Prostate Tumor Growth is Enhanced in SOCS3MyeKO

Mice. We next

investigated the functional importance of SOCS3 in myeloid cells in the prostate

microenvironment. TRAMP-C2 cells represent more advanced prostate cancer than

TRAMP-C1 cells, due to enhanced tumor growth and elevated expression of Ras and

Myc (34). TRAMP-C2 cells were injected intraprostatically into SOCS3fl/fl

and

SOCS3MyeKO

mice (30), and prostate weight compared amongst the two groups.

Orthotopic growth of TRAMP-C2 cells was significantly increased in SOCS3MyeKO

mice

(Fig. 3A). H&E staining of TRAMP-C2 tumors in SOCS3fl/fl

and SOCS3MyeKO

mice

showed poorly differentiated tumors compared to control prostate (Fig. 3B). Prostate

tumors from SOCS3MyeKO

mice have higher numbers of infiltrating CD45+ cells and Gr-

1+ cells (Fig. 3B). Orthotopic TRAMP-C2 tumors in SOCS3

MyeKO mice exhibited

increased Gr-1+CD11b

+ cell infiltration compared to SOCS3

fl/fl mice (Fig. 3C), and a

significant reduction in the percentage of infiltrating CD8+ T-cells (Fig. 3D).

Significantly higher levels of S100A8 and S100A9, inflammation and cancer-promoting

factors expressed by Gr-1+CD11b

+ MDSC (35), were detected in prostate tumors from

SOCS3MyeKO

mice compared with SOCS3fl/fl

mice (Fig. 3E).

Loss of SOCS3 Promotes Proliferation of Gr-1+CD11b

+ Cells. TRAMP tumor-bearing

mice developed splenomegaly compared to naïve mice, and SOCS3MyeKO

mice showed a

54

significant increase in spleen size and weight compared to SOCS3fl/fl

mice (Fig. 4A).

Perturbed myelopoiesis and spleen hematopoiesis (36) may account for the accumulation

of Gr-1+CD11b

+ cells

in tumors and increased spleen size. To address the role of SOCS3

on myeloid cell proliferation, EdU assays were performed. We observed no differences in

myelopoiesis (Fig. S3A), spleen size (Fig. S3B) and BM myeloid cell proliferation (Fig.

S3C) between naïve SOCS3fl/fl

and SOCS3MyeKO

mice. Gr-1+CD11b

+ cells from the

spleen of naïve SOCS3fl/fl

mice had ~6.4% proliferating cells, and both TRAMP-C1

tumor-bearing SOCS3fl/fl

and SOCS3MyeKO

mice showed increased proliferating cells

compared with naïve mice (Fig. 4B). Additionally, the percentage of proliferating Gr-

1+CD11b

+ cells from the spleen of SOCS3

MyeKO mice was higher than that of SOCS3

fl/fl

mice. Similarly, Gr-1High

CD11b+

cells and Gr-1Int

CD11b+

cells from the BM of TRAMP-

C1 tumor-bearing SOCS3MyeKO

mice exhibited an enhanced proliferation rate (Fig. 4C).

TRAMP tumor development was associated with a 2 - 3 fold increase in circulating white

blood cells (WBC) in SOCS3MyeKO

mice (Fig. 4D), due to a 3 - 5 fold increase in

granulocytes (Fig. 4E). This suggests that factors secreted by the tumor may increase the

mobility of BM progenitor cells to enter the bloodstream.

Next, we cultured BM cells from SOCS3fl/fl

and SOCS3MyeKO

mice in the absence

or presence of TRAMP conditioned media (CM) for 4 days, and total numbers of Gr-

1+CD11b

+ cells were determined by flow cytometry. The absolute number of Gr-

1+CD11b

+ cells generated from SOCS3

MyeKO mice was ~ 2-fold higher than SOCS3

fl/fl

mice (Fig. S3D). Cell cycle analysis was performed, and results indicate increased

numbers of SOCS3fl/fl

BM cells in S and M phase compared with SOCS3fl/fl

BM cells

55

(Fig. S3E). These results indicate that factors from the tumor microenvironment

contribute to the differentiation and proliferation of Gr-1+CD11b

+ cells, and that SOCS3

plays a critical role in regulating myeloid cell proliferative responses to these factors.

Absence of SOCS3 Enhances STAT3 Activation and BM Differentiation into Gr-

1+CD11b

+ MDSC. We utilized CM from TRAMP cells to assess functional effects on

MDSC. TRAMP CM, but not control medium, promoted BM cells to differentiate into

Gr-1+CD11b

+ cells, with higher percentages detected in SOCS3

MyeKO mice (Fig. 5A). In

addition, Gr-1+CD11b

+ cells from SOCS3

MyeKO mice incubated in TRAMP CM exhibited

a greater capacity to suppress antigen-specific T-cell proliferation (Fig. 5B) and IFN-γ

production (Fig. 5C) than did cells from SOCS3fl/fl

mice. Gr-1+CD11b

+ cells lacking

SOCS3 also produced elevated levels of nitrite (Fig. 5D) and had increased arginase

activity (Fig. 5E). Cells from SOCS3MyeKO

mice showed enhanced and prolonged STAT3

activation in the presence of TRAMP CM compared with SOCS3fl/fl

mice (Fig. 5F). Loss

of SOCS3 increased expression of Arginase 1, iNOS, S100A8, S100A9 and IDO1 (37)

by BM-derived Gr-1+CD11b

+ cells incubated with TRAMP CM (Fig. 5G). TRAMP CM

cultured Gr-1+CD11b

+ cells had low expression of MHC class II, with SOCS3

MyeKO cells

expressing less MHC class II compared to SOCS3fl/fl

cells (Fig. 5H), suggesting that loss

of SOCS3 maintains MDSC in a undifferentiated state. The majority of Gr-1+CD11b

+

cells generated in the presence of TRAMP CM express Ly6G (Fig. 5I). These results

demonstrate that prostate tumor cells secrete factors that drive the generation of Gr-

1+CD11b

+ cells from hematopoietic progenitor cells of the BM, and this effect is

amplified in the absence of SOCS3.

56

G-CSF Induces the Differentiation of BM Precursors into Functional MDSC in a

STAT3-dependent Manner. We next examined potential prostate tumor-derived soluble

factors that may contribute to the generation of Gr-1+CD11b

+ cells. Expression of G-CSF

in the serum of naïve and TRAMP tumor-bearing mice was examined, and elevated

levels of G-CSF were detected from mice with orthotopic or s.c. TRAMP tumors (Fig.

S4A). Prostate tissues from orthotopic tumor-bearing mice and flank tumor tissue were

evaluated for G-CSF concentrations. Higher amounts of G-CSF were detected in TRAMP

tumor-bearing mice compared to tissue from naïve mice (Fig. 6A), although the levels of

G-CSF were not different between tumor-bearing SOCS3MyeKO

and SOCS3fl/fl

mice. CM

from TRAMP cells also contained G-CSF (Fig. 6A). The expression of G-CSF was

validated at the mRNA level (Fig. S4B). Enhanced and prolonged activation of STAT3

was observed in G-CSF treated BM cells from SOCS3MyeKO

mice (Fig. 6B), which was

validated by flow cytometry (Fig. 6C). G-CSF also activated STAT5 in Gr-1+CD11b

+

cells, but SOCS3 deficiency did not affect this response (Fig. 6C). SOCS3 mRNA

expression was induced by G-CSF treatment of cells from SOCS3fl/fl

mice, but not in cells

from SOCS3MyeKO

mice (Fig. 6D). Inclusion of G-CSF in BM cultures promoted

differentiation into Gr-1+CD11b

+ cells, with SOCS3

MyeKO cells having higher levels (Fig.

6E, left). To validate the negative regulatory role of SOCS3 in G-CSF promotion of

MDSC, SOCS3 overexpression was utilized (29). Overexpression of SOCS3 in BM cells

from SOCS3fl/fl

and SOCS3MyeKO

mice diminished the generation of Gr-1+CD11b

+ cells

in response to G-CSF (Fig. 6E, right). To substantiate the involvement of STAT3 in

MDSC differentiation, JAK and STAT3 inhibitors were tested. A significant reduction in

57

Gr-1+CD11b

+ cells in the presence of the pan-JAK inhibitor P6 (38) or the STAT3

inhibitor Stattic (39) was observed (Fig. 6F). These results indicate that SOCS3

negatively regulates G-CSF induced generation of Gr-1+CD11b

+ cells by inhibiting

JAK/STAT3 activation. In the presence of G-CSF, loss of SOCS3 increased expression

of Arginase 1, iNOS, S100A8, S100A9 and IDO1 (Fig. 6G). Furthermore, Gr-1+CD11b

+

cells from SOCS3MyeKO

mice incubated in G-CSF exhibited a greater capacity to suppress

T-cell proliferation (Fig. 6H), and had increased arginase activity compared to Gr-

1+CD11b

+ cells from SOCS3

fl/fl mice (Fig. 6I). Thus, we identify G-CSF as an important

factor in the development of Gr-1+CD11b

+ MDSC via STAT3 activation, which is

regulated by SOCS3.

G-CSF Neutralization Limits Gr-1+CD11b

+ Cell Proliferation and Tumor Growth.

BM cells isolated from SOCS3fl/fl

and SOCS3MyeKO

mice were cultured in the presence of

TRAMP CM or G-CSF. Flow cytometric analysis demonstrates that addition of anti-G-

CSF Ab, but not isotype IgG, inhibits generation of Gr-1+CD11b

+ cells (Fig. S5A).

Neutralization of G-CSF partially inhibited BM cell proliferation in the presence of

TRAMP CM, and abolished proliferation induced by G-CSF (Fig. 7A).

The contribution of G-CSF to tumor growth was next assessed. TRAMP-C1 cells

were injected into the flanks of SOCS3fl/fl

and SOCS3MyeKO

mice, and anti-G-CSF mAb

or isotype control was administrated at day 22 (Fig. 7B). In both SOCS3fl/fl

and

SOCS3MyeKO

mice, tumor volume (Fig. 7B), weight (Fig. 7C) and serum G-CSF levels

(Fig. S5B) were significantly reduced after treatment with anti-G-CSF mAb. Circulating

WBC (Fig. S5C) and granulocytes (Fig. S5D) were also decreased after treatment with

58

neutralizing G-CSF mAb in SOCS3MyeKO

mice. Moreover, infiltration of MDSC in

tumors was significantly reduced after treatment with neutralizing G-CSF mAb (Fig. 7D),

as were levels of MDSC in the spleen of SOCS3MyeKO

mice, but not SOCS3fl/fl

mice (Fig.

7E). These data highlight the importance of myeloid SOCS3 in regulating prostate tumor

growth in response to G-CSF. To exclude a direct action of anti-G-CSF Ab on TRAMP-

C1 tumor cells, G-CSFR expression was assessed. We observed high levels of G-CSFR

expression on spleen or tumor Gr-1+ cells compared with TRAMP-C1 tumor cells and

non-myeloid CD11b- cells from tumor-bearing mice (Fig. S5E).

59

DISCUSSION

We demonstrate that SOCS3 expression in myeloid cells is an important determinant of

tumor growth, indicating a critical influence of the tumor microenvironment in cancer

progression. The loss of SOCS3 in myeloid cells promotes the differentiation of BM-

derived progenitor cells into Gr-1+CD11b

+ MDSC, and enhances the immunosuppressive

functions of these cells. Importantly, STAT3 activation is essential for this process.

Tumor-derived G-CSF is crucial for the mobilization and recruitment of Gr-1+CD11b

+

MDSC to tumors, where they decrease the presence of CD8+ T-cells. Neutralization of

G-CSF is effective in limiting the differentiation and functionality of MDSC, which in

vivo manifests as restricted tumor growth. These findings establish a circuitry between

MDSC, tumor cells and G-CSF in the tumor microenvironment; G-CSF secreted by

tumor cells promotes the recruitment and differentiation of MDSC, which is dependent

on STAT3 activation in MDSC. In the absence of SOCS3, this response and circuitry is

amplified (Fig. 7F). These results support a role for SOCS3 in repressing MDSC

differentiation, which ultimately relieves immunosuppression in the prostate tumor

microenvironment.

Persistent STAT3 activation is associated with poor prognosis in many cancer

types, including prostate cancer patients (40). Loss of the androgen receptor (AR) leads

to the development of prostate cancer stem cells, which requires STAT3 activation (41).

Stem-like cells from patients with prostate cancer secrete high levels of IL-6 and exhibit

hyperactivation of STAT3 (42). STAT3 activation in cells of the hematopoietic lineage is

60

also associated with creating a tumor microenvironment conducive to tumor growth. This

is especially true for MDSC, which rely on STAT3 for differentiation and functionality.

Ablation of STAT3 in multiple lineages of immune cells (neutrophils, NK cells, DCs)

enhanced their anti-tumor activity (39). While MDSC were not directly examined,

heightened activation of STAT3 in MDSC, as shown by our study, may contribute to

tumor immune tolerance. MDSC lacking SOCS3 with STAT3 hyperactivation have

potent immunosuppressive capabilities such as inhibition of antigen-specific CD8+ T-cell

proliferation and IFN-γ production, and enhanced nitrite production and arginase activity.

Also, SOCS3-deficient MDSC expressed MHC Class II at lower levels than SOCS3

wild-type MDSC, suggestive of an immature suppressive phenotype. Circulating MDSC

from prostate cancer patients displayed reduced expression of HLA-DR compared to age-

matched controls, and had more potent ability to suppress T-cell proliferation (43). It will

be informative to examine SOCS3 expression and STAT3 activation status in prostate

tumor cells, prostate stem cells and prostate tumor-associated MDSC to determine the

functional impact of SOCS3/STAT3 in patients with prostate cancer. Nonetheless, our

findings clearly demonstrate the importance of SOCS3 in restricting MDSC-mediated

anti-tumor immunity.

Using TRAMP C1/C2 cell lines in immune competent mice, elevated levels of

MDSC were detected in the tumors of SOCS3MyeKO

mice compared to SOCS3fl/fl

mice, in

both flank and orthotopic models. This was associated with a decrease of CD8+ T-cells

in the tumor, which supports the notion that MDSC interfere with T-cell activation and

proliferation. MDSC also selectively expand Foxp3+CD25

+ Treg, which promote tumor

61

growth by a variety of mechanisms (44). In our studies, Foxp3+ Treg were present at

comparable levels in tumors from both SOCS3MyeKO

and SOCS3fl/fl

mice, thus, Treg may

not have a prominent role in the TRAMP prostate cancer model (45).

G-CSF plays a crucial role in hematopoiesis by stimulating the proliferation,

differentiation and survival of myeloid progenitor cells, particularly cells within the

granulocytic lineage (28). JAK1, JAK2 and TYK2 are recruited to the G-CSF receptor

upon stimulation with G-CSF, and then in turn activate STAT1, STAT3 and STAT5, of

which STAT3 is most important. SOCS3 is induced by G-CSF in myeloid cells, and

serves as a negative regulator of G-CSF-induced cellular responses by binding to the G-

CSF receptor (28, 46). G-CSF is one of a number of soluble mediators that promote the

expansion and migration of MDSC from the bone marrow to tumors. While the levels of

G-CSF were comparable in SOCS3MyeKO

and SOCS3fl/fl

tumor-bearing mice,

SOCS3MyeKO

BM-derived cells are hyper-responsive to G-CSF, as documented by

increased duration and intensity of G-CSF-induced STAT3 phosphorylation, which led to

MDSC differentiation at a greater percentage than cells from SOCS3fl/fl

mice. In addition,

G-CSF-induced MDSC from SOCS3MyeKO

tumor-bearing mice exhibited a greater

capacity to suppress antigen-specific T-cell proliferation. Our in vivo findings

demonstrate that treatment of tumor-bearing mice with neutralizing G-CSF antibody

reduced circulating granulocytic cells as well as infiltration of MDSC in tumors. These

findings highlight the critical role of tumor-derived G-CSF in MDSC development, and

the importance of SOCS3 as an essential negative regulator of this process. SOCS3

expression in MDSC negatively regulates the expression of soluble mediators such as

62

S100A8 and S100A9 and products of iNOS and arginase 1 that support an

immunosuppressive milieu in tumors. In the absence of SOCS3, MDSC are hyper-

responsive to tumor-produced cytokines such as G-CSF, and aberrantly activate STAT3,

which in turn contributes to chronic cancer-related inflammation and suppression of anti-

tumor immune responses.

Clinical information regarding SOCS3 expression in prostate cancer is

inconclusive at this time. Pierconti et al, (9) demonstrated that methylation of the SOCS3

promoter was significantly associated with intermediate-high grade Gleason score and

with an unfavorable outcome. In benign prostate hyperplasia and normal controls, the

SOCS3 promoter was unmethylated. Analysis of the Oncomine database demonstrates

that SOCS3 mRNA expression is significantly lower in prostate carcinoma compared to

prostate cancer precursor (Luo dataset), but the Tomlins dataset shows the opposite

results (47). Data from cBioPortal (Prostate Adenocarcinoma MSKCC) indicates that

prostate cancer patients with SOCS3 mRNA overexpression trend towards a longer

disease-free time than patients with “normal” levels of SOCS3 mRNA (48). Thus, there

is clearly dysregulation of SOCS3 gene expression in patients with prostate cancer, but

the functional and clinical relevance is still under investigation. Nonetheless, it is clear

that a variety of mechanisms, including SOCS3 dysregulation and abundant IL-6

production, do contribute to hyperactivation of JAK/STAT pathway in animal models of

prostate cancer and patients (4, 42). Inhibitors of IL-6, JAKs and STAT3 are being

considered in the context of prostate cancer (4), and have already proven beneficial in

animal models (42). Furthermore, peptides that mimic the SOCS Kinase Inhibitory

63

Region (KIR), which is responsible for binding to JAKs and suppressing downstream

STAT3 activation (49), may prove beneficial in prostate cancer.

These studies not only highlight the significance of the STAT3/SOCS3 pathway

in regulating the differentiation and function of MDSC in cancer, but also identify this

intricate protein network as important therapeutic targets to eliminate MDSC-mediated

immunosuppression. This is especially important in light of the recent finding that MDSC

are responsible for resistance to immune check-point inhibitor, and that elimination of

MDSC led to cures of experimental, metastatic tumors (50).

64

ACKNOWLEDGMENTS

The assistance of the UAB Comprehensive Arthritis, Musculoskeletal, and

Autoimmunity Center Comprehensive Flow Cytometry Core (P30 AR48311) is

gratefully acknowledged.

Author contributions: H.Y., J.S.D., E.N.B., and H.Q. designed research; H.Y., Y.L.,

B.C.M, and H.Q. performed research; H.Y., E.N.B., and H.Q. analyzed data; and H.Y.,

J.S.D., D.R.H., S.P., E.N.B., and H.Q. wrote the manuscript.

65

REFERENCES

1. O'Shea JJ, Plenge R. JAK and STAT signaling molecules in immunoregulation

and immune-mediated disease. Immunity 2012;36:542-50.

2. Brantley EC, Benveniste EN. Signal transducer and activator of transcription-3: a

molecular hub for signaling pathways in gliomas. Mol Cancer Res 2008;6:675-84.

3. Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading

role for STAT3. Nat Rev Cancer 2009;9:798-809.

4. Guo Y, Xu F, Lu T, Duan Z, Zhang Z. Interleukin-6 signaling pathway in targeted

therapy for cancer. Cancer Treat Rev 2012;38:904-10.

5. Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and immune

regulation. Nat Rev Immunol 2007;7:454-65.

6. Li Y, de Haar C, Peppelenbosch MP, van der Woude CJ. SOCS3 in immune

regulation of inflammatory bowel disease and inflammatory bowel disease-related

cancer. Cytokine Growth Factor Rev 2012;23:127-38.

7. Babon JJ, Kershaw NJ, Murphy JM, Varghese LN, Laktyushin A, Young SN, et

al. Suppression of cytokine signaling by SOCS3: characterization of the mode of

inhibition and the basis of its specificity. Immunity 2012;36:239-50.

8. Inagaki-Ohara K, Kondo T, Ito M, Yoshimura A. SOCS, inflammation, and

cancer. Jakstat 2013;2:e24053.

9. Pierconti F, Martini M, Pinto F, Cenci T, Capodimonti S, Calarco A, et al.

Epigenetic silencing of SOCS3 identifies a subset of prostate cancer with an

aggressive behavior. Prostate 2011;71:318-25.

10. Calarco A, Pinto F, Pierconti F, Sacco E, Marrucci E, Totaro A, et al. Role of

SOCS3 evaluated by immunohistochemical analysis in a cohort of patients

affected by prostate cancer: preliminary results. Urologia 2012;79 Suppl 19:4-8.

11. Puhr M, Santer FR, Neuwirt H, Susani M, Nemeth JA, Hobisch A, et al. Down-

regulation of suppressor of cytokine signaling-3 causes prostate cancer cell death

through activation of the extrinsic and intrinsic apoptosis pathways. Cancer Res

2009;69:7375-84.

12. Kneitz B, Krebs M, Kalogirou C, Schubert M, Joniau S, van Poppel H, et al.

Survival in patients with high-risk prostate cancer is predicted by miR-221, which

regulates proliferation, apoptosis, and invasion of prostate cancer cells by

inhibiting IRF2 and SOCS3. Cancer Res 2014;74:2591-603.

13. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of

myeloid cells by tumours. Nat Rev Immunol 2012;12:253-68.

14. Khaled YS, Ammori BJ, Elkord E. Myeloid-derived suppressor cells in cancer:

recent progress and prospects. Immunol Cell Biol 2013;91:493-502.

15. Talmadge JE, Gabrilovich DI. History of myeloid-derived suppressor cells. Nat

Rev Cancer 2013;13:739-52.

16. Mauti LA, Le Bitoux MA, Baumer K, Stehle JC, Golshayan D, Provero P, et al.

Myeloid-derived suppressor cells are implicated in regulating permissiveness for

tumor metastasis during mouse gestation. J Clin Invest 2011;121:2794-807.

17. Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the

promotion of tumour angiogenesis. Nat Rev Cancer 2008;8:618-31.

18. Vanneman M, Dranoff G. Combining immunotherapy and targeted therapies in

cancer treatment. Nat Rev Cancer 2012;12:237-51.

66

19. Shojaei F, Wu X, Qu X, Kowanetz M, Yu L, Tan M, et al. G-CSF-initiated

myeloid cell mobilization and angiogenesis mediate tumor refractoriness to anti-

VEGF therapy in mouse models. Proc Natl Acad Sci U S A 2009;106:6742-7.

20. Waight JD, Hu Q, Miller A, Liu S, Abrams SI. Tumor-derived G-CSF facilitates

neoplastic growth through a granulocytic myeloid-derived suppressor cell-

dependent mechanism. PLoS One 2011;6:e27690.

21. Bayne LJ, Beatty GL, Jhala N, Clark CE, Rhim AD, Stanger BZ, et al. Tumor-

derived granulocyte-macrophage colony-stimulating factor regulates myeloid

inflammation and T cell immunity in pancreatic cancer. Cancer Cell 2012;21:822-

35.

22. Zhao X, Rong L, Zhao X, Li X, Liu X, Deng J, et al. TNF signaling drives

myeloid-derived suppressor cell accumulation. J Clin Invest 2012;122:4094-104.

23. Cheng P, Corzo CA, Luetteke N, Yu B, Nagaraj S, Bui MM, et al. Inhibition of

dendritic cell differentiation and accumulation of myeloid-derived suppressor

cells in cancer is regulated by S100A9 protein. J Exp Med 2008;205:2235-49.

24. Waight JD, Netherby C, Hensen ML, Miller A, Hu Q, Liu S, et al. Myeloid-

derived suppressor cell development is regulated by a STAT/IRF-8 axis. J Clin

Invest 2013;123:4464-78.

25. Qin H, Holdbrooks AT, Liu Y, Reynolds SL, Yanagisawa LL, Benveniste EN.

SOCS3 deficiency promotes M1 macrophage polarization and inflammation. J

Immunol 2012;189:3439-48.

26. Qin H, Yeh W-I, De Sarno P, Holdbrooks AT, Liu Y, Muldowney MT, et al.

Signal transducer and activator of transcription-3/suppressor of cytokine

signaling-3 (STAT3/SOCS3) axis in myeloid cells regulates neuroinflammation.

Proc Natl Acad Sci U S A 2012;109:5004-9.

27. Rodriguez PC, Quiceno DG, Zabaleta J, Ortiz B, Zea AH, Piazuelo MB, et al.

Arginase I production in the tumor microenvironment by mature myeloid cells

inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer

Res 2004;64:5839-49.

28. Croker BA, Metcalf D, Robb L, Wei W, Mifsud S, DiRago L, et al. SOCS3 is a

critical physiological negative regulator of G-CSF signaling and emergency

granulopoiesis. Immunity 2004;20:153-65.

29. Park KW, Nozell SE, Benveniste EN. Protective role of STAT3 in NMDA and

glutamate-induced neuronal death: negative regulatory effect of SOCS3. PLoS

One 2012;7:e50874.

30. Gurusamy D, Gray JK, Pathrose P, Kulkarni RM, Finkleman FD, Waltz SE.

Myeloid-specific expression of Ron receptor kinase promotes prostate tumor

growth. Cancer Res 2013;73:1752-63.

31. Liu Y, Holdbrooks AT, De Sarno P, Rowse AL, Yanagisawa LL, McFarland BC,

et al. Therapeutic efficacy of suppressing the JAK/STAT pathway in multiple

models of experimental autoimmune encephalomyelitis. J Immunol 2014;192:59-

72.

32. Greenberg NM, DeMayo F, Finegold MJ, Medina D, Tilley WD, Aspinall JO, et

al. Prostate cancer in a transgenic mouse. Proc Natl Acad Sci U S A

1995;92:3439-43.

67

33. Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of myeloid-derived

suppressor cells in tumor-bearing mice. J Immunol 2008;181:5791-802.

34. Isayeva T, Chanda D, Kallman L, Eltoum IE, Ponnazhagan S. Effects of sustained

antiangiogenic therapy in multistage prostate cancer in TRAMP model. Cancer

Res 2007;67:5789-97.

35. Acharyya S, Oskarsson T, Vanharanta S, Malladi S, Kim J, Morris PG, et al. A

CXCL1 paracrine network links cancer chemoresistance and metastasis. Cell

2012;150:165-78.

36. Sio A, Chehal MK, Tsai K, Fan X, Roberts ME, Nelson BH, et al. Dysregulated

hematopoiesis caused by mammary cancer is associated with epigenetic changes

and hox gene expression in hematopoietic cells. Cancer Res 2013;73:5892-904.

37. Smith C, Chang MY, Parker KH, Beury DW, DuHadaway JB, Flick HE, et al.

IDO is a nodal pathogenic driver of lung cancer and metastasis development.

Cancer Discov 2012;2:722-35.

38. Meares GP, Liu Y, Rajbhandari R, Qin H, Nozell SE, Mobley JA, et al. PERK

dependent activation of JAK1 and STAT3 contributes to ER stress induced

inflammation. Mol Cell Biol 2014;34:3911-25.

39. Vasquez-Dunddel D, Pan F, Zeng Q, Gorbounov M, Albesiano E, Fu J, et al.

STAT3 regulates arginase-I in myeloid-derived suppressor cells from cancer

patients. J Clin Invest 2013;123:1580-9.

40. Mora LB, Buettner R, Seigne J, Diaz J, Ahmad N, Garcia R, et al. Constitutive

activation of STAT3 in human prostate tumors and cell lines: direct inhibition of

STAT3 signaling induces apoptosis of prostate cancer cells. Cancer Res

2002;62:6659-66.

41. Schroeder A, Herrmann A, Cherryholmes G, Kowolik C, Buettner R, Pal S, et al.

Loss of androgen receptor expression promotes a stem-like cell phenotype in

prostate cancer through STAT3 signaling. Cancer Res 2014;74:1227-37.

42. Kroon P, Berry PA, Stower MJ, Rodrigues G, Mann VM, Simms M, et al. JAK-

STAT blockade inhibits tumor initiation and clonogenic recovery of prostate

cancer stem-like cells. Cancer research 2013;73:5288-98.

43. Vuk-Pavlovic S, Bulur PA, Lin Y, Qin R, Szumlanski CL, Zhao X, et al.

Immunosuppressive CD14+HLA-DRlow/- monocytes in prostate cancer. Prostate

2010;70:443-55.

44. Nagaraj S, Youn JI, Gabrilovich DI. Reciprocal relationship between myeloid-

derived suppressor cells and T cells. J Immunol 2013;191:17-23.

45. Degl'Innocenti E, Grioni M, Capuano G, Jachetti E, Freschi M, Bertilaccio MT, et

al. Peripheral T-cell tolerance associated with prostate cancer is independent from

CD4+CD25+ regulatory T cells. Cancer Res 2008;68:292-300.

46. Hortner M, Nielsch U, Mayr LM, Johnston JA, Heinrich PC, Haan S. Suppressor

of cytokine signaling-3 is recruited to the activated granulocyte-colony

stimulating factor receptor and modulates its signal transduction. J Immunol

2002;169:1219-27.

47. Oncomine. Prostate cancer 2015 [cited 2014 10 December]; Available from:

https://www.oncomine.org/resource/main.html#a%3A985%3Bd%3A46%3Bdso

%3AgeneOverex%3Bdt%3ApredefinedClass%3Bec%3A%5B2%5D%3Bepv%3

A150001.151078%2C2937%2C3508%3Bet%3Aover%3Bf%3A33306%3Bg%3A

68

9021%3Bgt%3Aboxplot%3Bp%3A200000762%3Bpg%3A1%3Bpvf%3A3483%

2C36061%2C150003%2C150004%3Bscr%3Adatasets%3Bss%3Aanalysis%3Bv

%3A18

48. Genomics cfC. Prostate Adenocarcinoma (MSKCC, Cancer Cell 2010). 2015;

Available from:

http://www.cbioportal.org/index.do?cancer_study_id=prad_mskcc&genetic_profil

e_ids_PROFILE_MUTATION_EXTENDED=prad_mskcc_mutations&genetic_p

rofile_ids_PROFILE_COPY_NUMBER_ALTERATION=prad_mskcc_cna&gen

etic_profile_ids_PROFILE_MRNA_EXPRESSION=prad_mskcc_mrna_zbynorm

&Z_SCORE_THRESHOLD=2.0&data_priority=0&case_set_id=prad_mskcc_co

mplete&case_ids=&gene_set_choice=user-defined-

list&gene_list=SOCS3%0D%0A&clinical_param_selection=null&tab_index=tab

_visualize&Action=Submit

49. Madonna S, Scarponi C, Doti N, Carbone T, Cavani A, Scognamiglio PL, et al.

Therapeutical potential of a peptide mimicking the SOCS1 kinase inhibitory

region in skin immune responses. Eur J Immunol 2013;43:1883-95.

50. Kim K, Skora AD, Li Z, Liu Q, Tam AJ, Blosser RL, et al. Eradication of

metastatic mouse cancers resistant to immune checkpoint blockade by

suppression of myeloid-derived cells. Proc Natl Acad Sci U S A 2014;111:11774-

9.

69

FIGURES

Figure 1. Myeloid-specific SOCS3 Loss Promotes Tumor Growth. (A) SOCS3fl/fl

(n=25) and SOCS3

MyeKO (n=22) mice were inoculated s.c. with 3.0 x 10

6 TRAMP-C1 or

70

C2 cells. Tumor size is indicated as volume (mm3 ± SD), and was evaluated up to 45 days.

(B) H&E staining of TRAMP-C1 tumors from mice at 45 days. Scale bar, 50 μm.

Immunofluorescence staining of tumor infiltrating CD45+ or Gr-1

+ cells, n = 3 for

SOCS3fl/fl

mice, n = 3 for SOCS3MyeKO

mice. Scale bar, 45 μm. (C) Flow cytometry plot

of Gr-1+CD11b

+ cells from tumors of TRAMP-C1 mice (top), and quantification of

tumor-infiltrating Gr-1+CD11b

+ cells (bottom) (n = 7 - 8 for SOCS3

fl/fl mice or

SOCS3MyeKO

mice). (D) SOCS3fl/fl

(n=3) and SOCS3

MyeKO (n=3) mice were inoculated s.c.

with 3.0 x 106 TRAMP-C1 cells. After 45 days of inoculation, tumor infiltrating CD45

+

cells were gated, and the percentage of the indicated cell subsets determined by flow

cytometry. (E) Flow cytometry plot of CD3+ and CD8

+ T-cells (gated in boxes) from

tumors of TRAMP-C1 mice (top), and quantification of tumor-infiltrating CD8+ T-cells

(bottom) (n = 8 - 10 for SOCS3fl/fl

mice or SOCS3MyeKO

mice). (F) SOCS3fl/fl

and

SOCS3MyeKO

mice were inoculated s.c with 3.0 x 106 TRAMP-C1 cells. At 24 days, mice

were randomly assigned to receive anti-Gr-1 or isotype antibodies (100 μg per mouse)

every two days for a total 9 treatments (n = 5 for SOCS3fl/fl

mice, n = 5 for SOCS3MyeKO

mice). Tumor size indicated as volume (mean mm3

± SD) was evaluated at 41 days.

*p<0.05 and **p<0.01.

71

Figure 2. Gr-1+CD11b

+ Cells from SOCS3

MyeKO Tumor-Bearing Mice Suppress

Antigen-specific T-cell Responses. (A) Diff-Quick stain of cytospin preparation of

CD45+Gr-1

+CD11b

+ cells from TRAMP-C1 tumors at 42 days. (B) Flow cytometric

analysis of myeloid cells in tumors formed by TRAMP-C1 cells at 42 days. Numbers

indicate the % of CD45+CD11b

+Ly6G

+ or CD45

+CD11b

+Ly6C

+ cells. Representative of

3 experiments. (C) CD45+Gr-1

+CD11b

+ cells were isolated from TRAMP-C1 tumors by

flow cytometry at 42 days, and antigen-specific suppression of CD8+ T-cells evaluated in

72

a co-culture assay. Splenocytes from OT-1 transgenic mice were cultured in the presence

of increasing ratios of Gr-1+CD11b

+ cells and stimulated with OVA derived peptide.

Proliferation of OVA-specific CD8+ T-cells was evaluated by flow cytometric analysis of

CFSE dilution after 72 h of co-culture. Data are mean ± SD; representative of 5

experiments. (D) Gene expression of CD45+Gr-1

+CD11b

+ cells from tumors of TRAMP-

C1 mice by qRT-PCR at day 42. Representative of 3 experiments. (E) SOCS3fl/fl

(n=3)

and SOCS3

MyeKO (n=3) mice were inoculated s.c. with 3.0 x 10

6 TRAMP-C1 cells.

Thirty-one days after inoculation, intracellular staining for p-STAT3 was performed in

Gr-1+CD11b

+ cells from BM, spleen, and tumor. Numbers indicate the quantification of

mean fluorescence intensity. *p<0.05.

73

Figure 3. Orthotopic Prostate Tumor Growth is Enhanced in SOCS3MyeKO

mice. (A)

One x 106 TRAMP-C2 cells were injected into the prostates of SOCS3

fl/fl (n=6) and

SOCS3MyeKO

(n=6) mice. Tumor weight was evaluated at 30 days. (B) H&E staining of

prostate at 30 days. Scale bar, 1 mm. Immunofluorescence staining of tumor infiltrating

CD45+ or Gr-1

+ cells at day 30. Scale bar, 45 μm. Prostate isolated from SOCS3

fl/fl mice

after intraprostatic injection of PBS was used as a control. Control mice (n = 2);

SOCS3fl/fl

mice (n = 4); SOCS3MyeKO

mice (n = 4). (C, D) Quantification of flow

cytometric analyses of prostate tumor-infiltrating Gr-1+CD11b

+ cells (C) and CD8

+ T-

cells (D) (n = 3 per group). (E) Expression of indicated genes from TRAMP-C2 prostate

74

tumors was evaluated by qRT-PCR at day 30 post implantation. Data are mean ± SD,

representative of 2 experiments. *p<0.05.

75

Figure 4. Loss of SOCS3 Promotes Proliferation of Gr-1+CD11b

+ Cells. (A) TRAMP-

C1 tumor-bearing SOCS3MyeKO

mice exhibit splenomegaly as indicated by spleen size

76

(n=5 for SOCS3fl/fl

mice, n=6 for SOCS3MyeKO

mice). Image of spleens (top) and

summary of spleen weight (bottom) are shown. (B) Naïve SOCS3fl/fl

mice and TRAMP-

C1 tumor-bearing SOCS3fl/fl

and SOCS3

MyeKO mice were injected i.p. with 100 μg of EdU,

and spleens were harvested 20 h after injection. Spleen cells were stained for Gr-1+ and

CD11b+, then detected by flow cytometry for EdU positive cells. Flow plot for one

mouse in each category. (C) BM cells were stained for Gr-1+ and CD11b

+, then detected

by flow cytometry for EdU positive cells. Number in each quadrant expressed as Gr-1High

and Gr-1Int

percentage of total cells (left), and histogram of EdU plot is shown (right),

with numbers expressed as percentage of total Gr-1+CD11b

+ cells. Representative of 2

experiments; Naïve mice (n = 2); SOCS3fl/fl

mice (n = 4); SOCS3MyeKO

mice (n = 4). (D,

E) Blood was obtained by cheek bleeding puncture from TRAMP-C1 tumor-bearing

mice 45 days after inoculation, and analyzed on the HEMAVET®950 hematology

analyzer. White blood cell (WBC) (103/μl) (D), and granulocyte counts (10

3/μl) (E) are

shown. *p<0.05 and **p<0.01.

77

Figure 5. Absence of SOCS3 Enhances STAT3 Activation and BM Differentiation

into Gr-1+CD11b

+ MDSC. (A) BM cells from SOCS3

fl/fl and

SOCS3

MyeKO mice were

78

cultured in TRAMP-C1 CM (100% volume), and Gr-1+CD11b

+ cells analyzed by flow

cytometry at day 4. Numbers are expressed as percentage of total CD45+ cells. (B) BM-

derived cells incubated with TRAMP-C1 CM for 4 days were co-cultured with CFSE (0.5

μM) labeled OT-1 splenocytes in the presence of OVA derived peptide. Proliferation of

CD8+ T-cells was evaluated by flow cytometric analysis of CFSE dilution after 48 h of

co-culture. Data are mean ± SD; representative of 3 experiments. (C) BM-derived cells

incubated with TRAMP CM (100% volume) for 4 days were co-cultured with OT-1

splenocytes in the presence of OVA derived peptide. Levels of IFN-γ in the supernatant

of co-cultured BM-derived Gr-1+CD11b

+ cells with OT-1 splenocytes (1:4 ratio) were

measured by ELISA. Data are representative of 3 experiments. (D) Nitrite concentration

in supernatant from co-cultured cells (1:4 ratio) was measured by Griess reaction.

Representative of 3 experiments. (E) Gr-1+CD11b

+ cells were sorted from cultured BM-

derived cells treated with TRAMP-C1 CM for 4 days, and arginase activity was measured

in lysates from sorted cells. Assay was performed in triplicate, data are the mean ± SD.

(F) BM-derived cells were treated with TRAMP-C1 CM (100% volume) for the indicated

times, and immunoblotting performed with the indicated antibodies. Representative of 4

experiments. (G) BM cells from SOCS3fl/fl

and SOCS3

MyeKO mice were treated with

TRAMP-C1 CM (100% volume) for 4 h, and expression of indicated genes evaluated by

qRT-PCR. (H) Expression of MHC class II by Gr-1+CD11b

+ cells was detected by flow

cytometry after 4 days of in vitro culture with TRAMP-C1 CM. Mature dendritic cells

generated by BM cells cultured in medium containing GM-CSF (10 ng/ml) for 7 days

were used as a positive control, and Rat IgG2b was used as isotype control. (I) BM cells

from SOCS3fl/fl

and SOCS3MyeKO

mice were cultured in the presence of TRAMP-C1 CM

79

and analyzed by flow cytometry at day 4. Numbers are expressed as percentage of total

CD45+CD11b

+ myeloid cells. Representative of 3 experiments. Data are mean ± SD;

*p<0.05 and **p<0.01.

80

Figure 6. G-CSF Induces the Differentiation of BM Precursors into Functional

MDSC in a STAT3-dependent Manner. (A) Supernatant of prostate tumors obtained

81

from mice with intraprostatic injection of TRAMP-C2 cells (at day 30), and flank tumors

from TRAMP-C1 or -C2 tumor-bearing mice (at day 42), as well as TRAMP-C1 or -C2

tumor cell CM were analyzed for G-CSF levels by ELISA. Data are mean ± SD;

representative of 3 experiments. (B) BM-derived cells were treated with G-CSF (10

ng/ml) for the indicated times and immunoblotting performed with the indicated

antibodies. Representative of 4 experiments. (C) BM-derived cells were incubated in the

absence or presence of G-CSF (10 ng/ml) for 2 h, then intracellular staining for p-STAT3

or p-STAT5 was performed. Representative of 2 experiments. (D) SOCS3 mRNA

expression from cultured BM-derived cells treated with G-CSF (10 ng/ml) for the

indicated times was evaluated by qRT-PCR. Data are mean ± SD; representative of 2

experiments. (E) BM cells from SOCS3fl/fl

and SOCS3

MyeKO mice were cultured in G-CSF

(20 ng/ml), G-CSF (20 ng/ml) with Lentivirus expressing GFP or Lentivirus expressing

SOCS3. Gr-1+CD11b

+ cells were analyzed by flow cytometry at day 4. Representative of

5 experiments. (F) BM cells from SOCS3fl/fl

and SOCS3

MyeKO mice were cultured in G-

CSF (20 ng/ml). Vehicle (DMSO), P6 (1 μM), or Stattic (1 μM) were added to the

cultures at days 0 and 2, and Gr-1+CD11b

+ cells analyzed by flow cytometry at day 4.

Representative of 2 experiments. (G) BM cells from SOCS3fl/fl

and SOCS3

MyeKO mice

were treated with G-CSF (10 ng/ml) for 4 h, and expression of indicated genes evaluated

by qRT-PCR. Data are mean ± SD; representative of 2 experiments. (H) BM cells

cultured for 4 days with G-CSF (20 ng/ml) were co-cultured with CFSE (0.5 μM) labeled

OT-1 splenocytes in the presence of OVA peptide. Proliferation of CD8+ T-cells was

evaluated by flow cytometric analysis of CFSE dilution after 48 h of co-culture. Numbers

indicates the ratio of OT-1 splenocytes:MDSC. Representative of 3 experiments. (I) Gr-

82

1+CD11b

+ cells were sorted from cultured BM-derived cells, then treated with G-CSF (10

ng/ml) for 4 h. Arginase activity was measured in lysates from sorted cells. Assay was

performed in triplicate, data are the mean ± SD. *p<0.05, **p<0.01.

83

Figure 7. G-CSF Neutralization Limits Gr-1+CD11b

+ Cell Proliferation In Vitro and

Tumor Growth In Vivo. (A) BM cells from SOCS3fl/fl

and SOCS3MyeKO

mice were

labeled with CFSE, then cultured in the presence of TRAMP CM or G-CSF (10 ng/ml)

with the indicated antibodies. Proliferation of Gr-1+CD11b

+ cells was evaluated by flow

cytometric analysis of CFSE dilution after 4 days. Numbers are expressed as percentage

proliferating cells of total Gr-1+CD11b

+ cells (Red, SOCS3

fl/fl; Black, SOCS3

MyeKO). (B)

SOCS3fl/fl

and SOCS3MyeKO

mice were inoculated s.c with 3.0 x 106 TRAMP-C1 cells.

84

Mice were randomly assigned to receive anti-G-CSF or isotype antibodies (10 μg per

mouse). Black arrows indicate Ab administration (n = 6 for SOCS3fl/fl

mice, n = 6 for

SOCS3MyeKO

mice). Tumor size indicated as volume (mean mm3

± SD) was evaluated up

to 39 days. (C) SOCS3fl/fl

and SOCS3MyeKO

mice were inoculated s.c. with 3.0 x 106

TRAMP-C1 cells. After 39 days, flank tumors were collected and weighed. Data

expressed as mean ± SD. (D, E) Quantification of flow cytometric analyses of tumor-

infiltrating (D) and spleen (E) Gr-1+CD11b

+ cells (n=3 per group). *p<0.05 and

**p<0.01. (F) Schematic model. Tumor cell-secreted G-CSF induces activation of the

JAK/STAT3 pathway in myeloid cells, which leads to induction of tumor promoting

genes, and to SOCS3 expression. SOCS3 is a feedback inhibitor of G-CSF induced

STAT3 activation. In the absence of SOCS3, MDSC are hyper-responsive to tumor-

produced G-CSF, aberrantly activate STAT3, and express enhanced levels of Arginase 1,

iNOS and S100A8/9. Elevated levels of MDSC suppress anti-tumor immune responses in

the tumor microenvironment, in part by inhibiting activity of CD8+ T-cells, and

eventually promote tumor growth.

85

Supplemental Figures

Y u , H . e t a l. , F ig . S 1

A B

S O C S3fl/fl

S O C S3M yeKO

CD25

Fo

xp

3C

D4

+F

ox

p3

+C

D2

5+

T-c

ell

s

(% o

f C

D4

5+

)

SO

CS

3f l/ fl

SO

CS

3M

yeK

O

0

2

4

6

8

1 0

5 .9 5 .4

% o

f C

D8

+ T

-ce

lls

(S

ple

en

)

TR

AM

P-C

1

TR

AM

P-C

2

TR

AM

P-C

1

TR

AM

P-C

2

Naiv

e

0

5

1 0

1 5

2 0

S O C S 3f l / f l

S O C S 3M y e K O

S O C S3fl/fl

S O C S3M yeKO

CD8

CD

3

1 2 .5 1 3 .2

Sp

lee

nC

S O C S3fl/fl

S O C S3M yeKO

Sp

lee

nG

r-1

C D 11b

6 .3 1 4 .9

Gr-1

+C

D1

1b

+ C

ell

s

(%

of

CD

45

+)

TR

AM

P-C

1

TR

AM

P-C

2

TR

AM

P-C

1

TR

AM

P-C

2

Naiv

e

0

1 0

2 0

3 0

*

* *

S O C S 3f l / f l

S O C S 3M y e K O

D a y s

Tu

mo

r v

olu

me

(m

m3

)

0 3 6 9 1 2 1 5 1 8 2 1 2 4 2 7 3 0 3 3 3 6 3 9 4 2

0

3 0 0

6 0 0

9 0 0

1 2 0 0S O C S 3

f l / f lIs o ty p e

S O C S 3f l / f l

A n ti-G r -1

S O C S 3M y e K O

Is o ty p e

S O C S 3M y e K O

A n ti-G r -1 * *

D

% o

f G

r-1

+ C

D1

1b

+ C

ell

s

Is o ty p e A n ti-G r-1

0

5

1 0

1 5

2 0

S O C S 3f l / f l

S O C S 3M y e K O*

*

E

Figure S1. Characterization of Immune Responses in TRAMP Tumor-bearing Mice.

(A) Representative flow cytometry plot of Gr-1+CD11b

+ cells (gated in boxes) from

86

spleen of TRAMP-C1 tumor-bearing mice (top), and quantification of spleen Gr-

1+CD11b

+ cells (bottom) (n = 7 - 8 for SOCS3

fl/fl mice or SOCS3

MyeKO mice). Spleen

isolated from naïve mice was used as a control (n = 3). (B) Representative flow

cytometry plot of CD8+ T-cells (gated in box) from spleens of TRAMP-C1 tumor-bearing

mice (top), and quantification of spleen CD8+ T-cells (bottom) (n = 9-10 per group). (C)

Representative flow cytometry plot of CD4+Foxp3

+CD25

+ Treg from tumors of TRAMP-

C1 mice (top), and quantification (bottom) (n = 3 per group). Data are mean ± SD,

representative of 2 experiments. (D) SOCS3fl/fl

and SOCS3MyeKO

mice were inoculated

s.c with 3.0 x 106 TRAMP-C1 cells. Mice were randomly assigned to receive anti-Gr-1 or

isotype Ab (100 μg per mouse). Black arrows indicate Ab administration (n = 5 for

SOCS3fl/fl

mice, n = 5 for SOCS3MyeKO

mice). Tumor size indicated as volume (mean

mm3

± SD) was evaluated up to 41 days. (E) Quantification of flow cytometric analyses

of spleen Gr-1+CD11b

+ cells at day 41 (n=2 - 3 per group).*p<0.05 and **p<0.01.

87

% o

f P

ro

life

ra

tin

g C

ell

s

O T -1 a lo n e 8 :1 4 :1 2 :1 1 :1

0

2 0

4 0

6 0

8 0

1 0 0

1 2 0

S O C S 3f l / f l

S O C S 3M y e K O

**

Y u , H . e t a l. , F ig . S 2

A B

Arg

ina

se

1 (

mU

pe

r 1

06

Ce

lls

)

S p le e n

0

1 0 0

2 0 0

3 0 0

4 0 0

S O C S 3f l / f l

S O C S 3M y e K O

*

Nit

rite

Co

nc

. (

M)

OT

-1 a

lone

SO

CS

3f l/ fl

SO

CS

3M

yeK

O

0

5 0

1 0 0

1 5 0

2 0 0

2 5 0

* *

* *

C D

IFN

- (

pg

/ml)

O T -1 a lo n e S O C S 3f l/ f l

S O C S 3M y e K O

0

1 0 0 0

2 0 0 0

3 0 0 0

4 0 0 0

*

* *

Figure S2. Gr-1+CD11b

+ Cells Isolated from SOCS3

MyeKO Mice Exhibit Enhanced

MDSC Suppressive Function. (A) Levels of IFN-γ in the supernatant of co-cultured

tumor-derived CD45+Gr-1

+CD11b

+ cells with OT-1 splenocytes (1:2 ratio) were

measured by ELISA. Data are the mean ± SD; representative of 3 experiments. (B)

Nitrite concentration in supernatant from co-cultured tumor-derived CD45+Gr-1

+CD11b

+

cells with OT-1 splenocytes (1:1 ratio) was measured by Griess reaction. Data are the

mean ± SD, representative of 3 experiments. (C) Gr-1+CD11b

+ cells were isolated from

the spleen of TRAMP tumor-bearing mice by flow cytometry, and antigen-specific

suppression of CD8+ T-cells was evaluated. Numbers indicates the ratio of OT-1

splenocytes:MDSC from spleen. Assay was performed in triplicate and data are the mean

88

± SD. Representative of 5 experiments. (D) Arginase activity was measured in lysates of

106 sorted MDSC from the spleen of TRAMP tumor-bearing SOCS3

fl/fl and SOCS3

MyeKO

mice. Assay was performed in triplicate, data are the mean ± SD. *p<0.05 and **p<0.01.

89

A

Y u , H . e t a l. , F ig u re S 3

C

Gr-1

+ C

D1

1b

+ C

ell

Nu

mb

er

(1 x

10

5)

Contr

ol M

ediu

m

TR

AM

P-C

1 C

M

0

5

1 0

1 5

2 0

2 5

S O C S 3f l / f l

S O C S 3M y e K O

* *

*

% o

f T

ota

l C

ell

s

Contr

ol M

ediu

m

TR

AM

P-C

1 C

M

Contr

ol M

ediu

m

TR

AM

P-C

1 C

M

0

5

1 0

1 5

2 0

S O C S 3f l / f l

S O C S 3M y e K O

S P h a s e M P h a s e

* *

D E

SO

CS

3M

ye

KO

Gr-

1

C D 11b

Ce

ll C

ou

nt

EdU

Na

ive

SO

CS

3fl

/fl

2 .7 0 .7

3 .1 1 .1

2 .7 0 .5

2 .5 0 .5

H igh

Int.

Gr-

1

C D 11b

S O C S3M yeKO

N a ive S O C S3fl/fl

2 8 .3 2 .1

2 1 .9 1 .6

3 3 .3 3 .4

2 0 .4 2 .2

N a ive

EdU

EdU

S O C S3M yeKO

N a ive S O C S3fl/fl

N a ive

1 3 .2 2 .5

2 9 .6 3 .5

1 2 .7 2 .8

3 4 .6 4 .1

Sp

lee

n W

eig

ht

(mg

)

S O C S 3f l/ f l

S O C S 3M y e K O

0

2 0

4 0

6 0

8 0

1 0 0

n s

B

Figure S3. Effect of SOCS3 on Gr-1+CD11b

+ Cell Proliferation. (A) Naïve SOCS3

fl/fl

mice and SOCS3

MyeKO mice were injected i.p. with 100 μg of EdU, and spleens were

90

harvested 20 h after injection (n=3). Spleen cells were stained for Gr-1+ and CD11b

+,

then detected by flow cytometry for EdU positive cells. (B) Spleen size of naïve

SOCS3fl/fl

and SOCS3MyeKO

mice (n=3). (C) BM cells were stained for Gr-1+ and CD11b

+,

then detected by flow cytometry for EdU positive cells. Number in each quadrant

expressed as Gr-1High

and Gr-1Int

percentage of total cells (left), and histogram of EdU

plot is shown (right), with numbers expressed as percentage of total Gr-1+CD11b

+ cells.

Representative of 2 experiments; Naïve SOCS3fl/fl

mice (n = 3); Naïve SOCS3MyeKO

mice

(n = 3). (D) BM cells from SOCS3fl/fl

and SOCS3

MyeKO mice were cultured in TRAMP

CM (100% volume) for 4 days. Total number of Gr-1+CD11b

+ cells was determined by

CountBright™ Absolute Counting Beads at day 4. (E) BM cells from SOCS3fl/fl

and

SOCS3MyeKO

mice were cultured in the presence of TRAMP CM (100% volume) for 2

days. Cell cycle of Gr-1+CD11b

+ cells was determined by Propidium Iodide staining

using flow cytometry. *p<0.05 and **p<0.01.

91

G-C

SF

(p

g/m

l)

Naiv

e

Ort

hoto

pic

TR

AM

P-C

2

TR

AM

P-C

1 T

um

or

s.c

.

TR

AM

P-C

2 T

um

or

s.c

.

0

2 0 0

4 0 0

6 0 0

8 0 0

1 0 0 0

S O C S 3f l / f l

S O C S 3M y e K O

*

G-C

SF

mR

NA

(F

old

In

cre

as

e)

Naiv

e

Ort

hoto

pic

TR

AM

P-C

2

TR

AM

P-C

1 s

.c. T

um

or

TR

AM

P-C

2 s

.c. T

um

or

TR

AM

P-C

1

TR

AM

P-C

2

0

1 0

2 0

3 0

4 0

5 0

S O C S 3f l / f l

S O C S 3M y e K O

*

A

Y u , H . e t a l. , F ig u re S 4

B

Figure S4. G-CSF Expression by TRAMP Tumor-Bearing Mice. (A) Serum G-CSF

levels were measured by ELISA in the indicated tumor-bearing mice (n=3 - 4) 30 days

after orthotopic injection and 45 days after s.c. inoculation with TRAMP-C1 or TRAMP-

C2 cells. Serum samples obtained from naïve mice were used as control (n=3). (B) G-

CSF mRNA expression levels was determined by qRT-PCR in tumors from mice with

intraprostatic injection of TRAMP-C2 cells (n=3 - 4), flank tumors from TRAMP-C1

(n=3 - 4) or TRAMP-C2 (n= 3 - 4) tumor-bearing mice, and TRAMP-C1 or C2 tumor

cell lines. n=3 for naïve mice. *p<0.05.

92

Ce

ll C

ou

nt

G -CSFR

3 7 .7

2 2 .6

5 .2

5 .6

WB

C 1

03

/l

Is o ty p e A n ti-G -C S F

0

5

1 0

1 5

2 0

S O C S 3f l / f l

S O C S 3M y e K O

*

Gra

nu

loc

yte

s 1

03

/l

Isoty

pe

Anti-G

-CS

F

0

2

4

6

S O C S 3f l / f l

S O C S 3M y e K O

*

Y u , H . e t a l. , F ig u re S 5

A B

Se

rum

G-C

SF

(p

g/m

l)

Is o ty p e A n ti-G -C S F

0

2 0 0

4 0 0

6 0 0

8 0 0

S O C S 3f l / f l

S O C S 3M y e K O

*

*

% o

f G

r-1

+C

D1

1b

+ C

ell

s

Contr

ol M

ediu

m

TR

AM

P C

M +

Isoty

pe

TR

AM

P C

M +

Anti-G

-CS

F

G-C

SF

+ Isoty

pe

G-C

SF

+ A

nti-G

-CS

F

0

2 0

4 0

6 0

8 0

S O C S 3f l / f l

S O C S 3M y e K O

* * * *

C D

E

S p le e n G r-1+

c e lls

T um o r C D 11b-

c e lls

T u m o r G r-1+

c e lls

T R A M P -C 1 ce lls

Figure S5. Effects of G-CSF Neutralizing Antibody. (A) BM cells from SOCS3fl/fl

and

SOCS3MyeKO

mice were cultured for 4 days in the presence of TRAMP CM or G-CSF (10

ng/ml) with either anti-G-CSF Ab (50 μg/ml) or IgG isotype control (50 μg/ml), then

analyzed by flow cytometry. Bars indicate the percentage of Gr-1+CD11b

+ cells of total

CD45+ cells. (B) Serum samples of the indicated groups (n=3) were collected from cheek

93

bleeding puncture after 39 days of inoculation. G-CSF protein levels were detected by

ELISA. (C, D) Blood was obtained from the indicated groups (n=3) 39 days after

subcutaneous inoculation. WBC (C) and granulocyte counts (D) (103/μl) are shown. (E)

Cells were isolated from spleen and tumors of TRAMP-C1 tumor-bearing mice at day 41

after inoculation, and G-CSFR staining was performed. Numbers indicate percentage of

G-CSFR positive cells (n=2). *p<0.05 and **p<0.01.

94

CONCLUSIONS

The studies above focused on investigating the role of the JAK/STAT pathway in

myeloid cells with respect to promoting cancer development. First, we generated mice

with targeted deletion of SOCS3 in myeloid cells (LysMCre-SOCS3fl/fl

, SOCS3MyeKO

mice) to study how SOCS3 in myeloid cells regulates immune responses in the context of

the prostate cancer microenvironment. We demonstrate that SOCS3 expression in

myeloid cells is an important determinant of tumor growth, indicating a critical influence

of the tumor microenvironment in cancer progression. The loss of SOCS3 in myeloid

cells promotes the differentiation of BM-derived progenitor cells into Gr-1+CD11b

+

MDSCs, and enhances the immunosuppressive functions of these cells. Importantly,

STAT3 activation is essential for this process. Tumor-derived G-CSF is crucial for the

mobilization and recruitment of Gr-1+CD11b

+ MDSCs to tumors, where they decrease

the presence of CD8+ T-cells. Neutralization of G-CSF is effective in limiting the

differentiation and functionality of MDSC, which in vivo manifests as restricted tumor

growth. These findings establish a circuitry between MDSC, tumor cells and G-CSF in

the tumor microenvironment; G-CSF secreted by tumor cells promotes the recruitment

and differentiation of MDSC, which is dependent on STAT3 activation in MDSC. In the

absence of SOCS3, this response and circuitry is amplified. These results support a role

for SOCS3 in repressing MDSC differentiation, which ultimately relieves

immunosuppression in the prostate tumor microenvironment.

95

Persistent STAT3 activation is associated with poor prognosis in many cancer types,

including prostate cancer (278, 279). Loss of the androgen receptor (AR) leads to the

development of prostate cancer stem cells, which require STAT3 activation (276, 280).

Stem-like cells from patients with prostate cancer secrete high levels of IL-6 and exhibit

hyperactivation of STAT3 (65). STAT3 activation in cells of the hematopoietic lineage is

also associated with creating a tumor microenvironment conducive to tumor growth. This

is especially true for MDSC, which rely on STAT3 for differentiation and functionality.

Ablation of STAT3 in multiple lineages of immune cells (neutrophils, NK cells, DCs)

enhanced their anti-tumor activity (1). While MDSCs were not directly examined,

heightened activation of STAT3 in MDSCs, as shown by our study, may contribute to

tumor immune tolerance. MDSCs lacking SOCS3 with STAT3 hyperactivation have

potent immunosuppressive capabilities such as inhibition of antigen-specific CD8+ T-cell

proliferation and IFN-γ production, and enhanced nitrite production and arginase activity.

Also, SOCS3-deficient MDSCs expressed MHC Class II at lower levels than SOCS3

wild-type MDSCs, suggestive of an immature suppressive phenotype. Nonetheless, our

findings clearly demonstrate the importance of SOCS3 in restricting MDSCs-mediated

anti-tumor immunity. SOCS3 expression in MDSCs negatively regulates the expression

of soluble mediators such as S100A8 and S100A9 and products of iNOS and arginase 1

that support an immunosuppressive milieu in tumors (277). In the absence of SOCS3,

MDSCs are hyper-responsive to tumor-produced cytokines such as G-CSF, and

aberrantly activate STAT3, which in turn contributes to chronic cancer-related

inflammation and suppression of anti-tumor immune responses.

96

G-CSF plays a crucial role in hematopoiesis by stimulating the proliferation,

differentiation and survival of myeloid progenitor cells, particularly cells within the

granulocytic lineage (235). JAK1, JAK2 and TYK2 are recruited to the G-CSF receptor

upon stimulation with G-CSF, and then in turn activate STAT1, STAT3 and STAT5, of

which STAT3 is most important. SOCS3 is induced by G-CSF in myeloid cells, and

serves as a negative regulator of G-CSF-induced cellular responses by binding to the G-

CSF receptor (208, 281). G-CSF is one of a number of soluble mediators that promote

the expansion and migration of MDSC from the bone marrow to tumors. While the levels

of G-CSF were comparable in SOCS3MyeKO

and SOCS3fl/fl

tumor-bearing mice,

SOCS3MyeKO

BM-derived cells are hyper-responsive to G-CSF, as documented by

increased duration and intensity of G-CSF-induced STAT3 phosphorylation, which led to

MDSCs differentiation at a greater percentage than cells from SOCS3fl/fl

mice. In addition,

G-CSF-induced MDSCs from SOCS3MyeKO

tumor-bearing mice exhibited a greater

capacity to suppress antigen-specific T-cell proliferation. Our findings demonstrate that

treatment of tumor-bearing mice with neutralizing G-CSF antibody reduced circulating

granulocytic cells as well as infiltration of MDSCs in tumors. Thus tumor-derived G-CSF

is an important regulator of inflammation and immune suppression within the tumor

microenvironment. G-CSF may not be critical for the development of Gr-1+CD11b

+ cells

in every tumor model. In LSL-KrasG12D

knock-in mice, pancreatic ductal adenocarcinoma

exhibit an infiltrate of Gr-1+CD11b

+ leukocytes due to elevated levels of GM-CSF (282).

Moreover, cytokines and growth factors other than G-CSF have been implicated in the

pathophysiology of Gr-1+CD11b

+ leukocytes in other mouse models—including IL-1β,

IL-6, and VEGF (207, 229). Elevated levels of G-CSF were also identified in a mouse

97

breast cancer model (283). Identification of G-CSF in cancer models substantiates the

importance of this cytokine in regulating and maintaining the high frequency of MDSCs

in the tumor microenvironment.

Clinical information regarding SOCS3 expression in prostate cancer is inconclusive at

this time. Pierconti et al, demonstrated that methylation of the SOCS3 promoter was

significantly associated with intermediate-high grade Gleason score and with an

unfavorable outcome. In benign prostate hyperplasia and normal controls, the SOCS3

promoter was unmethylated (118). Analysis of the Oncomine database demonstrates that

SOCS3 mRNA expression is significantly lower in prostate carcinoma compared to

benign prostatic hyperplasia (277). Data from cBioPortal (Prostate Adenocarcinoma

MSKCC) indicates that some prostate cancer patients with SOCS3 mRNA

overexpression trend towards a longer disease-free time than patients with “normal”

levels of SOCS3 mRNA (277). However, SCOS3 was overexpressed in a relatively small

number of patients, and no correlations with patient stage status were found. Thus, there

is a dysregulation of SOCS3 gene expression in a subgroup of patients with prostate

cancer, but the functional and clinical relevance is still under investigation. Nonetheless,

it is clear that a variety of mechanisms, including SOCS3 dysregulation and abundant IL-

6 production, do contribute to hyperactivation of JAK/STAT pathway in animal models

of prostate cancer and patients. Inhibitors of IL-6, JAKs and STAT3 are being considered

in the context of prostate cancer, and have already proven beneficial in animal models.

Furthermore, peptides that mimic the SOCS Kinase Inhibitory Region (KIR), which is

responsible for binding to JAKs and suppressing downstream STAT3 activation, may

98

prove beneficial in prostate cancer. These findings highlight the importance of SOCS3 as

a promising target for studying prostate cancer microenvironment.

It has been proposed that myeloid cells are involved during the process of cancer

development before clinical manifestations become apparent, promoting treatment

resistance, and metastatic spreading (179, 225). Blockage of tumor-infiltrating myeloid

cells through the use of CSF1R inhibitor was able to improve the efficacy of radiotherapy

in prostate cancer (254). However, the mechanisms underlying the tumor promoting role

of myeloid cells still remains poorly understood in prostate cancer (272). The goal of this

study was to investigate the importance of JAK/STAT pathway in myeloid cells in the

prostate cancer microenvironment. Understanding the underlying mechanism of the

immune suppressive microenvironment is important for developing more efficient

therapies for fighting tumor progression. Induction and expansion of MDSCs and the

resultant immune suppression is expected to be similar between humans and mice,

making the results of murine experiments useful in developing new anti-MDSC agents

and testing them in clinical trials. Peripheral blood MDSC levels correlate with a higher

tumor burden and a worse prognosis in lung cancer (225). Inhibition of MDSC in murine

models may enhance anti-tumor immunity by increasing responsiveness to interferon

stimulation (284). Inhibition or depletion of MDSC enhances the activity of cancer

vaccines in animal models (226). We substantiate that MDSCs play a vital role in

TRAMP prostate cancer development and the STAT3/SOCS3 pathway is a critical

regulator in MDSC differentiation (277).

Although the results presented here have demonstrated the importance of SOCS3 in

regulating myeloid cell differentiation in the tumor microenvironment, it could be further

99

developed in a number of ways. First, to further explore the importance of myeloid

SOCS3 in regulating prostate cancer progression, it will be of interest to investigate the

expression of SOCS3 in myeloid cells isolated from patients. Methylation of the SOCS3

gene promotor was evidenced in a subtype of prostate cancer patients, and reduced

SOCS3 gene expression associated with an unfavorable outcome (285). However, what

the level of SOCS3 expression in myeloid cells from those prostate cancer patients is not

known. A finding of SOCS3 expression in myeloid cells would support a direct role for

myeloid SOCS3 in control myeloid cell immune suppressive function in the cancer

microenvironment.

Second, our studies implicate the role of G-CSF/STAT3/SOCS3 in regulating MDSC

development. Since the presence of elevated levels of MDSCs in tumor is critical in

maintain suppressive microenvironment, blockage of MDSC development would be

beneficial for prostate cancer patients. Our results suggest that neutralization G-CSF is

able to reduce MDSC levels and tumor growth. Future experiments to utilize JAK/STAT

inhibitors or SOCS3 mimic peptides on tumor bearing mice would need to be addressed

in detail. Furthermore, given the few options for treating castration-resistant disease,

targeting G-CSF/STAT3/SOCS3 may be additive to current blockage of AR signaling,

resulting in a further decrease of tumor development.

Last but not least, recently recognized check point blockers including PD-1/PD-L1 and

CTLA4 are successful in helping the immune system to fight cancer (286). PD-L1 also

known as CD274, is expressed in myeloid cells which are critical in maintaining

periphery immune suppression. It will be of interest to investigate whether PD-L1

expression is downstream of the G-CSF/STAT3/SOCS3 pathway. Our results suggest

100

that SOCS3 deficient MDSCs express reduced level of MHC II, and stay in immature

state (277). PD-L1 expression in myeloid cells was upregulated through

JAK/STAT/SOCS pathway after LPS treatment (209). These reports suggest that it is

worthwhile to investigate whether the G-CSF/STAT3/SOCS3 axis in myeloid cells is

involved in regulating immune check points and execute their immune suppressive

functions.

In conclusion, our findings have elucidated the role of SOCS3 in myeloid cells in prostate

cancer development. These studies not only highlight the significance of the

STAT3/SOCS3 pathway in regulating the differentiation and function of MDSC in

cancer, but also identify this intricate protein network as important therapeutic targets to

eliminate MDSCs-mediated immunosuppression. Insights gained from our current studies

help searching efficient treatments that target myeloid cells and might substantially

increase the efficacy of current cancer immunotherapies. This is especially important in

light of the recent finding that MDSCs are responsible for resistance to immune check-

point inhibitors, and that elimination of MDSCs led to cures of experimental, metastatic

tumors.

101

REFERENCES

1. Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading

role for STAT3. Nat Rev Cancer 2009;9:798-809.

2. O'Shea JJ, Plenge R. JAK and STAT signaling molecules in immunoregulation

and immune-mediated disease. Immunity 2012;36:542-50.

3. Nicolas CS, Peineau S, Amici M, Csaba Z, Fafouri A, Javalet C, et al. The

Jak/STAT pathway is involved in synaptic plasticity. Neuron 2012;73:374-90.

4. O'Shea JJ, Holland SM, Staudt LM. JAKs and STATs in Immunity,

Immunodeficiency, and Cancer. New Eng J Med 2013;368:161-70.

5. Sansone P, Bromberg J. Targeting the Interleukin-6/Jak/Stat Pathway in Human

Malignancies. J Clin Oncol 2012;784:121-32.

6. Haque SJ, Sharma P. Interleukins and STAT signaling. Vitam Horm

2006;74:165-206.

7. Frank DA. Targeting STATs for cancer therapy: "Undruggable" no more. JAK-

STAT 2012;1:261-2.

8. Yu H, Lee H, Herrmann A, Buettner R, Jove R. Revisiting STAT3 signalling in

cancer: new and unexpected biological functions. Nat Rev Cancer 2014;14:736-

46.

9. Reich NC. STAT dynamics. Cytokine Growth Factor Rev 2007;18:511-8.

10. Kisseleva T, Bhattacharya S, Braunstein J, Schindler CW. Signaling through the

JAK/STAT pathway, recent advances and future challenges. Gene 2002;285:1-24.

11. Swiatek-Machado K, Kaminska B. STAT signaling in glioma cells. Adv Exp Med

2013;986:189-208.

102

12. Adach A, Ellert-Miklaszewska A, Kaminska B. Molecular characterization of

STAT signaling in inflammation and tumorigenesis. Methods Mol Biol

2009;512:265-78.

13. Brierley MM, Fish EN. Stats: multifaceted regulators of transcription. J Interferon

Cytokine Res 2005;25:733-44.

14. Koppikar P, Bhagwat N, Kilpivaara O, Manshouri T, Adli M, Hricik T, et al.

Heterodimeric JAK-STAT activation as a mechanism of persistence to JAK2

inhibitor therapy. Nature 2012;489:155-9.

15. Inghirami G, Chiarle R, Simmons WJ, Piva R, Schlessinger K, Levy DE. New

and old functions of STAT3: a pivotal target for individualized treatment of

cancer. Cell Cycle 2005;4:1131-3.

16. Multhoff G, Molls M, Radons J. Chronic inflammation in cancer development.

Front Immunol 2011;2:98.

17. Schindler C, Strehlow I. Cytokines and STAT signaling. Adv Pharmacol

2000;47:113-74.

18. Smithgall TE, Briggs SD, Schreiner S, Lerner EC, Cheng H, Wilson MB. Control

of myeloid differentiation and survival by Stats. Oncogene 2000;19:2612-8.

19. Geissmann F, Gordon S, Hume DA, Mowat AM, Randolph GJ. Unravelling

mononuclear phagocyte heterogeneity. Nat Rev Immunol 2010;10:453-60.

20. Weaver CT, Hatton RD, Mangan PR, Harrington LE. IL-17 family cytokines and

the expanding diversity of effector T-cell lineages. Ann Rev Immunol

2007;25:821-52.

103

21. Cao H, Wolff RG, Meltzer MS, Crawford RM. Differential regulation of class II

MHC determinants on macrophages by IFN- and IL-4. J Immunol

1989;143:3524-31.

22. Minami M, Inoue M, Wei S, Takeda K, Matsumoto M, Kishimoto T, et al.

STAT3 activation is a critical step in gp130-mediated terminal differentiation and

growth arrest of a myeloid cell line. Proc Natl Acad Sci U S A 1996;93:3963-6.

23. Morgan KJ, Gilliland DG. A role for JAK2 mutations in myeloproliferative

diseases. Annu Rev Med 2008;59:213-22.

24. Carow B, Rottenberg ME. SOCS3, a Major Regulator of Infection and

Inflammation. Front Immunol 2014;5:58.

25. Chan KS, Sano S, Kiguchi K, Anders J, Komazawa N, Takeda J, et al. Disruption

of STAT3 reveals a critical role in both the initiation and the promotion stages of

epithelial carcinogenesis. J Clin Invest 2004;114:720-8.

26. Lin WW, Karin M. A cytokine-mediated link between innate immunity,

inflammation, and cancer. J Clin Invest 2007;117:1175-83.

27. Groner B, Lucks P, Borghouts C. The function of Stat3 in tumor cells and their

microenvironment. Semin Cell Dev Biol 2008.

28. Ruffell B, Affara NI, Coussens LM. Differential macrophage programming in the

tumor microenvironment. Trends Immunol 2012;33:119-26.

29. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation.

Nature 2008;454:436-44.

30. Dauer DJ, Ferraro B, Song L, Yu B, Mora L, Buettner R, et al. Stat3 regulates

genes common to both wound healing and cancer. Oncogene 2005;24:3397-408.

104

31. Schreiner SJ, Schiavone AP, Smithgall TE. Activation of STAT3 by the Src

family kinase Hck requires a functional SH3 domain. J Biol Chem

2002;277:45680-7.

32. Aziz MH, Hafeez BB, Sand JM, Pierce DB, Aziz SW, Dreckschmidt NE, et al.

Protein kinase Cvarepsilon mediates Stat3Ser727 phosphorylation, Stat3-

regulated gene expression, and cell invasion in various human cancer cell lines

through integration with MAPK cascade (RAF-1, MEK1/2, and ERK1/2).

Oncogene 2010;29:3100-9.

33. Zhang Y, Sif S, DeWille J. The mouse C/EBPδ gene promoter is regulated by

STAT3 and Sp1 transcriptional activators, chromatin remodeling and c-Myc

repression. J Cell Biochem 2007;102:1256-70.

34. Jung JE, Lee HG, Cho IH, Chung DH, Yoon SH, Yang YM, et al. STAT3 is a

potential modulator of HIF-1-mediated VEGF expression in human renal

carcinoma cells. Faseb J 2005;19:1296-8.

35. Benekli M, Baer MR, Baumann H, Wetzler M. Signal transducer and activator of

transcription proteins in leukemias. Blood 2003;101:2940-54.

36. Sen M, Joyce S, Panahandeh M, Li C, Thomas SM, Maxwell J, et al. Targeting

Stat3 abrogates EGFR inhibitor resistance in cancer. Clin Cancer

Res2012;18:4986-96.

37. Ranger JJ, Levy DE, Shahalizadeh S, Hallett M, Muller WJ. Identification of a

Stat3-dependent transcription regulatory network involved in metastatic

progression. Cancer Res 2009;69:6823-30.

105

38. Lieblein JC, Ball S, Hutzen B, Sasser AK, Lin HJ, Huang TH, et al. STAT3 can

be activated through paracrine signaling in breast epithelial cells. BMC Cancer

2008;8:302.

39. De Marzo AM, Platz EA, Sutcliffe S, Xu J, Gronberg H, Drake CG, et al.

Inflammation in prostate carcinogenesis. Nature reviews Cancer 2007;7:256-69.

40. Alvarez JV, Greulich H, Sellers WR, Meyerson M, Frank DA. Signal transducer

and activator of transcription 3 is required for the oncogenic effects of non-small-

cell lung cancer-associated mutations of the epidermal growth factor receptor.

Cancer Res 2006;66:3162-8.

41. Grandis JR, Drenning SD, Zeng Q, Watkins SC, Melhem MF, Endo S, et al.

Constitutive activation of Stat3 signaling abrogates apoptosis in squamous cell

carcinogenesis in vivo. Proc Natl Acad Sci U S A 2000;97:4227-32.

42. McFarland BC, Ma JY, Langford CP, Gillespie GY, Yu H, Zheng Y, et al.

Therapeutic potential of AZD1480 for the treatment of human glioblastoma. Mol

Cancer Ther 2011;10:2384-93.

43. Miklossy G, Hilliard TS, Turkson J. Therapeutic modulators of STAT signalling

for human diseases. Nat Rev Drug Discov 2013;12:611-29.

44. Kasprzycka M, Marzec M, Liu X, Zhang Q, Wasik MA.

nucleophosmin/anaplastic lymphoma kinase (NPM/ALK) oncoprotein induces the

T regulatory cell phenotype by activating STAT3. Proc Natl Acad Sci U S A

2006;103:9964-9.

106

45. Fukada T, Ohtani T, Yoshida Y, Shirogane T, Nishida K, Nakajima K, et al.

STAT3 orchestrates contradictory signals in cytokine-induced G1 to S cell-cycle

transition. EMBO J 1998;17:6670-7.

46. Zhang F, Li C, Halfter H, Liu J. Delineating an oncostatin M-activated STAT3

signaling pathway that coordinates the expression of genes involved in cell cycle

regulation and extracellular matrix deposition of MCF-7 cells. Oncogene

2003;22:894-905.

47. Kiuchi N, Nakajima K, Ichiba M, Fukada T, Narimatsu M, Mizuno K, et al.

STAT3 is required for the gp130-mediated full activation of the c-myc gene. J

Exp Med 1999;189:63-73.

48. Shirogane T, Fukada T, Muller JM, Shima DT, Hibi M, Hirano T. Synergistic

roles for Pim-1 and c-Myc in STAT3-mediated cell cycle progression and

antiapoptosis. Immunity 1999;11:709-19.

49. Hirano T, Ishihara K, Hibi M. Roles of STAT3 in mediating the cell growth,

differentiation and survival signals relayed through the IL-6 family of cytokine

receptors. Oncogene 2000;19:2548-56.

50. Aggarwal BB, Kunnumakkara AB, Harikumar KB, Gupta SR, Tharakan ST,

Koca C, et al. Signal transducer and activator of transcription-3, inflammation,

and cancer: how intimate is the relationship? Ann N Y Acad Sci 2009;1171:59-76.

51. Bhardwaj A, Sethi G, Vadhan-Raj S, Bueso-Ramos C, Takada Y, Gaur U, et al.

Resveratrol inhibits proliferation, induces apoptosis, and overcomes

chemoresistance through down-regulation of STAT3 and nuclear factor-κB-

107

regulated antiapoptotic and cell survival gene products in human multiple

myeloma cells. Blood 2007;109:2293-302.

52. Hedvat M, Huszar D, Herrmann A, Gozgit JM, Schroeder A, Sheehy A, et al. The

JAK2 inhibitor AZD1480 potently blocks Stat3 signaling and oncogenesis in solid

tumors. Cancer Cell 2009;16:487-97.

53. Aggarwal BB, Gehlot P. Inflammation and cancer: how friendly is the

relationship for cancer patients? Curr Opin Pharmacol 2009;9:351-69.

54. Devarajan E, Huang S. STAT3 as a central regulator of tumor metastases. Curr

Mol Med 2009;9:626-33.

55. Balanis N, Wendt MK, Schiemann BJ, Wang Z, Schiemann WP, Carlin CR.

Epithelial-to-mesenchymal transition promotes breast cancer progression via a

fibronectin-dependent Stat3 signaling pathway. J Biol Chem 2013;288:17954-67.

56. Dechow TN, Pedranzini L, Leitch A, Leslie K, Gerald WL, Linkov I, et al.

Requirement of matrix metalloproteinase-9 for the transformation of human

mammary epithelial cells by STAT3-C. Proc Natl Acad Sci U S A

2004;101:10602-7.

57. Chen F, Xu Y, Luo Y, Zheng D, Song Y, Yu K, et al. Down-regulation of Stat3

decreases invasion activity and induces apoptosis of human glioma cells. J Mol

Neurosci 2010;40:353-9.

58. Hamilton KE, Simmons JG, Ding S, Van Landeghem L, Lund PK. Cytokine-

Induction of Tumor Necrosis Factor Receptor 2 (TNFR2) is Mediated by STAT3

in Colon Cancer Cells. Mol Cancer Res 2011; 204:451-20.

108

59. Slattery ML, Lundgreen A, Kadlubar SA, Bondurant KL, Wolff RK.

JAK/STAT/SOCS-signaling pathway and colon and rectal cancer. Mol

Carcinogen 2013;52:155-66.

60. Ling X, Arlinghaus RB. Knockdown of STAT3 expression by RNA interference

inhibits the induction of breast tumors in immunocompetent mice. Cancer Res

2005;65:2532-6.

61. Cheng GZ, Zhang W, Sun M, Wang Q, Coppola D, Mansour M, et al. Twist is

transcriptionally induced by activation of STAT3 and mediates STAT3 oncogenic

function. J Biol Chem 2008;283:14665-73.

62. Boniface K, Diveu C, Morel F, Pedretti N, Froger J, Ravon E, et al. Oncostatin M

secreted by skin infiltrating T lymphocytes is a potent keratinocyte activator

Involved in skin inflammation. J Immunol 2007;178:4615-22.

63. Cheng F, Wang HW, Cuenca A, Huang M, Ghansah T, Brayer J, et al. A critical

role for Stat3 signaling in immune tolerance. Immunity 2003;19:425-36.

64. Jarnicki A, Putoczki T, Ernst M. Stat3: linking inflammation to epithelial cancer -

more than a "gut" feeling? Cell Div 2010;5:14.

65. Rokavec M, Oner MG, Li H, Jackstadt R, Jiang L, Lodygin D, et al. IL-

6R/STAT3/miR-34a feedback loop promotes EMT-mediated colorectal cancer

invasion and metastasis. J Clin Invest 2014;124:1853-67.

66. Koeberlein B, Hausen AZ, Bektas N, Zentgraf H, Chin R, Toan NL, et al.

Hepatitis B virus overexpresses suppressor of cytokine signaling-3 (SOCS3)

thereby contributing to severity of inflammation in the liver. Virus Res 2009.

109

67. Schroer N, Pahne J, Walch B, Wickenhauser C, Smola S. Molecular pathobiology

of human cervical high-grade lesions: paracrine STAT3 activation in tumor-

instructed myeloid cells drives local MMP-9 expression. Cancer Res 2011;71:87-

97.

68. Duechting A, Tschope C, Kaiser H, Lamkemeyer T, Tanaka N, Aberle S, et al.

Human parvovirus B19 NS1 protein modulates inflammatory signaling by

activation of STAT3/PIAS3 in human endothelial cells. J Virol 2008;82:7942-52.

69. Hino R, Uozaki H, Murakami N, Ushiku T, Shinozaki A, Ishikawa S, et al.

Activation of DNA methyltransferase 1 by EBV latent membrane protein 2A

leads to promoter hypermethylation of PTEN gene in gastric carcinoma. Cancer

Res 2009;69:2766-74.

70. Zhang L, Alizadeh D, Van Handel M, Kortylewski M, Yu H, Badie B. Stat3

inhibition activates tumor macrophages and abrogates glioma growth in mice.

Glia 2009;181:2451-60

71. Xie TX, Huang FJ, Aldape KD, Kang SH, Liu M, Gershenwald JE, et al.

Activation of stat3 in human melanoma promotes brain metastasis. Cancer Res

2006;66:3188-96.

72. Landen CN, Jr., Lin YG, Armaiz Pena GN, Das PD, Arevalo JM, Kamat AA, et al.

Neuroendocrine modulation of signal transducer and activator of transcription-3

in ovarian cancer. Cancer Res 2007;67:10389-96.

73. Iwata-Kajihara T, Sumimoto H, Kawamura N, Ueda R, Takahashi T, Mizuguchi

H, et al. Enhanced Cancer Immunotherapy Using STAT3-Depleted Dendritic

110

Cells with High Th1-Inducing Ability and Resistance to Cancer Cell-Derived

Inhibitory Factors. J Immunol 2011;187:27-36.

74. Hossain DM, Dos Santos C, Zhang Q, Kozlowska A, Liu H, Gao C, et al.

Leukemia cell-targeted STAT3 silencing and TLR9 triggering generate systemic

antitumor immunity. Blood 2014;123:15-25.

75. Yoon S, Woo SU, Kang JH, Kim K, Shin HJ, Gwak HS, et al. NF-kappaB and

STAT3 cooperatively induce IL6 in starved cancer cells. Oncogene 2011.

76. McFarland BC, Hong SW, Rajbhandari R, Twitty GB, Gray GK, Yu H, et al. NF-

κB induced IL-6 ensures STAT3 activation and tumor aggressiveness in

glioblastoma. PLoS One 2013;8:e78728.

77. Guzzo C, Che Mat NF, Gee K. Interleukin-27 Induces a STAT1/3- and NF-

{kappa}B-dependent Proinflammatory Cytokine Profile in Human Monocytes. J

Biol Chem 2010;285:24404-11.

78. Dai B, Meng J, Peyton M, Girard L, Bornmann WG, Ji L, et al. STAT3 Mediates

Resistance to MEK Inhibitor through MicroRNA miR-17. Cancer Res

2011;71:3658-68.

79. Collins AS, McCoy CE, Lloyd AT, O'Farrelly C, Stevenson NJ. miR-19a: An

Effective Regulator of SOCS3 and Enhancer of JAK-STAT Signalling. PloS one

2013;8:e69090.

80. Zhang M, Liu Q, Mi S, Liang X, Zhang Z, Su X, et al. Both miR-17-5p and miR-

20a Alleviate Suppressive Potential of Myeloid-Derived Suppressor Cells by

Modulating STAT3 Expression. J Immunol 2011;186:4716-24.

111

81. Pichiorri F, Suh SS, Ladetto M, Kuehl M, Palumbo T, Drandi D, et al.

MicroRNAs regulate critical genes associated with multiple myeloma

pathogenesis. Proc Natl Acad Sci U S A 2008;105:12885-90.

82. Wang F, Wei J, Gjyshi O, Kong L-Y, Xu S, Lang F, et al. The Microrna

Regulatory Axis of STAT3: Implications for Glioma Stem Cell Biology. Neuro-

Oncology 2012;14:vi45.

83. Xin H, Herrmann A, Reckamp K, Zhang W, Pal S, Hedvat M, et al. Anti-

angiogenic and anti-metastatic activity of JAK inhibitor AZD1480. Cancer Res

2011;21:6601-10.

84. Abad C, Nobuta H, Li J, Kasai A, Yong WH, Waschek JA. Targeted STAT3

disruption in myeloid cells alters immunosuppressor cell abundance in a murine

model of spontaneous medulloblastoma. J Leukoc Biol 2014;95:357-67.

85. Nefedova Y, Nagaraj S, Rosenbauer A, Muro-Cacho C, Sebti SM, Gabrilovich DI.

Regulation of dendritic cell differentiation and antitumor immune response in

cancer by pharmacologic-selective inhibition of the janus-activated kinase

2/signal transducers and activators of transcription 3 pathway. Cancer Res

2005;65:9525-35.

86. Zhang H, Nguyen-Jackson H, Panopoulos AD, Li HS, Murray PJ, Watowich SS.

STAT3 controls myeloid progenitor growth during emergency granulopoiesis.

Blood 2010;116:2462-71.

87. Rosborough BR, Mathews LR, Matta BM, Liu Q, Raich-Regue D, Thomson AW,

et al. Cutting Edge: Flt3 Ligand Mediates STAT3-Independent Expansion but

112

STAT3-Dependent Activation of Myeloid-Derived Suppressor Cells. J Immunol

2014;192:3470-3.

88. Yen BL, Yen ML, Hsu PJ, Liu KJ, Wang CJ, Bai CH, et al. Multipotent Human

Mesenchymal Stromal Cells Mediate Expansion of Myeloid-Derived Suppressor

Cells via Hepatocyte Growth Factor/c-Met and STAT3. Stem cell reports

2013;1:139-51.

89. Lee H, Pal SK, Reckamp K, Figlin RA, Yu H. STAT3: a target to enhance

antitumor immune response. Cur top microbiol Immunol 2011;344:41-59.

90. Wu L, Du H, Li Y, Qu P, Yan C. Signal transducer and activator of transcription

3 (Stat3C) promotes myeloid-derived suppressor cell expansion and immune

suppression during lung tumorigenesis. Amer J Pathol 2011;179:2131-41.

91. Bromberg J, Wang TC. Inflammation and cancer: IL-6 and STAT3 complete the

link. Cancer Cell 2009;15:79-80.

92. Shuai K, Liu B. Regulation of JAK-STAT signalling in the immune system. Nat

Rev Immunol 2003;3:900-11.

93. Mustelin T, Vang T, Bottini N. Protein tyrosine phosphatases and the immune

response. Nat Rev Immunol 2005;5:43-57.

94. Shuai K. Regulation of cytokine signaling pathways by PIAS proteins. Cell Res

2006;16:196-202.

95. Long J, Wang G, Matsuura I, He D, Liu F. Activation of Smad transcriptional

activity by protein inhibitor of activated STAT3 (PIAS3). Proc Natl Acad Sci U S

A 2004;101:99-104.

113

96. Bourdonnay E, Zaslona Z, Penke LR, Speth JM, Schneider DJ, Przybranowski S,

et al. Transcellular delivery of vesicular SOCS proteins from macrophages to

epithelial cells blunts inflammatory signaling. J Exp Med 2015;212:729-42.

97. Yoshimura A, Suzuki M, Sakaguchi R, Hanada T, Yasukawa H. SOCS,

Inflammation, and Autoimmunity. Frontiers Immunol 2012;3:1-9.

98. Baker BJ, Akhtar LN, Benveniste EN. SOCS1 and SOCS3 in the control of CNS

immunity. Trends Immunol 2009;30:392-400.

99. Lindeman GJ, Wittlin S, Lada H, Naylor MJ, Santamaria M, Zhang JG, et al.

SOCS1 deficiency results in accelerated mammary gland development and

rescues lactation in prolactin receptor-deficient mice. Genes Dev 2001;15:1631-6.

100. Greenhalgh CJ, Rico-Bautista E, Lorentzon M, Thaus AL, Morgan PO, Willson

TA, et al. SOCS2 negatively regulates growth hormone action in vitro and in vivo.

J Clin Invest 2005;115:397-406.

101. Roberts AW, Robb L, Rakar S, Hartley L, Cluse L, Nicola NA, et al. Placental

defects and embryonic lethality in mice lacking suppressor of cytokine signaling 3.

Proc Natl Acad Sci U S A 2001;98:9324-9.

102. Babon JJ, Nicola NA. The biology and mechanism of action of suppressor of

cytokine signaling 3. Growth Fac 2012;48:166-77.

103. Shouda T, Yoshida T, Hanada T, Wakioka T, Oishi M, Miyoshi K, et al.

Induction of the cytokine signal regulator SOCS3/CIS3 as a therapeutic strategy

for treating inflammatory arthritis. J Clin Invest 2001;108:1781-8.

104. Lang R, Pauleau A-L, Parganas E, Takahashi Y, Mages J, Ihle JN, et al. SOCS3

regulates the plasticity of gp130 signaling. Nat Immunol 2003;4:546-50.

114

105. Lehmann U, Schmitz J, Weissenbach M, Sobota RM, Hortner M, Friederichs K,

et al. SHP2 and SOCS3 contribute to Tyr-759-dependent attenuation of

interleukin-6 signaling through gp130. J Biol Chem 2003;278:661-71.

106. Kershaw NJ, Murphy JM, Liau NP, Varghese LN, Laktyushin A, Whitlock EL, et

al. SOCS3 binds specific receptor-JAK complexes to control cytokine signaling

by direct kinase inhibition. Nat Struc& Molecul biol 2013;20:469-76.

107. Hanada T, Yoshida T, Kinjyo I, Minoguchi S, Yasukawa H, Kato S, et al. A

mutant form of JAB/SOCS1 augments the cytokine-induced JAK/STAT pathway

by accelerating degradation of wild-type JAB/CIS family proteins through the

SOCS-box. J Biol Chem 2001;276:40746-54.

108. Haan S, Ferguson P, Sommer U, Hiremath M, McVicar DW, Heinrich PC, et al.

Tyrosine phosphorylation disrupts elongin interaction and accelerates SOCS3

degradation. Biol Chem 2003;278:31972-9.

109. Babon JJ, Sabo JK, Soetopo A, Yao S, Bailey MF, Zhang JG, et al. The SOCS

box domain of SOCS3: Structure and interaction with the elonginBC-cullin5

ubiquitin ligase. J Mol Biol 2008.

110. Fischer P, Lehmann U, Sobota RM, Schmitz J, Niemand C, Linnemann S, et al.

The role of the inhibitors of interleukin-6 signal transduction SHP2 and SOCS3

for desensitization of interleukin-6 signalling. Biochem J 2004;378:449-60.

111. Ozawa Y, Nakao K, Kurihara T, Shimazaki T, Shimmura S, Ishida S, et al. Roles

of STAT3/SOCS3 pathway in regulation of the visual function and ubiquitin

proteasome-dependent degradation of Rhodopsin during retinal inflammation. J

Biol Chem 2008;283:24561-70.

115

112. Kawazoe Y, Naka T, Fujimoto M, Kohzaki H, Morita Y, Narazaki M, et al.

Signal transducer and activation of transcription (STAT)-induced STAT inhibitor

1 (SSI-1)/suppressor of cytokine signaling 1 (SOCS1) inhibits insulin signal

transduction pathway through modulating insulin receptor substrate 1 (IRS-1)

phosphorylation. J Exp Med 2001;193:263-9.

113. Waiboci LW, Ahmed CM, Mujtaba MG, Flowers LO, Martin JP, Haider MI, et al.

Both the suppressor of cytokine signaling 1 (SOCS-1) kinase inhibitory region

and SOCS-1 mimetic bind to JAK2 autophosphorylation site: implications for the

development of a SOCS-1 antagonist. J Immunol 2007;178:5058-68.

114. Tamiya T, Kashiwagi I, Takahashi R, Yasukawa H, Yoshimura A. Suppressors of

Cytokine Signaling (SOCS) Proteins and JAK/STAT Pathways: Regulation of T-

Cell Inflammation by SOCS1 and SOCS3. Arterioscler Thromb Vasc Biol

2011;31:980-5.

115. Krebs DL, Hilton DJ. SOCS proteins: negative regulators of cytokine signaling.

Stem Cells 2001;19:378-87.

116. Ying M, Li D, Yang L, Wang M, Wang N, Chen Y, et al. Loss of SOCS3

expression is associated with an increased risk of recurrent disease in breast

carcinoma. J Cancer Res Clin Oncol 2010;48:166-77.

117. Tomita S, Ishibashi K, Hashimoto K, Sugino T, Yanagida T, Kushida N, et al.

Suppression of SOCS3 increases susceptibility of renal cell carcinoma to

interferon-alpha. Cancer Sci 2011;102:57-63.

116

118. Pierconti F, Martini M, Pinto F, Cenci T, Capodimonti S, Calarco A, et al.

Epigenetic silencing of SOCS3 identifies a subset of prostate cancer with an

aggressive behavior. Prostate 2011;71:318-25.

119. Wu ZS, Cheng XW, Wang XN, Song NJ. Prognostic significance of

phosphorylated signal transducer and activator of transcription 3 and suppressor

of cytokine signaling 3 expression in human cutaneous melanoma. Melanoma Res

2011;21:483-90.

120. Li Y, de Haar C, Peppelenbosch MP, van der Woude CJ. SOCS3 in immune

regulation of inflammatory bowel disease and inflammatory bowel disease-related

cancer. Cytokine Growth Factor Rev 2012;23:127-38.

121. Berger H, Vegran F, Chikh M, Gilardi F, Ladoire S, Bugaut H, et al. SOCS3

transactivation by PPARgamma prevents IL-17-driven cancer growth. Cancer Res

2013; 2005;48:186-97.

122. Marine J-C, McKay C, Wang D, Topham DJ, Parganas E, Kakajima H, et al.

SOCS3 is essential in the regulation of fetal liver erythropoiesis. Cell

1999;98:617-27.

123. Kimura A, Kinjyo I, Matsumura Y, Mori H, Mashima R, Harada M, et al. SOCS3

is a physiological negative regulator for granulopoiesis and granulocyte colony-

stimulating factor receptor signaling. J Biol Chem 2004;279:6905-10.

124. Campbell IL. Cytokine-mediated inflammation, tumorigenesis, and disease-

associated JAK/STAT/SOCS signaling circuits in the CNS. Brain Res Brain Res

Rev 2005;48:166-77.

117

125. Ernst M, Inglese M, Waring P, Campbell IK, Bao S, Clay FJ, et al. Defective

gp130-mediated signal transducer and activator of transcription (STAT) signaling

results in degenerative joint disease, gastrointestinal ulceration, and failure of

uterine implantation. J Exp Med 2001;194:189-203.

126. Croker BA, Kiu H, Pellegrini M, Toe J, Preston S, Metcalf D, et al. IL-6 promotes

acute and chronic inflammatory disease in the absence of SOCS3. Immunol Cell

Biol 2011;48:166-77.

127. Zhang Z, Zeng B, Jiao G, Li H, Jing Z, Ouyang J, et al. Suppressor of cytokine

signaling 3 promotes bone marrow cells to differentiate into CD8+ T lymphocytes

in lung tissue via up-regulating Notch1 expression. Cancer Res 2009;69:1578-86.

128. Owaki T, Asakawa M, Kamiya S, Takeda K, Fukai F, Mizuguchi J, et al. IL-27

suppresses CD28-medicated IL-2 production through suppressor of cytokine

signaling 3. J Immunol 2006;176:2773-80.

129. Seki Y, Inoue H, Nagata N, Hayashi K, Fukuyama S, Matsumoto K, et al. SOCS-

3 regulates onset and maintenance of T(H)2-mediated allergic responses. Nat Med

2003;9:1047-54.

130. Daegelmann C, Herberth G, Roder S, Herbarth O, Giese T, Kramer U, et al.

Association between suppressors of cytokine signalling, T-helper type 1/T-helper

type 2 balance and allergic sensitization in children. Clin Exp Allergy

2008;38:438-48.

131. Zhang X, Tao Y, Wang J, Garcia-Mata R, Markovic-Plese S. Simvastatin inhibits

secretion of Th17-polarizing cytokines and antigen presentation by DCs in

patients with relapsing remitting multiple sclerosis. Eur J Immunol 2013;43:281-9.

118

132. Le Y, Zhu BM, Harley B, Park SY, Kobayashi T, Manis JP, et al. SOCS3 protein

developmentally regulates the chemokine receptor CXCR4-FAK signaling

pathway during B lymphopoiesis. Immunity 2007;27:811-23.

133. Liu Y, Stewart KN, Bishop E, Marek CJ, Kluth DC, Rees AJ, et al. Unique

expression of suppressor of cytokine signaling 3 is essential for classical

macrophage activation in rodents in vitro and in vivo. J Immunol 2008;180:6270-

8.

134. Niemand C, Nimmesgern A, Haan S, Fischer P, Schaper F, Rossaint R, et al.

Activation of STAT3 by IL-6 and IL-10 in primary human macrophages is

differentially modulated by suppressor of cytokine signaling 3. J Immunol

2003;170:3263-72.

135. Ilangumaran S, Ramanathan S, Rottapel R. Regulation of the immune system by

SOCS family adaptor proteins. Semin Immunol 2004;16:351-65.

136. Narayana Y, Balaji KN. Notch1 upregulation and signaling involved in

Mycobacterium bovis BCG induced SOCS3 expression in macrophages. J Biol

Chem 2008;283:12501-11.

137. Qin H, Holdbrooks AT, Liu Y, Reynolds SL, Yanagisawa LL, Benveniste EN.

SOCS3 deficiency promotes M1 macrophage polarization and inflammation. J

Immunol 2012;189:3439-48.

138. Yan C, Ward PA, Wang X, Gao H. Myeloid depletion of SOCS3 enhances LPS-

induced acute lung injury through CCAAT/enhancer binding protein delta

pathway. FASEB journal 2013; 122:2317-25.

119

139. Qin H, Yeh W-I, De Sarno P, Holdbrooks AT, Liu Y, Muldowney MT, et al.

Signal transducer and activator of transcription-3/suppressor of cytokine

signaling-3 (STAT3/SOCS3) axis in myeloid cells regulates neuroinflammation.

Proc Natl Acad Sci U S A 2012;109:5004-9.

140. Jackson SH, Yu CR, Mahdi RM, Ebong S, Egwuagu CE. Dendritic cell

maturation requires STAT1 and is under feedback regulation by suppressors of

cytokine signaling. J Immunol 2004;172:2307-15.

141. Orabona C, Pallotta MT, Volpi C, Fallarino F, Vacca C, Bianchi R, et al. SOCS3

drives proteasomal degradation of indoleamine 2,3-dioxygenase (IDO) and

antagonizes IDO-dependent tolerogenesis. Proc Natl Acad Sci U S A

2008;105:20828-33.

142. Matsumura Y, Kobayashi T, Ichiyama K, Yoshida R, Hashimoto M, Takimoto T,

et al. Selective expansion of foxp3-positive regulatory T cells and

immunosuppression by suppressors of cytokine signaling 3-deficient dendritic

cells. J Immunol 2007;179:2170-9.

143. Carlier J, Martin H, Mariame B, Rauwel B, Mengelle C, Weclawiak H, et al.

Paracrine inhibition of GM-CSF signaling by human cytomegalovirus in

monocytes differentiating to dendritic cells. Blood 2011.

144. Cui Q, Jiang W, Wang Y, Lv C, Luo J, Zhang W, et al. Transfer of suppressor of

cytokine signaling 3 by an oncolytic adenovirus induces potential antitumor

activities in hepatocellular carcinoma. Hepatol 2008;47:105-12.

145. Fridman WH, Pages F, Sautes-Fridman C, Galon J. The immune contexture in

human tumours: impact on clinical outcome. Nature Rev Cancer 2012;12:298-306.

120

146. Vesely MD, Kershaw MH, Schreiber RD, Smyth MJ. Natural innate and adaptive

immunity to cancer. Annu Rev Immunol;29:235-71.

147. Ikeda H, Old LJ, Schreiber RD. The roles of IFN in protection against tumor

development and cancer immunoediting. Cytokine Growth Factor Rev

2002;13:95-109.

148. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting:

from immunosurveillance to tumor escape. Nat Immunol 2002;3:991-8.

149. Koebel CM, Vermi W, Swann JB, Zerafa N, Rodig SJ, Old LJ, et al. Adaptive

immunity maintains occult cancer in an equilibrium state. Nature 2007;343:1022-

28.

150. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's

roles in cancer suppression and promotion. Science 2011;331:1565-70.

151. Matsushita H, Vesely MD, Koboldt DC, Rickert CG, Uppaluri R, Magrini VJ, et

al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer

immunoediting. Nature 2012;482:400-4.

152. O'Sullivan T, Saddawi-Konefka R, Vermi W, Koebel CM, Arthur C, White JM, et

al. Cancer immunoediting by the innate immune system in the absence of

adaptive immunity. J Exp Med 2012;209:1869-82.

153. Whiteside TL. The tumor microenvironment and its role in promoting tumor

growth. Oncogene 2008;27:5904-12.

154. Zhu Z, Singh V, Watkins SK, Bronte V, Shoe JL, Feigenbaum L, et al. High-

avidity T cells are preferentially tolerized in the tumor microenvironment. Cancer

Res 2013;73:595-604.

121

155. Ozao-Choy J, Ma G, Kao J, Wang GX, Meseck M, Sung M, et al. The novel role

of tyrosine kinase inhibitor in the reversal of immune suppression and modulation

of tumor microenvironment for immune-based cancer therapies. Cancer Res

2009;69:2514-22.

156. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell

2011;144:646-74.

157. Haffner MC, Mosbruger T, Esopi DM, Fedor H, Heaphy CM, Walker DA, et al.

Tracking the clonal origin of lethal prostate cancer. J clinl Invest 2013;123:4918-

22.

158. Sasaki T, Ryo A, Uemura H, Ishiguro H, Inayama Y, Yamanaka S, et al. An

immunohistochemical scoring system of prolyl isomerase Pin1 for predicting

relapse of prostate carcinoma after radical prostatectomy. Pathol Res Pract

2006;202:357-64.

159. Ogino S, Galon J, Fuchs CS, Dranoff G. Cancer immunology--analysis of host

and tumor factors for personalized medicine. Nature Rev Clin oncol 2011;8:711-9.

160. Carro MS, Lim WK, Alvarez MJ, Bollo RJ, Zhao X, Snyder EY, et al. The

transcriptional network for mesenchymal transformation of brain tumours. Nature

2010;463:318-25.

161. Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nat Rev

Cancer 2009;9:239-52.

162. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell

2010;140:883-99.

122

163. Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C, Flavell RA. Inflammation-

induced cancer: crosstalk between tumours, immune cells and microorganisms.

Nat Rev Cancer 2013;13:759-71.

164. Tran Thang NN, Derouazi M, Philippin G, Arcidiaco S, Di Berardino-Besson W,

Masson F, et al. Immune infiltration of spontaneous mouse astrocytomas is

dominated by immunosuppressive cells from early stages of tumor development.

Cancer Res 2010;70:4829-39.

165. Inozume T, Hanada K, Wang QJ, Yang JC. IL-17 secreted by tumor reactive T

cells induces IL-8 release by human renal cancer cells. J Immunother

2009;32:109-17.

166. Donson AM, Birks DK, Schittone SA, Kleinschmidt-Demasters BK, Sun DY,

Hemenway MF, et al. Increased immune gene expression and immune cell

infiltration in high-grade astrocytoma distinguish long-term from short-term

survivors. J Immunol 2012;189:1920-7.

167. Critchley-Thorne RJ, Simons DL, Yan N, Miyahira AK, Dirbas FM, Johnson DL,

et al. Impaired interferon signaling is a common immune defect in human cancer.

Proc Natl Acad Sci U S A 2009;106:9010-5.

168. Griesinger AM, Birks DK, Donson AM, Amani V, Hoffman LM, Waziri A, et al.

Characterization of Distinct Immunophenotypes across Pediatric Brain Tumor

Types. J Immunol 2013;191:4880-8.

169. Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor

microenvironment. Nat Immunol 2013;14:1014-22.

123

170. Talmadge JE, Donkor M, Scholar E. Inflammatory cell infiltration of tumors:

Jekyll or Hyde. Cancer Metastasis Rev 2007;26:373-400.

171. Danese S, Mantovani A. Inflammatory bowel disease and intestinal cancer: a

paradigm of the Yin-Yang interplay between inflammation and cancer. Oncogene

2010;29:3313-23.

172. Fridman WH, Galon J, Pages F, Tartour E, Sautes-Fridman C, Kroemer G.

Prognostic and predictive impact of intra- and peritumoral immune infiltrates.

Cancer Res 2011;71:5601-5.

173. Broz Miranda L, Binnewies M, Boldajipour B, Nelson Amanda E, Pollack

Joshua L, Erle David J, et al. Dissecting the Tumor Myeloid Compartment

Reveals Rare Activating Antigen-Presenting Cells Critical for T Cell Immunity.

Cancer Cell;26:638-52.

174. Marx J. Cancer biology. All in the stroma: cancer's Cosa Nostra. Science

2008;320:38-41.

175. Jaiswal S, Chao MP, Majeti R, Weissman IL. Macrophages as mediators of tumor

immunosurveillance. Trends Immunol 2010;31:212-9.

176. Lawrence T, Hageman T, Balkwill F. Cancer. Sex, cytokines, and cancer. Science

2007;317:51-2.

177. Ruffell B, Au A, Rugo HS, Esserman LJ, Hwang ES, Coussens LM. Leukocyte

composition of human breast cancer. Proc Natl Acad Sci U S A 2012;109:2796-

801.

178. Marx J. Cancer immunology. Cancer's bulwark against immune attack: MDS cells.

Science 2008;319:154-6.

124

179. Ostrand-Rosenberg S, Sinha P. Myeloid-derived suppressor cells: linking

inflammation and cancer. J Immunol 2009;182:4499-506.

180. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of

myeloid cells by tumours. Nat Rev Immunol 2012;12:253-68.

181. Coussens LM, Zitvogel L, Palucka AK. Neutralizing tumor-promoting chronic

inflammation: a magic bullet? Science 2013;339:286-91.

182. Sawant A, Hensel JA, Chanda D, Harris BA, Siegal GP, Maheshwari A, et al.

Depletion of plasmacytoid dendritic cells inhibits tumor growth and prevents bone

metastasis of breast cancer cells. J Immunol 2012;189:4258-65.

183. Arima K, Watanabe N, Hanabuchi S, Chang M, Sun SC, Liu YJ. Distinct signal

codes generate dendritic cell functional plasticity. Sci Signal 2010;3:ra4.

184. Yang XD, Ai W, Asfaha S, Bhagat G, Friedman RA, Jin G, et al. Histamine

deficiency promotes inflammation-associated carcinogenesis through reduced

myeloid maturation and accumulation of CD11b+Ly6G+ immature myeloid cells.

Nat Med 2011;17:87-95.

185. Pang Y, Gara SK, Achyut BR, Li Z, Yan HH, Day CP, et al. TGF-beta signaling

in myeloid cells is required for tumor metastasis. Cancer Discov 2013;3:936-51.

186. Zhang Z, Liu Q, Che Y, Yuan X, Dai L, Zeng B, et al. Antigen presentation by

dendritic cells in tumors is disrupted by altered metabolism that involves pyruvate

kinase M2 and its interaction with SOCS3. Cancer Res 2010;70:89-98.

187. Sultan AS, Brim H, Sherif ZA. Co-overexpression of Janus kinase 2 and signal

transducer and activator of transcription 5a promotes differentiation of mammary

125

cancer cells through reversal of epithelial-mesenchymal transition. Cancer Sci

2008;99:272-9.

188. Ganguly D, Haak S, Sisirak V, Reizis B. The role of dendritic cells in

autoimmunity. Nature Rev Immunol 2013;13:566-77.

189. Palucka K, Ueno H, Banchereau J. Recent developments in cancer vaccines. J

Immunol 2011;186:1325-31.

190. Watkins SK, Hurwitz AA. FOXO3: A master switch for regulating tolerance and

immunity in dendritic cells. Oncoimmunol 2012;1:252-4.

191. Blankenstein T, Coulie PG, Gilboa E, Jaffee EM. The determinants of tumour

immunogenicity. Nature Rev Cancer 2012;12:307-13.

192. Shi C, Pamer EG. Monocyte recruitment during infection and inflammation.

Nature Rev Immunol 2011;11:762-74.

193. Ono M, Torisu H, Fukushi J-I, Nishie A, Kuwano M. Biological implications of

macrophage infiltration in human tumor angiogenesis. Cancer Chemother

Pharmacol 1999;43 Suppl:S69-s71.

194. Condeelis J, Pollard JW. Macrophages: obligate partners for tumor cell migration,

invasion, and metastasis. Cell 2006;124:263-6.

195. Sica A, Larghi P, Mancino A, Rubino L, Porta C, Totaro MG, et al. Macrophage

polarization in tumour progression. Semin Cancer Biol 2008;18:349-55.

196. Doedens AL, Stockmann C, Rubinstein MP, Liao D, Zhang N, DeNardo DG, et al.

Macrophage expression of hypoxia-inducible factor-1 alpha suppresses T-cell

function and promotes tumor progression. Cancer Res 2010;70:7465-75.

126

197. De Palma M, Lewis CE. Cancer: Macrophages limit chemotherapy. Nature

2011;472:303-4.

198. Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J

Clin Inv 2012;122:787-95.

199. Kershaw MH, Smyth MJ. Immunology. Making macrophages eat cancer. Science

2013;341:41-2.

200. Lesokhin AM, Hohl TM, Kitano S, Cortez C, Hirschhorn-Cymerman D, Avogadri

F, et al. Monocytic CCR2(+) myeloid-derived suppressor cells promote immune

escape by limiting activated CD8 T-cell infiltration into the tumor

microenvironment. Cancer Res 2012;72:876-86.

201. Mantovani A, Biswas SK, Galdiero MR, Sica A, Locati M. Macrophage plasticity

and polarization in tissue repair and remodelling. J Pathol 2013;229:176-85.

202. Jordan JT, Sun W, Hussain SF, Deangulo G, Prabhu SS, Heimberger AB.

Preferential migration of regulatory T cells mediated by glioma-secreted

chemokines can be blocked with chemotherapy. Cancer Immunol Immunother

2008;57:123-31.

203. Castellone MD, Teramoto H, Williams BO, Druey KM, Gutkind JS.

Prostaglandin E2 promotes colon cancer cell growth through a Gs-axin-beta-

catenin signaling axis. Science 2005;310:1504-10.

204. Amarnath S, Mangus CW, Wang JCM, Wei F, He A, Kapoor V, et al. The PDL1-

PD1 Axis Converts Human TH1 Cells into Regulatory T Cells. Sci Trans Med

2011;3:111ra20.

127

205. Min Y, Ren X, Vaught DB, Chen J, Donnelly E, Lynch CC, et al. Tie2 signaling

regulates osteoclastogenesis and osteolytic bone invasion of breast cancer. Cancer

Res 2010;70:2819-28.

206. Cortez-Retamozo V, Etzrodt M, Newton A, Rauch PJ, Chudnovskiy A, Berger C,

et al. Origins of tumor-associated macrophages and neutrophils. Proc Natl Acad

Sci U S A 2012;109:2491-6.

207. Talmadge JE, Gabrilovich DI. History of myeloid-derived suppressor cells. Nat

Rev Cancer 2013;13:739-52.

208. Croker BA, Metcalf D, Robb L, Wei W, Mifsud S, DiRago L, et al. SOCS3 is a

critical physiological negative regulator of G-CSF signaling and emergency

granulopoiesis. Immunity 2004;20:153-65.

209. Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of myeloid-derived

suppressor cells in tumor-bearing mice. J Immunol 2008;181:5791-802.

210. Movahedi K, Laoui D, Gysemans C, Baeten M, Stange G, Van den Bossche J, et

al. Different tumor microenvironments contain functionally distinct subsets of

macrophages derived from Ly6C(high) monocytes. Cancer Res 2010;70:5728-39.

211. Mantovani A, Cassatella MA, Costantini C, Jaillon S. Neutrophils in the

activation and regulation of innate and adaptive immunity. Nat Rev Immunol

2011;11:519-31.

212. Medina-Echeverz J, Fioravanti J, Zabala M, Ardaiz N, Prieto J, Berraondo P.

Successful colon cancer eradication after chemoimmunotherapy is associated with

profound phenotypic change of intratumoral myeloid cells. J Immunol

2011;186:807-15.

128

213. Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the

promotion of tumour angiogenesis. Nat Rev Cancer 2008;8:618-31.

214. Nguyen-Jackson H, Panopoulos AD, Zhang H, Li HS, Watowich SS. STAT3

controls the neutrophil migratory response to CXCR2 ligands by direct activation

of G-CSF-induced CXCR2 expression and via modulation of CXCR2 signal

transduction. Blood 2010;115:3354-63.

215. Yang L, Pang Y, Moses HL. TGF-beta and immune cells: an important regulatory

axis in the tumor microenvironment and progression. Trends Immunol

2010;31:220-7.

216. Park S, Zhao D, Hatanpaa KJ, Mickey BE, Saha D, Boothman DA, et al. RIP1

Activates PI3K-Akt via a Dual Mechanism Involving NF-{kappa}B-Mediated

Inhibition of the mTOR-S6K-IRS1 Negative Feedback Loop and Down-

regulation of PTEN. Cancer Res 2009;69:4107-11.

217. Britschgi A, Andraos R, Brinkhaus H, Klebba I, Romanet V, Muller U, et al.

JAK2/STAT5 inhibition circumvents resistance to PI3K/mTOR blockade: a

rationale for cotargeting these pathways in metastatic breast cancer. Cancer cell

2012;22:796-811.

218. Das Roy L, Pathangey LB, Tinder TL, Schettini JL, Gruber HE, Mukherjee P.

Breast-cancer-associated metastasis is significantly increased in a model of

autoimmune arthritis. Breast Cancer Res 2009;11:R56.

219. Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, et al. Polarization of

tumor-associated neutrophil phenotype by TGF-beta: "N1" versus "N2" TAN.

Cancer Cell 2009;16:183-94.

129

220. Smith C, Chang MY, Parker KH, Beury DW, DuHadaway JB, Flick HE, et al.

IDO is a nodal pathogenic driver of lung cancer and metastasis development.

Cancer Discov 2012;2:722-35.

221. Jia W, Jackson-Cook C, Graf MR. Tumor-infiltrating, myeloid-derived suppressor

cells inhibit T cell activity by nitric oxide production in an intracranial rat glioma

+ vaccination model. J Neuroimmunol 2010;223:20-30.

222. Bald T, Quast T, Landsberg J, Rogava M, Glodde N, Lopez-Ramos D, et al.

Ultraviolet-radiation-induced inflammation promotes angiotropism and metastasis

in melanoma. Nature 2014;507:109-13.

223. Sinha P, Clements VK, Bunt SK, Albelda SM, Ostrand-Rosenberg S. Cross-talk

between myeloid-derived suppressor cells and macrophages subverts tumor

immunity toward a type 2 response. J Immunol 2007;179:977-83.

224. Nagaraj S, Gupta K, Pisarev V, Kinarsky L, Sherman S, Kang L, et al. Altered

recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat

Med 2007;13:828-35.

225. Greten TF, Manns MP, Korangy F. Myeloid derived suppressor cells in human

diseases. International immunopharmacology 2011;11:802-7.

226. Khaled YS, Ammori BJ, Elkord E. Myeloid-derived suppressor cells in cancer:

recent progress and prospects. Immunol Cell Biol 2013;91:493-502.

227. Youn JI, Gabrilovich DI. The biology of myeloid-derived suppressor cells: the

blessing and the curse of morphological and functional heterogeneity. Eur J

Immunol 2010;40:2969-75.

130

228. Ostrand-Rosenberg S, Sinha P, Beury DW, Clements VK. Cross-talk between

myeloid-derived suppressor cells (MDSC), macrophages, and dendritic cells

enhances tumor-induced immune suppression. Semi Cancer bio 2012;22:275-81.

229. Ortiz ML, Lu L, Ramachandran I, Gabrilovich DI. Myeloid-derived suppressor

cells in the development of lung cancer. Cancer Immunol Res 2014;2:50-8.

230. Peranzoni E, Zilio S, Marigo I, Dolcetti L, Zanovello P, Mandruzzato S, et al.

Myeloid-derived suppressor cell heterogeneity and subset definition. Cur Opin in

Immunol 2010;22:238-44.

231. Chae M, Peterson TE, Parney IF, Johnson AJ. Increasing Intracranial Glioma-

Associated Macrophages Drives Systemic Myeloid-Derived Suppressor Cell

Expansion. Neuro-Oncology 2012;14:vi42.

232. Chatterjee S, Das S, Chakraborty P, Manna A, Chatterjee M, Choudhuri SK.

Myeloid derived suppressor cells (MDSCs) can induce the generation of Th17

response from naive CD4+ T cells. Immunobio 2013;218:718-24.

233. Lu T, Ramakrishnan R, Altiok S, Youn JI, Cheng P, Celis E, et al. Tumor-

infiltrating myeloid cells induce tumor cell resistance to cytotoxic T cells in mice.

The J Clin Invest 2011;121:4015-29.

234. Panopoulos AD, Zhang L, Snow JW, Jones DM, Smith AM, El Kasmi KC, et al.

STAT3 governs distinct pathways in emergency granulopoiesis and mature

neutrophils. Blood 2006;108:3682-90.

235. Panopoulos AD, Watowich SS. Granulocyte colony-stimulating factor: molecular

mechanisms of action during steady state and 'emergency' hematopoiesis.

Cytokine 2008;42:277-88.

131

236. Gordy C, Pua H, Sempowski GD, He YW. Regulation of steady-state neutrophil

homeostasis by macrophages. Blood 2011;117:618-29.

237. Eyles JL, Hickey MJ, Norman MU, Croker BA, Roberts AW, Drake SF, et al. A

key role for G-CSF-induced neutrophil production and trafficking during

inflammatory arthritis. Blood 2008;112:5193-201.

238. Irandoust MI, Aarts LH, Roovers O, Gits J, Erkeland SJ, Touw IP. Suppressor of

cytokine signaling 3 controls lysosomal routing of G-CSF receptor. Embo J

2007;26:1782-93.

239. von Vietinghoff S, Ley K. Homeostatic regulation of blood neutrophil counts. J

Immunol 2008;181:5183-8.

240. Haribabu B, Richardson RM, Fisher I, Sozzani S, Peiper SC, Horuk R, et al.

Regulation of human chemokine receptors CXCR4. J Biol Chem

1997;272:28726-31.

241. Moore MAS. The role of chemoattraction in cancer metastases. Bioessays

2001;23:674-6.

242. Burger J, Kipps T. CXCR4: a key receptor in the crosstalk between tumor cells

and their microenvironment. Blood 2006;107:1761-7.

243. Richert MM, Vaidya KS, Mills CN, Wong D, Korz W, Hurst DR, et al. Inhibition

of CXCR4 by CTCE-9908 inhibits breast cancer metastasis to lung and bone.

Oncol Rep 2009;21:761-7.

244. Lu M, Grove EA, Miller RJ. Abnormal development of the hippocampal dentate

gyrus in mice lacking the CXCR4 chemokine receptor. Proc Natl Acad Sci U S A

2002;99:7090-5.

132

245. Zhou Y, Larsen PH, Hao C, Yong VW. CXCR4 is a major chemokine receptor on

glioma cells and mediates their survival. J Biol Chem 2002;277:49481-7.

246. Srivastava MK, Sinha P, Clements VK, Rodriguez P, Ostrand-Rosenberg S.

Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine

and cysteine. Cancer research 2010;70:68-77.

247. King IL, Dickendesher TL, Segal BM. Circulating Ly-6C+ myeloid precursors

migrate to the CNS and play a pathogenic role during autoimmune demyelinating

disease. Blood 2009;113:3190-7.

248. Ramachandran IR, Martner A, Pisklakova A, Condamine T, Chase T, Vogl T, et

al. Myeloid-derived suppressor cells regulate growth of multiple myeloma by

inhibiting T cells in bone marrow. J Immunol 2013;190:3815-23.

249. Schlecker E, Stojanovic A, Eisen C, Quack C, Falk CS, Umansky V, et al.

Tumor-Infiltrating Monocytic Myeloid-Derived Suppressor Cells Mediate CCR5-

Dependent Recruitment of Regulatory T Cells Favoring Tumor Growth. J

Immunol 2012;189:5602-11.

250. Ko JS, Zea AH, Rini BI, Ireland JL, Elson P, Cohen P, et al. Sunitinib mediates

reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma

patients. Clin Cancer Res 2009;15:2148-57.

251. Condamine T, Gabrilovich DI. Molecular mechanisms regulating myeloid-

derived suppressor cell differentiation and function. Tren Immunol 2011;32:19-25.

252. Li L, Zhang J, Diao W, Wang D, Wei Y, Zhang CY, et al. MicroRNA-155 and

MicroRNA-21 Promote the Expansion of Functional Myeloid-Derived Suppressor

Cells. J Immunol 2014;192:1034-43.

133

253. Marigo I, Bosio E, Solito S, Mesa C, Fernandez A, Dolcetti L, et al. Tumor-

induced tolerance and immune suppression depend on the C/EBPbeta

transcription factor. Immunity 2010;32:790-802.

254. Xu J, Escamilla J, Mok S, David J, Priceman S, West B, et al. CSF1R signaling

blockade stanches tumor-infiltrating myeloid cells and improves the efficacy of

radiotherapy in prostate cancer. Cancer Res 2013;73:2782-94.

255. Sawant A, Schafer CC, Jin TH, Zmijewski J, Tse HM, Roth J, et al. Enhancement

of Antitumor Immunity in Lung Cancer by Targeting Myeloid-Derived

Suppressor Cell Pathways. Cancer Res 2013:1-12

256. Begemann M, Fuller GN, Holland EC. Genetic modeling of glioma formation in

mice. Brain Pathol 2002;12:117-32.

257. Greenberg NM, DeMayo F, Finegold MJ, Medina D, Tilley WD, Aspinall JO, et

al. Prostate cancer in a transgenic mouse. Proc Natl Acad Sci U S A

1995;92:3439-43.

258. Berger MF, Lawrence MS, Demichelis F, Drier Y, Cibulskis K, Sivachenko AY,

et al. The genomic complexity of primary human prostate cancer. Nature

2011;470:214-20.

259. Roychowdhury S, Chinnaiyan AM. Advancing precision medicine for prostate

cancer through genomics. J Clin Oncol 2013;31:1866-73.

260. Bartek J, Mistrik M, Bartkova J. Androgen receptor signaling fuels DNA repair

and radioresistance in prostate cancer. Cancer Discov 2013;3:1222-4.

134

261. Polkinghorn WR, Parker JS, Lee MX, Kass EM, Spratt DE, Iaquinta PJ, et al.

Androgen receptor signaling regulates DNA repair in prostate cancers. Cancer

Discov 2013;3:1245-53.

262. da Silva HB, Amaral EP, Nolasco EL, de Victo NC, Atique R, Jank CC, et al.

Dissecting Major Signaling Pathways throughout the Development of Prostate

Cancer. Prostate cancer 2013;2013:920612.

263. Pienta KJ, Walia G, Simons JW, Soule HR. Beyond the androgen receptor: New

approaches to treating metastatic prostate cancer. Report of the 2013 Prouts Neck

Prostate Cancer Meeting. The Prostate 2014;74:314-20.

264. McNutt M. Cancer immunotherapy. Science 2013;342:1417.

265. Couzin-Frankel J. Breakthrough of the year 2013. Cancer immunotherapy.

Science 2013;342:1432-3.

266. Nelson WG. Profiling prostate cancer. Proc Natl Acad Sci U S A

2011;108:20861-2.

267. Gladson CL, Welch DR. New insights into the role of CXCR4 in prostate cancer

metastasis. Cancer Biol Ther 2008;7.

268. Hong MK, Kong J, Namdarian B, Longano A, Grummet J, Hovens CM, et al.

Paraneoplastic syndromes in prostate cancer. Nat Rev Urol 2010;7:681-92.

269. Goldstein AS, Huang J, Guo C, Garraway IP, Witte ON. Identification of a cell of

origin for human prostate cancer. Science 2010;329:568-71.

270. Hurwitz AA, Foster BA, Allison JP, Greenberg NM, Kwon ED. The TRAMP

Mouse as a Model for Prostate Cancer. Current Protocols in Immunology: John

Wiley & Sons, Inc.; 2001.

135

271. Isayeva T, Chanda D, Kallman L, Eltoum IE, Ponnazhagan S. Effects of sustained

antiangiogenic therapy in multistage prostate cancer in TRAMP model. Cancer

Res 2007;67:5789-97.

272. Gurusamy D, Gray JK, Pathrose P, Kulkarni RM, Finkleman FD, Waltz SE.

Myeloid-specific expression of Ron receptor kinase promotes prostate tumor

growth. Cancer Res 2013;73:1752-63.

273. Irshad S, Abate-Shen C. Modeling prostate cancer in mice: something old,

something new, something premalignant, something metastatic. Cancer Met Rev

2013;32:109-22.

274. Liu Z, Eltoum IE, Guo B, Beck BH, Cloud GA, Lopez RD. Protective

immunosurveillance and therapeutic antitumor activity of gammadelta T cells

demonstrated in a mouse model of prostate cancer. J Immunol 2008;180:6044-53.

275. Irshad S, Bansal M, Castillo-Martin M, Zheng T, Aytes A, Wenske S, et al. A

molecular signature predictive of indolent prostate cancer. Sci Trans Med

2013;5:202ra122.

276. Kroon P, Berry PA, Stower MJ, Rodrigues G, Mann VM, Simms M, et al. JAK-

STAT blockade inhibits tumor initiation and clonogenic recovery of prostate

cancer stem-like cells. Cancer Res 2013;73:5288-98.

277. Yu H, Liu Y, McFarland BC, Deshane JS, Hurst DR, Ponnazhagan S, et al.

SOCS3 Deficiency in Myeloid Cells Promotes Tumor Development: Involvement

of STAT3 Activation and Myeloid-Derived Suppressor Cells. Cancer Immunol

Res 2015;3:727-40.

136

278. Shodeinde AL, Barton BE. Potential use of STAT3 inhibitors in targeted prostate

cancer therapy: future prospects. OncoTarg Ther 2012;5:119-25.

279. Yu H, Kortylewski M, Pardoll D. Crosstalk between cancer and immune cells:

role of STAT3 in the tumour microenvironment. Nat Rev Immunol 2007;7:41-51.

280. Schroeder A, Herrmann A, Cherryholmes G, Kowolik C, Buettner R, Pal S, et al.

Loss of androgen receptor expression promotes a stem-like cell phenotype in

prostate cancer through STAT3 signaling. Cancer Res 2014;74:1227-37.

281. Boyle K, Egan P, Rakar S, Willson TA, Wicks IP, Metcalf D, et al. The SOCS

box of suppressor of cytokine signaling-3 contributes to the control of G-CSF

responsiveness in vivo. Blood 2007;110:1466-74.

282. Bayne LJ, Beatty GL, Jhala N, Clark CE, Rhim AD, Stanger BZ, et al. Tumor-

derived granulocyte-macrophage colony-stimulating factor regulates myeloid

inflammation and T cell immunity in pancreatic cancer. Cancer Cell 2012;21:822-

35.

283. Waight JD, Netherby C, Hensen ML, Miller A, Hu Q, Liu S, et al. Myeloid-

derived suppressor cell development is regulated by a STAT/IRF-8 axis. J Clin

Invest 2013;123:4464-78.

284. Bekisz J, Sato Y, Johnson C, Husain SR, Puri RK, Zoon KC. Immunomodulatory

effects of interferons in malignancies. J Interf & Cyto Res 2013;33:154-61.

285. Calarco A, Pinto F, Pierconti F, Sacco E, Marrucci E, Totaro A, et al. Role of

SOCS3 evaluated by immunohistochemical analysis in a cohort of patients

affected by prostate cancer: preliminary results. Urologia 2012;79 Suppl 19:4-8.

137

286. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat

Rev Cancer 2012;12:252-64.

138

APPENDIX

INSTITUTIONAL ANIMAL CARE AND USE COMMITTEE APPROVAL

139

140


Recommended