Journal of Cancer Therapy
Vol.06 No.08(2015), Article ID:58709,17 pages
10.4236/jct.2015.68078

STAT3 Signaling in Cancer

Ioannis Gkouveris1*, Nikolaos Nikitakis1, John Sauk2

1Department of Oral Pathology and Medicine, Dental School, National and Kapodistrian University of Athens, Athens, Greece

2School of Dentistry, University of Louisville, Louisville, USA

Email: *igkouver@gmail.com, nnikitakis1@yahoo.com, john.sauk@louisville.edu

Copyright © 2015 by authors and Scientific Research Publishing Inc.

This work is licensed under the Creative Commons Attribution International License (CC BY).

http://creativecommons.org/licenses/by/4.0/

Received 15 July 2015; accepted 7 August 2015; published 10 August 2015

ABSTRACT

In recent years, signal transducers and activators of transcription (STAT) proteins have been recognized as cytoplasmic transcription factors that mediate extracellular signaling to the nucleus controlling fundamental functions, such as cell proliferation, apoptosis, differentiation, immune responses and angiogenesis. Among them, STAT3 is a major player, aberrant activation of which is involved in several diseases, including cancer. Among other upstream regulators, IL-6/Jak signaling can activate STAT3 and its role appears to be critical in various types of cancer. Although STAT3 has been traditionally recognized as amoncogene, more recently the dual role of STAT3 in cancer, either tumor inductive or suppressive, has been appreciated. The importance and differential effect of STAT3 on tyrosine or serine residues are also a matter of continuing debate. Interestingly, recent findings suggesting that STAT3 plays an important role in cancer stem cell regulation have gained significant attention. This review summarizes current literature focusing on the significance of STAT3 in several diseases as well as in cancer. Understanding the complexity of STAT3 function has the potential to elucidate important molecular aspects of cancer with significant therapeutic implications.

Keywords:

STAT3, Signaling, Cancer, Dual Role, Tyrosine and Serine Phosphorylation

1. Introduction

STAT proteins (signal transducers and activators of transcription) constitute a large family of transcription factors with a dual role as signal transduction and transcription activators. STATs were first described in 1994 [1] as members of interferon (IFN) signaling complex [2] [3] . They are found in the cytoplasm in a latent form and become active in response to stimulation by cytokines and growth factors, hormones and peptides. STAT3 is a significant member of STAT family and has been largely studied, in recent years. This review attempts to elucidate the role of STAT3 molecule signaling in occurrence of diseases including cancer.

2. STATs: The Members of the Family

STAT family consists of 7 members in mammals including STAT-1, STAT-2, STAT-3, STAT-4, STAT-5 a and b, and STAT-6 [4] . STAT proteins comprise 750 to 850 amino acids and some authors classify them according to their functional role into two groups [5] [6] : STAT2, STAT4, and STAT6 represent the first group which is involved in T-cell development and IFN-γ signaling and become activated through several cytokines. The second one consists of STAT1, STAT3, and STAT5, being activated in different tissues through a series of different ligands and considered to be involved in various processes, such as IFN-γ signaling, development of mammary glands, growth hormones response and embryogenesis. This latter group of STATs is supposed to play a crucial role in cancer development by controlling fundamental cellular functions including cell cycle and apoptosis [6] .

2.1. STAT1

Darnell et al. first proposed that the initial physical role of STAT1 is to mediate the antiviral and immune effects of IFNs. Both STAT1 and STAT2 directly mediate IFN-α and IFN-γ biological effects and play a significant role in the mechanisms that control cell growth and apoptosis [1] . The involvement of STAT1 in cancer is controversial; for example, it is suggested that STAT1 activity enhances breast tumor growth and immune suppression [7] , while other evidence indicates the loss of its expression in different types of malignant cells, such as breast cancer, head and neck cancer [8] , melanoma, leukemia, and lymphoma [9] - [12] .

2.2. STAT2

STAT2 is supposed to be vital in innate immunity and specific viruses are found to target STAT2 to surpass the IFN antiviral response. STAT2 plays also a crucial role in promoting IFN-induced apoptosis, while its transcriptional activity is controversial, potentially acting both as a repressor and activator. Moreover, even though STAT2 forms heterodimers with STAT1, recent data proposed an alternative STAT2 signaling pathway, independent of STAT1 [13] .

2.3. STAT3

STAT3 activation in normal conditions drives a well organized gene regulation schedule. After STAT3 is exposed to cytokine stimulation, the molecule can reach a maximum of phosphorylation within the first 15 - 60 minutes, but STAT3 activation gradually decreases in the following hours [6] . STAT3 activation is mediated by the JAK family of tyrosine-kinases, most notably by JAK1 [14] . STAT3 can be activated independent of JAKs by other non-receptor tyrosine kinases, mostly by c-Src kinases [3] . STAT3 is phosphorylated at tyrosine 705 and serine 727 residues with tyrosine phosphorylation being associated in many cases with disease progression and tumorigenic potential [15] . Upon activation, phosphorylated STAT3 molecules form dimers and translocate into the nucleus to regulate transcription of genes, controlling cell survival and proliferation [5] [16] . Moreover, activated STAT3 regulates the expression of anti-apoptotic, pro-proliferative and immune response genes [5] [17] .

2.4. STAT4

Several lines of evidence imply that STAT4 expression is exclusively found in myeloid cells (activated monocytes, macrophages, and dendritic cells [18] . STAT4 is mostly activated by IL-12, which in turn regulates tissue inflammation, fibrogenesis and antiviral defense [19] . The binding of IL-12 with its receptor, in CD4+ Th1 cells is followed by phosphorylation of tyrosine 693 and serine 721 of STAT4 [20] . Consequently, the activated STAT4 moves into the nucleus, binds to DNA and enhances the production of inflammatory cytokines such as IFN-γ [21] .

2.5. STAT5 (A and B)

Janus kinases can also phosphorylate STAT5 proteins when the latter bind to the phospho-tyrosine residues in their receptors. STAT5 is phosporylated at Y694 and Y699 residues for STAT5A and STAT5B, respectively, and these phosphorylations are essential for the steady formation of STAT5 dimers through their SH2 domains. Both STAT5 isoforms are activated by the same set of cytokines, nevertheless some cytokines selectively activate either STAT5A or STAT5B (e.g. prolactin activates STAT5A) [22] . Upon activation, STAT5 dimers move into the nucleus and bind to specific DNA sequences, mainly concerning IFN-γ [23] . Moreover, interactions between STAT5 N-terminal domains lead to stable tetramer formation of Stat5 [24] [25] , which is critical for cytokine regulation and immune responses [26] . Aberrant STAT5 signaling is often associated with leukemogenesis and other cancers [27] [28] .

2.6. STAT6

The seventh member of STAT family, STAT6, regulates gene expression in several cell types, a crucial function for maintaining the balance between host immune defense and allergic inflammatory responses [19] . STAT6 gene regulation in response to IL-4/IL-13 stimulation varies and depends on the cell type [29] [30] . For example STAT6 promotes IgE chain and CD23 gene expression in B cells, while in T cells enhances Th2 differentiation genes gata3 and crth2 [31] - [34] . Moreover, Lawrence et al. reported that stimulation of macrophages by IL-4/ IL-13 induces STAT6-dependent activation and transcription of arginase 1, Retnla, and Chi3L3 in mouse and indolamine 2,3-dioxygenase (IDO) in humans [31] . STAT6 not only acts as a transcriptional activator by regulating Th2 development, but also suppresses gene expression through steric hindrance of binding by other transcription factors [29] . This repressing activity leads to subsequent side effects and probably plays a significant role in Th2 cell programming [35] .

3. Role of STAT3 in Diseases

Normally, STAT3 functional role contributes to controlled gene regulation. Nevertheless, STAT3 aberrant activation has been involved not only in oncogenesis but also in several other types of disease. For example, previous studies suggested that Stat3 plays a crucial role in the pathogenesis of diabetic nephropathy [36] and is involved in cytokine- and nutrient-induced insulin resistance [37] . Moreover, excessive STAT3 signaling leads to development of skeletal muscle insulin resistance in type 2 diabetes [37] .

Aberrant IL-6/STAT3 signaling has been also studied in endometriosis, which is an estrogen-dependent inflammatory disease. Kim et al. found higher levels of phospho-STAT3 and HIF1α (Hypoxia-inducible factor 1- alpha, a downstream substrate of STAT3) in the endometrium of patients with endometriosis compared with healthy women and proposed that consistent activation of STAT3 contributes in the pathogenesis of endometriosis [38] .

The JAK/STAT signaling pathway is found to be active in a variety of renal diseases and it is proposed to contribute in the pathophysiology of renal fibrosis. Matsui et al. referred that inhibition of the JAK/STAT signaling pathway, especially JAK2 and STAT3, appeared to diminish renal fibrosis and protected renal activity [39] . As a result Tanq et al. [40] demonstrated that fluorofenidone (FD), a novel pyridone agent, exerts an anti- fibrotic effect through inhibiting STAT3 tyrosine phosphorylation in the JAK2/STAT3 pathway, consequently adding a new therapeutic strategy in renal fibrosis.

STAT3 disease involvement has been also linked to dyregulations of IL-6 signaling. Several studies suggested that IL6 trans-signaling (complex of IL6 and soluble IL6 receptor) played a pathogenic role in lung airway smooth muscle diseases [41] . Classical IL6 as well as IL6 trans-signaling in human airway smooth muscle involve activation of Stat3, but IL6 trans-signaling has been shown to specifically contribute to asthma pathogenesis and can be considered as a potential modifier of airway inflammation and remodeling [41] .

Furthermore, it has been shown that high levels of IL10 are found in serum and tissues of patients with systemic lupus erythematosus (SLE). Hedrich et al. reported that Stat3 and Stat5 regulate trans-activation and epigenetic remodeling of IL10 by interacting with the histone acetyltransferase p300. Specifically, the activation of Stat3 in T cells from SLE patients led to enhanced recruitment to regulatory regions and competitive replacement of Stat5, leading to enhanced IL-10 expression [42] . As a result, Wang et al. suggested that Natura-alpha (a novel STAT3-Y705 inhibitor) could be used as a potential SLE inhibitor [43] .

In addition STAT3 plays a significant role in some autoimmune disorders including inflammatory bowel disease (IBD). Several lines of evidence suggested that STAT3 activation posseses a dual and contradictory role between innate and acquired immune responses in colitis [44] . STAT3-mediated activation of acquired immune responses contributed in pathogenesis of colitis by supporting the survival of pathogenic T cells, while STAT3- mediated activation of innate responses suppressed the pathogenesis of colitis [44] . More recent data suggested that several interleukins, like IL-6, IL-11, and IL-22, are highly expressed in IBD cases, and that consequent activation of JAK/STAT3 pathway can ameliorate disease and protect the epithelial lining cells [45] .

Moreover, experimental evidence showed that several inflammatory cytokines, such as IL-1β, tumor necrosis factor alpha and IL-6, are highly expressed in patients with rheumatoid arthritis (RA), leading to direct or indirect activation of STAT3, which in turn enhances expression of IL-6 family cytokines and promotes sustained inflammation and joint destruction [46] . In addition, Gao et al. found that there is an interaction between HIF1α, STAT3 and Notch-1 signaling in the regulation of pro-inflammatory pathway in RA, which implied a role for targeting STAT3 in treatment of RA [47] .

STAT3 regulation has also been involved in the pathophysiology of behavior. Several lines of evidence suggested a relationship between changes in the immune system, mostly in IL6 expression, and depression [48] . Kong et al. examined the existence of an IL6-induced modulation of serotonergic neurotransmission through the STAT3 signaling pathway which may enhance the role of IL6 in depression. Indeed, they found that IL6 directly regulated Serotonin Transporter protein levels (SERT) and consequently affected serotonin reuptake, thus proposing that IL6 could be connected to depression through a potent STAT3-dependent regulation model of SERT [48] .

Moreover, JAK/STAT3 pathway has been connected with Alzheimer’s and Huntington’s diseases [49] . Experimental evidence suggested that astrocyte reactivity is a hallmark of neurodegenerative diseases and that JAK/STAT3 pathway correlates with reactive astrocytes in models of acute injury. Ben Haim et al. examined astrocyte reactivity in progressive pathological conditions such as Alzheimer’s and Huntington’s disease and showed that JAK/STAT3 pathway is a common inducer of astrocyte reactivity thus adding novel information to the pathogenesis of neurodegenerative diseases [49] (Figure 1).

4. Dual Role of STAT3 in Cancer

4.1. STAT3 Tumorigenic Role

Inappropriate STAT3 activation, mainly due to persistent tyrosine 705 phosphorylation signaling, has been convincingly shown to contribute to oncogenesis and to promote the acquisition of a malignant phenotype [50] - [53] . Consistent activation of STAT3 in cancer transfers signals from cytokines and growth factors [54] and stimulates specific target genes such as Fos, Cyclin-D, CDC25A, c-Myc or Pim1 that induce cell proliferation, suppress apoptotic genes (Fas) [55] and up-regulate antiapoptotic genes including BCL2 (B-Cell CLL/Lymphoma- 2), BCLXL and Beta2-Macroglobulin [56] .

Figure 1. STAT3 involvement in diseases other than cancer.

Persistent STAT3 activation has been described in various types of solid and hematological cancers and targeting STAT3 expression could be a useful strategy for cancer therapies in the future [57] . For instance, activation of STAT3 by Src kinase has been shown to be essential in prostate and ovarian cancer [58] . Moreover, enhanced expression of BRCA1 gene (breast cancer susceptible gene 1), which has been associated with breast, ovarian and prostate cancer, induced constitutive phosphorylation of STAT3 at serine and tyrosine residues in prostate cancer cell lines, also interacting with the upstream activators JAK1 and JAK2 [59] . In addition, STAT3 constitutive activation, mostly associated with aberrant TGF-α/EGFR signaling, contributed to HNSCC development and growth [53] [60] . Further, activated STAT3 expression was found to significantly correlate with extent of tumor invasion, lymph node metastasis and tumor grade in colorectal cancer [61] .

4.1.1. STAT3 Overactivation

Furthermore, somatic mutations in STAT3 have been diagnosed in several malignancies, including hepatocellular adenomas, T-cell large granular lymphocytic leukemia (T-cell LGL), chronic lymphoproliferative disorders of natural killer cells (CLPD-NKs), diffuse large B-cell lymphoma, and CD30+ T-cell lymphomas [76] - [80] .

4.1.2. Disruption of STAT3 Negative Regulation

STAT3 persistent activation exists when a disruption in negative regulations of STAT3 occurs. Suppressors of cytokine signaling (SOCS) and protein tyrosine phosphatases (PTPs) are known to control STAT3 homeostasis of phosphorylation [81] - [83] . Experimental data suggested that loss of SOCS3 by genetic disruptions induced STAT3 activation and increased proliferation, survival and motility in several cancer cells [50] .

SOCS1 can also be suppressed by aberrant gene methylation and this condition has been shown to result to persistent STAT3 activation in several types of cancer [84] - [87] . Similarly SHP-1, a member of tyrosine phosphatase family can be deregulated after epigenetic alterations, notably in hematologic malignancies [86] [88] [89] . Disruption of SHP-1 has been proposed to correlate with constitutive activation of STAT3 in cancer types including ALK-positive anaplastic large cell lymphoma, chronic myeloid leukemia and multiple myeloma.

4.2. STAT3 Tumor Suppressive Role

Using siRNA techniques in astrocytes derived from conditional knockout mice, De la Iglesia et al. demonstrated a tumor suppressive effect of STAT3 in the absence of PTEN expression in glioblastoma. On the other hand, they observed an oncogenic STAT3 effect following co-expression of EGFRvIII and the interaction between these molecules in the nuclei of glioblastoma cells [91] .

5. Role of Il6/STAT3 Signaling in Cancer

Interleukin-6 (IL-6) is a cytokine secreted by T-cells and macrophages and is involved in immune and inflammatory responses [99] . IL-6 signaling launches with ligand binding to its receptor IL-6R and a common receptor subunit gp130. By this interaction, a hexameric receptor complex of two IL-6, IL-6R, and gp130 hetero-trimers is formed [100] . This signaling pathway is triggered during early immune responses, consequently stimulating the expression of various acute-phase proteins, and is referred to as classical signaling of IL-6 [101] . In addition, IL- 6 can also bind to an existing soluble type of the IL-6 receptor (sIL-6R) and form a complex that interacts with gp130. This type of IL-6 signaling, called IL-6 trans-signaling, differs from classical IL-6 signaling and plays a significant role in the function of several cells including neutrophils, macrophages, epithelial cells and T cells [102] . Both IL-6 signaling pathways trigger responses including activation of JAK (Janus Kinase) kinases. JAKs are cytoplasmic tyrosine kinases which in mammalians consitute a protein family of four members. JAK1, JAK2 and TYK2 are expressed in several cells, while JAK3 is found only in cells of the hematopoietic system [103] . JAKs and especially JAK1 is involved in the activation of STAT3 via phosphorylation of a specific tyrosine residue [14] [104] . As previously described, when STAT3 becomes phosphorylated, it forms dimers and moves from the cytoplasm to the nucleus stimulating transcription of STAT3 target genes, including cyclin D1, Bcl-xL, c-myc, Mcl1 and vascular endothelial growth factor (VEGF) [104] (Figure 2).

IL-6 has been demonstrated to be implicated in several malignancies including prostate [105] , breast [106] , lung cancer [107] , and oral SCC [108] , by regulating critical cellular activities such as proliferation [105] [107] (asangari, lin2), apoptosis [105] , and invasion [106] (lin1). For example, Chang et al. has demonstrated that IL-6 induced transient increase of STAT3 tyrosine phosphorylation in a dose-dependent manner, which in turn resulted in neuroendocrine dedifferentiation and cell proliferation in non-small cell lung cancer cells [109] . More- over, Wan et al. observed that cancer stem cells played a role in progression and recurrence of hepatocellular carcinoma after therapy and that tumor-associated macrophages (TAMs) expression was associated with poor outcomes. They suggested that TAMs produce IL-6 and contributed to expansion of human hepatocellular carcinoma stem cells via STAT3 signaling [110] .

Patel et al. documented that, in colorectal cancer cells, colonic inflammation through IL-6 signaling can result in metabolic changes of epithelial cells by controlling expression of cytochrome enzymes including CYP2E1 and CYP1B1, through transcriptional and epigenetic mechanisms. Specifically CYP2E1 overexpression, as a result of STAT3 pathway, enhanced activation of dietary carcinogens and DNA damage, thus promoting colorectal carcinogenesis [111] . Similarly, modulation of the IL-6/JAK/STAT3 signaling pathway has been proposed as a potential therapeutic approach to treat patients with colorectal cancer [104] .

Figure 2. STAT3 activation and signaling.

Furthermore, Liu et al. showed that the IL6-Stat3-AR (androgen receptor) cascade is a significant regulator of enzalutamide (androgen receptor antagonist drug) resistance in prostate cancer. This study also demonstrated that the drug Niclosamide could target IL6-Stat3-AR pathway and consequently overcome enzalutamide resistance, resulting in inhibition of migration and invasion in advanced prostate cancer [114] . Finally, Cheng et al. have also proposed salivary IL6 and IL-8 as potential biomarkers for oral SCC [108] .

6. Role of STAT3 Serine Phosphorylation

As descried before, numerous investigations have demonstrated that overexpression of STAT3 is responsible for various oncogenic processes, such as solid tumor progression, pathological angiogenesis [115] , and promotion of cell growth and transformation [116] . As alluded previously, it is also widely known that the oncogenic potential of STAT3 depends mainly on the phosphorylation status of Tyr705 [117] .

In contrast, STAT3 serine phosphorylation may also arise as a result of growth factor and cytokine stimulation, but the role of this activation remains controversial [117] . Several studies suggested that serine activation could drive to both stimulating and inhibitory effects on gene transcription [118] - [122] , while others postulated that Ser-727 phosphorylation may inhibit Tyr-705 phosphorylation or, quite the opposite, result in further STAT activation [123] .

The negative impact of Ser727 residue phosphorylation on STAT3 activity has been proposed in several studies indicating that Ser727 phosphorylation downregulates STAT3 tyrosine phosphorylation and causes alterations in nuclear translocation and transcriptional activity [122] . Moreover, it has been suggested that Ser727 phosphorylation either inhibits tyrosine activation or increases tyrosine dephosphorylation [124] . Mandal et al. found that when Stat3 Ser727 phosphorylation was reduced, tumorigenicity of glioma cells was increased probably through a CK2-PP2A (casein kinase 2―Protein phosphatase 2A) pathway followed by conversely increased STAT3 tyrosine phosphorylation [15] . Moreover, Venkatasubbarao et al. [125] and Wakahara et al. [126] proposed that phospho-Ser727 regulated the direction of STAT3 activity by enhancing either tyrosine dephosphorylation or phosphorylation, mainly through TC45 phosphatase. An Erk1/2-STAT3 crosstalk in oral SCC has been also described by Gkouveris et al., who demonstrated that ERK1/2 inhibition is followed by increases in STAT3 serine phosphorylation and increases in tyrosine phosphorylation, while ERK1/2 induction had the opposite effects [53] .

On the other hand, other researchers have proposed that serine phosphorylation correlated with increased nuclear translocation and enhanced transcriptional activity [120] [127] . Moreover, MEK-ERK signaling has been shown to drive Ras-induced phosphorylation of STAT3 on Ser727 and mitochondrial STAT3 is a crucial substrate of the Ras-MEK-ERK pathway during cellular transformation [128] . In addition, Zhang et al. suggested that Ser727 phosphorylation status mediated the behavior of a variety of tumors, also demonstrating that Ser727 phospshorylation of mitochondrial Stat3 is required for Ras-mediated transformation of MEFs (mouse embryo fibroblasts) [116] . In addition, it has been shown that Ser727 phosphorylation may correlate with the growth and transformation of other malignancies, such as chronic lymphocytic leukemia, prostate and breast cancer [129] - [131] . Lee et al. reported that YB-1 (y box binding protein) prevented the apoptosis of breast cancer cells by AKT/mTOR signaling, resulting in STAT3 serine phosphorylation [132] . Hazan-Halevy et al. suggested that constitutive STAT3 Ser727 phosphorylation played a crucial role in chronic lymphocytic leukemia (CLL) and targeting serine phosphorylation could be used as a novel therapeutic strategy [120] .

Furthermore, Waitkus et al. described that activation of epidermal growth factor receptor (EGFR) and protease-activated receptor 1 (PAR-1) leads to Both Thr714 and Ser727 STAT3 phosphorylation and consequently in a STAT3-dependent gene induction in endothelial cells and found that this double phosphorylated STAT3 complex is highly expressed compared to tyrosine-STAT3 levels in clear-cell renal-cell carcinoma [115] .

In another study, Miyakoshi et al. revealed that MAPK activation through FBS treatment of mouse hepatic carcinoma cells enhanced STAT3 phosphorylation in Ser727 and increased STAT3 nuclear translocation and cell proliferation [133] . Finally, Sakaguchi et al. showed that constitutive Ser727 phosphorylation in melanoma cells, partially mediated by the B-Raf-MEK-Erk1/2 pathway, affected cell survival and nuclear translocation of STAT3 [134] .

7. STAT3 Activation in Cancer Stem Cells

Aberrant regulation and transformation of stem cells into cancer stem cells (CSCs) are found to correlate with cancer development, metastasis and drug resistance [135] . It is suggested that a critical event which causes these cellular alterations is the existence of high Reactive oxygen species (ROS), Reactive nitrogen species (RNS) (ROS/RNS) and Lipoma-preferred partner (LPPs) levels in the cellular microenvironment as a result of chronic systemic or local inflammation [136] . In turn, the presence of high ROS/RNS expression levels is postulated to lead to DNA damage and oncogene activation [136] . Tumor inflammation can exist in cases of chronic trauma or infections (viruses or parasitic infections), chemical carcinogens, or autoimmune disorders [137] . Considering the fact that STAT3 is well known to play a significant role in tumor inflammatory environment, it is plausible that STAT3 activation is involved in CSCs regulation [136] .

STAT3 is suggested to promote prostate tumorigenesis and high tyrosine phosphorylated STAT3 levels correlate with higher Gleason score and pathologic stage of the disease [138] [139] . In contrast, inhibition of STAT3 signaling appears to exert antitumor effects in patient-derived PCa xenograft models [140] . Noteworthy is that STAT3 activation by IL-6 [141] or stress factors like ROS [142] results in enhanced self-renewal and tumor-propagating capacity of prostate CSCs [143] . Moreover, Hossain et al. found that glioma-associated-human mesenchymal stem cells (GA-hMSCs) enhance tumorigenic activity of glioma stem cells (GSCs) by inducing their proliferation and self-renewal through the IL-6/gp130/STAT3 pathway [144] .

High levels of aldehyde dehydrogenase (ALDH), a detoxifying enzyme mostly expressed in progenitor and stem cells, in endometrial cancer patients are associated with relatively lower survival rates compared to patients with low levels of ALDH. Recently, van der Zee et al. reported that endometrial cancer cells with high levels of ALDH activity, accompanied by upregulation of IL-6 receptor subunits, exhibited CSC activities. Notably, inhibition of the IL-6 receptor and its downstream effectors JAK1 and STAT3 dramatically reduced tumor cell growth [145] .

Recently, Islam et al. examined the role of RhoC (a pro-metastatic oncogene) in CSCs formation in HNSCC cell lines. ShRNA inhibition of RhoC resulted in lower expression of ALDH and CD44 stem cell markers, while STAT3 serine and tyrosine levels were significantly downregulated in those RhoC-depleted HNSCC cell lines. The authors concluded that over activation of IL6/STAT3 pathway, mainly regulated by RhoC, controls CSC functions [146] .

Furthermore, Won et al. indicated that high levels of CSC marker CD133 correlate with tumor growth and poor prognosis in hepatocellular carcinoma (HCC) and reported that STAT3 activation via IL-6 stimulation increased protein levels of CD133 and promoted cancer progression. In contrast, silencing of CD133 resulted in cell cycle arrest and tumor suppression by causing downregulation of cytokine-related genes, including TACC1, ACF7 and CKAP5. Also, treatment with sorafenib and nifuroxazide inhibited STAT3 activation and CD133 expression leading to reduced HCC xenograft formation [147] .

Moreover, Chen et al. investigated the effect of the neuroleptic drug pimozide in HCC cells or stem-like cells and found that treatment with pimozide resulted in reduced STAT3 activity, mainly manifested by both lower luciferase activity and expression of STAT3 target genes. Furthermore, IL6-induced tumorigenic effect in stem- like cells was decreased after pimozide treatment [148] .

In a recent investigation, Zhao et al. showed that vascular endothelial growth factor-A (VEGF), promotes breast and lung CSC self-renewal via VEGFR-2/JAK2/STAT3 binding, resulting in STAT3 activation and ensuing upregulation of Myc and Sox2. These novel findings support the notion that, in addition to angiogesis, VEGF drives tumor-initiating CSC self-renewal through VEGFR-2/STAT3 signaling [149] . Finally, Thakur et al. studied the effect of Shikonin (Shk) on breast cancer and investigated the existence of a possible anti-CSC role. Treatment of cells with Shk drove to lower levels of various epithelial to mesenchymal transition (EMT) and CSC associated markers, accompanied by inhibition of STAT3, FAK (Focal adhesion kinase) and Src, proposing a tumor suppressive effect in breast cancer [135] .

8. Conclusions

In summary, a plethora of studies indicate that deregulation of STAT3 pathway is involved in various diseases, including many cancer types, revealing the significance of retaining normal STAT3 signaling for cellular stability.

STAT3 constitutive activation has been shown to contribute to tumor development and progression, while IL-6/JAK pathway plays a crucial role in aberrant STAT3 signaling cascades. In addition, the role of STAT3 serine phosphorylation, as supporting or opposing the effects of tyrosine phosphorylation in the mechanisms of cancer pathology, needs further elucidation. More intriguing is the recent evidence demonstrating divergent roles of STAT3 in cancer biology, which may on certain occasions function as a potent tumor suppressor. Even grater enthusiasm has been generated by the latest discoveries implicating STAT3 in the regulation of cancer stem cells in various types of malignancies.

Understanding the complexity of STAT3 activation, as well as the significance of this signaling pathway in cancer, holds great promise for the development of new therapeutic strategies in several disorders, including cancer.

Cite this paper

IoannisGkouveris,NikolaosNikitakis,JohnSauk, (2015) STAT3 Signaling in Cancer. Journal of Cancer Therapy,06,709-726. doi: 10.4236/jct.2015.68078

References

  1. 1. Darnell Jr., J.E., Kerr, I.M. and Stark, G.R. (1994) Jak-STAT Pathways and Transcriptional Activation in Response to IFNs and Other Extracellular Signaling Proteins. Science, 264, 1415-1421.
    http://dx.doi.org/10.1126/science.8197455

  2. 2. Lai, S.Y. and Johnson, F.M. (2010) Defining the Role of the JAK-STAT Pathway in Head and Neck and Thoracic Malignancies: Implications for Future Therapeutic Approaches. Drug Resistance Updates, 13, 67-78.
    http://dx.doi.org/10.1016/j.drup.2010.04.001

  3. 3. Mali, S.B. (2015) Review of STAT3 (Signal Transducers and Activators of Transcription) in Head and Neck Cancer. Oral Oncology, 51, 565-569.
    http://dx.doi.org/10.1016/j.oraloncology.2015.03.004

  4. 4. Xiong, A., Yang, Z., Shen, Y., Zhou, J. and Shen, Q. (2014) Transcription Factor STAT3 as a Novel Molecular Target for Cancer Prevention. Cancers (Basel), 6, 926-957.
    http://dx.doi.org/10.3390/cancers6020926

  5. 5. Siveen, K.S., Sikka, S., Surana, R., Dai, X., Zhang, J., Kumar, A.P., Tan, B.K., Sethi, G. and Bishayee, A. (2014) Targeting the STAT3 Signaling Pathway in Cancer: Role of Synthetic and Natural Inhibitors. Biochimica et Biophysica Acta, 1845, 136-154.

  6. 6. Subramaniam, A., Shanmugam, M.K., Perumal, E., Li, F., Nachiyappan, A., Dai, X., et al. (2013) Potential Role of Signal Transducer and Activator of Transcription (STAT)3 Signaling Pathway in Inflammation, Survival, Proliferation and Invasion of Hepatocellular Carcinoma. Biochimica et Biophysica Acta (BBA)—Reviews on Cancer, 1835, 46-60.
    http://dx.doi.org/10.1016/j.bbcan.2012.10.002

  7. 7. Hix, L.M., Karavitis, J., Khan, M.W., Shi, Y.H., Khazaie, K. and Zhang, M. (2013) Tumor STAT1 Transcription Factor Activity Enhances Breast Tumor Growth and Immune Suppression Mediated by Myeloid-Derived Suppressor Cells. Journal of Biological Chemistry, 288, 11676-11688.
    http://dx.doi.org/10.1074/jbc.M112.441402

  8. 8. Adámková, L., Soucková, K. and Kovarík, J. (2007) Transcription Protein STAT1: Biology and Relation to Cancer. Folia Biologica (Praha), 53, 1-6.

  9. 9. Boudny, V., Kocak, I., Lauerova, L. and Kovarik, J. (2003) Interferon Inducibility of STAT 1 Activation and Its Prognostic Significance in Melanoma Patients. Folia Biologica (Praha), 49, 142-146.

  10. 10. Bowman, T., Garcia, R., Turkson, J. and Jove, R. (2000) STATs in Oncogenesis. Oncogene, 19, 2474-2488.
    http://dx.doi.org/10.1038/sj.onc.1203527

  11. 11. Buettner, R., Mora, L.B. and Jove, R. (2002) Activated STAT Signaling in Human Tumors Provides Novel Molecular Targets for Therapeutic Intervention. Clinical Cancer Research, 8, 945-954.

  12. 12. Kovarik, A., Fojtova, M., Boudny, V., Adamkova, L., Lauerova, L. and Kovarik, J. (2005) Interferon γ but Not α Induce SOCS 3 Expression of Human Melanoma Cell Lines. Melanoma Research, 15, 481-488.
    http://dx.doi.org/10.1097/00008390-200512000-00001

  13. 13. Steen, H.C. and Gamero, A. (2012) The Role of Signal Transducer and Activator of Transcription-2 in the Interferon Response. Journal of Interferon & Cytokine Research, 32, 103-110.
    http://dx.doi.org/10.1089/jir.2011.0099

  14. 14. Hirano, T., Ishihara, K. and Hibi, M. (2000) Roles of STAT3 in Mediating the Cell Growth, Differentiation and Survival Signals Relayed through the IL-6 Family of Cytokine Receptors. Oncogene, 19, 2548-2556.
    http://dx.doi.org/10.1038/sj.onc.1203551

  15. 15. Mandal, T., Bhowmik, A., Chatterjee, A., Chatterjee, U., Chatterjee, S. and Ghosh, M.K. (2014) Reduced Phosphorylation of Stat3 at Ser-727 Mediated by Casein Kinase 2—Protein Phosphatase 2A Enhances Stat3 Tyr-705 Induced Tumorigenic Potential of Glioma Cells. Cellular Signalling, 26, 1725-1734.
    http://dx.doi.org/10.1016/j.cellsig.2014.04.003

  16. 16. Chen, R.J., Ho, Y.S., Guo, H.R. and Wang, Y.J. (2008) Rapid Activation of Stat3 and ERK1/2 by Nicotine Modulates Cell Proliferation in Human Bladder Cancer Cells. Toxicological Sciences, 104, 283-293.
    http://dx.doi.org/10.1093/toxsci/kfn086

  17. 17. Grivennikov, S.I. and Karin, M. (2010) Dangerous Liaisons: STAT3 and NF-κB Collaboration and Crosstalk in Cancer. Cytokine & Growth Factor Reviews, 21, 11-19.
    http://dx.doi.org/10.1016/j.cytogfr.2009.11.005

  18. 18. Frucht, D.M., Aringer, M., Galon, J., Danning, C., Brown, M., Fan, S., et al. (2000) Stat4 Is Expressed in Activated Peripheral Blood Monocytes, Dendritic Cells, and Macrophages at Sites of Th1-Mediated Inflammation. The Journal of Immunology, 164, 4659-4664.
    http://dx.doi.org/10.4049/jimmunol.164.9.4659

  19. 19. Wurster, A.L., Tanaka, T. and Grusby, M.J. (2000) The Biology of Stat4 and Stat6. Oncogene, 19, 2577-2584.
    http://dx.doi.org/10.1038/sj.onc.1203485

  20. 20. Visconti, R., Gadina, M., Chiariello, M., Chen, E.H., Stancato, L.F., Gutkind, J.S. and O’Shea, J.J. (2000) Importance of the MKK6/p38 Pathway for Interleukin-12-Induced STAT4 Serine Phosphorylation and Transcriptional Activity. Blood, 96, 1844-1852.

  21. 21. Wang, Y., Qu, A.J. and Wang, H. (2015) Signal Transducer and Activator of Transcription 4 in Liver Diseases. International Journal of Biological Sciences, 11, 448-455.
    http://dx.doi.org/10.7150/ijbs.11164

  22. 22. Schindler, C. and Plumlee, C. (2008) Inteferons Pen the JAK-STAT Pathway. Seminars in Cell & Developmental Biology, 19, 311-318.
    http://dx.doi.org/10.1016/j.semcdb.2008.08.010

  23. 23. Kosan, C., Ginter, T., Heinzel, T. and Kramer, O.H. (2013) STAT5 Acetylation: Mechanisms and Consequences for Immunological Control and Leukemogenesis. JAK-STAT, 2, Article ID: e26102.
    http://dx.doi.org/10.4161/jkst.26102

  24. 24. Soldaini, E., John, S., Moro, S., Bollenbacher, J., Schindler, U. and Leonard, W.J. (2000) DNA Binding Site Selection of Dimeric and Tetrameric Stat5 Proteins Reveals a Large Repertoire of Divergent Tetrameric Stat5a Binding Sites. Molecular and Cellular Biology, 20, 389-401.
    http://dx.doi.org/10.1128/MCB.20.1.389-401.2000

  25. 25. John, S., Vinkemeier, U., Soldaini, E., Darnell Jr., J.E. and Leonard, W.J. (1999) The Significance of Tetramerization in Promoter Recruitment by Stat5. Molecular and Cellular Biology, 19, 1910-1918.

  26. 26. Lin, J.X., Li, P., Liu, D., Jin, H.T., He, J., Ata Ur Rasheed, M., Rochman, Y., Wang, L., Cui, K., Liu, C., et al. (2012) Critical Role of STAT5 Transcription Factor Tetramerization for Cytokine Responses and Normal Immune Function. Immunity, 36, 586-599.
    http://dx.doi.org/10.1016/j.immuni.2012.02.017

  27. 27. Ferbeyre, G. and Moriggl, R. (2011) The Role of Stat5 Transcription Factors as Tumor Suppressors or Oncogenes. Biochimica et Biophysica Acta, 1815, 104-114.
    http://dx.doi.org/10.1016/j.bbcan.2010.10.004

  28. 28. Moriggl, R., Sexl, V., Kenner, L., Duntsch, C., Stangl, K., Gingras, S., Hoffmeyer, A., Bauer, A., Piekorz, R., Wang, D., et al. (2005) Stat5 Tetramer Formation Is Associated with Leukemogenesis. Cancer Cell, 7, 87-99.
    http://dx.doi.org/10.1016/j.ccr.2004.12.010

  29. 29. Walford, H.H. and Doherty, T.A. (2013) STAT6 and Lung Inflammation. JAK-STAT, 2, Article ID: e25301.
    http://dx.doi.org/10.4161/jkst.25301

  30. 30. Lee, J.H., Kaminski, N., Dolganov, G., Grunig, G., Koth, L., Solomon, C., et al. (2001) Interleukin-13 Induces Dramatically Different Transcriptional Programs in Three Human Airway Cell Types. American Journal of Respiratory Cell and Molecular Biology, 25, 474-485.
    http://dx.doi.org/10.1165/ajrcmb.25.4.4522

  31. 31. Lawrence, T. and Natoli, G. (2011) Transcriptional Regulation of Macrophage Polarization: Enabling Diversity with Identity. Nature Reviews Immunology, 11, 750-761.
    http://dx.doi.org/10.1038/nri3088

  32. 32. Kaplan, M.H., Schindler, U., Smiley, S.T. and Grusby, M.J. (1996) Stat6 Is Required for Mediating Responses to IL-4 and for Development of Th2 Cells. Immunity, 4, 313-319.
    http://dx.doi.org/10.1016/S1074-7613(00)80439-2

  33. 33. Shimoda, K., van Deursen, J., Sangster, M.Y., Sarawar, S.R., Carson, R.T., Tripp, R.A., et al. (1996) Lack of IL-4-Induced Th2 Response and IgE Class Switching in Mice with Disrupted Stat6 Gene. Nature, 380, 630-633.
    http://dx.doi.org/10.1038/380630a0

  34. 34. Takeda, K., Tanaka, T., Shi, W., Matsumoto, M., Minami, M., Kashiwamura, S., et al. (1996) Essential Role of Stat6 in IL-4 Signalling. Nature, 380, 627-630.
    http://dx.doi.org/10.1038/380627a0

  35. 35. Maier, E., Duschl, A. and Horejs-Hoeck, J. (2012) STAT6-Dependent and -Independent Mechanisms in Th2 Polarization. European Journal of Immunology, 42, 2827-2833.
    http://dx.doi.org/10.1002/eji.201242433

  36. 36. Lu, T.C., Wang, Z.H., Feng, X., Chuang, P.Y., Fang, W., Shen, Y., Levy, D.E., Xiong, H., Chen, N. and He, J.C. (2009) Knockdown of Stat3 Activity in Vivo Prevents Diabetic Glomerulopathy. Kidney International, 76, 63-71.
    http://dx.doi.org/10.1038/ki.2009.98

  37. 37. Mashili, F., Chibalin, A.V., Krook, A. and Zierath, J.R. (2013) Constitutive STAT3 Phosphorylation Contributes to Skeletal Muscle Insulin Resistance in Type 2 Diabetes. Diabetes, 62, 457-465.
    http://dx.doi.org/10.2337/db12-0337

  38. 38. Kim, B.G., Yoo, J.Y., Kim, T.H., Shin, J.H., Langenheim, J.F., Ferguson, S.D., et al. (2015) Aberrant Activation of Signal Transducer and Activator of Transcription-3 (STAT3) Signaling in Endometriosis. Human Reproduction, 30, 1069-1078.
    http://dx.doi.org/10.1093/humrep/dev050

  39. 39. Matsui, F. and Meldrum, K.K. (2012) The Role of the Janus Kinase Family/Signal Transducer and Activator of Transcription Signaling Pathway in Fibrotic Renal Disease. Journal of Surgical Research, 178, 339-345.
    http://dx.doi.org/10.1016/j.jss.2012.06.050

  40. 40. Tang, J., Liu, C.Y., Lu, M.M., Zhang, J., Mei, W.J., Yang, W.J., et al. (2015) Fluorofenidone Protects against Renal Fibrosis by Inhibiting STAT3 Tyrosine Phosphorylation. Molecular and Cellular Biochemistry. (In Press)
    http://dx.doi.org/10.1007/s11010-015-2456-5

  41. 41. Robinson, M.B., Deshpande, D.A., Chou, J., Cui, W., Smith, S., Langefeld, C., Hastie, A.T., Bleecker, E.R. and Hawkins, G.A. (2015) IL6 Trans-Signaling Increases Expression of Airways Disease Genes in Airway Smooth Muscle. American Journal of Physiology—Lung Cellular and Molecular Physiology, 309, L129-L138.

  42. 42. Hedrich, C.M., Rauen, T., Apostolidis, S.A., Grammatikos, A.P., Rodriguez, N.R., Ioannidis, C., et al. (2014) Stat3 Promotes IL-10 Expression in Lupus T Cells through Trans-Activation and Chromatin Remodeling. Proceedings of the National Academy of Sciences of the United States of America, 111, 13457-13462.
    http://dx.doi.org/10.1073/pnas.1408023111

  43. 43. Wang, L.G., Han, L.Y., Fallon, J., Tsao, A. and Chiao, J.W. (2013) Natura-Alpha, a Novel STAT3-Y705 Inhibitor in Treating Systemic Lupus Erythematosus in NZB/W Female Mice (P5157). The Journal of Immunology, 190, 68.6.

  44. 44. Sugimoto, K. (2008) Role of STAT3 in Inflammatory Bowel Disease. World Journal of Gastroenterology, 14, 5110-5114.
    http://dx.doi.org/10.3748/wjg.14.5110

  45. 45. Nguyen, P.M., Putoczki, T.L. and Ernst, M. (2015) STAT3-Activating Cytokines: A Therapeutic Opportunity for Inflammatory Bowel Disease? Journal of Interferon & Cytokine Research, 35, 340-350.
    http://dx.doi.org/10.1089/jir.2014.0225

  46. 46. Mori, T., Miyamoto, T., Yoshida, H., Asakawa, M., Kawasumi, M., Kobayashi, T., et al. (2011) IL-1β and TNFα-Initiated IL-6-STAT3 Pathway Is Critical in Mediating Inflammatory Cytokines and RANKL Expression in Inflammatory Arthritis. International Immunology, 23, 701-712.
    http://dx.doi.org/10.1093/intimm/dxr077

  47. 47. Gao, W., McCormick, J., Connolly, M., Balogh, E., Veale, D.J. and Fearon, U. (2015) Hypoxia and STAT3 Signalling Interactions Regulate Pro-Inflammatory Pathways in Rheumatoid Arthritis. Annals of the Rheumatic Diseases, 74, 1275-1283.
    http://dx.doi.org/10.1136/annrheumdis-2013-204105

  48. 48. Kong, E., Sucic, S., Monje, F.J., Savalli, G., Diao, W., Khan, D., et al. (2015) STAT3 Controls IL6-Dependent Regulation of Serotonin Transporter Function and Depression-Like Behavior. Scientific Reports, 5, 9009.

  49. 49. Ben Haim, L., Ceyzériat, K., Carrillo-de Sauvage, M.A., Aubry, F., Auregan, G., Guillermier, M., et al. (2015) The JAK/STAT3 Pathway Is a Common Inducer of Astrocyte Reactivity in Alzheimer’s and Huntington’s Diseases. The Journal of Neuroscience, 35, 2817-2829.
    http://dx.doi.org/10.1523/JNEUROSCI.3516-14.2015

  50. 50. Zhang, H.F. and Lai, R. (2014) STAT3 in Cancer-Friend or Foe? Cancers (Basel), 6, 1408-1440.
    http://dx.doi.org/10.3390/cancers6031408

  51. 51. Vultur, A., Cao, J., Arulanandam, R., Turkson, J., Jove, R., Greer, P., et al. (2004) Cell-to-Cell Adhesion Modulates Stat3 Activity in Normal and Breast Carcinoma Cells. Oncogene, 23, 2600-2616.
    http://dx.doi.org/10.1038/sj.onc.1207378

  52. 52. Steinman, R.A., Wentzel, A., Lu, Y., Stehle, C. and Grandis, J.R. (2003) Activation of Stat3 by Cell Confluence Reveals Negative Regulation of Stat3 by Cdk2. Oncogene, 22, 3608-3615.
    http://dx.doi.org/10.1038/sj.onc.1206523

  53. 53. Gkouveris, I., Nikitakis, N., Karanikou, M., Rassidakis, G. and Sklavounou, A. (2014) Erk1/2 Activation and Modulation of STAT3 Signaling in Oral Cancer. Oncology Reports, 32, 2175-2182.
    http://dx.doi.org/10.3892/or.2014.3440

  54. 54. Yan, S., Li, Z. and Thiele, C.J. (2013) Inhibition of STAT3 with Orally Active JAK Inhibitor AZD1480 Decreases Tumor Growth in Neuroblastoma and Pediatric Sarcomas in Vitro and in Vivo. Oncotarget, 4, 433-445.

  55. 55. Ivanov, V.N., Bhoumik, A., Krasilnikov, M., Raz, R., Owen-Schaub, L.B., Levy, D., Horvath, C.M. and Ronai, Z. (2001) Cooperation between STAT3 and c-Jun Suppresses Fas Transcription. Molecular Cell, 7, 517-528.
    http://dx.doi.org/10.1016/S1097-2765(01)00199-X

  56. 56. Barre, B., Avril, S. and Coqueret, O. (2003) Opposite Regulation of Myc and p21waf1 Transcription by STAT3 Proteins. The Journal of Biological Chemistry, 278, 2990-2996.
    http://dx.doi.org/10.1074/jbc.M210422200

  57. 57. Bournazou, E. and Bromberg, J. (2013) Targeting the Tumor Microenvironment: JAK-STAT3 Signaling. JAK-STAT, 2, Article ID: e23828.
    http://dx.doi.org/10.4161/jkst.23828

  58. 58. Garcia, R., Yu, C.L., Hudnall, A., Catlett, R., Nelson, K.L., Smithgall, T., et al. (1997) Constitutive Activation of Stat3 in Fibroblasts Transformed by Diverse Oncoproteins and in Breast Carcinoma Cells. Cell Growth & Differentiation, 8, 1267-1276.

  59. 59. Gao, B., Shen, X., Kunos, G., Meng, Q.H., Goldberg, I.D., Rosen, E.M. and Fan, S.J. (2001) Constitutive Activation of JAK-STAT3 Signaling by BRCA1 in Human Prostate Cancer Cells. FEBS Letters, 488, 179-184.
    http://dx.doi.org/10.1016/S0014-5793(00)02430-3

  60. 60. Nikitakis, N.G., Siavash, H. and Sauk, J.J. (2004) Targeting the STAT Pathway in Head and Neck Cancer: Recent Advances and Future Prospects. Current Cancer Drug Targets, 4, 637-651.
    http://dx.doi.org/10.2174/1568009043332736

  61. 61. Kusaba, T., Nakayama, T., Yamazumi, K., Yakata, Y., Yoshizaki, A., Inoue, K., Nagayasu, T. and Sekine, I. (2006) Activation of STAT3 Is a Marker of Poor Prognosis in Human Colorectal Cancer. Oncology Reports, 15, 1445-1451.
    http://dx.doi.org/10.3892/or.15.6.1445

  62. 62. Hirano, T., Ishihara, K. and Hibi, M. (2000) Roles of STAT3 in Mediating the Cell Growth, Differentiation and Survival Signals Relayed through the IL-6 Family of Cytokine Receptors. Oncogene, 19, 2548-2556.
    http://dx.doi.org/10.1038/sj.onc.1203551

  63. 63. Rawat, R., Rainey, G.J., Thompson, C.D., Frazier-Jessen, M.R., Brown, R.T. and Nordan, R.P. (2000) Constitutive Activation of STAT3 Is Associated with the Acquisition of an Interleukin 6-Independent Phenotype by Murine Plasmacytomas and Hybridomas. Blood, 96, 3514-3521.

  64. 64. Catlett-Falcone, R., Landowski, T.H., Oshiro, M.M., Turkson, J., Levitzki, A., Savino, R., et al. (1999) Constitutive Activation of Stat3 Signaling Confers Resistance to Apoptosis in Human U266 Myeloma Cells. Immunity, 10, 105-115.
    http://dx.doi.org/10.1016/S1074-7613(00)80011-4

  65. 65. Skinnider, B.F., Elia, A.J., Gascoyne, R.D., Patterson, B., Trumper, L., Kapp, U. and Mak, T.W. (2002) Signal Transducer and Activator of Transcription 6 Is Frequently Activated in Hodgkin and Reed-Sternberg Cells of Hodgkin Lymphoma. Blood, 99, 618-626.
    http://dx.doi.org/10.1182/blood.V99.2.618

  66. 66. Lowenberg, B. and Touw, I.P. (1993) Hematopoietic Growth Factors and Their Receptors in Acute Leukemia. Blood, 81, 281-292.

  67. 67. Redell, M.S., Ruiz, M.J., Alonzo, T.A., Gerbing, R.B. and Tweardy, D.J. (2011) Stat3 Signaling in Acute Myeloid Leukemia: Ligand-Dependent and -Independent Activation and Induction of Apoptosis by a Novel Small-Molecule Stat3 Inhibitor. Blood, 117, 5701-5709.
    http://dx.doi.org/10.1182/blood-2010-04-280123

  68. 68. Azare, J., Leslie, K., Al-Ahmadie, H., Gerald, W., Weinreb, P.H., Violette, S.M. and Bromberg, J. (2007) Constitutively Activated Stat3 Induces Tumorigenesis and Enhances Cell Motility of Prostate Epithelial Cells through Integrin Beta 6. Molecular and Cellular Biology, 27, 4444-4453.
    http://dx.doi.org/10.1128/MCB.02404-06

  69. 69. Bromberg, J.F., Wrzeszczynska, M.H., Devgan, G., Zhao, Y., Pestell, R.G., Albanese, C. and Darnell Jr., J.E. (1999) STAT3 as an Oncogene. Cell, 98, 295-303.
    http://dx.doi.org/10.1016/S0092-8674(00)81959-5

  70. 70. Turkson, J., Bowman, T., Garcia, R., Caldenhoven, E., de Groot, R.P. and Jove, R. (1998) Stat3 Activation by Src Induces Specific Gene Regulation and Is Required for Cell Transformation. Molecular and Cellular Biology, 18, 2545-2552.

  71. 71. Gao, S.P., Mark, K.G., Leslie, K., Pao, W., Motoi, N., Gerald, W.L., Travis, W.D., Bornmann, W., Veach, D., Clarkson, B., et al. (2007) Mutations in the EGFR Kinase Domain Mediate STAT3 Activation via IL-6 Production in Human Lung Adenocarcinomas. Journal of Clinical Investigation, 117, 3846-3856.
    http://dx.doi.org/10.1172/JCI31871

  72. 72. Inda, M.M., Bonavia, R., Mukasa, A., Narita, Y., Sah, D.W., Vandenberg, S., Brennan, C., Johns, T.G., Bachoo, R., Hadwiger, P., et al. (2010) Tumor Heterogeneity Is an Active Process Maintained by a Mutant EGFR-Induced Cytokine Circuit in Glioblastoma. Genes & Development, 24, 1731-1745.
    http://dx.doi.org/10.1101/gad.1890510

  73. 73. Yu, H., Pardoll, D. and Jove, R. (2009) STATs in Cancer Inflammation and Immunity: A Leading Role for STAT3. Nature Reviews Cancer, 9, 798-809.
    http://dx.doi.org/10.1038/nrc2734

  74. 74. Iliopoulos, D., Jaeger, S.A., Hirsch, H.A., Bulyk, M.L. and Struhl, K. (2010) STAT3 Activation of miR-21 and miR-181b-1 via PTEN and CYLD Are Part of the Epigenetic Switch Linking Inflammation to Cancer. Molecular Cell, 39, 493-506.
    http://dx.doi.org/10.1016/j.molcel.2010.07.023

  75. 75. Lee, H., Deng, J., Kujawski, M., Yang, C., Liu, Y., Herrmann, A., Kortylewski, M., Horne, D., Somlo, G., Forman, S., et al. (2010) STAT3-Induced S1PR1 Expression Is Crucial for Persistent STAT3 Activation in Tumors. Nature Medicine, 16, 1421-1428.
    http://dx.doi.org/10.1038/nm.2250

  76. 76. Ohgami, R.S., Ma, L., Merker, J.D., Martinez, B., Zehnder, J.L. and Arber, D.A. (2013) STAT3 Mutations Are Frequent in CD30+ T-Cell Lymphomas and T-Cell Large Granular Lymphocytic Leukemia. Leukemia, 27, 2244-2247.
    http://dx.doi.org/10.1038/leu.2013.104

  77. 77. Pilati, C., Amessou, M., Bihl, M.P., Balabaud, C., Nhieu, J.T., Paradis, V., Nault, J.C., Izard, T., Bioulac-Sage, P., Couchy, G., et al. (2011) Somatic Mutations Activating STAT3 in Human Inflammatory Hepatocellular Adenomas. Journal of Experimental Medicine, 208, 1359-1366.
    http://dx.doi.org/10.1084/jem.20110283

  78. 78. Fasan, A., Kern, W., Grossmann, V., Haferlach, C., Haferlach, T. and Schnittger, S. (2013) STAT3 Mutations Are Highly Specific for Large Granular Lymphocytic Leukemia. Leukemia, 27, 1598-1600.
    http://dx.doi.org/10.1038/leu.2012.350

  79. 79. Koskela, H.L., Eldfors, S., Ellonen, P., van Adrichem, A.J., Kuusanmaki, H., Andersson, E.I., Lagstrom, S., Clemente, M.J., Olson, T., Jalkanen, S.E., et al. (2012) Somatic STAT3 Mutations in Large Granular Lymphocytic Leukemia. The New England Journal of Medicine, 366, 1905-1913.
    http://dx.doi.org/10.1056/NEJMoa1114885

  80. 80. Jerez, A., Clemente, M.J., Makishima, H., Koskela, H., Leblanc, F., Peng Ng, K., Olson, T., Przychodzen, B., Afable, M., Gomez-Segui, I., et al. (2012) STAT3 Mutations Unify the Pathogenesis of Chronic Lymphoproliferative Disorders of NK Cells and T-Cell Large Granular Lymphocyte Leukemia. Blood, 120, 3048-3057.
    http://dx.doi.org/10.1182/blood-2012-06-435297

  81. 81. Julien, S.G., Dube, N., Hardy, S. and Tremblay, M.L. (2011) Inside the Human Cancer Tyrosine Phosphatome. Nature Reviews Cancer, 11, 35-49.
    http://dx.doi.org/10.1038/nrc2980

  82. 82. Ostman, A., Hellberg, C. and Bohmer, F.D. (2006) Protein-Tyrosine Phosphatases and Cancer. Nature Reviews Cancer, 6, 307-320.
    http://dx.doi.org/10.1038/nrc1837

  83. 83. Yoshimura, A., Naka, T. and Kubo, M. (2007) SOCS Proteins, Cytokine Signalling and Immune Regulation. Nature Reviews Cancer, 7, 454-465.
    http://dx.doi.org/10.1038/nri2093

  84. 84. Yoshikawa, H., Matsubara, K., Qian, G.S., Jackson, P., Groopman, J.D., Manning, J.E., et al. (2001) SOCS-1, a Negative Regulator of the JAK/STAT Pathway, Is Silenced by Methylation in Human Hepatocellular Carcinoma and Shows Growth-Suppression Activity. Nature Genetics, 28, 29-35.
    http://dx.doi.org/10.1038/ng0501-29

  85. 85. To, K.F., Chan, M.W., Leung, W.K., Ng, E.K., Yu, J., Bai, A.H., Lo, A.W., Chu, S.H., Tong, J.H., Lo, K.W., et al. (2004) Constitutional Activation of IL-6-Mediated JAK/STAT Pathway through Hypermethylation of SOCS-1 in Human Gastric Cancer Cell Line. British Journal of Cancer, 91, 1335-1341.
    http://dx.doi.org/10.1038/sj.bjc.6602133

  86. 86. Chim, C.S., Fung, T.K., Cheung, W.C., Liang, R. and Kwong, Y.L. (2004) SOCS1 and SHP1 Hypermethylation in Multiple Myeloma: Implications for Epigenetic Activation of the JAK/STAT Pathway. Blood, 103, 4630-4635.
    http://dx.doi.org/10.1182/blood-2003-06-2007

  87. 87. Galm, O., Yoshikawa, H., Esteller, M., Osieka, R. and Herman, J.G. (2003) SOCS-1, a Negative Regulator of Cytokine Signaling, Is Frequently Silenced by Methylation in Multiple Myeloma. Blood, 101, 2784-2788.
    http://dx.doi.org/10.1182/blood-2002-06-1735

  88. 88. Zhang, Q., Wang, H.Y., Marzec, M., Raghunath, P.N., Nagasawa, T. and Wasik, M.A. (2005) STAT3- and DNA Methyltransferase 1-Mediated Epigenetic Silencing of SHP-1 Tyrosine Phosphatase Tumor Suppressor Gene in Malignant T Lymphocytes. Proceedings of the National Academy of Sciences of the United States of America, 102, 6948-6953.
    http://dx.doi.org/10.1073/pnas.0501959102

  89. 89. Oka, T., Ouchida, M., Koyama, M., Ogama, Y., Takada, S., Nakatani, Y., Tanaka, T., Yoshino, T., Hayashi, K., Ohara, N., et al. (2002) Gene Silencing of the Tyrosine Phosphatase SHP1 Gene by Aberrant Methylation in Leukemias/ Lymphomas. Cancer Research, 62, 6390-6394.

  90. 90. Brantley, E.C., Nabors, L.B., Gillespie, G.Y., Choi, Y.H., Palmer, C.A., Harrison, K., Roarty, K. and Benveniste, E.N. (2008) Loss of Protein Inhibitors of Activated STAT-3 Expression in Glioblastoma Multiforme Tumors: Implications for STAT-3 Activation and Gene Expression. Clinical Cancer Research, 14, 4694-4704.
    http://dx.doi.org/10.1158/1078-0432.ccr-08-0618

  91. 91. de la Iglesia, N., Konopka, G., Puram, S.V., Chan, J.A., Bachoo, R.M., You, M.J., et al. (2008) Identification of a PTEN-Regulated STAT3 Brain Tumor Suppressor Pathway. Genes & Development, 22, 449-462.
    http://dx.doi.org/10.1101/gad.1606508

  92. 92. Musteanu, M., Blaas, L., Mair, M., Schlederer, M., Bilban, M., Tauber, S., et al. (2010) Stat3 Is a Negative Regulator of Intestinal Tumor Progression in Apc(Min) Mice. Gastroenterology, 138, 1003-1011.
    http://dx.doi.org/10.1053/j.gastro.2009.11.049

  93. 93. Lee, J., Kim, J.C., Lee, S.E., Quinley, C., Kim, H., Herdman, S., Corr, M. and Raz, E. (2012) Signal Transducer and Activator of Transcription 3 (STAT3) Protein Suppresses Adenoma-to-Carcinoma Transition in Apcmin/+ Mice via Regulation of Snail-1 (SNAI) Protein Stability. The Journal of Biological Chemistry, 287, 18182-18189.
    http://dx.doi.org/10.1074/jbc.M111.328831

  94. 94. Wang, H., Lafdil, F., Wang, L., Park, O., Yin, S., Niu, J.Y., et al. (2011) Hepatoprotective versus Oncogenic Functions of STAT3 in Liver Tumorigenesis. American Journal of Pathology, 179, 714-724.
    http://dx.doi.org/10.1016/j.ajpath.2011.05.005

  95. 95. Schneller, D., Machat, G., Sousek, A., Proell, V., van Zijl, F., Zulehner, G., Huber, H., et al. (2011) p19(ARF)/ p14(ARF) Controls Oncogenic Functions of Signal Transducer and Activator of Transcription 3 in Hepatocellular Carcinoma. Hepatology, 54, 164-172.
    http://dx.doi.org/10.1002/hep.24329

  96. 96. Pectasides, E., Egloff, A.M., Sasaki, C., Kountourakis, P., Burtness, B., Fountzilas, G., et al. (2010) Nuclear Localization of Signal Transducer and Activator of Transcription 3 in Head and Neck Squamous Cell Carcinoma Is Associated with a Better Prognosis. Clinical Cancer Research, 16, 2427-2434.
    http://dx.doi.org/10.1158/1078-0432.CCR-09-2658

  97. 97. Ettl, T., Stiegler, C., Zeitler, K., Agaimy, A., Zenk, J., Reichert, T.E., et al. (2012) EGFR, HER2, Survivin, and Loss of pSTAT3 Characterize High-Grade Malignancy in Salivary Gland Cancer with Impact on Prognosis. Human Pathology, 43, 921-931.
    http://dx.doi.org/10.1016/j.humpath.2011.08.006

  98. 98. Sato, T., Neilson, L.M., Peck, A.R., Liu, C., Tran, T.H., Witkiewicz, A., et al. (2011) Signal Transducer and Activator of Transcription-3 and Breast Cancer Prognosis. American Journal of Cancer Research, 1, 347-355.

  99. 99. Liu, Q., Li, G., Li, R., Shen, J., He, Q., Deng, L., Zhang, C. and Zhang, J. (2010) IL-6 Promotion of Glioblastoma Cell Invasion and Angiogenesis in U251 and T98G Cell Lines. The Journal of Neuro-Oncology, 100, 165-176.
    http://dx.doi.org/10.1007/s11060-010-0158-0

  100. 100. Varghese, J.N., Moritz, R.L., Lou, M.Z., Van Donkelaar, A., Ji, H., Ivancic, N., Branson, K.M., Hall, N.E. and Simpson, R.J. (2002) Structure of the Extracellular Domains of the Human Interleukin-6 Receptor Alpha-Chain. Proceedings of the National Academy of Sciences of the United States of America, 99, 15959-15964.
    http://dx.doi.org/10.1073/pnas.232432399

  101. 101. Rose-John, S. (2001) Coordination of Interleukin-6 Biology by Membrane Bound and Soluble Receptors. Advances in Experimental Medicine and Biology, 495, 145-151.
    http://dx.doi.org/10.1007/978-1-4615-0685-0_19

  102. 102. Culiq, Z. (2011) Cytokine Disbalance in Common Human Cancers. Biochimica et Biophysica Acta, 1813, 308-314.
    http://dx.doi.org/10.1016/j.bbamcr.2010.12.010

  103. 103. Haan, C., Kreis, S., Margue, C. and Behrmann, I. (2006) Jaks and Cytokine Receptors—An Intimate Relationship. Biochemical Pharmacology, 72, 1538-1546.
    http://dx.doi.org/10.1016/j.bcp.2006.04.013

  104. 104. Wanq, S.W. and Sun, Y.M. (2014) The IL-6/JAK/STAT3 Pathway: Potential Therapeutic Strategies in Treating Colorectal Cancer (Review). International Journal of Oncology, 44, 1032-1040.

  105. 105. Asangani, I.A., Dommeti, V.L., Wang, X., Malik, R., Cieslik, M., Yang, R., et al. (2014) Therapeutic Targeting of BET Bromodomain Proteins in Castration-Resistant Prostate Cancer. Nature, 510, 278-282.
    http://dx.doi.org/10.1038/nature13229

  106. 106. Lin, T.H., Izumi, K., Lee, S.O., Lin, W.J., Yeh, S. and Chang, C. (2013) Anti-Androgen Receptor ASC-J9 versus Anti-Androgens MDV3100 (Enzalutamide) or Casodex (Bicalutamide) Leads to Opposite Effects on Prostate Cancer Metastasis via Differential Modulation of Macrophage Infiltration and STAT3-CCL2 Signaling. Cell Death & Disease, 4, e764.
    http://dx.doi.org/10.1038/cddis.2013.270

  107. 107. Lin, T.H., Lee, S.O., Niu, Y., Xu, D., Liang, L., Li, L., et al. (2013) Differential Androgen Deprivation Therapies with Anti-Androgens Casodex/Bicalutamide or MDV3100/Enzalutamide versus Anti-Androgen Receptor ASC-J9(R) Lead to Promotion versus Suppression of Prostate Cancer Metastasis. The Journal of Biological Chemistry, 288, 19359-19369.
    http://dx.doi.org/10.1074/jbc.M113.477216

  108. 108. Lisa Cheng, Y.S., Jordan, L., Gorugantula, L.M., Schneiderman, E., Chen, H.S. and Rees, T. (2014) Salivary Interleukin-6 and -8 in Patients with Oral Cancer and Patients with Chronic Oral Inflammatory Diseases. Journal of Periodontology, 85, 956-965.
    http://dx.doi.org/10.1902/jop.2013.130320

  109. 109. Chang, K.T., Tsai, C.M., Chiou, Y.C., Chiu, C.H., Jeng, K.S. and Huang, C.Y. (2005) IL-6 Induces Neuroendocrine Dedifferentiation and Cell Proliferation in Non-Small Cell Lung Cancer Cells. American Journal of Physiology—Lung Cellular and Molecular Physiology, 289, L446-L453.
    http://dx.doi.org/10.1152/ajplung.00089.2005

  110. 110. Wan, S., Zhao, E., Kryczek, I., Vatan, L., Sadovskaya, A., Ludema, G., et al. (2014) Tumor-Associated Macrophages Produce Interleukin 6 and Signal via STAT3 to Promote Expansion of Human Hepatocellular Carcinoma Stem Cells. Gastroenterology, 147, 1393-1404.
    http://dx.doi.org/10.1053/j.gastro.2014.08.039

  111. 111. Patel, S.A., Bhambra, U., Charalambous, M.P., David, R.M., Edwards, R.J., Lightfoot, T., et al. (2014) Interleukin-6 Mediated Upregulation of CYP1B1 and CYP2E1 in Colorectal Cancer Involves DNA Methylation, miR27b and STAT3. British Journal of Cancer, 111, 2287-2296.
    http://dx.doi.org/10.1038/bjc.2014.540

  112. 112. Yang, L., Han, S. and Sun, Y. (2014) An IL6-STAT3 Loop Mediates Resistance to PI3K Inhibitors by Inducing Epithelial-Mesenchymal Transition and Cancer Stem Cell Expansion in Human Breast Cancer Cells. Biochemical and Biophysical Research Communications, 453, 582-587.
    http://dx.doi.org/10.1016/j.bbrc.2014.09.129

  113. 113. Zheng, T., Hong, X., Wang, J., Pei, T., Liang, Y., Yin, D., et al. (2014) Gankyrin Promotes Tumor Growth and Metastasis through Activation of IL-6/STAT3 Signaling in Human Cholangiocarcinoma. Hepatology, 59, 935-946.
    http://dx.doi.org/10.1002/hep.26705

  114. 114. Liu, C., Lou, W., Armstrong, C., Zhu, Y., Evans, C.P. and Gao, A.C. (2015) Niclosamide Suppresses Cell Migration and Invasion in Enzalutamide Resistant Prostate Cancer Cells via Stat3-AR Axis Inhibition. The Prostate. (Electronic Publish ahead of Print)
    http://dx.doi.org/10.1002/pros.23015

  115. 115. Waitkus, M.S., Chandrasekharan, U.M., Willard, B., Tee, T.L., Hsieh, J.K., Przybycin, C.G., et al. (2014) Signal Integration and Gene Induction by a Functionally Distinct STAT3 Phosphoform. Molecular and Cellular Biology, 34, 1800-1811.
    http://dx.doi.org/10.1128/MCB.00034-14

  116. 116. Zhang, Q., Raje, V., Yakovlev, V.A., Yacoub, A., Szczepanek, K., Meier, J., et al. (2013) Mitochondrial Localized Stat3 Promotes Breast Cancer Growth via Phosphorylation of Serine 727. The Journal of Biological Chemistry, 288, 31280-31288.
    http://dx.doi.org/10.1074/jbc.M113.505057

  117. 117. Reich, N.C. and Liu, L. (2006) Tracking STAT Nuclear Traffic. Nature Reviews Immunology, 6, 602-612.
    http://dx.doi.org/10.1038/nri1885

  118. 118. Wen, Z. and Darnell Jr., J.E. (1997) Mapping of Stat3 Serine Phosphorylation to a Single Residue (727) and Evidence That Serine Phosphorylation Has No Influence on DNA Binding of Stat1 and Stat3. Nucleic Acids Research, 25, 2062-2067.
    http://dx.doi.org/10.1093/nar/25.11.2062

  119. 119. Wen, Z., Zhong, Z. and Darnell Jr., J.E. (1995) Maximal Activation of Transcription by Stat1 and Stat3 Requires Both Tyrosine and Serine Phosphorylation. Cell, 82, 241-250.
    http://dx.doi.org/10.1016/0092-8674(95)90311-9

  120. 120. Hazan-Halevy, I., Harris, D., Liu, Z., Liu, J., Li, P., Chen, X., Shanker, S., Ferrajoli, A., Keating, M.J. and Estrov, Z. (2010) STAT3 Is Constitutively Phosphorylated on Serine 727 Residues, Binds DNA, and Activates Transcription in CLL Cells. Blood, 115, 2852-2863.
    http://dx.doi.org/10.1182/blood-2009-10-230060

  121. 121. Lim, C.P. and Cao, X. (1999) Serine Phosphorylation and Negative Regulation of Stat3 by JNK. The Journal of Biological Chemistry, 274, 31055-31061.
    http://dx.doi.org/10.1074/jbc.274.43.31055

  122. 122. Chung, J., Uchida, E., Grammer, T.C. and Blenis, J. (1997) STAT3 Serine Phosphorylation by ERK-Dependent and -Independent Pathways Negatively Modulates Its Tyrosine Phosphorylation. Molecular and Cellular Biology, 17, 6508-6516.

  123. 123. O’Shea, J.J., Gadina, M. and Schreiber, R.D. (2002) Cytokine Signaling in 2002: New Surprises in the Jak/Stat Pathway. Cell, 109, S121-S131.
    http://dx.doi.org/10.1016/s0092-8674(02)00701-8

  124. 124. Decker, T. and Kovarik, P. (2000) Serine Phosphorylation of STATs. Oncogene, 19, 2628-2637.
    http://dx.doi.org/10.1038/sj.onc.1203481

  125. 125. Venkatasubbarao, K., Choudary, A. and Freeman, J.W. (2005) Farnesyl Transferase Inhibitor (R115777)-Induced Inhibition of STAT3(Tyr705) Phosphorylation in Human Pancreatic Cancer Cell Lines Require Extracellular Signal-Regulated Kinases. Cancer Research, 65, 2861-2871.
    http://dx.doi.org/10.1158/0008-5472.CAN-04-2396

  126. 126. Wakahara, R., Kunimoto, H., Tanino, K., Kojima, H., Inoue, A., Shintaku, H. and Nakajima, K. (2012) Phospho-Ser727 of STAT3 Regulates STAT3 Activity by Enhancing Dephosphorylation of Phospho-Tyr705 Largely through TC45. Genes Cells, 17, 132-145.
    http://dx.doi.org/10.1111/j.1365-2443.2011.01575.x

  127. 127. Aggarwal, B.B., Kunnumakkara, A.B., Harikumar, K.B., Gupta, S.R., Tharakan, S.T., Koca, C., et al. (2009) Signal Transducer and Activator of Transcription-3, Inflammation, and Cancer: How Intimate Is the Relationship? Annals of the New York Academy of Sciences, 1171, 59-76.
    http://dx.doi.org/10.1111/j.1749-6632.2009.04911.x

  128. 128. Gough, D., Koetz, L. and Levy, D. (2013) The MEK-ERK Pathway Is Necessary for Serine Phosphorylation of Mitochondrial STAT3 and Ras-Mediated Transformation. PLoS ONE, 8, e83395.
    http://dx.doi.org/10.1371/journal.pone.0083395

  129. 129. Chapman, R.S., Lourenco, P.C., Tonner, E., Flint, D.J., Selbert, S., Takeda, K., Akira, S., Clarke, A.R. and Watson, C.J. (1999) Suppression of Epithelial Apoptosis and Delayed Mammary Gland Involution in Mice with a Conditional Knockout of Stat3. Genes & Development, 13, 2604-2616.
    http://dx.doi.org/10.1101/gad.13.19.2604

  130. 130. Sano, S., Takahama, Y., Sugawara, T., Kosaka, H., Itami, S., Yoshikawa, K., Miyazaki, J., van Ewijk, W. and Takeda, J. (2001) Stat3 in Thymic Epithelial Cells Is Essential for Postnatal Maintenance of Thymic Architecture and Thymocyte Survival. Immunity, 15, 261-273.
    http://dx.doi.org/10.1016/S1074-7613(01)00180-7

  131. 131. Takeda, K., Kaisho, T., Yoshida, N., Takeda, J., Kishimoto, T. and Akira, S. (1998) Stat3 Activation Is Responsible for IL-6-Dependent T Cell Proliferation through Preventing Apoptosis: Generation and Characterization of T Cell-Specific Stat3-Deficient Mice. The Journal of Immunology, 161, 4652-4660.

  132. 132. Lee, C., Dhillon, J., Wang, M.Y., Gao, Y., Hu, K., Park, E., et al. (2008) Targeting YB-1 in HER-2 Overexpressing Breast Cancer Cells Induces Apoptosis via the mTOR/STAT3 Pathway and Suppresses Tumor Growth in Mice. Cancer Research, 68, 8661-8666.
    http://dx.doi.org/10.1158/0008-5472.CAN-08-1082

  133. 133. Miyakoshi, M., Yamamoto, M., Tanaka, H. and Ogawa, K. (2014) Serine 727 Phosphorylation of STAT3: An Early Change in Mouse Hepatocarcinogenesis Induced by Neonatal Treatment with Diethylnitrosamine. Molecular Carcinogenesis, 53, 67-76.
    http://dx.doi.org/10.1002/mc.21949

  134. 134. Sakaguchi, M., Oka, M., Iwasaki, T., Fukami, Y. and Nishigori, C. (2012) Role and Regulation of STAT3 Phosphorylation at Ser727 in Melanocytes and Melanoma Cells. Journal of Investigative Dermatology, 132, 1877-1885.
    http://dx.doi.org/10.1038/jid.2012.45

  135. 135. Thakur, R., Trivedi, R., Rastogi, N., Singh, M. and Mishra, D.P. (2015) Inhibition of STAT3, FAK and Src Mediated Signaling Reduces Cancer Stem Cell Load, Tumorigenic Potential and Metastasis in Breast Cancer. Scientific Reports, 5, Article ID: 10194.
    http://dx.doi.org/10.1038/srep10194

  136. 136. Blaylock, R.L. (2015) Cancer Microenvironment, Inflammation and Cancer Stem Cells: A Hypothesis for a Paradigm Change and New Targets in Cancer Control. Surgical Neurology International, 6, 92.
    http://dx.doi.org/10.4103/2152-7806.157890

  137. 137. Liou, G.Y. and Storz, P. (2010) Reactive Oxygen Species in Cancer. Free Radical Research, 44, 479-496.
    http://dx.doi.org/10.3109/10715761003667554

  138. 138. Horinaga, M., Okita, H., Nakashima, J., Kanao, K., Sakamoto, M. and Murai, M. (2005) Clinical and Pathologic Significance of Activation of Signal Transducer and Activator of Transcription 3 in Prostate Cancer. Urology, 66, 671-675.
    http://dx.doi.org/10.1016/j.urology.2005.03.066

  139. 139. Liu, X., He, Z., Li, C., Huang, G., Ding, C. and Liu, H. (2012) Correlation Analysis of JAK-STAT Pathway Components on Prognosis of Patients with Prostate Cancer. Pathology Oncology Research: POR, 18, 17-23.

  140. 140. Han, Z., Wang, X., Ma, L., Chen, L., Xiao, M., Huang, L., et al. (2014) Inhibition of STAT3 Signaling Targets Both Tumor-Initiating and Differentiated Cell Populations in Prostate Cancer. Oncotarget, 5, 8416-8428.

  141. 141. Kroon, P., Berry, P.A., Stower, M.J., Rodrigues, G., Mann, V.M., Simms, M., et al. (2013) JAK-STAT Blockade Inhibits Tumor Initiation and Clonogenic Recovery of Prostate Cancer Stem-Like Cells. Cancer Research, 73, 5288-5298.
    http://dx.doi.org/10.1158/0008-5472.CAN-13-0874

  142. 142. Qu, Y., Oyan, A.M., Liu, R., Hua, Y., Zhang, J., Hovland, R., et al. (2013) Generation of Prostate Tumor-Initiating Cells Is Associated with Elevation of Reactive Oxygen Species and IL-6/STAT3 Signaling. Cancer Research, 73, 7090-7100.
    http://dx.doi.org/10.1158/0008-5472.CAN-13-1560

  143. 143. Rybak, A.P., Bristow, R.G. and Kapoor, A. (2015) Prostate Cancer Stem Cells: Deciphering the Origins and Pathways Involved in Prostate Tumorigenesis and Aggression. Oncotarget, 6, 1900-1919.

  144. 144. Hossain, A., Gumin, J., Gao, F., Figueroa, J., Shinojima, N., Takezaki, T., et al. (2015) Mesenchymal Stem Cells Isolated from Human Gliomas Increase Proliferation and Maintain Stemness of Glioma Stem Cells through the IL-6/ gp130/STAT3 Pathway. Stem Cells, 33, 2400-2415.

  145. 145. van der Zee, M., Sacchetti, A., Cansoy, M., Joosten, R., Teeuwssen, M., Heijmans-Antonissen, C., et al. (2015). IL6/JAK1/STAT3 Signaling Blockade in Endometrial Cancer Affects the ALDHHICD126+-Like Component and Reduces Tumor Burden. Cancer Research, Epub ahead of Print.
    http://dx.doi.org/10.1158/0008-5472.can-14-2498

  146. 146. Islam, M., Sharma, S. and Teknos, T. (2014) RhoC Regulates Cancer Stem Cells in Head and Neck Squamous Cell Carcinoma by Overexpressing IL-6 and Phosphorylation of STAT3. PLoS ONE, 9, e88527.
    http://dx.doi.org/10.1371/journal.pone.0088527

  147. 147. Won, C., Kim, B.H., Hee, Y.E., Choi, K.J., Kim, E.K., et al. (2015) STAT3-Mediated CD133 Upregulation Contributes to Promotion of Hepatocellular Carcinoma. Hepatology, Epub ahead of Print.
    http://dx.doi.org/10.1002/hep.27968

  148. 148. Chen, J.J., Cai, N., Chen, G.Z., Jia, C.C., Qiu, D.B., Du, C., et al. (2015) The Neuroleptic Drug Pimozide Inhibits Stem-Like Cell Maintenance and Tumorigenicity in Hepatocellular Carcinoma. Oncotarget, Epub ahead of Print.

  149. 149. Zhao, D., Pan, C., Sun, J., Gilbert, C., Drews-Elger, K., Azzam, D.J., et al. (2015) VEGF Drives Cancer-Initiating Stem Cells through VEGFR-2/Stat3 Signaling to Upregulate Myc and Sox2. Oncogene, 34, 3107-3119.
    http://dx.doi.org/10.1038/onc.2014.257

Abbreviations

ALDH: Aldehyde dehydrogenase

AR: Androgen receptor

BRCA1: Breast cancer susceptible gene 1

CK2-PP2A: Casein kinase 2―protein phosphatase 2A

CLL: Chronic lymphocytic leukemia

CSCs: Cancer stem cells

EGFR: Epidermal growth factor receptor

EMT: Epithelial to mesenchymal transition

FD: Fluorofenidone

GSCs: Glioma stem cells

HCC: Hepatocellular carcinoma

HIF1α: Hypoxia-inducible factor 1-alpha

HNSCC: Head and neck squamous cell carcinoma

IBD: Inflammatory bowel disease

IFN: Interferon

JAK: Janus kinase

LPPs: Lipoma-preferred partner

MEFs: Mouse embryo fibroblasts

PAR-1: Protease-activated receptor 1

PIAS: Protein inhibitors of activated STAT

PTKs: Protein tyrosine kinases

PTPs: Protein tyrosine phosphatases

RA: Rheumatoid arthritis

RNS: Reactive nitrogen species

ROS: Reactive oxygen species

SERT: Serotonin transporter

Shk: Shikonin

SLE: Systemic lupus erythematosus

SOCS: Suppressors of cytokine signaling

STAT: Signal transducers and activators of transcription

TAMs: Tumor-associated macrophages

VEGF: Vascular endothelial growth factor

YB-1: y box binding protein

NOTES

*Corresponding author.