STATs in cancer inflammation and immunity: a leading role for STAT3 (original) (raw)

Nat Rev Cancer. Author manuscript; available in PMC 2016 May 4.

Published in final edited form as:

PMCID: PMC4856025

NIHMSID: NIHMS779912

Hua Yu

*Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, USA.

Drew Pardoll

‡Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland 20892, USA.

Richard Jove

*Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, USA.

*Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, USA.

‡Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland 20892, USA.

Abstract

Commensurate with their roles in regulating cytokine-dependent inflammation and immunity, signal transducer and activator of transcription (STAT) proteins are central in determining whether immune responses in the tumour microenvironment promote or inhibit cancer. Persistently activated STAT3 and, to some extent, STAT5 increase tumour cell proliferation, survival and invasion while suppressing anti-tumour immunity. The persistent activation of STAT3 also mediates tumour-promoting inflammation. STAT3 has this dual role in tumour inflammation and immunity by promoting pro-oncogenic inflammatory pathways, including nuclear factor-κB (NF-κB) and interleukin-6 (IL-6)–GP130–Janus kinase (JAK) pathways, and by opposing STAT1- and NF-κB-mediated T helper 1 anti-tumour immune responses. Consequently, STAT3 is a promising target to redirect inflammation for cancer therapy.

The importance of inflammation in tumour initiation and malignant progression has become the focus of attention with good reason. Inflammatory conditions can initiate or promote oncogenic transformation, and genetic and epigenetic changes in malignant cells can also generate an inflammatory microenvironment that further supports tumour progression1. Cancer-associated inflammation is marked by the presence of specific inflammatory cells and inflammatory mediators, including cytokines and chemokines1. Recent evidence suggests a crucial role for signal transducer and activator of transcription (STAT) family proteins — especially STAT3 — in selectively inducing and maintaining a pro-carcinogenic inflammatory microenvironment, both at the initiation of malignant transformation and during cancer progression19. STAT3 is linked to inflammation-associated tumorigenesis that is initiated by genetic alterations in malignant cells1013, as well as by many environmental factors, including chemical carcinogens, sunlight, infection, cigarette smoking and stress1422.

Because of its ability to induce the expression of a large array of inflammatory mediators and its role as a core transcription factor in diverse immune responses, nuclear factor-κB (NF-κB) signalling has been recognized as a major pathway responsible for both inflammation-induced carcinogenesis and anti-tumour immunity1,2325. Given their central roles in inflammation and cancer23,2531, it is not surprising that signalling by various STATs, particularly STAT3, is highly interconnected with NF-κB signalling3,7,3235. There are striking parallels, as well as contrasts, between NF-κB and STAT3. Both proteins are not only persistently activated in cancer and essential for transducing cytoplasmic signals from extracellular stimuli, but they also function as nuclear transcription factors required for regulating genes involved in tumour proliferation, survival, angiogenesis and invasion, in addition to genes encoding key cancer-promoting inflammatory mediators23,2528,36,37. It is mechanistically relevant that STAT3 interacts with NF-κB at several levels in a highly context-dependent manner. For example, several inflammatory factors encoded by NF-κB target genes, most notably interleukin-6 (IL-6), are important STAT3 activators2,3,7,29,3840. In tumours, STAT3 directly interacts with the NF-κB family member RELA, trapping it in the nucleus and thereby contributing to constitutive NF-κB activation in cancer32. Ultimately, STAT3 and NF-κB also co-regulate numerous oncogenic and inflammatory genes27,28,36,39. Continuous deregulation of these genes in tumour cells and the tumour microenvironment by persistently activated STAT3 and NF-κB — in contrast to their tightly controlled regulation in normal physiology — is crucial for inflammation and malignant progression.

Aside from the tumour-promoting role of inflammation, many murine studies and clinical findings have underscored the importance of immune responses and inflammatory mediators — both naturally occurring and therapeutically induced — in suppressing tumorigenesis and tumour growth4147. STAT3 and, to some extent, STAT5 and STAT6 are involved in inhibiting anti-tumour immunity5,48,49. Although crucial for inducing oncogenic inflammatory conditions, NF-κB is also indispensible for mediating anti-tumour immune responses1,25,28. By contrast, STAT3 activation restrains anti-tumour immune responses46,28,5052 by antagonizing NF-κB- and STAT1-mediated expression of anti-tumour T helper 1 (TH1) cytokines such as IL-12 and interferon-γ (IFNγ), which are necessary for both innate and T cell-mediated anti-tumour immunity5,28,50,53,54. STAT3 signalling in innate immune cells is required for the immunosuppressive and tumour-promoting effects of myeloid-derived suppressor cells (MDSCs) and tumour-associated macrophages4,5,5052. STAT3 also mediates T regulatory cell expansion in tumours and is necessary for the development of TH17 T cells5,50,5557, which can promote tumour growth6,58. Because STAT3 induces the expression of cytokines, growth factors and angiogenic factors, and the associated receptors in turn activate STAT3, a feedforward loop is established between tumour cells and immune cells in the tumour microenvironment4,27,28. As a consequence, persistent activation of several STATs, especially STAT3, mediates both the propagation of tumour-promoting inflammation and the suppression of anti-tumour immunity, and so provides a promising molecular target for modulating immune responses to improve cancer therapy.

STAT proteins in immune modulation

The STAT protein family consists of seven members, which are encoded by distinct genes: STAT1, STAT2, STAT3, STAT4, STAT6, and the closely related STAT5A and STAT5B27,59,60. One distinguishing feature of the proteins encoded by these genes is their dual roles — they both transduce signals through the cytoplasm and function as transcription factors in the nucleus30,38,61. STATs were originally discovered through their capacity to mediate signalling from IFN and IL-6 receptors following engagement with their cognate cytokines29,30,38,40,59,6164. Cytokine receptors do not usually have intrinsic tyrosine kinase activity: instead, their engagement activates receptor-associated tyrosine kinases, most prominently the Janus kinase (JAK) family kinases (JAK1, JAK2, JAK3 and TYK2)27,30,38,59,65. Following phosphorylation of specific tyrosine residues in STAT proteins, they form stable homodimers or heterodimers with other STAT proteins through reciprocal phosphotyrosine–SRC homology 2 (SH2) domain interactions. Each STAT family protein responds to a defined set of cytokines (TABLE 1), and each also regulates, with other transcription factors and/or cofactors, a group of specific genes (TABLES 1,​2). Importantly, many of the downstream target genes of STATs encode cytokines and growth factors, the receptors of which signal through the same STATs, thereby providing a mechanism for autocrine and paracrine STAT activation. For example, in response to IL-6 signalling through GP130 (also known as IL6ST)–JAK, STAT3 forms homodimers that translocate to the nucleus. In the nucleus STAT3–STAT3 homodimers modulate the expression of genes encoding IL-6 itself and other mediators crucial for the classic physiological acute phase response and cancer-promoting inflammatory conditions2,3,7,29,39,63,66 (FIG. 1; TABLE 2). By contrast, on stimulation by type 1 IFNs, STAT1–STAT1 homodimers or STAT1–STAT2 heterodimers accumulate in the nucleus and regulate the expression of genes that promote growth arrest and apoptosis67,68. Type 1 IFNs also activate certain types of innate immunity, including natural killer cell responses, as well as TH1-type adaptive immunity, which is characterized by the production of IFNγ, the receptor of which further activates STAT1 and STAT2 (REFS 59,62,65,69). This loop is crucial for controlling viral infection and mediating anti-tumour immunity (TABLE 1).

An external file that holds a picture, illustration, etc. Object name is nihms-779912-f0001.jpg

STAT3 connects inflammation and cancer

Inflammation and cancer are linked by both oncogenic (intrinsic) and environmental (extrinsic) pathways. The intrinsic pathway is activated by genetic or epigenetic alterations in transformed cells. Such alterations include those that cause the overexpression or the persistent activation of growth factor receptors with intrinsic tyrosine kinase activity and cytokine receptors with associated Janus kinase (JAK) family tyrosine kinases. Oncogenic mutations in receptor-associated JAK family members also underlie some types of cancer. These receptors (several examples are shown), as well as non-receptor tyrosine kinases such as SRC, can be activated by extrinsic pathways — environmental factors that are associated with cancer inflammation — which include ultraviolet (UV) radiation, chemical carcinogens, infection, stress and cigarette smoke. Activated tyrosine kinases induced by both intrinsic and extrinsic pathways phosphorylate signal transducer and activator of transcription 3 (STAT3), which in turn forms dimers that translocate to the nucleus, where they directly regulate gene expression. In addition to upregulating numerous genes involved in proliferation, survival, invasion and metastasis, STAT3 induces the expression of many cytokines, chemokines and other mediators, such as interleukin-6 and cyclooxygenase 2, that are associated with cancer-promoting inflammation. Importantly, receptors for many of these cytokines, chemokines and mediators in turn further activate STAT3, thus forming autocrine and paracrine feedforward loops that result in a stable change to the genetic programme and the promotion of cancer inflammation. P, phosphorylation.

Table 1

STAT family activation by specific cytokines and growth factors, and their target genes

STAT protein Key activators Main target genes Example genes Refs
STAT1 IFNγ, IFNα and IFNβ TH1-type immunostimulatory,and pro-apoptosis TBX21, CD80, CD40, IL-12,CDKN1A and severalcaspases 59
STAT2 IFNα and IFNβ TH1-type immunostimulatory,and pro-apoptosis CD80 and CD40 59,69
STAT3 IL-6, IL-10, IL-23, IL-21,IL-11, LIF and OSM TH17-type,anti-apoptosis,pro-proliferation,angiogenic, and metastatic IL-17, IL-23, BCL-XL, BCL-2,MCL1, CCND1 and VEGF 28,38,57,66,142145
STAT4 IL-12 TH1-type, especially IFNγ _IFN_γ 59,100
STAT5A andSTAT5B IL-2, GM-CSF, IL-15, IL-7,IL-3, IL-5, growth homonesand prolactin Anti-apoptosis,pro-proliferation, anddifferentiation BCL-XL, CCND2 and FOXP3 27,146,147
STAT6 IL-4 and IL-13 TH2-type, and anti-apoptosis GATA3 and BCL-2 59,148

Table 2

STAT3 and NF-κB target genes

Gene Regulated byRELA? Regulated byREL? Upregulatedby STAT3?* Downregulatedby STAT3?* STAT3activators? Refs
BCL-XL 28,36
MYC 28,36
BIRC5 (which encodes survivin) 28,36
MMP9 28,36
MMP2 28,36
CCND1 (which encodes cyclinD1) 28,36
HIF1α 28,36
ICAM1 36,149
TWIST1 36,150
VIM (which encodes vimentin) 36,151
MCL1 28,152
HSP70 and HSP90 153
IL-10 85,101,143
VEGF 4,28,36,154
FGF2 (also known as BFGF) 4,84
COX2 36,87
CXCL12 (also known as SDF1) 155
IL-11 13
IL-23 5,56
IL-21 142
IL-17 6,39
IL-6 39,85,86,101,111
IL-12A (also known as P35) 5,108
CD80 28,111
CD86 28,111
CXCL10 (also known as IP-10) 28,156
IFNγ 101,111
IFNβ 86,111
CCL5 (also known as RANTES) 86,156
NOS2 36,78,101
IL-8 32,75,111
IL-1β 4,101,111
CCL2 (also known as MCP1) 36,98,156,157

STAT3 induces cancer inflammation

Epidemiological studies of chronic inflammatory conditions at specific organ sites provide some of the most compelling evidence that inflammation promotes malignant transformation. Virtually all liver and gastric cancers arise from infections that cause chronic hepatitis (for which the infectious agents are hepatitis B virus and hepatitis C virus) and chronic gastritis (for which the infectious agent is Helicobacter pylori), respectively1. Crohn’s colitis and ulcerative colitis are associated with dramatically increased colon cancer incidence1. However, the microenvironment of cancers with no prior underlying inflammatory history is also laden with haematopoietic-derived cells in various states of activation1. These observations suggest that cancer and inflammation are linked by both extrinsic (environmental) and intrinsic pathways, the latter of which originate from genetic and epigenetic events in the malignant cells1. STAT proteins, especially STAT3, are crucial for both the extrinsic and the intrinsic pathways underlying cancer inflammation2,3,57,1013,29,40,66. Owing to its oncogenic properties, which include causing increased tumour cell proliferation and survival, STAT3 drives the malignant properties that are associated with chronic inflammation3,7,27,31,7073.

STAT3 signalling 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 (TABLE 2). Nevertheless, the ongoing contribution of persistently activated STAT3, as with other overactive transcription factors in cancer, to the continuous transcriptional regulation of any specific gene in tumour cells remains to be determined experimentally. In addition to being downstream of cytokine receptors, STAT3 is activated by growth factor receptor and non-receptor tyrosine kinases (FIG. 1). For example, epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor (PDGFR), among many other polypeptide growth factor receptors, as well as non-receptor tyrosine kinases such as SRC, are upstream of STAT3 and frequently overexpressed or overactivated in solid tumour cells27,29,65,7478.

STAT5 and STAT6 are persistently activated in various haematopoietic malignancies27,7982. In the case of chronic myelogenous leukaemia, the oncogenic BCR–ABL chromosomal translocation causes persistent activation of STAT5 in the malignant cells79,83. IL-13–STAT6 signalling is crucial for the development of certain lymphomas and leukaemias81,82. Both STAT5 and STAT6 have been shown to upregulate genes important for haematopoietic tumour survival and proliferation when persistently activated in tumour cells. A role for these two STATs in tumour immune evasion has also been indicated, which we discuss in later sections. Although site-directed mutations can create constitutively active oncogenic forms of certain STATs, which demonstrates the intrinsic oncogenic potential of STAT3 in particular, the increased activation of STATs in naturally occurring cancers is almost exclusively secondary to the activation of upstream kinases27,74,76,81,82,84. The crucial roles for persistent STAT activation in oncogenesis have been well established in many different studies by a large number of laboratories over the past decade.

Cytokines, chemokines and other mediators, such as IL-6, IL-1β, macrophage colony-stimulating factor, prostaglandins and cyclooxygenase 2 (COX2, which is required for the production of prostaglandins), are crucial for inducing and maintaining a cancer-promoting inflammatory environment1, and STAT3 is crucial for regulating their expression (TABLE 2). Although tumour cells are known to produce some of these mediators, they are mainly produced by the stromal inflammatory cells46,8,9,28,50. Importantly, the persistent activation of STAT3 intrinsic to tumour cells is transmitted to stromal inflammatory cells in the tumour microenvironment85,86. This is because, in tumour cells, STAT3 is a transcription factor for numerous genes encoding cytokines, chemo kines and growth factors, the associated receptors of which in turn activate STAT3 in stromal cells27,29,38,87 (FIG. 1). Therefore, a STAT3 feedforward loop is established between tumour cells and non-transformed cells in the microenvironment, including immune cells28. This feedforward loop is exemplified in multiple myeloma, which provided the first direct evidence for a mechanistic link between persistent STAT3 signalling and human cancer. In myeloma cells, IL-6 drives JAK and STAT3 activation, leading to tumour cell survival through the upregulation of anti-apoptotic genes2. Activation of STAT3 is also observed in multiple myeloma bone marrow stromal cells, which are a major source of IL-6 that induces persistent activation of STAT3 in the tumour cells, thereby establishing a feedforward loop8. Although whether transformed cells and/or non-transformed stromal cells initiate IL-6 production in multiple myeloma remains to be fully characterized, autocrine and paracrine feedforward loops formed by cytokine–STAT3 signalling are recurrent themes in many human cancers9,10,28. Furthermore, the stable establishment of this feedforward loop probably represents an indirect epigenetic mechanism, as there is continuous deregulation of gene expression without gene mutations, which is not unlike changes in DNA methylation88. In this regard, it is notable that STAT3 can directly promote DNA methylation89.

STAT3 in inflammation-induced carcinogenesis

Although the IL-6–JAK–STAT3 pathway is an important mediator of cancer inflammation that is initiated by genetic changes in transformed cells (intrinsic pathway), as discussed above, it is also crucial for inflammatory conditions caused by environmental and other aetiological factors that are associated with increased cancer risk (extrinsic pathway). Several infectious agents that are known to cause inflammation-induced cancer involve STAT3 activation and probably depend on STAT3 for their oncogenic potential. For instance, infection with H. pylori, which is associated with gastric cancer, activates STAT3 through its cytotoxin-associated gene A in host cells17. Many tumour viruses are also known to activate STAT3 by various distinct mechanisms, including hepatitis B virus90, human papillomavirus91, human T-lymphotropic virus type 1 (REF. 21) and Epstein–Barr virus92. A human enterotoxigenic strain of Bacteroides fragilis (termed ETBF) has been shown to induce colitis in both mice and humans; ETBF colonization of mice results in STAT3 activation in the colon and dramatically increases colon tumorigenesis58. Both lipopolysaccharide (which mimics bacterial infection) and live bacteria are able to activate STAT3, resulting in the production of IL-1β and IL-6 (REF. 19), which are major mediators of inflammation-induced cancer. Lipopolysaccharide is known to function through Toll-like receptor 4 (TLR4), and it has been shown that the engagement of TLR4 and TLR9 can also directly activate STAT3 (REF. 93). These and other studies provide a potential mechanistic explanation for why TLRs, which are the sensors of infection, can promote tumour growth94.

STAT3-mediated inflammation may also be central to non-infectious pathways of carcinogenesis. Importantly, a crucial role of STAT3 signalling in mediating ultraviolet light-induced skin cancer has been demonstrated in a transgenic mouse model22; cigarette smoke-associated cancer development may also be linked to STAT3 activation through the nicotinic receptor14. Chronic stress is another contributer 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 adrenaline18. Chemical carcinogens also induce tumour growth and incidence through STAT3, as shown by the examples below.

Owing to its central role in mediating inflammatory signals, the NF-κB pathway has been a major focus of studies on inflammation-induced carcino-genesis. Conditionally ablating inhibitor of NF-κB (IκB) kinase-β (Ikkβ), which blocks NF-κB activation by preventing IκB phosphorylation and degradation (BOX 1), abrogated colorectal cancer resulting from inflammatory bowel disease and ulcerative colitis induced by chemical carcinogens, azoxymethane and dextran sulfate95. More recent studies in colitis-associated colorectal cancer mouse models have provided direct evidence that STAT3 signalling is required for inflammation-induced cancer3,7. Furthermore, IL-6, which is downstream of NF-κB and primarily produced by bone marrow-derived myeloid cells, activates STAT3 in both inflammatory cells and the epithelial cells from which the tumours arose. Activation of STAT3 induced the upregulation of key genes involved in cell proliferation and survival, and increased the nuclear localization of β-catenin, which contributes to colorectal carcinogenesis3. Importantly, in the absence of the Stat3 alleles, carcinogens continued to induce inflammation but not tumour incidence3. Increased IL-6 expression is also observed in human colorectal cancer, suggesting the importance of IL-6 in mediating inflammatory conditions that are associated with this cancer96. Furthermore, IL-6 seems to be a crucial mediator of gender bias in susceptibility to hepatocellular carcinoma. This disease mainly occurs in men, and gender bias linked to IL-6 has been shown in a carcinogen-induced liver cancer mouse model97. The administration of the carcinogen diethylnitrosamine led to a higher serum IL-6 concentration in male mice than it did in female mice. Moreover, without IL-6, the gender difference in tumour development was abrogated. Interestingly, in the liver tissues in which the tumours developed, it was not IKK but STAT3 that was persistently activated. This supports a crucial role for IL-6–STAT3 signalling in mediating the gender bias in liver cancer susceptibility97.

Box 1

NF-κB signalling and regulation of inflammatory genes

The nuclear factor-κB (NF-κB) family consists of five Rel proteins RELA, REL, RELB, p50 and p52. Among these proteins, only RELA, REL and RELB contain carboxy-terminal transactivation domains that are required for transcriptional activation. All of these proteins contain an amino-terminal Rel-homology domain crucial for dimerization, DNA binding and interaction with inhibitor of NF-κB (IκB) family inhibitors. NF-κB (both the homodimer and the heterodimer forms) is sequestered in the cytoplasm by IκB family proteins. After being phosphorylated by the IκB kinase IKK, IκB undergoes ubiqutin-dependent degradation and releases NF-κB, which then translocates to the nucleus. NF-κB dimers can also form complexes with IκB in the nucleus, and these are exported to the cytoplasm. The RELA–p50 heterodimer is the prototype NF-κB transcription factor. It is crucial for the expression of genes encoding pro-inflammatory mediators, such as interleukin-6 (IL-6), IL-1β, cyclooxygenase 2 and IL-23, which can also be cancer promoting. By contrast, the REL–p50 heterodimer mainly activates expression of T helper 1-immunostimulatory genes, including _IL_-12, CD40 and CD80.

Chronic inflammatory conditions that promote tumour formation can also be attributed to genetic alterations that directly affect the STAT3 pathway1013. The importance of constitutively active mutations in GP130, which encodes a subunit of the IL-6 receptor, has been demonstrated in human inflammatory hepatocellular adenomas12. Constitutively active mutant GP130 causes the persistent activation of JAKs and STAT3 in the absence of cytokine ligands. A crucial role for STAT3 in inflammation-induced adenocarcinomas was also demonstrated in a transgenic mouse model with a constitutively active GP130 in epithelial cells70. Studies in mice with Gp130 mutations demonstrated that an increase in GP130 and STAT3 signalling led to inflammation-associated gastric tumorigenesis13. Mutations at specific sites in JAK2 have also been observed in some human cancers, including myeloid leukaemias11. These human and mouse genetic studies show that the persistent activation of the IL6–GP130–JAK–STAT3 signalling axis is an important contributor to inflammation-induced cancers, making it an attractive target for treating and/or preventing inflammation that favours cancer progression.

STATs in anti-cancer or pro-cancer inflammation

One of the most debated topics in cancer biology is whether the immune system suppresses or promotes cancer. Many experimental data suggest a crucial role of the immune system in controlling tumour incidence and growth, and in certain human cancers or subsets of cancers the presence of immune cells predicts better prognosis4147. Evidence suggests that innate immune cells, such as macrophages, natural killer cells and dendritic cells, can destroy tumour cells when appropriately activated54 (FIG. 2). T cells from the adaptive arm of the immune system can also attack tumours when activated in a TH1 immunological milieu (FIG. 3). However, tumour-associated macrophages have long been known to promote cancer, partly through their ability to secrete angiogenic, metastatic and growth factors98,99 (FIG. 2). Another related myeloid lineage of cells, MDSCs, which are increased in cancer, not only suppresses anti-tumour immunity but also directly contributes to tumour growth49 (FIG. 2).

An external file that holds a picture, illustration, etc. Object name is nihms-779912-f0002.jpg

Different STAT proteins modulate pro-cancer and anti-tumour responses by myeloid cells

Myeloid cells, such as dendritic cells (DCs) and macrophages, can stimulate anti-tumour T helper 1 (TH1) adaptive immunity and even cause direct tumour cell death, which is associated with the production of TH1 cytokines, including interleukin-12 (IL-12) and interferon-γ (IFNγ). Activation of signal transducer and activator of transcription 1 (STAT1) (p-STAT1 denotes the activated phosphorylated form of STAT1) and STAT4 is important for anti-tumour TH1 responses. However, tumour-associated macrophages (TAMs) harbouring activated STAT3 (p-STAT3) no longer exhibit anti-tumour effects in the tumour microenvironment. Instead, along with myeloid-derived suppressor cells (MDSCs), TAMs promote cancer progression when STAT3 is activated. Moreover, STAT3 activity in the tumour microenvironment contributes to the expression of pro-cancer inflammatory mediators and angiogenic and growth factors, leading to increased tumour growth. Both activated STAT3 and STAT6 also promote MDSC expansion. Similar STAT3-dependent factors are produced by tumour cells and endothelial cells, forming a crosstalk network among tumour myeloid cells, tumour cells and tumour endothelial cells important for tumour angiogenesis and metastasis.

An external file that holds a picture, illustration, etc. Object name is nihms-779912-f0003.jpg

STAT proteins regulate cancer adaptive immunity

Signal transducer and activator of transcription 1 (STAT1) signalling in T cells promotes T helper 1 (TH1) differentiation and interleukin-12 receptor (IL-12R) expression. IL-12R signalling through STAT4 (p-STAT4 denotes the activated phosphorylated form of STAT4) further promotes TH1 expansion, which leads to interferon-γ (IFNγ) expression. In the TH1 milieu, adaptive immune responses control tumour growth. CD8+ T cells have a crucial role in cytotoxicity against tumour cells. Regulatory T (TReg) cells are important negative regulators of TH1 anti-tumour immunity. STAT5 is crucial for TReg cell differentiation and expansion. Both STAT5 and STAT3 contribute to the expression of forkhead box P3, a marker for TReg cells. At the same time, STAT3 activation in tumour TReg cells promotes the expression of IL-10 and transforming growth factor-β (TGFβ), which are the mediators of TReg cell suppressive functions, leading to the downregulation of TH1 anti-tumour immune responses. In the presence of increased IL-6, TGFβ and IL-23, all of which can be regulated by STAT3 in the tumour microenvironment, TH17 T cells expand and produce IL-17. This IL-17 activates STAT3 in diverse immune cells and other stromal cells in the tumour microenvironment through IL-6, which can further promote tumour growth.

The distinction between immune responses that inhibit and those that promote cancer is, to a large extent, regulated by members of the STAT family of proteins. In particular, STAT1 activation leads to IL-12 production; STAT4 activation induced by IL-12 receptor (IL-12R) engagement in turn promotes TH1 responses that produce IFNγ, the receptor of which itself signals through STAT1 to increase anti-tumour responses through the activation of natural killer cells, macrophages and CD8+ T cell-mediated cytolytic activity60,100 (FIG. 3). Ablating the Stat3 gene in natural killer cells and macrophages in mice can boost expression of TH1 mediators34,101, leading to STAT1 upregulation and increased anti-tumour immune responses28,50,85,86. Both type 1 interferons and IFNγ mediate anti-tumour immunity through STAT1-dependent innate effectors (such as natural killer cells), TH1-type responses and cytotoxic T lymphocyte activation45,47. The ability of STAT3 to antagonize STAT1 has been well documented50,53,68. STAT3 and STAT6 have also been implicated in promoting immunosuppressive MDSCs49,52. Recent evidence points to a role for STAT3 in mediating the cancer-promoting properties, such as angiogenesis, of tumour-associated MDSCs and macrophages4,52,98,102 (FIG. 2). STAT3 and STAT5 also have major roles in the expansion of T regulatory cells5,50,55, which promote tumour progression by inhibiting anti-tumour immune responses that are mediated by TH1-type CD4+ T cells and CD8+ T cells103 (FIG. 3).

New insights into STAT-dependent regulation of the balance between distinct immune responses have come from the recent discovery of a distinct TH17 T helper cell subset (FIG. 3). Although TH1 development involves type 1 interferons and is further promoted by IL-12, TH17 development requires a combination of IL-6 and transforming growth factor-β (TGFβ) and is further promoted by the IL-12 family member IL-23 (REFS 54,104). STAT3 activation is central to TH17 development56 and functions both as the major IL-6R-dependent transcription factor in the T cell and as a transcriptional activator of Il-23a (also known as p19) that encodes part of IL-23 (REF. 5). Recent evidence supports a cancer-promoting role for both IL-23 and TH17 responses (FIG. 3). In a cutaneous carcinogenesis model, _Il_-23a knockout resulted in decreased skin tumour formation, and _Il_-12a knockout resulted in increased tumour formation105. Antibody blockade of IL-23R in vivo also decreased the growth rates of some transplanted tumours5. Inducible myeloid-specific Stat3 gene ablation abrogated IL-23 production by tumour-infiltrating macrophages but induced the release of IL-12 by tumour-infiltrating dendritic cells (which was accompanied by a decrease in tumour growth)5. In addition, expression of Il-23r was increased in tumour-promoting regulatory T cells in tumours5. IL-23 also increases intra-tumoural TH17 activity, which can further promote tumour growth. For example, IL-17 (secreted by TH17 cells) positively affected tumour STAT3 activity and tumour growth in some transplanted models6. The importance of IL-17–STAT3 in tumorigenesis was further illustrated in the ETBF bacterial pro-carcinogenesis model, in which both _Il_-17 expression and STAT3 activity were increased. STAT3 activation is required for IL-17 production by the lamina propria CD4+ T cells, and IL-17 blockade abrogates most of the colon tumorigenesis induced by ETBF colonization58. However, IL-17 can both promote and inhibit tumorigenesis6,58,106,107; further studies are therefore required to understand its role in cancer.

STAT3–NF-κB in cancer inflammation

As discussed above, both STAT3 and NF-κB have crucial and integrated roles in inflammatory responses that promote cancer development and growth28,2325,27,28,31,32,50. However, NF-κB is involved not only in pro-carcinogenic inflammation but also in anti-tumour immune responses1,25,28,32. STAT3, conversely, is a specific regulator of pro-carcinogenic inflammation — driving cancer-promoting inflammatory conditions —while simultaneously suppressing anti-tumour immune responses28,50,85,86. Intriguingly, STAT3 and NF-κB are frequently persistently activated in the same tumour cells, and in tumour-associated myeloid cells. Both transcription factors induce the expression of a highly overlapping repertoire of proliferative, anti-apoptotic, angiogenic and metastatic genes that promote tumour development and growth (TABLE 2). However, this is where the similarity between the two proteins ends. STAT3 inhibits the expression of NF-κB target genes involved in TH1 innate immunity and adaptive immune responses important for controlling microbial infections and tumour growth28 (TABLE 2). In this way, STAT3 opposes the activation of anti-tumour immunity programmes by NF-κB when both are activated in the same cells. Therefore, STAT3 interacts with NF-κB at multiple levels and the consequence of this interaction is highly context dependent. Recent studies (which are described below) have shed light on the molecular basis of these interactions.

The prototypical NF-κB complex is a RELA–p50 heterodimer (BOX 1), which is crucial for both the induction of inflammatory responses and NF-κB-mediated pro-oncogenic effects108,109. Another NF-κB family protein, REL (BOX 1), regulates the transcription of many immunostimulatory cytokines and chemokines that are involved in anti-pathogen and anti-tumour immune responses. Unlike RELA, which is persistently activated in tumours, including tumour-associated immune cells, REL is not usually activated in cancer. Recent evidence suggests that accumulation of RELA in the nucleus can be promoted by acetylation by p300 (also known as EP300)110 (FIG. 4). Importantly, STAT3 is required for p300-mediated RELA acetylation; consequently, persistent activation of RELA in tumour cells, as well as in non-transformed cells in the tumour microenvironment, requires continuous STAT3 signalling32 (FIG. 4).

An external file that holds a picture, illustration, etc. Object name is nihms-779912-f0004.jpg

Multilevel interactions between STAT3 and NF-κB

In normal physiology, nuclear factor-κB (NF-κB) is sequestered in the cytoplasm by inhibitor of NF-κBα (IκBα). When IκB kinases (IKKs) are activated by various pathogens and/or proinflammatory cytokines, IκBα is phosphorylated and degraded, allowing NF-κB nuclear translocation (BOX 1). As a transcription factor, NF-κB (especially REL) upregulates many genes involved in stimulating T helper 1 (TH1) immune responses that are necessary to control pathogens and to mediate anti-tumour immune responses. Several RELA downstream target genes encode factors that in turn activate signal transducer and activator of transcription 3 (STAT3) (TABLE 2). STAT3, which is also activated by intrinsic and extrinsic pathways, has the ability to inhibit IKK and thereby reduce NF-κB-associated TH1 immunity. In addition, STAT3 contributes to the persistent activation of NF-κB during chronic inflammation and in transformed cells by prolonging nuclear retention of RELA. NF-κB–IκBα complexes can shuttle in and out of the nucleus in the absence of stimuli, although the rate of nuclear export is greater than that of nuclear import. STAT3 facilitates nuclear localization of RELA through p300 (also known as EP300)-mediated acetylation, which interferes with the NF-κB–IκBα interaction and prevents their nuclear export. When STAT3 activity is increased in tumours, NF-κB prefers the STAT3–p300 interaction. Although their cognate DNA-binding sites need not be immediately adjacent, NF-κB and STAT3 regulate the expression of numerous oncogenic and inflammatory mediators (TABLE 2). Many of these gene products in turn further activate STAT3. Therefore, STAT3 and NF-κB interact at multiple levels, thereby promoting cancer inflammation, restraining anti-tumour TH1 immune responses and increasing tumour cell proliferation and survival as well as tumour angiogenesis and metastasis. P, phosphorylation; Ub, ubiquitylation.

Just as STAT3 can increase NF-κB activity in cancer, persistent activation of STAT3 in tumours, especially in immune cells of the tumour microenvironment, is dependent on NF-κB (specifically RELA). This reciprocal relationship with RELA stems from the fact that several cytokines and growth factors encoded by RELA target genes are STAT3 activators (TABLE 2). Among the STAT3 activators downstream of NF-κB, the most crucial ones are IL-6 and related cytokines. As discussed earlier, the importance of IL-6 and other members of the IL-6 family of cytokines, including IL-11, in activating the JAK–STAT3 pathway leading to cancer-promoting inflammation has been widely documented. COX2, another factor downstream of NF-κB that mediates cancer-promoting inflammation, also activates STAT3 through IL-6 (REF. 87). Several other inflammatory cytokines, including IL-17, activate STAT3 through NF-κB–IL-6 signalling6,39. Additional examples of STAT3 activators that are regulated by NF-κB are IL-23 and IL-21 (REF. 57), both of which contribute to inflammation-induced cancer. A striking feature of STAT3–NF-κB interactions is that many of these inflammatory mediators, including IL-6, COX2, IL-17 and IL-23 (all of which activate STAT3), also require STAT3 as a co-transcription factor with RELA for their expression (FIG. 4; TABLE 2). Therefore, not only do these two transcription factors need each other for their persistent activation in cancer, they also ensure the continued expression of their activators, facilitating the stable establishment of this feedforward loop and a permanent change in the genetic programme (FIG. 4).

One perplexing question that remains to be fully resolved is how STAT3 inhibits NF-κB-mediated transcription of TH1 immunostimulatory genes in tumours and their microenvironment (TABLE 2). Although constitutive STAT3 activation in cancer maintains nuclear NF-κB levels32, genetic ablation of Stat3 revealed an inhibitory role of STAT3 on IKK, and so NF-κB activation, in normal immune cells34 and tumour cells32 (FIG. 4). IKK in turn activates various members of the NF-κB family of proteins, including RELA and REL (BOX 1; FIG. 4), and also RELB. Although RELA–p50 and REL–p50 both upregulate many TH1 immunostimulatory genes, RELA additionally increases the expression of numerous genes involved in chronic inflammation and cancer progression (TABLE 2). By contrast, REL is mainly involved in regulating TH1 immunostimulatory genes111. For example, IL-12A (also known as P35), which encodes a component of IL-12, is mostly regulated by REL. It is notable that STAT3 inhibits _IL_-12 expression50, at least in part, by downregulating REL5. Several RELA-regulated genes, such as _IL_-6, _IL_-8 and inducible nitric oxide synthase 2 (NOS2), can be either upregulated or downregulated by STAT3 (TABLE 2). The finding that STAT3 can both inhibit IKK during acute inflammation and cooperate with RELA as a co-transcription factor for these genes in chronic inflammation-associated cancer provides a potential mechanistic explanation for this apparent paradox (FIG. 4). Further studies are required to clarify why under cancer-promoting chronic inflammatory conditions persistently activated RELA and STAT3 regulate one set of genes, which are mostly oncogenic, while leaving many of the TH1 immunostimulatory genes inactive (FIG. 4).

Targeting STAT3 for cancer therapy

Oncogenic transcription factors are prime targets for cancer therapy, partly because multiple cytoplasmic signalling pathways converge on a limited number of nuclear transcription factors26. STAT3 and NF-κB (specifically RELA) are among the most frequently activated oncogenic transcription factors and, as discussed here, both regulate cancer-associated inflammation23,2528. However, owing to the crucial role of NF-κB in inducing anti-tumour immune responses, its inhibition could actually promote tumour growth, and long-term blockade might cause substantial immune suppression25. Genetic studies demonstrate that ablation of the Stat3 gene in either tumour cells or tumour-associated macrophages and other immune cells, even under chronic inflammatory conditions, inhibits carcinogenesis as well as the growth of established tumours3,20,50,112. Moreover, mice reconstituted with _Stat3_-deficient immune cells can mount potent anti-tumour immune responses50. Although prolonged Stat3 gene ablation in mice ultimately causes TH1-associated autoimmunity, a therapeutic window has been demonstrated50. Additionally, genetic studies in patients with hyper-IgE syndrome identified dominant-negative STAT3 gene mutations — effectively abolishing STAT3 signalling — as the cause of the disease in some of the patients113115. These studies in mice and humans suggest that although completely blocking STAT3 can lead to severe disease either directly or indirectly, partial STAT3 inhibition for a limited duration — as in drug-based cancer therapy — can potentially convert tumour-promoting inflammation to anti-tumour immune responses without generating severe side effects.

Direct targeting of STAT3 as a new approach for more effective cancer therapy is highly desirable26,27. Although the inhibition of tyrosine kinases could potentially activate alternative pathways leading to increased metastasis and tumour growth116,117, Stat3 gene ablation in diverse tumour models results in the inhibition of tumour growth20,50,112. However, unlike tyrosine kinases and other molecular targets, transcription factors do not possess intrinsic enzymatic activity. This makes the development of small-molecule STAT3 inhibitors considerably more challenging, requiring alternative approaches such as the disruption of protein–protein or protein–DNA interactions. Nevertheless, two major approaches have been taken along these lines: structure-based design and random screening of compound libraries. In terms of structure-based design, the primary molecular target has so far been the phosphotyrosine–SH2 domain interactions that stabilize active STAT3 dimers. Phosphopeptides and their derivative peptidomimetics with reduced peptide properties have been designed that block STAT3 dimerization and DNA-binding activity118120. In addition, screening of virtual compound libraries in silico against molecular models of STAT3 on the basis of its crystal structure has led to both peptide and non-peptide small molecules that disrupt STAT3 dimerization and transcriptional activity121. As a complementary approach, random screening of chemical libraries in vitro has yielded additional non-peptide small molecules with activity against STAT3 (REFS 122124). These small molecules inhibit STAT3-mediated gene regulation, block tumour cell proliferation and selectively induce the apoptosis of tumour cells with activated STAT3 (REFS 121124). In some cases, STAT3 inhibitors possess anti-tumour activity in mouse tumour models, including reducing cancer-promoting immune responses, while promoting anti-tumour immunity50. Although none of the STAT3 inhibitors developed to date is considered a clinical candidate, and further work is required to attain this goal, these studies have established proof-of-concept that direct targeting of STAT3 is feasible.

Oligonucleotides, including antisense RNA, small interfering RNA and decoy DNA-binding sites, have also been developed to inhibit STAT3 (REFS 112,125127). A recent study also demonstrated the feasibility of targeting STAT3, using a conjugate of small interfering RNA linked to a Toll-like receptor ligand, in myeloid, dendritic and B cells through both local and systemic delivery. Knocking down STAT3 in the immune cells by this approach led to anti-tumour effects128. Although the clinical applicability of oligonucleotide-based approaches remains to be tested, several studies in animals suggested their potential use to target STAT3 for cancer therapy128,129. Another class of STAT3 inhibitors includes natural products and their derivatives with anti-tumour activities, such as cucurbitacin, resveratrol, galiellalactone, curcumin and indirubin130132. The molecular mechanisms of action of these natural product inhibitors, which probably inhibit other oncogenic signalling pathways in addition to STAT3, remain to be fully determined. It is notable that some of these natural products inhibit JAK activity either directly or indirectly. Owing to the importance of IL-6–JAK signalling in inducing STAT3 activation, JAK family kinases (particularly JAK1 and JAK2) are crucial targets for inflammation-induced cancer and cancer-associated inflammation133,134. Some of the JAK family kinase inhibitors that block STAT3 signalling can convert cancer-promoting inflammation to anti-tumour immunity, therefore providing proof-of-concept for this approach135,136. Recently, new classes of tyrosine kinase inhibitors that target JAK2 have been identified for clinical development137.

Several US Food and Drug Administration-approved tyrosine kinase inhibitors already in the clinic, including sorafenib and sunitinib, can inhibit STAT3 signalling indirectly, resulting in tumour cell cycle arrest and apoptosis138,139. Sunitinib has further been shown to inhibit STAT3 activity in tumour-associated immune cells, modulating the tumour immunological microenvironment in favour of cancer therapy139,140. It inhibits immunosuppressive MDSCs and T regulatory cells in both mouse models and human clinical trials139141. Although the inhibition of STAT3 signalling contributes to the anti-tumour activities of sorafenib and sunitinib, their precise molecular mechanisms of action in terms of STAT3 inhibition remain to be determined.

On the basis of the foregoing discussion, tyrosine kinase inhibitors that modulate the tumour immuno-logical environment by inhibiting STAT3 activity, combined with immunotherapeutic approaches, may lead to increased anti-tumour immune responses. In sum, targeting STAT3 — a central regulatory node on which many oncogenic and inflammatory pathways converge — holds as yet untapped promise for future cancer therapy. In addition to the inhibition of tumour cell proliferation and survival, such STAT3 inhibitors would be predicted to convert inflammation in the tumour microenvironment from tumour promoting to tumour suppressing.

Conclusions

From the earliest pathology studies dating back to the 1800s, cancer has been known to be associated with inflammation. Indeed, many human cancers arise and progress in the setting of chronic inflammatory states. Conversely, there are numerous examples in which induced inflammation and immunity can inhibit tumour growth or even eliminate established tumours. It has become clear that distinct types of inflammation can either promote or inhibit cancer induction and growth. STATs represent central regulators of cancer-associated inflammation and influence interactions between tumour cells and their immune microenvironment that determine whether the inflammation promotes or inhibits cancer. This regulation is accomplished largely through the induction or suppression of specific cytokines and growth factors, as well as through signalling by their cognate receptors. Together with NF-κB, STAT3 is a major mediator of pro-carcinogenic inflammation, and it also opposes NF-κB and STAT1-dependent immune responses that promote anti-tumour immunity. Because of its pivotal role in determining the nature of cancer-associated inflammation, STAT3 represents a promising therapeutic target for converting cancer-promoting inflammation to anti-tumour immunity.

At a glance

Acknowledgements

We would like to thank members of our laboratories who have made major discoveries in the topics discussed here. We also thank the many laboratories that have contributed important publications relevant to the topics in this Review but which could not be cited owing to space limitations. Special thanks go to L. Wang and J. Deng for helping to prepare the figures and references, R. Buettner for compiling data on STAT3 inhibitors, and M. Kortylewski for critical reading of the manuscript.

Glossary

TH1 A TH1 response is mediated by CD4+ T cells and promoted by type 1 interferons and IL-12, this response mediates its effect through cytokines, particularly IFNγ, promoting cellular immune responses against intracellular infections and tumours.
Innate immune cells Innate immune cells include natural killer cells, macrophages, neutrophils and dendritic cells, which provide immediate non-specific defence against pathogens. These cells identify and destroy virus-, bacteria- and fungus-infected cells and malignant cells.
Myeloid-derived suppressor cells MDSCs. Heterogenous and plastic cells. When isolated from normal bone marrow, they do not exhibit imunosuppresive effects. However, when exposed to the tumour microenvironment, they inhibit both CD4+ and CD8+ T cells.
TH17 T cells Defined by the secretion of IL-17A but not IFNγ or IL-4. TH17 cells have been implicated in protective immunity to intestinal and pulmonary bacterial infections, as well as pathological immunity in several autoimmune diseases.
Type 1 IFNs Include multiple family members of the IFNα group and the related IFNβ. All type 1 IFNs bind to a single receptor, termed the type 1 or IFNα receptor. They mediate direct anti-viral activity against infected cells, and can also inhibit tumour growth and promote anti-tumour immune responses.
Natural killer cell A type of lymphocyte that protects against infectious microbes and kills tumour cells through the recognition of specific cell membrane molecules that are upregulated under conditions of cell stress, such as infection or carcinogenic transformation.
Adaptive immunity Mediated by antigen-specific T lymphocytes and antibodies produced by B cells. It takes longer to develop than innate immunity but has greater antigen specificity and includes the development of immunological memory.
Dendritic cells Specialized and the most efficient antigen-presenting cells, which can activate T cells and thereby induce antigen-specific immune responses.
CD8+ T cell The T cell subset from which cytolytic T cells develop. These in turn directly recognize target cells based on the surface expression of antigenic peptide complexed with MHC I molecules and kill their targets by injecting granzymes that induce apoptosis.
Lamina propria A constituent of the moist linings of mucous membranes, which line different tubes of the body, including the gastrointestinal tract.
Hyper-IgE syndrome A rare immune and connective tissue disorder characterized by dermatitis, cyst-forming pneumonia and increased serum levels of immunoglobulin E antibody. In some patients it is caused by autosomal dominant STAT3 mutations.
Decoy DNA-binding sites DNA oligonucleotides with sequence-specific binding sites that sequester their cognate binding proteins, thereby preventing them from binding to the regulatory sequences of nuclear genes.

References

1. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436–444. [PubMed] [Google Scholar]

2. Catlett-Falcone R, et al. Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity. 1999;10:105–115. [PubMed] [Google Scholar] This report was the first to document that IL-6–JAK–STAT3 signalling, which increases inflammation-induced cancer, is important for the growth and survival of human tumour cells.

3. Bollrath J, et al. gp130-mediated Stat3 activation in enterocytes regulates cell survival and cell-cycle progression during colitis-associated tumorigenesis. Cancer Cell. 2009;15:91–102. [PubMed] [Google Scholar]

4. Kujawski M, et al. Stat3 mediates myeloid cell-dependent tumor angiogenesis in mice. J. Clin. Invest. 2008;118:3367–3377. [PMC free article] [PubMed] [Google Scholar] This study demonstrated that STAT3 is a crucial mediator for myeloid cell-induced tumour angiogenesis.

5. Kortylewski M, et al. Regulation of the IL-23 and IL-12 balance by Stat3 signaling in the tumor microenvironment. Cancer Cell. 2009;15:114–123. [PMC free article] [PubMed] [Google Scholar]

6. Wang L, Yi T, Kortylewski M, Pardoll D, Zeng D, Yu H. IL-17 can promote tumor growth through an IL-6/Stat3 signaling pathway. J. Exp. Med. 2009;206:1457–1464. [PMC free article] [PubMed] [Google Scholar]

7. Grivennikov S, et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell. 2009;15:103–113. [PMC free article] [PubMed] [Google Scholar] Studies reported in references 3 and 7 identified STAT3 as a central node for inflammation-induced cancer, suggesting that it is a crucial target for treating and preventing cancer-promoting inflammation.

8. Shain KH, et al. β1 integrin adhesion enhances IL-6-mediated STAT3 signaling in myeloma cells: implications for microenvironment influence on tumor survival and proliferation. Cancer Res. 2009;69:1009–1015. [PMC free article] [PubMed] [Google Scholar]

9. Ara T, et al. Interleukin-6 in the bone marrow microenvironment promotes the growth and survival of neuroblastoma cells. Cancer Res. 2009;69:329–337. [PMC free article] [PubMed] [Google Scholar]

10. Gao SP, et al. Mutations in the EGFR kinase domain mediate STAT3 activation via IL-6 production in human lung adenocarcinomas. J. Clin. Invest. 2007;117:3846–3856. [PMC free article] [PubMed] [Google Scholar]

11. Olcaydu D, et al. A common JAK2 haplotype confers susceptibility to myeloproliferative neoplasms. Nature Genet. 2009;41:450–454. [PubMed] [Google Scholar]

12. Rebouissou S, et al. Frequent in-frame somatic deletions activate gp130 in inflammatory hepatocellular tumours. Nature. 2009;457:200–204. [PMC free article] [PubMed] [Google Scholar] This study provides excellent examples of genetic mutations activating the IL-6–GP130–JAK–STAT3 pathway leading to inflammation-associated human cancer.

13. Ernst M, et al. STAT3 and STAT1 mediate IL-11-dependent and inflammation-associated gastric tumorigenesis in gp130 receptor mutant mice. J. Clin. Invest. 2008;118:1727–1738. [PMC free article] [PubMed] [Google Scholar]

14. Arredondo J, Chernyavsky AI, Jolkovsky DL, Pinkerton KE, Grando SA. Receptor-mediated tobacco toxicity: cooperation of the Ras/Raf-1/MEK1/ERK and JAK-2/STAT-3 pathways downstream of alpha7 nicotinic receptor in oral keratinocytes. FASEB J. 2006;20:2093–2101. [PubMed] [Google Scholar]

15. Gu M, Singh RP, Dhanalakshmi S, Agarwal C, Agarwal R. Silibinin inhibits inflammatory and angiogenic attributes in photocarcinogenesis in SKH-1 hairless mice. Cancer Res. 2007;67:3483–3491. [PubMed] [Google Scholar]

16. Aziz MH, Manoharan HT, Verma AK. Protein kinase C epsilon, which sensitizes skin to sun’s UV radiation-induced cutaneous damage and development of squamous cell carcinomas, associates with Stat3. Cancer Res. 2007;67:1385–1394. [PubMed] [Google Scholar]

17. Bronte-Tinkew DM, et al. Helicobacter pylori cytotoxin-associated gene A activates the signal transducer and activator of transcription 3 pathway in vitro and in vivo. Cancer Res. 2009;69:632–639. [PMC free article] [PubMed] [Google Scholar]

18. Landen CN, Jr, et al. Neuroendocrine modulation of signal transducer and activator of transcription-3 in ovarian cancer. Cancer Res. 2007;67:10389–10396. [PubMed] [Google Scholar]

19. Samavati L, et al. STAT3 tyrosine phosphorylation is critical for interleukin 1β and interleukin-6 production in response to lipopolysaccharide and live bacteria. Mol. Immunol. 2009;46:1867–1877. [PubMed] [Google Scholar]

20. Chan KS, et al. Disruption of Stat3 reveals a critical role in both the initiation and the promotion stages of epithelial carcinogenesis. J. Clin. Invest. 2004;114:720–728. [PMC free article] [PubMed] [Google Scholar]

21. Migone TS, et al. Constitutively activated Jak-STAT pathway in T cells transformed with HTLV-I. Science. 1995;269:79–81. [PubMed] [Google Scholar]

22. Sano S, et al. Signal transducer and activator of transcription 3 is a key regulator of keratinocyte survival and proliferation following UV irradiation. Cancer Res. 2005;65:5720–5729. [PubMed] [Google Scholar]

23. Karin M, Greten FR. NF-κB: linking inflammation and immunity to cancer development and progression. Nature Rev. Immunol. 2005;5:749–759. [PubMed] [Google Scholar]

24. Pikarsky E, et al. NF-κB functions as a tumour promoter in inflammation-associated cancer. Nature. 2004;431:461–466. [PubMed] [Google Scholar]

25. Baud V, Karin M. Is NF-κB a good target for cancer therapy? Hopes and pitfalls. Nature Rev. Drug Discov. 2009;8:33–40. [PMC free article] [PubMed] [Google Scholar]

26. Darnell JE., Jr. Transcription factors as targets for cancer therapy. Nature Rev. Cancer. 2002;2:740–749. [PubMed] [Google Scholar]

27. Yu H, Jove R. The STATs of cancer — new molecular targets come of age. Nature Rev. Cancer. 2004;4:97–105. [PubMed] [Google Scholar]

28. Yu H, Kortylewski M, Pardoll D. Crosstalk between cancer and immune cells: role of STAT3 in the tumour microenvironment. Nature Rev. Immunol. 2007;7:41–51. [PubMed] [Google Scholar]

29. Zhong Z, Wen Z, Darnell JE., Jr. Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science. 1994;264:95–98. [PubMed] [Google Scholar]

30. Darnell JE, Jr, Kerr IM, Stark GR. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science. 1994;264:1415–1421. [PubMed] [Google Scholar]

31. Bromberg JF, et al. Stat3 as an oncogene. Cell. 1999;98:295–303. [PubMed] [Google Scholar] This report was the first to demonstrate that Stat3 has intrinsic oncogenic potential.

32. Lee H, et al. Persistently activated Stat3 maintains constitutive NF-κB activity in tumors. Cancer Cell. 2009;15:283–293. [PMC free article] [PubMed] [Google Scholar] This study revealed how STAT3 directly interacts with RELA and facilitates its acetylation by p300, leading to prolonged NF-κB nuclear retention in both tumour cells and tumour-associated immune cells.

33. Lu T, Stark GR. Cytokine overexpression and constitutive NFκB in cancer. Cell Cycle. 2004;3:1114–1117. [PubMed] [Google Scholar]

34. Welte T, et al. STAT3 deletion during hematopoiesis causes Crohn’s disease-like pathogenesis and lethality: a critical role of STAT3 in innate immunity. Proc. Natl Acad. Sci. USA. 2003;100:1879–1884. [PMC free article] [PubMed] [Google Scholar]

35. Yang J, et al. Unphosphorylated STAT3 accumulates in response to IL-6 and activates transcription by binding to NFκB. Genes Dev. 2007;21:1396–1408. [PMC free article] [PubMed] [Google Scholar]

36. Basseres DS, Baldwin AS. Nuclear factor-κB and inhibitor of κB kinase pathways in oncogenic initiation and progression. Oncogene. 2006;25:6817–6830. [PubMed] [Google Scholar]

37. Haura EB, Turkson J, Jove R. Mechanisms of disease: insights into the emerging role of signal transducers and activators of transcription in cancer. Nature. Clin. Pract. Oncol. 2005;2:315–324. [PubMed] [Google Scholar]

38. Heinrich PC, Behrmann I, Muller-Newen G, Schaper F, Graeve L. Interleukin-6-type cytokine signalling through the gp130/Jak/STAT pathway. Biochem. J. 1998;334(Pt 2):297–314. [PMC free article] [PubMed] [Google Scholar]

39. Ogura H, et al. Interleukin-17 promotes autoimmunity by triggering a positive-feedback loop via interleukin-6 induction. Immunity. 2008;29:628–636. [PubMed] [Google Scholar]

40. Akira S, et al. Molecular cloning of APRF, a novel IFN-stimulated gene factor 3 p91-related transcription factor involved in the gp130-mediated signaling pathway. Cell. 1994;77:63–71. [PubMed] [Google Scholar]

41. Dunn GP, et al. A critical function for type I interferons in cancer immunoediting. Nature Immunol. 2005;6:722–729. [PubMed] [Google Scholar]

42. Blattman JN, Greenberg PD. Cancer immunotherapy: a treatment for the masses. Science. 2004;305:200–205. [PubMed] [Google Scholar]

43. Pardoll DM. Spinning molecular immunology into successful immunotherapy. Nature Rev. Immunol. 2002;2:227–238. [PubMed] [Google Scholar]

44. Kaplan DH, et al. Demonstration of an interferon γ-dependent tumor surveillance system in immunocompetent mice. Proc. Natl Acad. Sci. USA. 1998;95:7556–7561. [PMC free article] [PubMed] [Google Scholar]

45. Koebel CM, et al. Adaptive immunity maintains occult cancer in an equilibrium state. Nature. 2007;450:903–907. [PubMed] [Google Scholar] This paper demonstrates the important role of adaptive immunity in controlling tumour outgrowth.

46. Galon J, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313:1960–1964. [PubMed] [Google Scholar]

47. Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nature Rev. Immunol. 2006;6:836–848. [PubMed] [Google Scholar]

48. Sasaki K, et al. Stat6 signaling suppresses VLA-4 expression by CD8+ T cells and limits their ability to infiltrate tumor lesions in vivo. J. Immunol. 2008;181:104–108. [PMC free article] [PubMed] [Google Scholar]

49. Ostrand-Rosenberg S, Sinha P. Myeloid-derived suppressor cells: linking inflammation and cancer. J. Immunol. 2009;182:4499–4506. [PMC free article] [PubMed] [Google Scholar]

50. Kortylewski M, et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nature Med. 2005;11:1314–1321. [PubMed] [Google Scholar] This was the first report to show that STAT3 is persistently activated in tumour-associated immune cells, leading to the suppression of anti-tumour innate and adaptive immune responses.

51. Cheng F, et al. A critical role for Stat3 signaling in immune tolerance. Immunity. 2003;19:425–436. [PubMed] [Google Scholar]

52. Cheng P, 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. [PMC free article] [PubMed] [Google Scholar]

53. Ho HH, Ivashkiv LB. Role of STAT3 in type I interferon responses. Negative regulation of STAT1-dependent inflammatory gene activation. J. Biol. Chem. 2006;281:14111–14118. [PubMed] [Google Scholar]

54. Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nature Rev. Immunol. 2003;3:133–146. [PubMed] [Google Scholar]

55. Matsumura Y, et al. Selective expansion of foxp3-positive regulatory T cells and immunosuppression by suppressors of cytokine signaling 3-deficient dendritic cells. J. Immunol. 2007;179:2170–2179. [PubMed] [Google Scholar]

56. Chen Z, et al. Selective regulatory function of Socs3 in the formation of IL-17-secreting T cells. Proc. Natl Acad. Sci. USA. 2006;103:8137–8142. [PMC free article] [PubMed] [Google Scholar]

57. Chen Z, O’Shea JJ. Th17 cells: a new fate for differentiating helper T cells. Immunol. Res. 2008;41:87–102. [PubMed] [Google Scholar]

58. Wu S, et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nature Med. 2009;15:1016–1022. [PMC free article] [PubMed] [Google Scholar]

59. Schindler C, Levy DE, Decker T. JAK-STAT signaling: from interferons to cytokines. J. Biol. Chem. 2007;282:20059–20063. [PubMed] [Google Scholar]

60. Takeda K, Akira S. STAT family of transcription factors in cytokine-mediated biological responses. Cytokine Growth Factor Rev. 2000;11:199–207. [PubMed] [Google Scholar]

61. Stark GR, Kerr IM, Williams BR, Silverman RH, Schreiber RD. How cells respond to interferons. Annu. Rev. Biochem. 1998;67:227–264. [PubMed] [Google Scholar]

62. Darnell JE., Jr. Studies of IFN-induced transcriptional activation uncover the Jak-Stat pathway. J. Interferon Cytokine Res. 1998;18:549–554. [PubMed] [Google Scholar]

63. Taga T, Kishimoto T. Gp130 and the interleukin-6 family of cytokines. Annu. Rev. Immunol. 1997;15:797–819. [PubMed] [Google Scholar]

64. 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;19:2548–2556. [PubMed] [Google Scholar]

65. Levy DE, Darnell JE., Jr. Stats: transcriptional control and biological impact. Nature Rev. Mol. Cell Biol. 2002;3:651–662. [PubMed] [Google Scholar]

66. Heinrich PC, et al. Principles of interleukin (IL)-6-type cytokine signalling and its regulation. Biochem. J. 2003;374:1–20. [PMC free article] [PubMed] [Google Scholar]

67. Chin YE, et al. Cell growth arrest and induction of cyclin-dependent kinase inhibitor p21 WAF1/CIP1 mediated by STAT1. Science. 1996;272:719–722. [PubMed] [Google Scholar]

68. Shen Y, Devgan G, Darnell JE, Jr, Bromberg JF. Constitutively activated Stat3 protects fibroblasts from serum withdrawal and UV-induced apoptosis and antagonizes the proapoptotic effects of activated Stat1. Proc. Natl Acad. Sci. USA. 2001;98:1543–1548. [PMC free article] [PubMed] [Google Scholar]

69. Leung S, Qureshi SA, Kerr IM, Darnell JE, Jr, Stark GR. Role of STAT2 in the alpha interferon signaling pathway. Mol. Cell. Biol. 1995;15:1312–1317. [PMC free article] [PubMed] [Google Scholar]

70. Li Y, et al. Activation of the signal transducers and activators of the transcription 3 pathway in alveolar epithelial cells induces inflammation and adenocarcinomas in mouse lung. Cancer Res. 2007;67:8494–8503. [PubMed] [Google Scholar]

71. Judd LM, et al. STAT3 activation regulates growth, inflammation, and vascularization in a mouse model of gastric tumorigenesis. Gastroenterology. 2006;131:1073–1085. [PubMed] [Google Scholar]

72. Ogata H, et al. Deletion of the SOCS3 gene in liver parenchymal cells promotes hepatitis-induced hepatocarcinogenesis. Gastroenterology. 2006;131:179–193. [PubMed] [Google Scholar]

73. Dauer DJ, et al. Stat3 regulates genes common to both wound healing and cancer. Oncogene. 2005;24:3397–3408. [PubMed] [Google Scholar]

74. Grandis JR, et al. Requirement of Stat3 but not Stat1 activation for epidermal growth factor receptor-mediated cell growth in vitro. J. Clin. Invest. 1998;102:1385–1392. [PMC free article] [PubMed] [Google Scholar]

75. Trevino JG, et al. Src activation of Stat3 is an independent requirement from NF-κB activation for constitutive IL-8 expression in human pancreatic adenocarcinoma cells. Angiogenesis. 2006;9:101–110. [PubMed] [Google Scholar]

76. Bowman T, et al. Stat3-mediated Myc expression is required for Src transformation and PDGF-induced mitogenesis. Proc. Natl Acad. Sci. USA. 2001;98:7319–7324. [PMC free article] [PubMed] [Google Scholar]

77. Yu CL, et al. Enhanced DNA-binding activity of a Stat3-related protein in cells transformed by the Src oncoprotein. Science. 1995;269:81–83. [PubMed] [Google Scholar] This was the first report of persistent activation of STAT3 signalling by a specific oncoprotein in transformed cells.

78. Lo HW, et al. Nuclear interaction of EGFR and STAT3 in the activation of the iNOS/NO pathway. Cancer Cell. 2005;7:575–589. [PubMed] [Google Scholar]

79. Lin TS, Mahajan S, Frank DA. STAT signaling in the pathogenesis and treatment of leukemias. Oncogene. 2000;19:2496–2504. [PubMed] [Google Scholar]

80. Battle TE, Frank DA. The role of STATs in apoptosis. Curr. Mol. Med. 2002;2:381–392. [PubMed] [Google Scholar]

81. Bruns HA, Kaplan MH. The role of constitutively active Stat6 in leukemia and lymphoma. Crit. Rev. Oncol. Hematol. 2006;57:245–253. [PubMed] [Google Scholar]

82. Skinnider BF, Kapp U, Mak TW. The role of interleukin 13 in classical Hodgkin lymphoma. Leuk. Lymphoma. 2002;43:1203–1210. [PubMed] [Google Scholar]

83. Huang M, et al. Inhibition of Bcr-Abl kinase activity by PD180970 blocks constitutive activation of Stat5 and growth of CML cells. Oncogene. 2002;21:8804–8816. [PubMed] [Google Scholar]

84. Xie TX, et al. Activation of stat3 in human melanoma promotes brain metastasis. Cancer Res. 2006;66:3188–3196. [PubMed] [Google Scholar]

85. Sumimoto H, Imabayashi F, Iwata T, Kawakami Y. The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J. Exp. Med. 2006;203:1651–1656. [PMC free article] [PubMed] [Google Scholar]

86. Wang T, et al. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nature Med. 2004;10:48–54. [PubMed] [Google Scholar]

87. Dalwadi H, et al. Cyclooxygenase-2-dependent activation of signal transducer and activator of transcription 3 by interleukin-6 in non-small cell lung cancer. Clin. Cancer Res. 2005;11:7674–7682. [PubMed] [Google Scholar]

88. Russo V, Martienssen RA, Riggs AD. Epigenetic Mechanisms of Gene Regulation. Cold Spring Harbor Laboratory Press; USA: 1996. p. 662. [Google Scholar]

89. Zhang Q, Wang HY, Liu X, Wasik MA. STAT5A is epigenetically silenced by the tyrosine kinase NPM1-ALK and acts as a tumor suppressor by reciprocally inhibiting NPM1-ALK expression. Nature Med. 2007;13:1341–1348. [PubMed] [Google Scholar]

90. Choudhari SR, et al. Deactivation of Akt and STAT3 signaling promotes apoptosis, inhibits proliferation, and enhances the sensitivity of hepatocellular carcinoma cells to an anticancer agent, Atiprimod. Mol. Cancer Ther. 2007;6:112–121. [PubMed] [Google Scholar]

91. Sun S, Steinberg BM. PTEN is a negative regulator of STAT3 activation in human papillomavirus-infected cells. J. Gen. Virol. 2002;83:1651–1658. [PubMed] [Google Scholar]

92. Muromoto R, et al. Epstein-Barr virus-derived EBNA2 regulates STAT3 activation. Biochem. Biophys. Res. Commun. 2009;378:439–443. [PubMed] [Google Scholar]

93. Kortylewski M, et al. Toll-like receptor 9 activation of signal transducer and activator of transcription 3 constrains its agonist-based immunotherapy. Cancer Res. 2009;69:2497–2505. [PMC free article] [PubMed] [Google Scholar]

94. Chiron D, Bekeredjian-Ding I, Pellat-Deceunynck C, Bataille R, Jego G. Toll-like receptors: lessons to learn from normal and malignant human B cells. Blood. 2008;112:2205–2213. [PMC free article] [PubMed] [Google Scholar]

95. Greten FR, et al. IKKβ links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell. 2004;118:285–296. [PubMed] [Google Scholar]

96. Groblewska M, et al. Serum interleukin 6 (IL-6) and C-reactive protein (CRP) levels in colorectal adenoma and cancer patients. Clin. Chem. Lab. Med. 2008;46:1423–1428. [PubMed] [Google Scholar]

97. Naugler WE, et al. Gender disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science. 2007;317:121–124. [PubMed] [Google Scholar]

98. Matsukawa A, et al. Stat3 in resident macrophages as a repressor protein of inflammatory response. J. Immunol. 2005;175:3354–3359. [PubMed] [Google Scholar]

99. Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nature Rev. Cancer. 2004;4:71–78. [PubMed] [Google Scholar]

100. Chang HC, et al. Impaired development of human Th1 cells in patients with deficient expression of STAT4. Blood. 2009;113:5887–5890. [PMC free article] [PubMed] [Google Scholar]

101. Takeda K, et al. Enhanced Th1 activity and development of chronic enterocolitis in mice devoid of Stat3 in macrophages and neutrophils. Immunity. 1999;10:39–49. [PubMed] [Google Scholar]

102. Kubo M, Hanada T, Yoshimura A. Suppressors of cytokine signaling and immunity. Nature Immunol. 2003;4:1169–1176. [PubMed] [Google Scholar]

103. Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nature Rev. Immunol. 2006;6:295–307. [PubMed] [Google Scholar]

104. Gerosa F, et al. Differential regulation of interleukin 12 and interleukin 23 production in human dendritic cells. J. Exp. Med. 2008;205:1447–1461. [PMC free article] [PubMed] [Google Scholar]

105. Langowski JL, et al. IL-23 promotes tumour incidence and growth. Nature. 2006;442:461–465. [PubMed] [Google Scholar]

106. Numasaki M, et al. Interleukin-17 promotes angiogenesis and tumor growth. Blood. 2003;101:2620–2627. [PubMed] [Google Scholar]

107. Kryczek I, Wei S, Szeliga W, Vatan L, Zou W. Endogenous IL-17 contributes to reduced tumor growth and metastasis. Blood. 2009;114:357–359. [PMC free article] [PubMed] [Google Scholar]

108. Ghosh S, May MJ, Kopp EB. NF-κB and Rel proteins: evolutionarily conserved mediators of immune responses. Annu. Rev. Immunol. 1998;16:225–260. [PubMed] [Google Scholar]

109. Karin M, Cao Y, Greten FR, Li ZW. NF-κB in cancer: from innocent bystander to major culprit. Nature Rev. Cancer. 2002;2:301–310. [PubMed] [Google Scholar]

110. Chen LF, Greene WC. Shaping the nuclear action of NF-κB. Nature Rev. Mol. Cell Biol. 2004;5:392–401. [PubMed] [Google Scholar]

111. Wang J, et al. Distinct roles of different NF-κB subunits in regulating inflammatory and T cell stimulatory gene expression in dendritic cells. J. Immunol. 2007;178:6777–6788. [PubMed] [Google Scholar]

112. Chiarle R, et al. Stat3 is required for ALK-mediated lymphomagenesis and provides a possible therapeutic target. Nature Med. 2005;11:623–629. [PubMed] [Google Scholar]

113. Milner JD, et al. Impaired TH17 cell differentiation in subjects with autosomal dominant hyper-IgE syndrome. Nature. 2008;452:773–776. [PMC free article] [PubMed] [Google Scholar] This paper demonstrates that STAT3 dominant-negative mutation in individuals with hyper-IgE syndrome is associated with impaired TH17 cell differentiation.

114. Minegishi Y, et al. Dominant-negative mutations in the DNA-binding domain of STAT3 cause hyper-IgE syndrome. Nature. 2007;448:1058–1062. [PubMed] [Google Scholar]

115. Al Khatib S, et al. Defects along the TH17 differentiation pathway underlie genetically distinct forms of the hyper IgE syndrome. J. Allergy Clin. Immunol. 2009;124:342–348. [PMC free article] [PubMed] [Google Scholar]

116. Sen B, Saigal B, Parikh N, Gallick G, Johnson FM. Sustained Src inhibition results in signal transducer and activator of transcription 3 (STAT3) activation and cancer cell survival via altered Janus-activated kinase-STAT3 binding. Cancer Res. 2009;69:1958–1965. [PMC free article] [PubMed] [Google Scholar]

117. Paez-Ribes M, et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. 2009;15:220–231. [PMC free article] [PubMed] [Google Scholar]

118. Turkson J, et al. Novel peptidomimetic inhibitors of signal transducer and activator of transcription 3 dimerization and biological activity. Mol. Cancer Ther. 2004;3:261–269. [PubMed] [Google Scholar]

119. Mandal PK, et al. Conformationally constrained peptidomimetic inhibitors of signal transducer and activator of transcription. 3: Evaluation and molecular modeling. J. Med. Chem. 2009;52:2429–2442. [PMC free article] [PubMed] [Google Scholar]

120. Turkson J, et al. Phosphotyrosyl peptides block Stat3-mediated DNA binding activity, gene regulation, and cell transformation. J. Biol. Chem. 2001;276:45443–45455. [PubMed] [Google Scholar]

121. Siddiquee K, et al. Selective chemical probe inhibitor of Stat3, identified through structure-based virtual screening, induces antitumor activity. Proc. Natl Acad. Sci. USA. 2007;104:7391–7396. [PMC free article] [PubMed] [Google Scholar]

122. Schust J, Sperl B, Hollis A, Mayer TU, Berg T. Stattic: a small-molecule inhibitor of STAT3 activation and dimerization. Chem. Biol. 2006;13:1235–1242. [PubMed] [Google Scholar]

123. Turkson J, et al. Inhibition of constitutive signal transducer and activator of transcription 3 activation by novel platinum complexes with potent antitumor activity. Mol. Cancer Ther. 2004;3:1533–1542. [PubMed] [Google Scholar]

124. Turkson J, et al. A novel platinum compound inhibits constitutive Stat3 signaling and induces cell cycle arrest and apoptosis of malignant cells. J. Biol. Chem. 2005;280:32979–32988. [PubMed] [Google Scholar]

125. Ling X, Arlinghaus RB. Knockdown of STAT3 expression by RNA interference inhibits the induction of breast tumors in immunocompetent mice. Cancer Res. 2005;65:2532–2536. [PubMed] [Google Scholar]

126. Leong PL, et al. Targeted inhibition of Stat3 with a decoy oligonucleotide abrogates head and neck cancer cell growth. Proc. Natl Acad. Sci. USA. 2003;100:4138–4143. [PMC free article] [PubMed] [Google Scholar]

127. Barton BE, Karras JG, Murphy TF, Barton A, Huang HF. Signal transducer and activator of transcription 3 (STAT3) activation in prostate cancer: Direct STAT3 inhibition induces apoptosis in prostate cancer lines. Mol. Cancer Ther. 2004;3:11–20. [PubMed] [Google Scholar]

128. Kortylewski M, et al. In vivo delivery of siRNA to immune cells by conjugation to a TLR9 agonist enhances antitumor immune responses. Nature Biotechnol. 2009;27:925–932. [PMC free article] [PubMed] [Google Scholar]

129. Sen M, et al. Lack of toxicity of a STAT3 decoy oligonucleotide. Cancer Chemother. Pharmacol. 2009;63:983–995. [PMC free article] [PubMed] [Google Scholar]

130. Blaskovich MA, et al. Discovery of JSI-124 (cucurbitacin I), a selective Janus kinase/signal transducer and activator of transcription 3 signaling pathway inhibitor with potent antitumor activity against human and murine cancer cells in mice. Cancer Res. 2003;63:1270–1279. [PubMed] [Google Scholar]

131. Kotha A, et al. Resveratrol inhibits Src and Stat3 signaling and induces the apoptosis of malignant cells containing activated Stat3 protein. Mol. Cancer Ther. 2006;5:621–629. [PubMed] [Google Scholar]

132. Nam S, et al. Indirubin derivatives inhibit Stat3 signaling and induce apoptosis in human cancer cells. Proc. Natl Acad. Sci. USA. 2005;102:5998–6003. [PMC free article] [PubMed] [Google Scholar]

133. Ghoreschi K, Laurence A, O’Shea JJ. Selectivity and therapeutic inhibition of kinases: to be or not to be? Nature Immunol. 2009;10:356–360. [PMC free article] [PubMed] [Google Scholar]

135. Kong LY, et al. A novel inhibitor of signal transducers and activators of transcription 3 activation is efficacious against established central nervous system melanoma and inhibits regulatory T cells. Clin. Cancer Res. 2008;14:5759–5768. [PMC free article] [PubMed] [Google Scholar]

136. Nefedova Y, et al. 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;65:9525–9535. [PMC free article] [PubMed] [Google Scholar]

137. Gozgit JM, et al. Effects of the JAK2 inhibitor, AZ960, on Pim/BAD/BCL-xL survival signaling in the human JAK2 V617F cell line SET-2. J. Biol. Chem. 2008;283:32334–32343. [PubMed] [Google Scholar]

138. Yang F, et al. Sorafenib inhibits signal transducer and activator of transcription 3 signaling associated with growth arrest and apoptosis of medulloblastomas. Mol. Cancer Ther. 2008;7:3519–3526. [PMC free article] [PubMed] [Google Scholar]

139. Xin H, et al. Sunitinib inhibition of Stat3 induces renal cell carcinoma tumor cell apoptosis and reduces immunosuppressive cells. Cancer Res. 2009;69:2506–2513. [PMC free article] [PubMed] [Google Scholar]

140. Ozao-Choy J, 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;69:2514–2522. [PMC free article] [PubMed] [Google Scholar]

141. Ko JS, et al. Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients. Clin. Cancer Res. 2009;15:2148–2157. [PubMed] [Google Scholar]

142. Caprioli F, et al. Autocrine regulation of IL-21 production in human T lymphocytes. J. Immunol. 2008;180:1800–1807. [PubMed] [Google Scholar]

143. Donnelly RP, Dickensheets H, Finbloom DS. The interleukin-10 signal transduction pathway and regulation of gene expression in mononuclear phagocytes. J. Interferon Cytokine Res. 1999;19:563–573. [PubMed] [Google Scholar]

144. Lamprecht B, et al. Aberrant expression of the Th2 cytokine IL-21 in Hodgkin lymphoma cells regulates STAT3 signaling and attracts Treg cells via regulation of MIP-3α Blood. 2008;112:3339–3347. [PubMed] [Google Scholar]

145. Takatori H, Kanno Y, Chen Z, O’Shea JJ. New complexities in helper T cell fate determination and the implications for autoimmune diseases. Mod. Rheumatol. 2008;18:533–541. [PMC free article] [PubMed] [Google Scholar]

146. Stout BA, Bates ME, Liu LY, Farrington NN, Bertics PJ. IL-5 and granulocyte-macrophage colony-stimulating factor activate STAT3 and STAT5 and promote Pim-1 and cyclin D3 protein expression in human eosinophils. J. Immunol. 2004;173:6409–6417. [PubMed] [Google Scholar]

147. Zorn E, et al. IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood. 2006;108:1571–1579. [PMC free article] [PubMed] [Google Scholar]

148. Finkelman FD, et al. Interleukin-4- and interleukin-13-mediated host protection against intestinal nematode parasites. Immunol. Rev. 2004;201:139–155. [PubMed] [Google Scholar]

149. Schuringa JJ, Timmer H, Luttickhuizen D, Vellenga E, Kruijer W. c-Jun and c-Fos cooperate with STAT3 in IL-6-induced transactivation of the IL-6 respone element (IRE) Cytokine. 2001;14:78–87. [PubMed] [Google Scholar]

150. Lo HW, et al. Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelial-mesenchymal transition in cancer cells via up-regulation of TWIST gene expression. Cancer Res. 2007;67:9066–9076. [PMC free article] [PubMed] [Google Scholar]

151. Wu Y, Diab I, Zhang X, Izmailova ES, Zehner ZE. Stat3 enhances vimentin gene expression by binding to the antisilencer element and interacting with the repressor protein, ZBP-89. Oncogene. 2004;23:168–178. [PubMed] [Google Scholar]

152. Liu H, et al. Serine phosphorylation of STAT3 is essential for Mcl-1 expression and macrophage survival. Blood. 2003;102:344–352. [PubMed] [Google Scholar]

153. Madamanchi NR, Li S, Patterson C, Runge MS. Thrombin regulates vascular smooth muscle cell growth and heat shock proteins via the JAK-STAT pathway. J. Biol. Chem. 2001;276:18915–18924. [PubMed] [Google Scholar]

154. Bartoli M, et al. VEGF differentially activates STAT3 in microvascular endothelial cells. FASEB J. 2003;17:1562–1564. [PubMed] [Google Scholar]

155. Gao H, Priebe W, Glod J, Banerjee D. Activation of signal transducers and activators of transcription 3 and focal adhesion kinase by stromal cell-derived factor 1 is required for migration of human mesenchymal stem cells in response to tumor cell-conditioned medium. Stem Cells. 2009;27:857–865. [PubMed] [Google Scholar]

156. Leung TH, Hoffmann A, Baltimore D. One nucleotide in a κB site can determine cofactor specificity for NF-κB dimers. Cell. 2004;118:453–464. [PubMed] [Google Scholar]

157. Burysek L, Syrovets T, Simmet T. The serine protease plasmin triggers expression of MCP-1 and CD40 in human primary monocytes via activation of p38 MAPK and janus kinase (JAK)/STAT signaling pathways. J. Biol. Chem. 2002;277:33509–33517. [PubMed] [Google Scholar]